1
|
Früholz I, Meyer-Luehmann M. The intricate interplay between microglia and adult neurogenesis in Alzheimer's disease. Front Cell Neurosci 2024; 18:1456253. [PMID: 39360265 PMCID: PMC11445663 DOI: 10.3389/fncel.2024.1456253] [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: 06/28/2024] [Accepted: 08/26/2024] [Indexed: 10/04/2024] Open
Abstract
Microglia, the resident immune cells of the central nervous system, play a crucial role in regulating adult neurogenesis and contribute significantly to the pathogenesis of Alzheimer's disease (AD). Under physiological conditions, microglia support and modulate neurogenesis through the secretion of neurotrophic factors, phagocytosis of apoptotic cells, and synaptic pruning, thereby promoting the proliferation, differentiation, and survival of neural progenitor cells (NPCs). However, in AD, microglial function becomes dysregulated, leading to chronic neuroinflammation and impaired neurogenesis. This review explores the intricate interplay between microglia and adult neurogenesis in health and AD, synthesizing recent findings to provide a comprehensive overview of the current understanding of microglia-mediated regulation of adult neurogenesis. Furthermore, it highlights the potential of microglia-targeted therapies to modulate neurogenesis and offers insights into potential avenues for developing novel therapeutic interventions.
Collapse
Affiliation(s)
- Iris Früholz
- Department of Neurology, Medical Center ˗ University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Melanie Meyer-Luehmann
- Department of Neurology, Medical Center ˗ University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
2
|
Wang EY, Chen HS, Wu MC, Yang YL, Wang HL, Liu CW, Lai TW. Microglia through MFG-E8 signaling decrease the density of degenerating neurons and protect the brain from the development of cortical infarction after stroke. PLoS One 2024; 19:e0308464. [PMID: 39110702 PMCID: PMC11305554 DOI: 10.1371/journal.pone.0308464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/22/2024] [Indexed: 08/10/2024] Open
Abstract
Neuronal loss is a hallmark of stroke and other neurodegenerative diseases, and as such, neuronal loss caused by microglia has been thought to be a contributing factor to disease progression. Here, we show that microglia indeed contribute significantly to neuronal loss in a mouse model of stroke, but this microglial-dependent process of neuronal clearance specifically targets stressed and degenerating neurons in the ischemic cortical region and not healthy non-ischemic neurons. Nonspecific stimulation of microglia decreased the density of neurons in the ischemic cortical region, whereas specific inhibition of MFG-E8 signaling, which is required for microglial phagocytosis of neurons, had the opposite effect. In both scenarios, the effects were microglia specific, as the same treatments had no effect in mice whose microglia were depleted prior to stroke. Finally, even though the inhibition of MFG-E8 signaling increased neuronal density in the ischemic brain region, it substantially exacerbated the development of cortical infarction. In conclusion, microglia through MFG-E8 signaling contribute to the loss of ischemic neurons and, in doing so, minimize the development of cortical infarction after stroke.
Collapse
Affiliation(s)
- Eric Yuhsiang Wang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | | | - Meng-Chih Wu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Ya Lan Yang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Hwai-Lee Wang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
| | - Che-Wei Liu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
- Department of Plastic and Reconstructive Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Ted Weita Lai
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Neuroscience and Brain Disease Center, China Medical University, Taichung, Taiwan
- Drug Development Center, China Medical University, Taichung, Taiwan
- Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
3
|
Surala M, Soso-Zdravkovic L, Munro D, Rifat A, Ouk K, Vida I, Priller J, Madry C. Lifelong absence of microglia alters hippocampal glutamatergic networks but not synapse and spine density. EMBO Rep 2024; 25:2348-2374. [PMID: 38589666 PMCID: PMC11094096 DOI: 10.1038/s44319-024-00130-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/13/2024] [Accepted: 03/20/2024] [Indexed: 04/10/2024] Open
Abstract
Microglia sculpt developing neural circuits by eliminating excess synapses in a process called synaptic pruning, by removing apoptotic neurons, and by promoting neuronal survival. To elucidate the role of microglia during embryonic and postnatal brain development, we used a mouse model deficient in microglia throughout life by deletion of the fms-intronic regulatory element (FIRE) in the Csf1r locus. Surprisingly, young adult Csf1rΔFIRE/ΔFIRE mice display no changes in excitatory and inhibitory synapse number and spine density of CA1 hippocampal neurons compared with Csf1r+/+ littermates. However, CA1 neurons are less excitable, receive less CA3 excitatory input and show altered synaptic properties, but this does not affect novel object recognition. Cytokine profiling indicates an anti-inflammatory state along with increases in ApoE levels and reactive astrocytes containing synaptic markers in Csf1rΔFIRE/ΔFIRE mice. Notably, these changes in Csf1rΔFIRE/ΔFIRE mice closely resemble the effects of acute microglial depletion in adult mice after normal development. Our findings suggest that microglia are not mandatory for synaptic pruning, and that in their absence pruning can be achieved by other mechanisms.
Collapse
Affiliation(s)
- Michael Surala
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117, Berlin, Germany
| | - Luna Soso-Zdravkovic
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117, Berlin, Germany
| | - David Munro
- University of Edinburgh and UK Dementia Research Institute, Edinburgh, EH16 4TJ, UK
| | - Ali Rifat
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Koliane Ouk
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Neuropsychiatry and Laboratory of Molecular Psychiatry, Charitéplatz 1, 10117, Berlin, Germany
| | - Imre Vida
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute for Integrative Neuroanatomy, Charitéplatz 1, 10117, Berlin, Germany
| | - Josef Priller
- University of Edinburgh and UK Dementia Research Institute, Edinburgh, EH16 4TJ, UK.
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Neuropsychiatry and Laboratory of Molecular Psychiatry, Charitéplatz 1, 10117, Berlin, Germany.
- DZNE Berlin, 10117, Berlin, Germany.
- Department of Psychiatry and Psychotherapy; School of Medicine and Health, Technical University of Munich and German Center for Mental Health (DZPG), 81675, Munich, Germany.
| | - Christian Madry
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
4
|
Kazis D, Chatzikonstantinou S, Ciobica A, Kamal FZ, Burlui V, Calin G, Mavroudis I. Epidemiology, Risk Factors, and Biomarkers of Post-Traumatic Epilepsy: A Comprehensive Overview. Biomedicines 2024; 12:410. [PMID: 38398011 PMCID: PMC10886732 DOI: 10.3390/biomedicines12020410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
This paper presents an in-depth exploration of Post-Traumatic Epilepsy (PTE), a complex neurological disorder following traumatic brain injury (TBI), characterized by recurrent, unprovoked seizures. With TBI being a global health concern, understanding PTE is crucial for effective diagnosis, management, and prognosis. This study aims to provide a comprehensive overview of the epidemiology, risk factors, and emerging biomarkers of PTE, thereby informing clinical practice and guiding future research. The epidemiological aspect of the study reveals PTE as a significant contributor to acquired epilepsies, with varying incidence influenced by injury severity, age, and intracranial pathologies. The paper delves into the multifactorial nature of PTE risk factors, encompassing clinical, demographic, and genetic elements. Key insights include the association of injury severity, intracranial hemorrhages, and early seizures with increased PTE risk, and the roles of age, gender, and genetic predispositions. Advancements in neuroimaging, electroencephalography, and molecular biology are presented, highlighting their roles in identifying potential PTE biomarkers. These biomarkers, ranging from radiological signs to electroencephalography EEG patterns and molecular indicators, hold promise for enhancing PTE pathogenesis understanding, early diagnosis, and therapeutic guidance. The paper also discusses the critical roles of astrocytes and microglia in PTE, emphasizing the significance of neuroinflammation in PTE development. The insights from this review suggest potential therapeutic targets in neuroinflammation pathways. In conclusion, this paper synthesizes current knowledge in the field, emphasizing the need for continued research and a multidisciplinary approach to effectively manage PTE. Future research directions include longitudinal studies for a better understanding of TBI and PTE outcomes, and the development of targeted interventions based on individualized risk profiles. This research contributes significantly to the broader understanding of epilepsy and TBI.
Collapse
Affiliation(s)
- Dimitrios Kazis
- Third Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (D.K.)
| | - Symela Chatzikonstantinou
- Third Department of Neurology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (D.K.)
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, Alexandru Ioan Cuza University of Iasi, 20th Carol I Avenue, 700506 Iasi, Romania;
- Center of Biomedical Research, Romanian Academy, Iasi Branch, Teodor Codrescu 2, 700481 Iasi, Romania
- Academy of Romanian Scientists, 3 Ilfov, 050044 Bucharest, Romania
| | - Fatima Zahra Kamal
- Higher Institute of Nursing Professions and Health Technical (ISPITS), Marrakech 40000, Morocco
- Laboratory of Physical Chemistry of Processes and Materials, Faculty of Sciences and Techniques, Hassan First University, Settat 26000, Morocco
| | - Vasile Burlui
- Department of Biomaterials, Faculty of Dental Medicine, Apollonia University, 700511 Iasi, Romania;
| | - Gabriela Calin
- Department of Biomaterials, Faculty of Dental Medicine, Apollonia University, 700511 Iasi, Romania;
| | - Ioannis Mavroudis
- Department of Neuroscience, Leeds Teaching Hospitals, Leeds LS2 9JT, UK
- Faculty of Medicine, Leeds University, Leeds LS2 9JT, UK
| |
Collapse
|
5
|
Luo Y, Wang Z. The Impact of Microglia on Neurodevelopment and Brain Function in Autism. Biomedicines 2024; 12:210. [PMID: 38255315 PMCID: PMC10813633 DOI: 10.3390/biomedicines12010210] [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: 11/28/2023] [Revised: 12/29/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Microglia, as one of the main types of glial cells in the central nervous system (CNS), are widely distributed throughout the brain and spinal cord. The normal number and function of microglia are very important for maintaining homeostasis in the CNS. In recent years, scientists have paid widespread attention to the role of microglia in the CNS. Autism spectrum disorder (ASD) is a highly heterogeneous neurodevelopmental disorder, and patients with ASD have severe deficits in behavior, social skills, and communication. Most previous studies on ASD have focused on neuronal pathological changes, such as increased cell proliferation, accelerated neuronal differentiation, impaired synaptic development, and reduced neuronal spontaneous and synchronous activity. Currently, more and more research has found that microglia, as immune cells, can promote neurogenesis and synaptic pruning to maintain CNS homeostasis. They can usually reduce unnecessary synaptic connections early in life. Some researchers have proposed that many pathological phenotypes of ASD may be caused by microglial abnormalities. Based on this, we summarize recent research on microglia in ASD, focusing on the function of microglia and neurodevelopmental abnormalities. We aim to clarify the essential factors influenced by microglia in ASD and explore the possibility of microglia-related pathways as potential research targets for ASD.
Collapse
Affiliation(s)
- Yuyi Luo
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China;
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China
| | - Zhengbo Wang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China;
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China
| |
Collapse
|
6
|
Fernandes VM, Auld V, Klämbt C. Glia as Functional Barriers and Signaling Intermediaries. Cold Spring Harb Perspect Biol 2024; 16:a041423. [PMID: 38167424 PMCID: PMC10759988 DOI: 10.1101/cshperspect.a041423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Glia play a crucial role in providing metabolic support to neurons across different species. To do so, glial cells isolate distinct neuronal compartments from systemic signals and selectively transport specific metabolites and ions to support neuronal development and facilitate neuronal function. Because of their function as barriers, glial cells occupy privileged positions within the nervous system and have also evolved to serve as signaling intermediaries in various contexts. The fruit fly, Drosophila melanogaster, has significantly contributed to our understanding of glial barrier development and function. In this review, we will explore the formation of the glial sheath, blood-brain barrier, and nerve barrier, as well as the significance of glia-extracellular matrix interactions in barrier formation. Additionally, we will delve into the role of glia as signaling intermediaries in regulating nervous system development, function, and response to injury.
