401
|
Cell-type-specific epigenetic effects of early life stress on the brain. Transl Psychiatry 2022; 12:326. [PMID: 35948532 PMCID: PMC9365848 DOI: 10.1038/s41398-022-02076-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 02/08/2023] Open
Abstract
Early life stress (ELS) induces long-term phenotypic adaptations that contribute to increased vulnerability to a host of neuropsychiatric disorders. Epigenetic mechanisms, including DNA methylation, histone modifications and non-coding RNA, are a proposed link between environmental stressors, alterations in gene expression, and phenotypes. Epigenetic modifications play a primary role in shaping functional differences between cell types and can be modified by environmental perturbations, especially in early development. Together with contributions from genetic variation, epigenetic mechanisms orchestrate patterns of gene expression within specific cell types that contribute to phenotypic variation between individuals. To date, many studies have provided insights into epigenetic changes resulting from ELS. However, most of these studies have examined heterogenous brain tissue, despite evidence of cell-type-specific epigenetic modifications in phenotypes associated with ELS. In this review, we focus on rodent and human studies that have examined epigenetic modifications induced by ELS in select cell types isolated from the brain or associated with genes that have cell-type-restricted expression in neurons, microglia, astrocytes, and oligodendrocytes. Although significant challenges remain, future studies using these approaches can enable important mechanistic insight into the role of epigenetic variation in the effects of ELS on brain function.
Collapse
|
402
|
Aronica E, Binder DK, Drexel M, Ikonomidou C, Kadam SD, Sperk G, Steinhäuser C. A companion to the preclinical common data elements and case report forms for neuropathology studies in epilepsy research. A report of the TASK3 WG2 Neuropathology Working Group of the ILAE/AES Joint Translational Task Force. Epilepsia Open 2022. [PMID: 35938285 DOI: 10.1002/epi4.12638] [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: 11/27/2021] [Accepted: 01/28/2022] [Indexed: 11/06/2022] Open
Abstract
The International League Against Epilepsy/American Epilepsy Society (ILAE/AES) Joint Translational Task Force initiated the TASK3 working group to create common data elements (CDEs) for various aspects of preclinical epilepsy research studies, which could help improve the standardization of experimental designs. This article addresses neuropathological changes associated with seizures and epilepsy in rodent models of epilepsy. We discuss CDEs for histopathological parameters for neurodegeneration, changes in astrocyte morphology and function, mechanisms of inflammation, and changes in the blood-brain barrier and myelin/oligodendrocytes resulting from recurrent seizures in rats and mice. We provide detailed CDE tables and case report forms (CRFs), and with this companion manuscript, we discuss the rationale and methodological aspects of individual neuropathological examinations. The CDEs, CRFs, and companion paper are available to all researchers, and their use will benefit the harmonization and comparability of translational preclinical epilepsy research. The ultimate hope is to facilitate the development of rational therapy concepts for treating epilepsies, seizures, and comorbidities and the development of biomarkers assessing the pathological state of the disease.