Collapse
Affiliation(s)
- Vilaiwan M Fernandes
- Department of Cell and Developmental Biology, University College London, London UC1E 6DE, United Kingdom
| | - Vanessa Auld
- Department of Zoology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Christian Klämbt
- Institute for Neuro- and Behavioral Biology, University of Münster, Münster 48149, Germany
| |
Collapse
|
7
|
Wheeler S, Rai-Bhogal R, Crawford DA. Abnormal Microglial Density and Morphology in the Brain of Cyclooxygenase 2 Knockin Mice. Neuroscience 2023; 534:66-81. [PMID: 37863307 DOI: 10.1016/j.neuroscience.2023.10.009] [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: 08/18/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 10/22/2023]
Abstract
Prostaglandin E2 (PGE2) is a signaling molecule produced by cyclooxygenase-2 (COX-2) that is important in healthy brain development. Anomalies in the COX-2/PGE2 pathway due to genetic or environmental factors have been linked to Autism Spectrum Disorders (ASD). Our previous studies showed that COX-2 deficient (COX-2-KI) mice exhibit sex-dependent molecular changes in the brain and associated autism-related behaviors. Here, we aim to determine the effect of COX-2-KI on microglial density and morphology in the developing brain. Microglia normally transition between an amoeboid or ramified morphology depending on their surroundings and are important for the development of the healthy brain, assisting with synaptogenesis, synaptic pruning, and phagocytosis. We use COX-2-KI male and female mice to evaluate microglia density, morphology, and branch length and number in five brain regions (cerebellum, hippocampus, olfactory bulb, prefrontal cortex, and thalamus) at the gestational day 19 (G19) and postnatal day 25 (PN25). We discovered that COX2-KI females were affected at G19 with increased microglial density, altered percentage of amoeboid and ramified microglia, affected branch length, and decreased branching networks in a region-specific manner; these effects persisted to PN25 in select regions. Interestingly, while limited changes were found in G19 COX-2-KI males, at PN25 we found increased microglial density, higher percentages of ramified microglia, and increased branch counts, and length observed in nearly all brain regions tested. Overall, we show for the first time that the COX-2 deficiency in our ASD mouse model influences microglia morphology in a sex- and region- and stage-dependent manner.
Collapse
Affiliation(s)
- Sarah Wheeler
- School of Kinesiology and Health Science, York University, Toronto, ON M3J 1P3, Canada; Neuroscience Graduate Diploma Program, York University, Toronto, ON M3J 1P3, Canada
| | | | - Dorota A Crawford
- School of Kinesiology and Health Science, York University, Toronto, ON M3J 1P3, Canada; Neuroscience Graduate Diploma Program, York University, Toronto, ON M3J 1P3, Canada; Department of Biology, York University, Toronto, ON M3J 1P3, Canada.
| |
Collapse
|
8
|
Reid KM, Brown GC. LRPAP1 is released from activated microglia and inhibits microglial phagocytosis and amyloid beta aggregation. Front Immunol 2023; 14:1286474. [PMID: 38035103 PMCID: PMC10687467 DOI: 10.3389/fimmu.2023.1286474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Low-density lipoprotein receptor-related protein-associated protein 1 (LRPAP1), also known as receptor associated protein (RAP), is an endoplasmic reticulum (ER) chaperone and inhibitor of LDL receptor related protein 1 (LRP1) and related receptors. These receptors have dozens of physiological ligands and cell functions, but it is not known whether cells release LRPAP1 physiologically at levels that regulate these receptors and cell functions. We used mouse BV-2 and human CHME3 microglial cell lines, and found that microglia released nanomolar levels of LRPAP1 when inflammatory activated by lipopolysaccharide or when ER stressed by tunicamycin. LRPAP1 was found on the surface of live activated and non-activated microglia, and anti-LRPAP1 antibodies induced internalization. Addition of 10 nM LRPAP1 inhibited microglial phagocytosis of isolated synapses and cells, and the uptake of Aβ. LRPAP1 also inhibited Aβ aggregation in vitro. Thus, activated and stressed microglia release LRPAP1 levels that can inhibit phagocytosis, Aβ uptake and Aβ aggregation. We conclude that LRPAP1 release may regulate microglial functions and Aβ pathology, and more generally that extracellular LRPAP1 may be a physiological and pathological regulator of a wide range of cell functions.
Collapse
Affiliation(s)
| | - Guy C. Brown
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
9
|
Jiménez Peinado P, Urbach A. From Youthful Vigor to Aging Decline: Unravelling the Intrinsic and Extrinsic Determinants of Hippocampal Neural Stem Cell Aging. Cells 2023; 12:2086. [PMID: 37626896 PMCID: PMC10453598 DOI: 10.3390/cells12162086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Since Joseph Altman published his pioneering work demonstrating neurogenesis in the hippocampus of adult rats, the number of publications in this field increased exponentially. Today, we know that the adult hippocampus harbors a pool of adult neural stem cells (NSCs) that are the source of life-long neurogenesis and plasticity. The functions of these NSCs are regulated by extrinsic cues arising from neighboring cells and the systemic environment. However, this tight regulation is subject to imbalance with age, resulting in a decline in adult NSCs and neurogenesis, which contributes to the progressive deterioration of hippocampus-related cognitive functions. Despite extensive investigation, the mechanisms underlying this age-related decline in neurogenesis are only incompletely understood, but appear to include an increase in NSC quiescence, changes in differentiation patterns, and NSC exhaustion. In this review, we summarize recent work that has improved our knowledge of hippocampal NSC aging, focusing on NSC-intrinsic mechanisms as well as cellular and molecular changes in the niche and systemic environment that might be involved in the age-related decline in NSC functions. Additionally, we identify future directions that may advance our understanding of NSC aging and the concomitant loss of hippocampal neurogenesis and plasticity.
Collapse
Affiliation(s)
| | - Anja Urbach
- Department of Neurology, Jena University Hospital, 07747 Jena, Germany
- Jena Center for Healthy Aging, Jena University Hospital, 07747 Jena, Germany
- Aging Research Center Jena, Leibniz Institute on Aging, 07745 Jena, Germany
| |
Collapse
|
10
|
Wang H, Li J, Zhang H, Wang M, Xiao L, Wang Y, Cheng Q. Regulation of microglia polarization after cerebral ischemia. Front Cell Neurosci 2023; 17:1182621. [PMID: 37361996 PMCID: PMC10285223 DOI: 10.3389/fncel.2023.1182621] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Stroke ranks second as a leading cause of death and permanent disability globally. Microglia, innate immune cells in the brain, respond rapidly to ischemic injury, triggering a robust and persistent neuroinflammatory reaction throughout the disease's progression. Neuroinflammation plays a critical role in the mechanism of secondary injury in ischemic stroke and is a significant controllable factor. Microglia activation takes on two general phenotypes: the pro-inflammatory M1 type and the anti-inflammatory M2 type, although the reality is more complex. The regulation of microglia phenotype is crucial to controlling the neuroinflammatory response. This review summarized the key molecules and mechanisms of microglia polarization, function, and phenotypic transformation following cerebral ischemia, with a focus on the influence of autophagy on microglia polarization. The goal is to provide a reference for the development of new targets for the treatment for ischemic stroke treatment based on the regulation of microglia polarization.
Collapse
Affiliation(s)
- Hao Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Jingjing Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Han Zhang
- School of Medicine, Nantong University, Nantong, China
| | - Mengyao Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Lifang Xiao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Yitong Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Qiong Cheng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Jiangsu Province Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| |
Collapse
|
11
|
Wu C, Pan Y, Wang L, Liu M, Wu M, Wang J, Yang G, Guo Y, Ma Y. A new method for primary culture of microglia in rats with spinal cord injury. Biochem Biophys Res Commun 2022; 599:63-68. [PMID: 35176626 DOI: 10.1016/j.bbrc.2022.02.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/08/2022] [Accepted: 02/08/2022] [Indexed: 11/29/2022]
Abstract
At present, the primary culture method of microglia is complicated, and the culture of spinal cord microglia is rare, so we will explore to establish a new and efficient primary culture method of microglia in rats with spinal cord injury (SCI). The SCI model of SD rats was established by modified A11en's method, and the model of SCI was performed on 1 d, 3 d, 7 d and 14 d respectively. Then the injured spinal cord was removed, mechanically separated and filtered. The morphology of microglia was observed the next day and its purity was identified by CD11b and Iba1 immunofluorescence labeling. According to the above results, the morphological changes of microglia after 3 d of SCI were observed at 1 d, 2 d and 4 d. The results showed that the purity of microglia was 98%. The number of microglia after 3 d of SCI was the most. After SCI, the migration ability of microglia was enhanced, the number of microglia in the injured area increased, and the number was the highest at 3 d, then gradually decreased. In addition, the microglia after SCI would gradually change from active state to resting state with the passage of time. Therefore, we can use a simple and efficient mechanical separation method to extract primary microglia, which provides the basis for the study of microglia.
Collapse
Affiliation(s)
- Chengjie Wu
- Department of Traumatology and Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yalan Pan
- Laboratory of Chinese Medicine Nursing Intervention for Chronic Diseases, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lining Wang
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing, China
| | - Mengmin Liu
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing, China
| | - Mao Wu
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Jianwei Wang
- Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| | - Guanglu Yang
- Department of Traumatology and Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Guo
- Department of Traumatology and Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Yong Ma
- Department of Traumatology and Orthopedics, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; Laboratory of New Techniques of Restoration & Reconstruction, Institute of Traumatology & Orthopedics, Nanjing University of Chinese Medicine, Nanjing, China; School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing, China.
| |
Collapse
|
12
|
Li SY, Johnson R, Smyth LC, Dragunow M. Platelet-derived growth factor signalling in neurovascular function and disease. Int J Biochem Cell Biol 2022; 145:106187. [PMID: 35217189 DOI: 10.1016/j.biocel.2022.106187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/08/2022] [Accepted: 02/21/2022] [Indexed: 11/25/2022]
Abstract
Platelet-derived growth factors are critical for cerebrovascular development and homeostasis. Abnormalities in this signalling pathway are implicated in neurological diseases, especially those where neurovascular dysfunction and neuroinflammation plays a prominent role in disease pathologies, such as stroke and Alzheimer's disease; the angiogenic nature of this pathway also draws its significance in brain malignancies such as glioblastoma where tumour angiogenesis is profuse. In this review, we provide an updated overview of the actions of the platelet-derived growth factors on neurovascular function, their role in the regulation of perivascular cell types expressing the cognate receptors, neurological diseases associated with aberrance in signalling, and highlight the clinical relevance and therapeutic potentials of this pathway for central nervous system diseases.
Collapse
Affiliation(s)
- Susan Ys Li
- Department of Pharmacology and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| | - Rebecca Johnson
- Department of Pharmacology and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| | - Leon Cd Smyth
- Center for Brain Immunology and Glia, Department of Pathology and Immunology, Washington University in St Louis, MO, USA.
| | - Mike Dragunow
- Department of Pharmacology and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
13
|
Gao T, Huang F, Wang W, Xie Y, Wang B. Interleukin-10 genetically modified clinical-grade mesenchymal stromal cells markedly reinforced functional recovery after spinal cord injury via directing alternative activation of macrophages. Cell Mol Biol Lett 2022; 27:27. [PMID: 35300585 PMCID: PMC8931978 DOI: 10.1186/s11658-022-00325-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/22/2022] [Indexed: 12/19/2022] Open
Abstract
Background After spinal cord injury (SCI), dysregulated or nonresolving inflammatory processes can severely disturb neuronal homeostasis and drive neurodegeneration. Although mesenchymal stromal cell (MSC)-based therapies have showed certain therapeutic efficacy, no MSC therapy has reached its full clinical goal. In this study, we examine interleukin-10 (IL10) genetically modified clinical-grade MSCs (IL10-MSCs) and evaluate their clinical safety, effectiveness, and therapeutic mechanism in a completely transected SCI mouse model. Methods We established stable IL10-overexpressing human umbilical-cord-derived MSCs through electric transduction and screened out clinical-grade IL10-MSCs according to the criteria of cell-based therapeutic products, which were applied to mice with completely transected SCI by repeated tail intravenous injections. Then we comprehensively investigated the motor function, histological structure, and nerve regeneration in SCI mice, and further explored the potential therapeutic mechanism after IL10-MSC treatment. Results IL10-MSC treatment markedly reinforced locomotor improvement, accompanied with decreased lesion volume, regeneration of axons, and preservation of neurons, compared with naïve unmodified MSCs. Further, IL10-MSC transplantation increased the ratio of microglia to infiltrated alternatively activated macrophages (M2), and reduced the ratio of classically activated macrophages (M1) at the injured spinal cord, meanwhile increasing the percentage of Treg and Th2 cells, and reducing the percentage of Th1 cells in the peripheral circulatory system. In addition, IL10-MSC administration could prevent apoptosis and promote neuron differentiation of neural stem cells (NSCs) under inflammatory conditions in vitro. Conclusions IL10-MSCs exhibited a reliable safety profile and demonstrated promising therapeutic efficacy in SCI compared with naïve MSCs, providing solid support for future clinical application of genetically engineered MSCs. Supplementary Information The online version contains supplementary material available at 10.1186/s11658-022-00325-9.
Collapse
Affiliation(s)
- Tianyun Gao
- Center for Clinic Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Feifei Huang
- Center for Clinic Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Wenqing Wang
- Center for Clinic Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Yuanyuan Xie
- Center for Clinic Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Bin Wang
- Center for Clinic Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
14
|
Jin N, Sha W, Gao L. Shaping the Microglia in Retinal Degenerative Diseases Using Stem Cell Therapy: Practice and Prospects. Front Cell Dev Biol 2021; 9:741368. [PMID: 34966736 PMCID: PMC8710684 DOI: 10.3389/fcell.2021.741368] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 11/29/2021] [Indexed: 12/11/2022] Open
Abstract
Retinal degenerative disease (RDD) refers to a group of diseases with retinal degeneration that cause vision loss and affect people's daily lives. Various therapies have been proposed, among which stem cell therapy (SCT) holds great promise for the treatment of RDDs. Microglia are immune cells in the retina that have two activation phenotypes, namely, pro-inflammatory M1 and anti-inflammatory M2 phenotypes. These cells play an important role in the pathological progression of RDDs, especially in terms of retinal inflammation. Recent studies have extensively investigated the therapeutic potential of stem cell therapy in treating RDDs, including the immunomodulatory effects targeting microglia. In this review, we substantially summarized the characteristics of RDDs and microglia, discussed the microglial changes and phenotypic transformation of M1 microglia to M2 microglia after SCT, and proposed future directions for SCT in treating RDDs.
Collapse
Affiliation(s)
- Ni Jin
- Senior Department of Ophthalmology, The Third Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China.,Department of Endocrinology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Weiwei Sha
- Department of Endocrinology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Lixiong Gao
- Senior Department of Ophthalmology, The Third Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
15
|
Carbon Monoxide Modulation of Microglia-Neuron Communication: Anti-Neuroinflammatory and Neurotrophic Role. Mol Neurobiol 2021; 59:872-889. [PMID: 34796462 DOI: 10.1007/s12035-021-02643-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/09/2021] [Indexed: 10/19/2022]
Abstract
Microglia, the 'resident immunocompetent cells' of the central nervous system (CNS), are key players in innate immunity, synaptic refinement and homeostasis. Dysfunctional microglia contribute heavily to creating a toxic inflammatory milieu, a driving factor in the pathophysiology of several CNS disorders. Therefore, strategies to modulate the microglial function are required to tackle exacerbated tissue inflammation. Carbon monoxide (CO), an endogenous gaseous molecule produced by the degradation of haem, has anti-inflammatory, anti-apoptotic, and pro-homeostatic and cytoprotective roles, among others. ALF-826A, a novel molybdenum-based CO-releasing molecule, was used for the assessment of neuron-microglia remote communication. Primary cultures of rat microglia and neurons, or the BV-2 microglial and CAD neuronal murine cell lines, were used to study the microglia-neuron interaction. An approach based on microglial-derived conditioned media in neuronal culture was applied. Medium derived from CO-treated microglia provided indirect neuroprotection against inflammation by limiting the lipopolysaccharide (LPS)-induced expression of reactivity markers (CD11b), the production of reactive oxygen species (ROS) and the secretion of inflammatory factors (TNF-α, nitrites). This consequently prevented neuronal cell death and maintained neuronal morphology. In contrast, in the absence of inflammatory stimulus, conditioned media from CO-treated microglia improved neuronal morphological complexity, which is an indirect manner of assessing neuronal function. Likewise, the microglial medium also prevented neuronal cell death induced by pro-oxidant tert-Butyl hydroperoxide (t-BHP). ALF-826 treatment reinforced microglia secretion of Interleukin-10 (IL-10) and adenosine, mediators that may protect against t-BHP stress in this remote communication model. Chemical inhibition of the adenosine receptors A2A and A1 reverted the CO-derived neuroprotective effect, further highlighting a role for CO in regulating neuron-microglia communication via purinergic signalling. Our findings indicate that CO has a modulatory role on microglia-to-neuron communication, promoting neuroprotection in a non-cell autonomous manner. CO enhances the microglial release of neurotrophic factors and blocks exacerbated microglial inflammation. CO improvement of microglial neurotrophism under non-inflammatory conditions is here described for the first time.
Collapse
|
16
|
Wang XL, Li L. Microglia Regulate Neuronal Circuits in Homeostatic and High-Fat Diet-Induced Inflammatory Conditions. Front Cell Neurosci 2021; 15:722028. [PMID: 34720877 PMCID: PMC8549960 DOI: 10.3389/fncel.2021.722028] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
Microglia are brain resident macrophages, which actively survey the surrounding microenvironment and promote tissue homeostasis under physiological conditions. During this process, microglia participate in synaptic remodeling, neurogenesis, elimination of unwanted neurons and cellular debris. The complex interplay between microglia and neurons drives the formation of functional neuronal connections and maintains an optimal neural network. However, activation of microglia induced by chronic inflammation increases synaptic phagocytosis and leads to neuronal impairment or death. Microglial dysfunction is implicated in almost all brain diseases and leads to long-lasting functional deficiency, such as hippocampus-related cognitive decline and hypothalamus-associated energy imbalance (i.e., obesity). High-fat diet (HFD) consumption triggers mediobasal hypothalamic microglial activation and inflammation. Moreover, HFD-induced inflammation results in cognitive deficits by triggering hippocampal microglial activation. Here, we have summarized the current knowledge of microglial characteristics and biological functions and also reviewed the molecular mechanism of microglia in shaping neural circuitries mainly related to cognition and energy balance in homeostatic and diet-induced inflammatory conditions.
Collapse
Affiliation(s)
- Xiao-Lan Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lianjian Li
- Department of Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China.,Hubei Province Academy of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
17
|
Mallach A, Gobom J, Arber C, Piers TM, Hardy J, Wray S, Zetterberg H, Pocock J. Differential Stimulation of Pluripotent Stem Cell-Derived Human Microglia Leads to Exosomal Proteomic Changes Affecting Neurons. Cells 2021; 10:cells10112866. [PMID: 34831089 PMCID: PMC8616378 DOI: 10.3390/cells10112866] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 10/07/2021] [Accepted: 10/13/2021] [Indexed: 01/22/2023] Open
Abstract
Microglial exosomes are an emerging communication pathway, implicated in fulfilling homeostatic microglial functions and transmitting neurodegenerative signals. Gene variants of triggering receptor expressed on myeloid cells-2 (TREM2) are associated with an increased risk of developing dementia. We investigated the influence of the TREM2 Alzheimer’s disease risk variant, R47Hhet, on the microglial exosomal proteome consisting of 3019 proteins secreted from human iPS-derived microglia (iPS-Mg). Exosomal protein content changed according to how the iPS-Mg were stimulated. Thus lipopolysaccharide (LPS) induced microglial exosomes to contain more inflammatory signals, whilst stimulation with the TREM2 ligand phosphatidylserine (PS+) increased metabolic signals within the microglial exosomes. We tested the effect of these exosomes on neurons and found that the exosomal protein changes were functionally relevant and influenced downstream functions in both neurons and microglia. Exosomes from R47Hhet iPS-Mg contained disease-associated microglial (DAM) signature proteins and were less able to promote the outgrowth of neuronal processes and increase mitochondrial metabolism in neurons compared with exosomes from the common TREM2 variant iPS-Mg. Taken together, these data highlight the importance of microglial exosomes in fulfilling microglial functions. Additionally, variations in the exosomal proteome influenced by the R47Hhet TREM2 variant may underlie the increased risk of Alzheimer’s disease associated with this variant.
Collapse
Affiliation(s)
- Anna Mallach
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, University College, London WC1N 1PJ, UK; (A.M.); (T.M.P.)
| | - Johan Gobom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, S-43180 Molndal, Sweden; (J.G.); (H.Z.)
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-431 80 Molndal, Sweden
| | - Charles Arber
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK; (C.A.); (J.H.); (S.W.)
| | - Thomas M. Piers
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, University College, London WC1N 1PJ, UK; (A.M.); (T.M.P.)
| | - John Hardy
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK; (C.A.); (J.H.); (S.W.)
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
| | - Selina Wray
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK; (C.A.); (J.H.); (S.W.)
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, S-43180 Molndal, Sweden; (J.G.); (H.Z.)
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, S-431 80 Molndal, Sweden
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London WC1N 1PJ, UK; (C.A.); (J.H.); (S.W.)
- UK Dementia Research Institute at UCL, London WC1E 6BT, UK
| | - Jennifer Pocock
- Department of Neuroinflammation, UCL Queen Square Institute of Neurology, University College, London WC1N 1PJ, UK; (A.M.); (T.M.P.)
- Correspondence:
| |
Collapse
|
18
|
Soares NL, Vieira HLA. Microglia at the Centre of Brain Research: Accomplishments and Challenges for the Future. Neurochem Res 2021; 47:218-233. [PMID: 34586585 DOI: 10.1007/s11064-021-03456-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 02/08/2023]
Abstract
Microglia are the immune guardians of the central nervous system (CNS), with critical functions in development, maintenance of homeostatic tissue balance, injury and repair. For a long time considered a forgotten 'third element' with basic phagocytic functions, a recent surge in interest, accompanied by technological progress, has demonstrated that these distinct myeloid cells have a wide-ranging importance for brain function. This review reports microglial origins, development, and function in the healthy brain. Moreover, it also targets microglia dysfunction and how it contributes to the progression of several neurological disorders, focusing on particular molecular mechanisms and whether these may present themselves as opportunities for novel, microglia-targeted therapeutic approaches, an ever-enticing prospect. Finally, as it has been recently celebrated 100 years of microglia research, the review highlights key landmarks from the past century and looked into the future. Many challenging problems have arisen, thus it points out some of the most pressing questions and experimental challenges for the ensuing century.