Collapse
Affiliation(s)
- Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| | - Devin K Binder
- Center for Glial-Neuronal Interactions, Division of Biomedical Sciences, School of Medicine, University of California, Riverside, California, USA
| | - Meinrad Drexel
- Department of Genetics and Pharmacology, Institute of Molecular and Cellular Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| | | | - Shilpa D Kadam
- The Hugo Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Guenther Sperk
- Department of Pharmacology, Medical University Innsbruck, Innsbruck, Austria
| | - Christian Steinhäuser
- Institute of Cellular Neurosciences, Medical School, University of Bonn, Bonn, Germany
| |
Collapse
|
403
|
Pischiutta F, Caruso E, Cavaleiro H, Salgado AJ, Loane DJ, Zanier ER. Mesenchymal stromal cell secretome for traumatic brain injury: Focus on immunomodulatory action. Exp Neurol 2022; 357:114199. [PMID: 35952763 DOI: 10.1016/j.expneurol.2022.114199] [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: 03/18/2022] [Revised: 06/14/2022] [Accepted: 08/03/2022] [Indexed: 11/15/2022]
Abstract
The severity and long-term consequences of brain damage in traumatic brain injured (TBI) patients urgently calls for better neuroprotective/neuroreparative strategies for this devastating disorder. Mesenchymal stromal cells (MSCs) hold great promise and have been shown to confer neuroprotection in experimental TBI, mainly through paracrine mechanisms via secreted bioactive factors (i.e. secretome), which indicates significant potential for a cell-free neuroprotective approach. The secretome is composed of cytokines, chemokines, growth factors, proteins, lipids, nucleic acids, metabolites, and extracellular vesicles; it may offer advantages over MSCs in terms of delivery, safety, and variability of therapeutic response for brain injury. Immunomodulation by molecular factors secreted by MSCs is considered to be a key mechanism involved in their multi-potential therapeutic effects. Regulated neuroinflammation is required for healthy remodeling of central nervous system during development and adulthood. Moreover, immune cells and their secreted factors can also contribute to tissue repair and neurological recovery following acute brain injury. However, a chronic and maladaptive neuroinflammatory response can exacerbate TBI and contribute to progressive neurodegeneration and long-term neurological impairments. Here, we review the evidence for MSC-derived secretome as a therapy for TBI. Our framework incorporates a detailed analysis of in vitro and in vivo studies investigating the effects of the secretome on clinically relevant neurological and histopathological outcomes. We also describe the activation of immune cells after TBI and the immunomodulatory properties exerted by mediators released in the secretome. We then describe how ageing modifies central and systemic immune responses to TBI and discuss challenges and opportunities of developing secretome based neuroprotective therapies for elderly TBI populations. Finally, strategies aimed at modulating the secretome in order to boost its efficacy for TBI will also be discussed.
Collapse
Affiliation(s)
- Francesca Pischiutta
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Neuroscience, Milan, Italy
| | - Enrico Caruso
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Neuroscience, Milan, Italy; Neuroscience Intensive Care Unit, Department of Anesthesia and Critical Care, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Helena Cavaleiro
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Neuroscience, Milan, Italy; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal; Stemmatters, Biotechnology and Regenerative Medicine, Guimarães, Portugal
| | - Antonio J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - David J Loane
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Elisa R Zanier
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Neuroscience, Milan, Italy.
| |
Collapse
|
404
|
Llorente-Ovejero A, Bengoetxea de Tena I, Martínez-Gardeazabal J, Moreno-Rodríguez M, Lombardero L, Manuel I, Rodríguez-Puertas R. Cannabinoid Receptors and Glial Response Following a Basal Forebrain Cholinergic Lesion. ACS Pharmacol Transl Sci 2022; 5:791-802. [PMID: 36110372 PMCID: PMC9469185 DOI: 10.1021/acsptsci.2c00069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Indexed: 11/28/2022]
Abstract
The endocannabinoid system modulates learning, memory, and neuroinflammatory processes, playing a key role in neurodegeneration, including Alzheimer's disease (AD). Previous results in a rat lesion model of AD showed modulation of endocannabinoid receptor activity in the basalo-cortical pathway following a specific lesion of basal forebrain cholinergic neurons (BFCNs), indicating that the glial neuroinflammatory response accompanying the lesion is related to endocannabinoid signaling. In this study, 7 days after the lesion, decreased astrocyte and increased microglia immunoreactivities (GFAP and Iba-1) were observed, indicating microglia-mediated neuroinflammation. Using autoradiographic studies, the density and functional coupling to G-proteins of endocannabinoid receptor subtypes were studied in tissue sections from different brain areas where microglia density increased, using CB1 and CB2 selective agonists and antagonists. In the presence of the specific CB1 receptor antagonist, SR141716A, [3H]CP55,940 binding (receptor density) was completely blocked in a dose-dependent manner, while the selective CB2 receptor antagonist, SR144528, inhibited binding to 25%, at best. [35S]GTPγS autoradiography (receptor coupling to Gi/0-proteins) evoked by CP55,940 (CB1/CB2 agonist) and HU308 (more selective for CB2) was abolished by SR141716A in all areas, while SR144528 blocked up to 51.8% of the coupling to Gi/0-proteins evoked by CP55,940 restricted to the nucleus basalis magnocellularis. Together, these results demonstrate that there are increased microglia and decreased astrocyte immunoreactivities 1 week after a specific deletion of BFCNs, which projects to cortical areas, where the CB1 receptor coupling to Gi/0-proteins is upregulated. However, at the lesion site, the area with the highest neuroinflammatory response, there is also a limited contribution of CB2.