Collapse
Affiliation(s)
- Nuno L Soares
- Chronic Diseases Research Center (CEDOC) - Faculdade de Ciências Médicas/NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria 130, 1169-056, Lisboa, Portugal.
| | - Helena L A Vieira
- Chronic Diseases Research Center (CEDOC) - Faculdade de Ciências Médicas/NOVA Medical School, Universidade Nova de Lisboa, Campo dos Mártires da Pátria 130, 1169-056, Lisboa, Portugal.,Department of Chemistry, UCIBIO, Applied Molecular Biosciences Unit, NOVA School of Science and Technology, Universidade Nova de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, Lisboa, Portugal
| |
Collapse
|
19
|
He GL, Wang ZZ, Yu XT, Shen TT, Luo Z, Li P, Luo X, Tan YL, Gao P, Yang XS. The involvement of microglial CX3CR1 in heat acclimation-induced amelioration of adult hippocampal neurogenesis impairment in EMF-exposed mice. Brain Res Bull 2021; 177:181-193. [PMID: 34555433 DOI: 10.1016/j.brainresbull.2021.09.018] [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: 08/05/2021] [Accepted: 09/17/2021] [Indexed: 12/12/2022]
Abstract
Microglial CX3C chemokine receptor 1 (CX3CR1) has been implicated in numerous cellular mechanisms, including signalling pathways that regulate brain homoeostasis and adult hippocampal neurogenesis. Specific environmental conditions can impair hippocampal neurogenesis-related cognition, learning and memory. However, the role of CX3CR1 in the neurogenic alterations resulting from the cross-tolerance protection conferred by heat acclimation (HA) against the effects of electromagnetic field (EMF) exposure is less well understood. Here, we investigated the role of microglial CX3CR1 signalling in adult hippocampal neurogenesis induced by HA in EMF-exposed mice. We found that EMF exposure significantly decreased the number of proliferating and differentiating cells in the dentate gyrus (DG) of the hippocampus, resulting in a reduced neurogenesis rate. Moreover, alterations in the phenotypes of activated microglia and decreased expression levels of CX3CR1, but not sirtuin 1 (SIRT1), were observed in the brains of EMF-exposed mice. Remarkably, HA treatment improved microglial phenotypes, restored the expression of CX3CR1, and ameliorated the decrease in the adult hippocampal neurogenesis rate following EMF exposure. Moreover, pharmacological inhibition of CX3CR1 and SIRT1 failed to restore CX3CR1 expression and ameliorate hippocampal neurogenesis impairment following HA plus EMF stimulation. These results indicate that microglial CX3CR1 is involved in the cross-tolerance protective effect of HA on adult hippocampal neurogenesis upon EMF exposure.
Collapse
Affiliation(s)
- Gen-Lin He
- Department of Tropical Medicine, Army Medical University, Chongqing, China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| | - Ze-Ze Wang
- Department of Tropical Medicine, Army Medical University, Chongqing, China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| | - Xue-Ting Yu
- Department of Tropical Medicine, Army Medical University, Chongqing, China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| | - Ting-Ting Shen
- Department of Tropical Medicine, Army Medical University, Chongqing, China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| | - Zhen Luo
- Department of Tropical Medicine, Army Medical University, Chongqing, China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| | - Ping Li
- Department of Tropical Medicine, Army Medical University, Chongqing, China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| | - Xue Luo
- Department of Tropical Medicine, Army Medical University, Chongqing, China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| | - Yu-Long Tan
- Department of Tropical Medicine, Army Medical University, Chongqing, China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China
| | - Peng Gao
- Key Laboratory of Medical Protection for Electromagnetic Radiation Ministry of Education, Army Medical University, Chongqing, China
| | - Xue-Sen Yang
- Department of Tropical Medicine, Army Medical University, Chongqing, China; Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Army Medical University, Chongqing, China.
| |
Collapse
|
20
|
Vega-Rivera NM, González-Monroy E, Morelos-Santana E, Estrada-Camarena E. The relevance of the endocrine condition in microglia morphology and dendrite complexity of doublecortin-associated neurons in young adult and middle-aged female rats exposed to acute stress. Eur J Neurosci 2021; 54:5293-5309. [PMID: 34302304 DOI: 10.1111/ejn.15398] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 07/19/2021] [Accepted: 07/19/2021] [Indexed: 01/04/2023]
Abstract
Menopause, natural or surgical, might facilitate the onset of psychiatric pathologies. Some reports suggest that their severity could increase if the decline of ovarian hormones occurs abruptly and before natural endocrine senescence. Therefore, we compared the effects of ovariectomy on microglia's morphological alterations, the complexity of newborn neurons, and the animal's ability to cope with stress. Young adult (3 months) and middle-aged (15 months) female Wistar rats were subjected to an ovariectomy (OVX) or were sham-operated. After 3 weeks, animals were assigned to one of the following independent groups: (1) young adult OVX + no stress; (2) young adult sham + no stress; (3) young adult OVX + stress; (4) young adult sham + stress; (5) middle-aged OVX + no stress; (6) middle-aged sham + no stress; (7) middle-aged OVX + stress; (8) middle-aged sham + stress. Acute stress was induced by forced swimming test (FST) exposure. Immobility behavior was scored during FST and 30 min after; animals were euthanized, their brains collected and prepared for immunohistochemical detection of Iba-1 to analyze morphological alterations in microglia, and doublecortin (DCX) detection to evaluate the dendrite complexity of newborn neurons. OVX increased immobility behavior, induced microglia morphological alterations, and reduced dendrite complexity of newborn neurons in young adult rats. FST further increased this effect. In middle-aged rats, the main effects were related to the aging process without OVX or stress exposure. In conclusion, surgical menopause favors in young adult rats, but not in middle-aged, the vulnerability to develop immobility behavior, retracted morphology of microglial cells, and decreased dendrite complexity of newborn neurons.
Collapse
Affiliation(s)
- Nelly Maritza Vega-Rivera
- Laboratory of Neuropsychopharmacology, Division of Neurosciences, National Institute of Psychiatry, Mexico City, Mexico
| | - Edgar González-Monroy
- Laboratory of Neuropsychopharmacology, Division of Neurosciences, National Institute of Psychiatry, Mexico City, Mexico
| | - Erik Morelos-Santana
- Division of Clinical Investigations, National Institute of Psychiatry, Mexico City, Mexico
| | - Erika Estrada-Camarena
- Laboratory of Neuropsychopharmacology, Division of Neurosciences, National Institute of Psychiatry, Mexico City, Mexico
| |
Collapse
|
21
|
Saddala MS, Yang X, Tang S, Huang H. Transcriptome-wide analysis reveals core sets of transcriptional regulators of sensome and inflammation genes in retinal microglia. Genomics 2021; 113:3058-3071. [PMID: 34242709 DOI: 10.1016/j.ygeno.2021.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 06/15/2021] [Accepted: 07/02/2021] [Indexed: 01/18/2023]
Abstract
BACKGROUND Retinal microglial cells (RMCs) play crucial roles in maintaining normal visual functions in a healthy eye. However, the underlying mechanisms of RMCs over-activation manifesting the alterations of sensome profile and inflammation state, which contribute to various retinal neurodegenerative diseases, remain elusive. Here, we aimed to identify the core set of sensome and pro-inflammatory genes and their regulators using transcriptome and data mining approaches. METHODS We performed paired-end RNA-sequencing in primary microglial cell cultures treated with TNFα/IFNϒ (10 ng/ml for 12 h) and PBS as a control. Gene enrichment analysis and hierarchical clustering for the differentially expressed transcripts highlight functional pathways and network perturbations. We examined overlaps of the mouse microglial gene expression profiles with the data-mined human sensome and pro-inflammatory marker genes. The core sets of sensome and pro-inflammatory genes were selected and predicted for transcription factors (TFs). The identified TFs in RNA-Seq are validated by the quantitative PCR method. RESULTS TNFα/IFNϒ induced 668 differentially expressed transcripts in retinal microglial cells relative to the control. Furthermore, gene enrichment analysis and the gene expression network revealed activated microglial genes, biological, molecular and inflammatory pathways. The overlapping analysis of the TNFα/IFNϒ-activated microglia genes and the data-mined human gene sets revealed 22 sensome and 61 pro-inflammatory genes. Based on network analysis, we determined 10 genes as the core sets of sensome and pro-inflammatory genes and predicted the top ten TFs that regulate them. The SP110, IRF1, FLI1, SP140 (sensome) and RELB, BATF2, NFKB2, TRAFD1, SP100, NFKB1 (inflammation) are differentially expressed between the TNFα/IFNϒ activated and the non-activated microglia which were validated by quantitative PCR. The outcomes indicate that these transcriptional regulators are highly expressed and may regulate the sensome and inflammatory genes of RMCs and switch them to over-activation. CONCLUSION Our results comprise a powerful, cross-species functional genomics resource for sensome and inflammation of RMCs, which may provide novel therapeutic approaches to prevent retinal neurodegenerative diseases.
Collapse
Affiliation(s)
- Madhu Sudhana Saddala
- University of Missouri School of Medicine, Columbia, Missouri, United States of America; Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xu Yang
- University of Missouri School of Medicine, Columbia, Missouri, United States of America; Aier Eye Institute, Aier Eye Hospital Group, Changsha, Hunan, China
| | - Shibo Tang
- Aier Eye Institute, Aier Eye Hospital Group, Changsha, Hunan, China
| | - Hu Huang
- University of Missouri School of Medicine, Columbia, Missouri, United States of America.
| |
Collapse
|
22
|
McCarty MF, Lerner A. The second phase of brain trauma can be controlled by nutraceuticals that suppress DAMP-mediated microglial activation. Expert Rev Neurother 2021; 21:559-570. [PMID: 33749495 DOI: 10.1080/14737175.2021.1907182] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION A delayed second wave of brain trauma is mediated in large part by microglia that are activated to a pro-inflammatory M1 phenotype by DAMP proteins released by dying neurons. These microglia can promote apoptosis or necrosis in neighboring neurons by producing a range of pro-inflammatory cytokines and the deadly oxidant peroxynitrite. This second wave could therefore be mitigated with agents that blunt the post-traumatic M1 activation of microglia and that preferentially promote a pro-healing M2 phenotype. AREAS COVERED The literature on nutraceuticals that might have clinical potential in this regard. EXPERT OPINION The chief signaling pathway whereby DAMPs promote M1 microglial activation involves activation of toll-like receptor 4 (TLR4), NADPH oxidase, NF-kappaB, and the stress activated kinases JNK and p38. The green tea catechin EGCG can suppress TLR4 expression. Phycocyanobilin can inhibit NOX2-dependent NADPH oxidase, ferulate and melatonin can oppose pro-inflammatory signal modulation by NADPH oxidase-derived oxidants. Long-chain omega-3 fatty acids, the soy isoflavone genistein, the AMPK activator berberine, glucosamine, and ketone bodies can down-regulate NF-kappaB activation. Vitamin D activity can oppose JNK/p38 activation. A sophisticated program of nutraceutical supplementation may have important potential for mitigating the second phase of neuronal death and aiding subsequent healing.
Collapse
Affiliation(s)
- Mark F McCarty
- Department of research, Catalytic Longevity Foundation, San Diego, California, USA
| | - Aaron Lerner
- Chaim Sheba Medical Center, The Zabludowicz Research Center for Autoimmune Diseases, Tel Hashomer, Israel
| |
Collapse
|
23
|
Fujita Y, Yamashita T. Mechanisms and significance of microglia-axon interactions in physiological and pathophysiological conditions. Cell Mol Life Sci 2021; 78:3907-3919. [PMID: 33507328 PMCID: PMC11072252 DOI: 10.1007/s00018-021-03758-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/28/2020] [Accepted: 01/06/2021] [Indexed: 12/15/2022]
Abstract
Microglia are the resident immune cells of the central nervous system, and are important for cellular processes. In addition to their classical roles in pathophysiological conditions, these immune cells also dynamically interact with neurons and influence their structure and function in physiological conditions. Microglia have been shown to contact neurons at various points, including the dendrites, cell bodies, synapses, and axons, and support various developmental functions, such as neuronal survival, axon elongation, and maturation of the synaptic circuit. This review summarizes the current knowledge regarding the roles of microglia in brain development, with particular emphasis on microglia-axon interactions. We will review recent findings regarding the functions and signaling pathways involved in the reciprocal interactions between microglia and neurons. Moreover, as these interactions are altered in disease and injury conditions, we also discuss the effect and alteration of microglia-axon interactions in disease progression and the potential role of microglia in developmental brain disorders.
Collapse
Affiliation(s)
- Yuki Fujita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan.
- WPI Immunology Frontier Research Center, Osaka University, 3-1, Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan.
- WPI Immunology Frontier Research Center, Osaka University, 3-1, Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Graduate School of Frontier Bioscience, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
24
|
Rahman AA, Amruta N, Pinteaux E, Bix GJ. Neurogenesis After Stroke: A Therapeutic Perspective. Transl Stroke Res 2021; 12:1-14. [PMID: 32862401 PMCID: PMC7803692 DOI: 10.1007/s12975-020-00841-w] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/14/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022]
Abstract
Stroke is a major cause of death and disability worldwide. Yet therapeutic strategies available to treat stroke are very limited. There is an urgent need to develop novel therapeutics that can effectively facilitate functional recovery. The injury that results from stroke is known to induce neurogenesis in penumbra of the infarct region. There is considerable interest in harnessing this response for therapeutic purposes. This review summarizes what is currently known about stroke-induced neurogenesis and the factors that have been identified to regulate it. Additionally, some key studies in this field have been highlighted and their implications on future of stroke therapy have been discussed. There is a complex interplay between neuroinflammation and neurogenesis that dictates stroke outcome and possibly recovery. This highlights the need for a better understanding of the neuroinflammatory process and how it affects neurogenesis, as well as the need to identify new mechanisms and potential modulators. Neuroinflammatory processes and their impact on post-stroke repair have therefore also been discussed.
Collapse
Affiliation(s)
- Abir A Rahman
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, Room 1349, 131 S. Robertson, Ste 1300, New Orleans, LA, 70112, USA
| | - Narayanappa Amruta
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, Room 1349, 131 S. Robertson, Ste 1300, New Orleans, LA, 70112, USA
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, University of Manchester, A.V. Hill Building, Oxford Road, Manchester, M13 9PT, UK
| | - Gregory J Bix
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, Room 1349, 131 S. Robertson, Ste 1300, New Orleans, LA, 70112, USA.
- Tulane Brain Institute, Tulane University, New Orleans, LA, 70112, USA.
| |
Collapse
|
25
|
Ghasemi M, Keyhanian K, Douthwright C. Glial Cell Dysfunction in C9orf72-Related Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. Cells 2021; 10:cells10020249. [PMID: 33525344 PMCID: PMC7912327 DOI: 10.3390/cells10020249] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/14/2021] [Accepted: 01/25/2021] [Indexed: 12/17/2022] Open
Abstract
Since the discovery of the chromosome 9 open reading frame 72 (C9orf72) repeat expansion mutation in 2011 as the most common genetic abnormality in amyotrophic lateral sclerosis (ALS, also known as Lou Gehrig's disease) and frontotemporal dementia (FTD), progress in understanding the signaling pathways related to this mutation can only be described as intriguing. Two major theories have been suggested-(i) loss of function or haploinsufficiency and (ii) toxic gain of function from either C9orf72 repeat RNA or dipeptide repeat proteins (DPRs) generated from repeat-associated non-ATG (RAN) translation. Each theory has provided various signaling pathways that potentially participate in the disease progression. Dysregulation of the immune system, particularly glial cell dysfunction (mainly microglia and astrocytes), is demonstrated to play a pivotal role in both loss and gain of function theories of C9orf72 pathogenesis. In this review, we discuss the pathogenic roles of glial cells in C9orf72 ALS/FTD as evidenced by pre-clinical and clinical studies showing the presence of gliosis in C9orf72 ALS/FTD, pathologic hallmarks in glial cells, including TAR DNA-binding protein 43 (TDP-43) and p62 aggregates, and toxicity of C9orf72 glial cells. A better understanding of these pathways can provide new insights into the development of therapies targeting glial cell abnormalities in C9orf72 ALS/FTD.
Collapse
Affiliation(s)
- Mehdi Ghasemi
- Correspondence: ; Tel.: +1-774-441-7726; Fax: +1-508-856-4485
| | | | | |
Collapse
|
26
|
Liang TY, Peng SY, Ma M, Li HY, Wang Z, Chen G. Protective effects of sevoflurane in cerebral ischemia reperfusion injury: a narrative review. Med Gas Res 2021; 11:152-154. [PMID: 34213497 PMCID: PMC8374460 DOI: 10.4103/2045-9912.318860] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Ischemia/reperfusion (I/R) injury is a phenomenon that the reperfusion of ischemic organs or tissues aggravates their damage, which poses a serious health threat and economic burden to the world. I/R gives rise to a series of physiological and pathological world, including inflammatory response, oxidative stress, brain edema, blood-brain barrier destruction, and neuronal death. Therefore, finding effective treatment measures is extremely important to the recovery of I/R patients and the improvement of long-term quality of life. Sevoflurane is an important volatile anesthetic which has been reported to reduce myocardial I/R damage and infarct size. Sevoflurane also has anti-inflammatory and neuroprotective effects. As reported sevoflurane treatment could reduce nerve function injury, cerebral infarction volume and the level of inflammatory factors. At the same time, there is evidence that sevoflurane can reduce neuron apoptosis and antioxidant stress. The protective effect of sevoflurane in brain injury has been proved to be existed in several aspects, so that a comprehensive understanding of its neuroprotective effect is helpful to exploit new treatment paths for I/R, provide clinicians with new clinical treatment decisions, contribute to the effective treatment of I/R patients and the improvement of quality of life after I/R healing.
Collapse
Affiliation(s)
- Tian-Yu Liang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Song-Yang Peng
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Mian Ma
- Department of Neurosurgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, Jiangsu Province, China
| | - Hai-Ying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
27
|
Lago-Baldaia I, Fernandes VM, Ackerman SD. More Than Mortar: Glia as Architects of Nervous System Development and Disease. Front Cell Dev Biol 2020; 8:611269. [PMID: 33381506 PMCID: PMC7767919 DOI: 10.3389/fcell.2020.611269] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022] Open
Abstract
Glial cells are an essential component of the nervous system of vertebrates and invertebrates. In the human brain, glia are as numerous as neurons, yet the importance of glia to nearly every aspect of nervous system development has only been expounded over the last several decades. Glia are now known to regulate neural specification, synaptogenesis, synapse function, and even broad circuit function. Given their ubiquity, it is not surprising that the contribution of glia to neuronal disease pathogenesis is a growing area of research. In this review, we will summarize the accumulated evidence of glial participation in several distinct phases of nervous system development and organization-neural specification, circuit wiring, and circuit function. Finally, we will highlight how these early developmental roles of glia contribute to nervous system dysfunction in neurodevelopmental and neurodegenerative disorders.
Collapse
Affiliation(s)
- Inês Lago-Baldaia
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Vilaiwan M. Fernandes
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Sarah D. Ackerman
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR, United States
| |
Collapse
|
28
|
Zhang Z, Ishrat S, O'Bryan M, Klein B, Saraswati M, Robertson C, Kannan S. Pediatric Traumatic Brain Injury Causes Long-Term Deficits in Adult Hippocampal Neurogenesis and Cognition. J Neurotrauma 2020; 37:1656-1667. [DOI: 10.1089/neu.2019.6894] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Zhi Zhang
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan, USA
| | - Samiha Ishrat
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan, USA
| | - Megan O'Bryan
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan, USA
| | - Brandon Klein
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan, USA
| | - Manda Saraswati
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Courtney Robertson
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
29
|
Mukherjee S, Arisi GM, Mims K, Hollingsworth G, O'Neil K, Shapiro LA. Neuroinflammatory mechanisms of post-traumatic epilepsy. J Neuroinflammation 2020; 17:193. [PMID: 32552898 PMCID: PMC7301453 DOI: 10.1186/s12974-020-01854-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 05/25/2020] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) occurs in as many as 64-74 million people worldwide each year and often results in one or more post-traumatic syndromes, including depression, cognitive, emotional, and behavioral deficits. TBI can also increase seizure susceptibility, as well as increase the incidence of epilepsy, a phenomenon known as post-traumatic epilepsy (PTE). Injury type and severity appear to partially predict PTE susceptibility. However, a complete mechanistic understanding of risk factors for PTE is incomplete. MAIN BODY From the earliest days of modern neuroscience, to the present day, accumulating evidence supports a significant role for neuroinflammation in the post-traumatic epileptogenic progression. Notably, substantial evidence indicates a role for astrocytes, microglia, chemokines, and cytokines in PTE progression. Although each of these mechanistic components is discussed in separate sections, it is highly likely that it is the totality of cellular and neuroinflammatory interactions that ultimately contribute to the epileptogenic progression following TBI. CONCLUSION This comprehensive review focuses on the neuroinflammatory milieu and explores putative mechanisms involved in the epileptogenic progression from TBI to increased seizure-susceptibility and the development of PTE.
Collapse
Affiliation(s)
- Sanjib Mukherjee
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Gabriel M Arisi
- Department of Physiology, Federal University of Sao Paulo - Escola Paulista de Medicina, Sao Paulo, Brazil.
| | - Kaley Mims
- Texas A&M University, College Station, TX, USA
| | | | | | - Lee A Shapiro
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M University Health Science Center, Bryan, TX, USA.
| |
Collapse
|
30
|
Brezzo G, Simpson J, Ameen-Ali KE, Berwick J, Martin C. Acute effects of systemic inflammation upon the neuro-glial-vascular unit and cerebrovascular function. Brain Behav Immun Health 2020; 5:100074. [PMID: 32685933 PMCID: PMC7357601 DOI: 10.1016/j.bbih.2020.100074] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 04/17/2020] [Indexed: 12/30/2022] Open
Abstract
Brain health relies on a tightly regulated system known as neurovascular coupling whereby the cellular constituents of the neuro-glial-vascular unit (NGVU) regulate cerebral haemodynamics in accordance with brain metabolic demand. Disruption of neurovascular coupling impairs brain health and is associated with the development of a number for neurological conditions, including Alzheimer's disease. The NGVU is also a key site of action for neuroinflammatory responses and contributes to the transition of systemic inflammation to neuroinflammatory processes. Thus, systemic inflammatory challenges may cause a shift in NGVU operation towards prioritising neuroinflammatory action and thus altering neurovascular coupling and resultant cerebrovascular changes. To investigate this, rats were injected with lipopolysaccharide (LPS) (2 mg/kg) to induce a systemic inflammatory response, or vehicle, and brain haemodynamic responses to sensory and non-sensory (hypercapnia) stimuli were assessed in vivo using optical imaging techniques. Following imaging, animals were perfused and their brains extracted to histologically characterise components of the NGVU to determine the association between underlying cellular changes and in vivo blood flow regulation. LPS-treated animals showed changes in haemodynamic function and cerebrovascular dynamics 6 hours after LPS administration. Histological assessment identified a significant increase in astrogliosis, microgliosis and endothelial activation in LPS-treated animals. Our data shows that an acutely induced systemic inflammatory response is able to rapidly alter in vivo haemodynamic function and is associated with significant changes in the cellular constituents of the NGVU. We suggest that these effects are initially mediated by endothelial cells, which are directly exposed to the circulating inflammatory stimulus and have been implicated in regulating functional hyperaemia.