Collapse
Affiliation(s)
| | | | - Jonatan Martínez-Gardeazabal
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa 48940, Spain
- Neurodegenerative Diseases, Biocruces Bizkaia Health Research Institute, Barakaldo 48903, Spain
| | - Marta Moreno-Rodríguez
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa 48940, Spain
| | - Laura Lombardero
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa 48940, Spain
| | - Iván Manuel
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa 48940, Spain
- Neurodegenerative Diseases, Biocruces Bizkaia Health Research Institute, Barakaldo 48903, Spain
| | - Rafael Rodríguez-Puertas
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa 48940, Spain
- Neurodegenerative Diseases, Biocruces Bizkaia Health Research Institute, Barakaldo 48903, Spain
| |
Collapse
|
405
|
Doughty D, Rajpurohit SK, Trang A, Alptekin A, Korkaya AK, Achyut BR, Arbab AS, Bradford JW. Development of a novel purification protocol to isolate and identify brain microglia. Exp Biol Med (Maywood) 2022; 247:1433-1446. [PMID: 35666093 PMCID: PMC9493764 DOI: 10.1177/15353702221096060] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Microglia, the tissue-resident macrophage of the central nervous system (CNS), play a paramount role in brain health and disease status. Here, we describe a novel method for enriching and isolating primary microglia from mouse brain tissue. This isolation method yields a high number of cells from either young or adult mice, and importantly, maintains the health and function of the cells for subsequent cell culture. We also describe flow cytometry methods using novel cell surface markers, including CX3CR1 and Siglec-H, to specifically label microglia while avoiding other bone marrow and/or non-CNS derived macrophages and monocytes, which has been historically difficult to achieve. As microglia are crucial in multiple aspects of biology, such as in normal brain development/function, immune response, neurodegeneration, and cancer, this isolation technique could greatly benefit a wide range of studies in human CNS biology, health, and disease mechanisms. Being able to isolate a largely pure population of microglia could also allow for a more comprehensive understanding of their functional dynamics and role in disease mechanisms, advancement of potential biomarkers, and development of novel therapeutic targets to improve prognosis and quality of life in multiple diseases.
Collapse
Affiliation(s)
- Deanna Doughty
- Department of Biological Sciences,
Augusta University, Augusta, GA 30912, USA,D. D. is Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
| | | | - Amy Trang
- Department of Biological Sciences,
Augusta University, Augusta, GA 30912, USA
| | - Ahmet Alptekin
- Georgia Cancer Center, Augusta
University, Augusta, GA 30912, USA
| | - Ahmet K Korkaya
- Department of Biological Sciences,
Augusta University, Augusta, GA 30912, USA
| | - Bhagelu R Achyut
- Winship Cancer Institute, Emory
University, Atlanta, GA 30322, USA
| | - Ali S Arbab
- Georgia Cancer Center, Augusta
University, Augusta, GA 30912, USA
| | - Jennifer W Bradford
- Department of Biological Sciences,
Augusta University, Augusta, GA 30912, USA,Jennifer W Bradford.
| |
Collapse
|
406
|
Andrijevic D, Vrselja Z, Lysyy T, Zhang S, Skarica M, Spajic A, Dellal D, Thorn SL, Duckrow RB, Ma S, Duy PQ, Isiktas AU, Liang D, Li M, Kim SK, Daniele SG, Banu K, Perincheri S, Menon MC, Huttner A, Sheth KN, Gobeske KT, Tietjen GT, Zaveri HP, Latham SR, Sinusas AJ, Sestan N. Cellular recovery after prolonged warm ischaemia of the whole body. Nature 2022; 608:405-412. [PMID: 35922506 PMCID: PMC9518831 DOI: 10.1038/s41586-022-05016-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 06/23/2022] [Indexed: 02/05/2023]
Abstract
After cessation of blood flow or similar ischaemic exposures, deleterious molecular cascades commence in mammalian cells, eventually leading to their death1,2. Yet with targeted interventions, these processes can be mitigated or reversed, even minutes or hours post mortem, as also reported in the isolated porcine brain using BrainEx technology3. To date, translating single-organ interventions to intact, whole-body applications remains hampered by circulatory and multisystem physiological challenges. Here we describe OrganEx, an adaptation of the BrainEx extracorporeal pulsatile-perfusion system and cytoprotective perfusate for porcine whole-body settings. After 1 h of warm ischaemia, OrganEx application preserved tissue integrity, decreased cell death and restored selected molecular and cellular processes across multiple vital organs. Commensurately, single-nucleus transcriptomic analysis revealed organ- and cell-type-specific gene expression patterns that are reflective of specific molecular and cellular repair processes. Our analysis comprises a comprehensive resource of cell-type-specific changes during defined ischaemic intervals and perfusion interventions spanning multiple organs, and it reveals an underappreciated potential for cellular recovery after prolonged whole-body warm ischaemia in a large mammal.