Collapse
Affiliation(s)
- Gaia Brezzo
- The University of Sheffield, Department of Psychology, Cathedral Court, 1 Vicar Lane, Sheffield, S1 2LT, UK
| | - Julie Simpson
- The University of Sheffield, Sheffield Institute for Translational Neuroscience (SITraN), 385a Glossop Road, Sheffield, S10 2HQ, UK
| | - Kamar E. Ameen-Ali
- The University of Sheffield, Department of Psychology, Cathedral Court, 1 Vicar Lane, Sheffield, S1 2LT, UK
| | - Jason Berwick
- The University of Sheffield, Department of Psychology, Cathedral Court, 1 Vicar Lane, Sheffield, S1 2LT, UK
| | - Chris Martin
- The University of Sheffield, Department of Psychology, Cathedral Court, 1 Vicar Lane, Sheffield, S1 2LT, UK
| |
Collapse
|
31
|
Accelerated brain aging towards transcriptional inversion in a zebrafish model of the K115fs mutation of human PSEN2. PLoS One 2020; 15:e0227258. [PMID: 31978074 PMCID: PMC6980398 DOI: 10.1371/journal.pone.0227258] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 12/16/2019] [Indexed: 12/26/2022] Open
Abstract
Background The molecular changes involved in Alzheimer’s disease (AD) progression remain unclear since we cannot easily access antemortem human brains. Some non-mammalian vertebrates such as the zebrafish preserve AD-relevant transcript isoforms of the PRESENILIN genes lost from mice and rats. One example is PS2V, the alternative transcript isoform of the PSEN2 gene. PS2V is induced by hypoxia/oxidative stress and shows increased expression in late onset, sporadic AD brains. A unique, early onset familial AD mutation of PSEN2, K115fs, mimics the PS2V coding sequence suggesting that forced, early expression of PS2V-like isoforms may contribute to AD pathogenesis. Here we use zebrafish to model the K115fs mutation to investigate the effects of forced PS2V-like expression on the transcriptomes of young adult and aged adult brains. Methods We edited the zebrafish genome to model the K115fs mutation. To explore its effects at the molecular level, we analysed the brain transcriptome and proteome of young (6-month-old) and aged (24-month-old) wild type and heterozygous mutant female sibling zebrafish. Finally, we used gene co-expression network analysis (WGCNA) to compare molecular changes in the brains of these fish to human AD. Results Young heterozygous mutant fish show transcriptional changes suggesting accelerated brain aging and increased glucocorticoid signalling. These early changes precede a transcriptional ‘inversion’ that leads to glucocorticoid resistance and other likely pathological changes in aged heterozygous mutant fish. Notably, microglia-associated immune responses regulated by the ETS transcription factor family are altered in both our zebrafish mutant model and in human AD. The molecular changes we observe in aged heterozygous mutant fish occur without obvious histopathology and possibly in the absence of Aβ. Conclusions Our results suggest that forced expression of a PS2V-like isoform contributes to immune and stress responses favouring AD pathogenesis. This highlights the value of our zebrafish genetic model for exploring molecular mechanisms involved in AD pathogenesis.
Collapse
|
32
|
High-Fat Diet-Induced Obesity Causes Sex-Specific Deficits in Adult Hippocampal Neurogenesis in Mice. eNeuro 2020; 7:ENEURO.0391-19.2019. [PMID: 31871124 PMCID: PMC6946541 DOI: 10.1523/eneuro.0391-19.2019] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/26/2019] [Accepted: 12/01/2019] [Indexed: 12/13/2022] Open
Abstract
Adult hippocampal neurogenesis (AHN) is suppressed by high-fat (HF) diet and metabolic disease, including obesity and type 2 diabetes. Deficits in AHN may contribute to cognitive decline and increased risk of dementia and mood disorders, which have higher prevalence in women. However, sex differences in the effects of HF diet/metabolic disease on AHN have yet to be thoroughly investigated. Herein, male and female C57BL/6J mice were fed an HF or control (CON) diet from ∼2 to 6 months of age. After 3 months on the diet, mice were injected with 5-ethynyl-2′-deoxyuridine (EdU) then killed 4 weeks later. Cell proliferation, differentiation/maturation, and survival of new neurons in the dentate gyrus were assessed with immunofluorescence for EdU, Ki67, doublecortin (DCX), and NeuN. CON females had more proliferating cells (Ki67+) and neuroblasts/immature neurons (DCX+) compared with CON males; however, HF diet reduced these cells in females to the levels of males. Diet did not affect neurogenesis in males. Further, the numbers of proliferating cells and immature neurons were inversely correlated with both weight gain and glucose intolerance in females only. These effects were robust in the dorsal hippocampus, which supports cognitive processes. Assessment of microglia in the dentate gyrus using immunofluorescence for Iba1 and CD68 uncovered sex-specific effects of diet, which may contribute to observed differences in neurogenesis. These findings demonstrate sex-specific effects of HF diet/metabolic disease on AHN, and highlight the potential for targeting neurogenic deficits to treat cognitive decline and reduce the risk of dementia associated with these conditions, particularly in females.
Collapse
|
33
|
Microglia Actively Remodel Adult Hippocampal Neurogenesis through the Phagocytosis Secretome. J Neurosci 2020; 40:1453-1482. [PMID: 31896673 PMCID: PMC7044727 DOI: 10.1523/jneurosci.0993-19.2019] [Citation(s) in RCA: 190] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 12/15/2022] Open
Abstract
During adult hippocampal neurogenesis, most newborn cells undergo apoptosis and are rapidly phagocytosed by resident microglia to prevent the spillover of intracellular contents. Here, we propose that phagocytosis is not merely passive corpse removal but has an active role in maintaining neurogenesis. First, we found that neurogenesis was disrupted in male and female mice chronically deficient for two phagocytosis pathways: the purinergic receptor P2Y12, and the tyrosine kinases of the TAM family Mer tyrosine kinase (MerTK)/Axl. In contrast, neurogenesis was transiently increased in mice in which MerTK expression was conditionally downregulated. Next, we performed a transcriptomic analysis of the changes induced by phagocytosis in microglia in vitro and identified genes involved in metabolism, chromatin remodeling, and neurogenesis-related functions. Finally, we discovered that the secretome of phagocytic microglia limits the production of new neurons both in vivo and in vitro Our data suggest that microglia act as a sensor of local cell death, modulating the balance between proliferation and survival in the neurogenic niche through the phagocytosis secretome, thereby supporting the long-term maintenance of adult hippocampal neurogenesis.SIGNIFICANCE STATEMENT Microglia are the brain professional phagocytes and, in the adult hippocampal neurogenic niche, they remove newborn cells naturally undergoing apoptosis. Here we show that phagocytosis of apoptotic cells triggers a coordinated transcriptional program that alters their secretome, limiting neurogenesis both in vivo and in vitro In addition, chronic phagocytosis disruption in mice deficient for receptors P2Y12 and MerTK/Axl reduces adult hippocampal neurogenesis. In contrast, inducible MerTK downregulation transiently increases neurogenesis, suggesting that microglial phagocytosis provides a negative feedback loop that is necessary for the long-term maintenance of adult hippocampal neurogenesis. Therefore, we speculate that the effects of promoting engulfment/degradation of cell debris may go beyond merely removing corpses to actively promoting regeneration in development, aging, and neurodegenerative diseases.
Collapse
|
34
|
Bhattrai A, Irimia A, Van Horn JD. Neuroimaging of traumatic brain injury in military personnel: An overview. J Clin Neurosci 2019; 70:1-10. [PMID: 31331746 PMCID: PMC6861663 DOI: 10.1016/j.jocn.2019.07.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 07/04/2019] [Indexed: 02/02/2023]
Abstract
BACKGROUND The incidence of blunt-force traumatic brain injury (TBI) is especially prevalent in the military, where the emergency care admission rate has been reported to be 24.6-41.8 per 10,000 soldier-years. Given substantial advancements in modern neuroimaging techniques over the past decade in terms of structural, functional, and connectomic approaches, this mode of exploration can be viewed as best suited for understanding the underlying pathology and for providing proper intervention at effective time-points. APPROACH Here we survey neuroimaging studies of mild-to-severe TBI in military veterans with the intent to aid the field in the creation of a roadmap for clinicians and researchers whose aim is to understand TBI progression. DISCUSSION Recent advancements on the quantification of neurocognitive dysfunction, cellular dysfunction, intracranial pressure, cerebral blood flow, inflammation, post-traumatic neuropathophysiology, on blood serum biomarkers and on their correlation to neuroimaging findings are reviewed to hypothesize how they can be used in conjunction with one another. This may allow clinicians and scientists to comprehensively study TBI in military service members, leading to new treatment strategies for both currently-serving as well as veteran personnel, and to improve the study of TBI more broadly.
Collapse
Affiliation(s)
- Avnish Bhattrai
- USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, 2025 Zonal Avenue, SHN, Los Angeles, CA 90033, USA
| | - Andrei Irimia
- Ethel Percy Andrus Gerontology Center, USC Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave., Room 228C, Los Angeles, CA 90089-0191, USA.
| | - John Darrell Van Horn
- USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, 2025 Zonal Avenue, SHN, Los Angeles, CA 90033, USA.
| |
Collapse
|
35
|
VanRyzin JW, Marquardt AE, Pickett LA, McCarthy MM. Microglia and sexual differentiation of the developing brain: A focus on extrinsic factors. Glia 2019; 68:1100-1113. [PMID: 31691400 DOI: 10.1002/glia.23740] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/09/2019] [Accepted: 10/11/2019] [Indexed: 12/16/2022]
Abstract
Microglia, the innate immune cells of the brain, have recently been removed from the position of mere sentinels and promoted to the role of active sculptors of developing circuits and cells. Alongside their functions in normal brain development, microglia coordinate sexual differentiation of the brain, a set of processes which vary by region and endpoint like that of microglia function itself. In this review, we highlight the ways microglia are both targets and drivers of brain sexual differentiation. We examine the factors that may drive sex differences in microglia, with a special focus on how changing microenvironments in the developing brain dictate microglia phenotypes and discuss how their diverse functions sculpt lasting sex-specific changes in the brain. Finally, we consider how sex-specific early life environments contribute to epigenetic programming and lasting sex differences in microglia identity.
Collapse
Affiliation(s)
- Jonathan W VanRyzin
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ashley E Marquardt
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland
| | - Lindsay A Pickett
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland
| | - Margaret M McCarthy
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland.,Program in Neuroscience, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
36
|
Subhramanyam CS, Wang C, Hu Q, Dheen ST. Microglia-mediated neuroinflammation in neurodegenerative diseases. Semin Cell Dev Biol 2019; 94:112-120. [PMID: 31077796 DOI: 10.1016/j.semcdb.2019.05.004] [Citation(s) in RCA: 507] [Impact Index Per Article: 101.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/07/2019] [Accepted: 05/07/2019] [Indexed: 12/11/2022]
Abstract
Microglia, being the resident immune cells of the central nervous system, play an important role in maintaining tissue homeostasis and contributes towards brain development under normal conditions. However, when there is a neuronal injury or other insult, depending on the type and magnitude of stimuli, microglia will be activated to secrete either proinflammatory factors that enhance cytotoxicity or anti-inflammatory neuroprotective factors that assist in wound healing and tissue repair. Excessive microglial activation damages the surrounding healthy neural tissue, and the factors secreted by the dead or dying neurons in turn exacerbate the chronic activation of microglia, causing progressive loss of neurons. It is the case observed in many neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease and amyotrophic lateral sclerosis. This review gives a detailed account of the microglia-mediated neuroinflammation in various neurodegenerative diseases. Hence, resolving chronic inflammation mediated by microglia bears great promise as a novel treatment strategy to reduce neuronal damage and to foster a permissive environment for further regeneration effort.
Collapse
Affiliation(s)
| | - Cheng Wang
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, 4 Medical Drive, 117594, Singapore
| | - Qidong Hu
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, 4 Medical Drive, 117594, Singapore.
| | - S Thameem Dheen
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, 4 Medical Drive, 117594, Singapore.
| |
Collapse
|
37
|
Neuregulin-1 Fosters Supportive Interactions between Microglia and Neural Stem/Progenitor Cells. Stem Cells Int 2019; 2019:8397158. [PMID: 31089334 PMCID: PMC6476022 DOI: 10.1155/2019/8397158] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/31/2018] [Accepted: 02/13/2019] [Indexed: 01/23/2023] Open
Abstract
Microglia play diverse roles in homeostasis and pathology of the central nervous system (CNS). Their response to injury or insult is critical for initiating neuroinflammation and tissue damage as well as resolution of inflammation and wound healing. Changes to the microenvironment of microglia appear to be a key determinant of their phenotype and their role in the endogenous repair process in the injured or diseased CNS. Our recent findings have identified a positive role for neuregulin-1 (Nrg-1) in regulating immune response in spinal cord injury and focal demyelinating lesions. We show that increasing the tissue availability of Nrg-1 after injury can promote endogenous repair by modulating neuroinflammation. In the present study, we sought to elucidate the specific role of Nrg-1 in regulating microglial activity and more importantly their influence on the behavior of neural stem/progenitor cells (NPCs). Using injury-relevant in vitro systems, we demonstrate that Nrg-1 attenuates the expression of proinflammatory mediators in activated microglia. Moreover, we provide novel evidence that availability of Nrg-1 can restore the otherwise suppressed phagocytic ability of proinflammatory microglia. Interestingly, the presence of Nrg-1 in the microenvironment of proinflammatory microglia mitigates their inhibitory effects on NPC proliferation. Nrg-1 treated proinflammatory microglia also augment mobilization of NPCs, while they had no influence on their suppressive effects on NPC differentiation. Mechanistically, we show that Nrg-1 enhances the interactions of proinflammatory microglia and NPCs, at least in part, through reduction of TNF-α expression in microglia. These findings provide new insights into the endogenous regulation of microglia-NPC interactions and identify new potential targets for optimizing this important crosstalk during the regenerative process after CNS injury and neuroinflammatory conditions.
Collapse
|
38
|
Anderson SR, Vetter ML. Developmental roles of microglia: A window into mechanisms of disease. Dev Dyn 2019; 248:98-117. [PMID: 30444278 PMCID: PMC6328295 DOI: 10.1002/dvdy.1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/21/2018] [Accepted: 10/21/2018] [Indexed: 12/12/2022] Open
Abstract
Microglia are engineers of the central nervous system (CNS) both in health and disease. In addition to the canonical immunological roles of clearing damaging entities and limiting the spread of toxicity and death, microglia remodel the CNS throughout life. While they have been extensively studied in disease and injury, due to their highly variable functions, their precise role in these contexts still remains uncertain. Over the past decade, we have greatly expanded our understanding of microglial function, including their essential homeostatic roles during development. Here, we review these developmental roles, identify parallels in disease, and speculate whether developmental mechanisms re-emerge in disease and injury. Developmental Dynamics 248:98-117, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sarah R Anderson
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, Utah
| | - Monica L Vetter
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah
| |
Collapse
|
39
|
Caspases orchestrate microglia instrumental functions. Prog Neurobiol 2018; 171:50-71. [DOI: 10.1016/j.pneurobio.2018.09.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 09/21/2018] [Accepted: 09/29/2018] [Indexed: 12/16/2022]
|
40
|
Kreisel T, Wolf B, Keshet E, Licht T. Unique role for dentate gyrus microglia in neuroblast survival and in VEGF-induced activation. Glia 2018; 67:594-618. [DOI: 10.1002/glia.23505] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 06/24/2018] [Accepted: 06/25/2018] [Indexed: 12/23/2022]
Affiliation(s)
- Tirzah Kreisel
- Department of Developmental Biology and Cancer Research; Hadassah Medical School, The Hebrew University; Jerusalem Israel
- Edmond and Lily Safra Center for Brain Sciences (ELSC); The Hebrew University; Jerusalem Israel
| | - Brachi Wolf
- Department of Developmental Biology and Cancer Research; Hadassah Medical School, The Hebrew University; Jerusalem Israel
| | - Eli Keshet
- Department of Developmental Biology and Cancer Research; Hadassah Medical School, The Hebrew University; Jerusalem Israel
| | - Tamar Licht
- Department of Developmental Biology and Cancer Research; Hadassah Medical School, The Hebrew University; Jerusalem Israel
| |
Collapse
|
41
|
Abstract
Microglia, the primary resident immune cell type, constitute a key population of glia in the retina. Recent evidence indicates that microglia play significant functional roles in the retina at different life stages. During development, retinal microglia regulate neuronal survival by exerting trophic influences and influencing programmed cell death. During adulthood, ramified microglia in the plexiform layers interact closely with synapses to maintain synaptic structure and function that underlie the retina's electrophysiological response to light. Under pathological conditions, retinal microglia participate in potentiating neurodegeneration in diseases such as glaucoma, retinitis pigmentosa, and age-related neurodegeneration by producing proinflammatory neurotoxic cytokines and removing living neurons via phagocytosis. Modulation of pathogenic microglial activation states and effector mechanisms has been linked to neuroprotection in animal models of retinal diseases. These findings have led to the design of early proof-of-concept clinical trials with microglial modulation as a therapeutic strategy.
Collapse
Affiliation(s)
- Sean M. Silverman
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;,
| | - Wai T. Wong
- Unit on Neuron-Glia Interactions in Retinal Disease, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;,
| |
Collapse
|
42
|
Dang D, Saiyin H, Yu Q, Liang W. Effects of sevoflurane preconditioning on microglia/macrophage dynamics and phagocytosis profile against cerebral ischemia in rats. CNS Neurosci Ther 2018; 24:564-571. [PMID: 29427321 PMCID: PMC6490012 DOI: 10.1111/cns.12823] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 01/07/2018] [Accepted: 01/19/2018] [Indexed: 01/17/2023] Open
Abstract
AIM The effects of sevoflurane on microglia/macrophages, promoting or suppressing their activation, remains controversy. We aimed to determine whether sevoflurane preconditioning can protect brain via changing microglia/macrophage dynamics and phagocytosis profile after ischemia. METHODS The impact of sevoflurane preconditioning was evaluated on microglia/macrophage migration, phagocytosis and proliferation altogether from day 1 to day 7 after transient middle cerebral arterial occlusion (tMCAO) in rats. RESULTS Sevoflurane preconditioning was identified to accelerate microglia/macrophage migrating to and invasion in the ischemic core from day 1 to day 5 after damage. Significant accumulation of amoeboid and phagocytic microglia/macrophages was observed in sevoflurane group from day 3 to day 5 after ischemia injury. In addition, sevoflurane pretreatment also promoted the proliferation of microglia/macrophage (Iba1+ /Ki67+ ) dramatically in ischemic core on day 3 postinsult. CONCLUSIONS Our current study has identified the impact of sevoflurane preconditioning on microglia/macrophage dynamics, including its migration, phagocytosis, and proliferation at early stage after brain ischemia and reperfusion. Sevoflurane might enhance microglia/macrophage activation and promote brain repair. These results could help to approach more relevant microglia/macrophage cell-based strategy for human stroke therapy.
Collapse
Affiliation(s)
- Dan‐Dan Dang
- Department of AnesthesiologyHuashan HospitalFudan UniversityShanghaiChina
| | - Hexige Saiyin
- State Key Laboratory of Genetic EngineeringSchool of Life SciencesFudan UniversityShanghaiChina
| | - Qiong Yu
- Department of AnesthesiologyHuashan HospitalFudan UniversityShanghaiChina
| | - Wei‐Min Liang
- Department of AnesthesiologyHuashan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
43
|
New Insights into Microglia-Neuron Interactions: A Neuron's Perspective. Neuroscience 2018; 405:103-117. [PMID: 29753862 DOI: 10.1016/j.neuroscience.2018.04.046] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 04/26/2018] [Accepted: 04/28/2018] [Indexed: 01/16/2023]
Abstract
Microglia are the primary immune cells of the central nervous system. However, recent data indicate that microglia also contribute to diverse physiological and pathophysiological processes that extend beyond immune-related functions and there is a growing interest to understand the mechanisms through which microglia interact with other cells in the brain. In particular, the molecular processes that contribute to microglia-neuron communication in the healthy brain and their role in common brain diseases have been intensively studied during the last decade. In line with this, fate-mapping studies, genetic models and novel pharmacological approaches have revealed the origin of microglial progenitors, demonstrated the role of self-maintaining microglial populations during brain development or in adulthood, and identified the unexpectedly long lifespan of microglia that may profoundly change our view about senescence and age-related human diseases. Despite the exponentially increasing knowledge about microglia, the role of these cells in health and disease is still extremely controversial and the precise molecular targets for intervention are not well defined. This is in part due to the lack of microglia-specific manipulation approaches until very recently and to the high level of complexity of the interactions between microglia and other cells in the brain that occur at different temporal and spatial scales. In this review, we briefly summarize the known physiological roles of microglia-neuron interactions in brain homeostasis and attempt to outline some major directions and challenges of future microglia research.
Collapse
|
44
|
VanRyzin JW, Pickett LA, McCarthy MM. Microglia: Driving critical periods and sexual differentiation of the brain. Dev Neurobiol 2018; 78:580-592. [PMID: 29243403 DOI: 10.1002/dneu.22569] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 12/05/2017] [Accepted: 12/13/2017] [Indexed: 12/12/2022]
Abstract
The proverbial role of microglia during brain development is shifting from passive members of the brain's immune system to active participants that are able to dictate enduring outcomes. Despite these advances, little attention has been paid to one of the most critical components of early brain development-sexual differentiation. Mounting evidence suggests that the normal developmental functions microglia perform-cell number regulation and synaptic connectivity-may be involved in the sex-specific patterning of the brain during these early sensitive periods, and may have lasting sex-dependent and sex-independent effects on behavior. In this review, we outline the known functions of microglia during developmental sensitive periods, and highlight the role they play in the establishment of sex differences in brain and behavior. We also propose a framework for how researchers can incorporate microglia in their study of sex differences and vice versa. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 580-592, 2018.
Collapse
Affiliation(s)
- Jonathan W VanRyzin
- Department of Pharmacology, The University of Maryland School of Medicine, Baltimore, Maryland, 21201.,Program in Neuroscience, The University of Maryland School of Medicine, Baltimore, Maryland, 21201
| | - Lindsay A Pickett
- Department of Pharmacology, The University of Maryland School of Medicine, Baltimore, Maryland, 21201.,Program in Neuroscience, The University of Maryland School of Medicine, Baltimore, Maryland, 21201
| | - Margaret M McCarthy
- Department of Pharmacology, The University of Maryland School of Medicine, Baltimore, Maryland, 21201.,Program in Neuroscience, The University of Maryland School of Medicine, Baltimore, Maryland, 21201
| |
Collapse
|
45
|
Leung JWH, Lau BWM, Chan VSF, Lau CS, So KF. Abnormal increase of neuronal precursor cells and exacerbated neuroinflammation in the corpus callosum in murine model of systemic lupus erythematosus. Restor Neurol Neurosci 2018; 34:443-53. [PMID: 27163251 PMCID: PMC4927870 DOI: 10.3233/rnn-160638] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Purpose: Systemic Lupus Erythematosus (SLE) is an autoimmune disease which is characterised by elevated levels of autoantibodies and cytokines in the body. Via alteration of the regulation of inflammation, damage to different organ systems, including the central nervous system (CNS), was found in SLE patients. Patients diagnosed with SLE were reported to suffer from different kinds of psychiatric signs and symptoms. As neurogenesis has been suggested to be a potential key player of psychiatric symptoms and emotional behavior disturbances, this study aims to investigate whether neurogenesis is altered in an animal model of SLE. Also, neuroinflammation was studied. Methods: Female NZB/W F1 mice were used as an animal model of SLE. Animals were divided into two groups: 1. pre-diseased mice (lupus-prone NZB/W F1 female mice, age 10–15 weeks, negative for proteinuria and with basal levels of serum anti-dsDNA autoantibodies) and 2. diseased mice (NZB/W F1 female mice, > 25 weeks of age, with elevated serum levels of anti-dsDNA autoantibodies and with persistent proteinuria of > 3 mg/ml for more than 2 weeks). Comparisons of the levels of neurogenesis and neuroinflammtion between two groups of mice were studied by the immunohistochemistry. Results: After the onset of SLE symptoms, a reduction of neurogenesis in the hippocampus was found, while there was a dramatic increase of doublecortin (DCX+) neuronal precursor cells in the corpus callosum (CC) and in the subventricular zone (SVZ). Meanwhile, exacerbated inflammation was present in the corpus callosum of the diseased mice, which was suggested by the increased number of GFAP+ cells and IBA-1+ cells. Conclusions: To the best of our knowledge, this is the first study showing an increase of neuronal precursor cells in the corpus callosum of the female NZB/W F1 mice. The present study suggests a coincidence but not a causal relationship between neurogenesis and neuroinflammation. The present results have also provided new insight showing that the altered neurogenesis and neuroinflammation may be a potential neurological mechanism for the cognitive and mood disturbance found in the SLE patients.
Collapse
Affiliation(s)
- Joseph Wai-Hin Leung
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Benson Wui-Man Lau
- Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong
| | - Vera Sau-Fong Chan
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Chak-Sing Lau
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Kwok-Fai So
- Department of Ophthalmology, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong.,State Key Laboratory of Brain and Cognitive Science, The University of Hong Kong, Hong Kong.,GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China.,Co-innovation Center of Neuroregeneration, Nantong University, Jiangsu, China.,Ministry of Education CNS Regeneration International Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| |
Collapse
|
46
|
Abstract
Although autism spectrum disorder (ASD) has a strong genetic basis, its etiology is complex, with several genetic factors likely to be involved as well as environmental factors. Immune dysregulation has gained significant attention as a causal mechanism in ASD pathogenesis. ASD has been associated with immune abnormalities in the brain and periphery, including inflammatory disorders and autoimmunity in not only the affected individuals but also their mothers. Prenatal exposure to maternal immune activation (MIA) has been implicated as an environmental risk factor for ASD. In support of this notion, animal models have shown that MIA results in offspring with behavioral, neurological, and immunological abnormalities similar to those observed in ASD. This raises the question of how MIA exposure can lead to ASD in susceptible individuals. Recent evidence points to a potential inflammation pathway linking MIA-associated ASD with the activity of T helper 17 (Th17) lymphocytes and their effector cytokine interleukin-17A (IL-17A). IL-17A has been implicated from human studies and elevated IL-17A levels in the blood have been found to correlate with phenotypic severity in a subset of ASD individuals. In MIA model mice, elevated IL-17A levels also have been observed. Additionally, antibody blockade to inhibit IL-17A signaling was found to prevent ASD-like behaviors in offspring exposed to MIA. Therefore, IL-17A dysregulation may play a causal role in the development of ASD. The source of increased IL-17A in the MIA mouse model was attributed to maternal Th17 cells because genetic removal of the transcription factor RORγt to selectively inhibit Th17 differentiation in pregnant mice was able to prevent ASD-like behaviors in the offspring. Similar to ASD individuals, the MIA-exposed offspring also displayed cortical dysplasia which could be prevented by inhibition of IL-17A signaling in pregnant mice. This finding reveals one possible cellular mechanism through which ASD-related cognitive and behavioral deficits may emerge following maternal inflammation. IL-17A can exert strong effects on cell survival and differentiation and the activity of signal transduction cascades, which can have important consequences during cortical development on neural function. This review examines IL-17A signaling pathways in the context of both immunity and neural function that may contribute to the development of ASD associated with MIA.
Collapse
Affiliation(s)
- Helen Wong
- Institute for Behavioral Genetics, University of Colorado-Boulder, CO 80303, United States; Department of Integrative Physiology, University of Colorado-Boulder, Boulder, CO 80303, United States; Linda Crnic Institute, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Charles Hoeffer
- Institute for Behavioral Genetics, University of Colorado-Boulder, CO 80303, United States; Department of Integrative Physiology, University of Colorado-Boulder, Boulder, CO 80303, United States; Linda Crnic Institute, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, United States.
| |
Collapse
|
47
|
Osman AM, Rodhe J, Shen X, Dominguez CA, Joseph B, Blomgren K. The Secretome of Microglia Regulate Neural Stem Cell Function. Neuroscience 2017; 405:92-102. [PMID: 29101080 DOI: 10.1016/j.neuroscience.2017.10.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 10/02/2017] [Accepted: 10/24/2017] [Indexed: 12/22/2022]
Abstract
Brain injury is associated with neuroinflammation, and microglia are key players in this process. Microglia can acquire pro-inflammatory or anti-inflammatory properties, but how this affects neural stem cells (NSCs) remains controversial. Here, NSCs were grown in conditioned media collected from either non-stimulated microglia, or microglia stimulated with pro- or anti-inflammatory agents. NSC survival, proliferation, migration, and differentiation were investigated thereafter. We found that NSCs kept in conditioned medium from the anti-inflammatory microglial subtype had better survival, increased migration, and lower astrocytic differentiation compared to NSCs grown in conditioned medium collected from the pro-inflammatory subtype. Finally, we found that NSCs differentiated in microglial conditioned media generated cells expressing the pro-inflammatory chemokine CCL2, most pronounced when differentiated in medium from the pro-inflammatory microglia subtype. Our results show that microglial subtypes regulate NSCs differently and induce generation of cells with inflammatory properties.
Collapse
Affiliation(s)
- Ahmed M Osman
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Johanna Rodhe
- Department of Oncology Pathology, Cancer Centrum Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Xianli Shen
- Department of Oncology Pathology, Cancer Centrum Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Cecilia A Dominguez
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Bertrand Joseph
- Department of Oncology Pathology, Cancer Centrum Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden; Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
48
|
Kaur C, Rathnasamy G, Ling EA. Biology of Microglia in the Developing Brain. J Neuropathol Exp Neurol 2017; 76:736-753. [PMID: 28859332 DOI: 10.1093/jnen/nlx056] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Microglia exist in different morphological forms in the developing brain. They show a small cell body with scanty cytoplasm with many branching processes in the grey matter of the developing brain. However, in the white matter such as the corpus callosum where the unmyelinated axons are loosely organized, they appear in an amoeboid form having a round cell body endowed with copious cytoplasm rich in organelles. The amoeboid cells eventually transform into ramified microglia in the second postnatal week when the tissue becomes more compact with the onset of myelination. Microglia serve as immunocompetent macrophages that act as neuropathology sensors to detect and respond swiftly to subtle changes in the brain tissues in pathological conditions. Microglial functions are broadly considered as protective in the normal brain development as they phagocytose dead cells and sculpt neuronal connections by pruning excess axons and synapses. They also secrete a number of trophic factors such as insulin-like growth factor-1 and transforming growth factor-β among many others that are involved in neuronal and oligodendrocyte survival. On the other hand, microglial cells when activated produce a plethora of molecules such as proinflammatory cytokines, chemokines, reactive oxygen species, and nitric oxide that are implicated in the pathogenesis of many pathological conditions such as epilepsy, cerebral palsy, autism, and perinatal hypoxic-ischemic brain injury. Although many studies have investigated the origin and functions of the microglia in the developing brain, in-depth in vivo studies along with analysis of their transcriptome and epigenetic changes need to be undertaken to elucidate their full potential be it protective or neurotoxic. This would lead to a better understanding of their roles in the healthy and diseased developing brain and advancement of therapeutic strategies to target microglia-mediated neurotoxicity.
Collapse
Affiliation(s)
- Charanjit Kaur
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; and Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Gurugirijha Rathnasamy
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; and Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Eng-Ang Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; and Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
49
|
Ramos E, Patiño P, Reiter RJ, Gil-Martín E, Marco-Contelles J, Parada E, de Los Rios C, Romero A, Egea J. Ischemic brain injury: New insights on the protective role of melatonin. Free Radic Biol Med 2017; 104:32-53. [PMID: 28065781 DOI: 10.1016/j.freeradbiomed.2017.01.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 12/20/2016] [Accepted: 01/04/2017] [Indexed: 12/15/2022]
Abstract
Stroke represents one of the most common causes of brain's vulnerability for many millions of people worldwide. The plethora of physiopathological events associated with brain ischemia are regulate through multiple signaling pathways leading to the activation of oxidative stress process, Ca2+ dyshomeostasis, mitochondrial dysfunction, proinflammatory mediators, excitotoxicity and/or programmed neuronal cell death. Understanding this cascade of molecular events is mandatory in order to develop new therapeutic strategies for stroke. In this review article, we have highlighted the pleiotropic effects of melatonin to counteract the multiple processes of the ischemic cascade. Additionally, experimental evidence supports its actions to ameliorate ischemic long-term behavioural and neuronal deficits, preserving the functional integrity of the blood-brain barrier, inducing neurogenesis and cell proliferation through receptor-dependent mechanism, as well as improving synaptic transmission. Consequently, the synthesis of melatonin derivatives designed as new multitarget-directed products has focused a great interest in this area. This latter has been reinforced by the low cost of melatonin and its reduced toxicity. Furthermore, its spectrum of usages seems to be wide and with the potential for improving human health. Nevertheless, the molecular and cellular mechanisms underlying melatonin´s actions need to be further exploration and accordingly, new clinical studies should be conducted in human patients with ischemic brain pathologies.
Collapse
Affiliation(s)
- Eva Ramos
- Department of Toxicology & Pharmacology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Paloma Patiño
- Paediatric Unit, La Paz University Hospital, Paseo de la Castellana 261, 28046 Madrid, Spain
| | - Russel J Reiter
- Department of Cellular and Structural Biology. University of Texas Health Science Center at San Antonio, USA
| | - Emilio Gil-Martín
- Department of Biochemistry, Genetics and Immunology, Faculty of Biology, University of Vigo, Vigo, Spain
| | - José Marco-Contelles
- Medicinal Chemistry Laboratory, Institute of General Organic Chemistry (CSIC), Juan de la Cierva, 3, 28006 Madrid, Spain
| | - Esther Parada
- Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain; Instituto de I+D del Medicamento Teófilo Hernando (ITH), Facultad de Medicina, Universidad Autónoma de Madrid, Spain
| | - Cristobal de Los Rios
- Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain; Instituto de I+D del Medicamento Teófilo Hernando (ITH), Facultad de Medicina, Universidad Autónoma de Madrid, Spain
| | - Alejandro Romero
- Department of Toxicology & Pharmacology, Faculty of Veterinary Medicine, Complutense University of Madrid, 28040 Madrid, Spain.
| | - Javier Egea
- Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, 28006 Madrid, Spain; Instituto de I+D del Medicamento Teófilo Hernando (ITH), Facultad de Medicina, Universidad Autónoma de Madrid, Spain.
| |
Collapse
|
50
|
Gross A, Benninger F, Madar R, Illouz T, Griffioen K, Steiner I, Offen D, Okun E. Toll-like receptor 3 deficiency decreases epileptogenesis in a pilocarpine model of SE-induced epilepsy in mice. Epilepsia 2017; 58:586-596. [DOI: 10.1111/epi.13688] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/27/2016] [Indexed: 12/28/2022]
Affiliation(s)
- Adi Gross
- The Mina and Everard Goodman Faculty of Life Sciences; Bar-Ilan University; Ramat-Gan Israel
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center; Bar-Ilan University; Ramat-Gan Israel
- The Paul Feder Laboratory on Alzheimer's disease research; Tel-Aviv University; Tel Aviv Israel
| | - Felix Benninger
- The Mina and Everard Goodman Faculty of Life Sciences; Bar-Ilan University; Ramat-Gan Israel
- The Neuroscience Laboratory; Felsenstein Medical Research Center; Tel-Aviv University; Tel Aviv Israel
- Department of Neurology; Rabin Medical Center; Petach Tikva Israel
| | - Ravit Madar
- The Mina and Everard Goodman Faculty of Life Sciences; Bar-Ilan University; Ramat-Gan Israel
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center; Bar-Ilan University; Ramat-Gan Israel
- The Paul Feder Laboratory on Alzheimer's disease research; Tel-Aviv University; Tel Aviv Israel
| | - Tomer Illouz
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center; Bar-Ilan University; Ramat-Gan Israel
- The Paul Feder Laboratory on Alzheimer's disease research; Tel-Aviv University; Tel Aviv Israel
| | - Kathleen Griffioen
- Department of Biology and Chemistry; Liberty University; Lynchburg Virginia U.S.A
| | - Israel Steiner
- Department of Neurology; Rabin Medical Center; Petach Tikva Israel
| | - Daniel Offen
- The Neuroscience Laboratory; Felsenstein Medical Research Center; Tel-Aviv University; Tel Aviv Israel
| | - Eitan Okun
- The Mina and Everard Goodman Faculty of Life Sciences; Bar-Ilan University; Ramat-Gan Israel
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center; Bar-Ilan University; Ramat-Gan Israel
- The Paul Feder Laboratory on Alzheimer's disease research; Tel-Aviv University; Tel Aviv Israel
| |
Collapse
|