Collapse
Affiliation(s)
- David Andrijevic
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA.,These authors contributed equally: David Andrijevic, Zvonimir Vrselja, Taras Lysyy, Shupei Zhang
| | - Zvonimir Vrselja
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA.,These authors contributed equally: David Andrijevic, Zvonimir Vrselja, Taras Lysyy, Shupei Zhang
| | - Taras Lysyy
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA.,Department of Surgery, Yale School of Medicine New Haven, New Haven, CT, USA.,These authors contributed equally: David Andrijevic, Zvonimir Vrselja, Taras Lysyy, Shupei Zhang
| | - Shupei Zhang
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA.,Department of Genetics, Yale School of Medicine, New Haven, CT, USA.,These authors contributed equally: David Andrijevic, Zvonimir Vrselja, Taras Lysyy, Shupei Zhang
| | - Mario Skarica
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Ana Spajic
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - David Dellal
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA.,Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Stephanie L. Thorn
- Yale Translational Research Imaging Center, Department of Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Robert B. Duckrow
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Shaojie Ma
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Phan Q. Duy
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA.,Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA.,Medical Scientist Training Program (MD-PhD), Yale School of Medicine, New Haven, CT, USA
| | - Atagun U. Isiktas
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Dan Liang
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Mingfeng Li
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Suel-Kee Kim
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Stefano G. Daniele
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA.,Medical Scientist Training Program (MD-PhD), Yale School of Medicine, New Haven, CT, USA
| | - Khadija Banu
- Department of Nephrology, Yale School of Medicine, New Haven, CT, USA
| | - Sudhir Perincheri
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Madhav C. Menon
- Department of Nephrology, Yale School of Medicine, New Haven, CT, USA
| | - Anita Huttner
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Kevin N. Sheth
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA.,Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Kevin T. Gobeske
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Gregory T. Tietjen
- Department of Surgery, Yale School of Medicine New Haven, New Haven, CT, USA.,Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Hitten P. Zaveri
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Stephen R. Latham
- Interdisciplinary Center for Bioethics, Yale University, New Haven, CT, USA
| | - Albert J. Sinusas
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA.,Department of Biomedical Engineering, Yale University, New Haven, CT, USA.,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.,Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
| | - Nenad Sestan
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA. .,Department of Genetics, Yale School of Medicine, New Haven, CT, USA. .,Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA. .,Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA. .,Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT, USA. .,Yale Child Study Center, New Haven, CT, USA.
| |
Collapse
|
407
|
Klein L, Ophelders DR, van den Hove D, Damoiseaux M, Rutten BP, Reutelingsperger CP, Schurgers LJ, Wolfs TG. Prenatal administration of multipotent adult progenitor cells modulates the systemic and cerebral immune response in an ovine model of chorioamnionitis. Brain Behav Immun Health 2022; 23:100458. [PMID: 35647567 PMCID: PMC9136278 DOI: 10.1016/j.bbih.2022.100458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/17/2022] [Accepted: 03/31/2022] [Indexed: 11/30/2022] Open
Abstract
Systemic and cerebral inflammation following antenatal infection (e.g. chorioamnionitis) and dysregulation of the blood brain barrier (BBB) are major risk factors for abnormal neonatal brain development. Administration of multipotent adult progenitor cells (MAPCs) represents an interesting pharmacological strategy as modulator of the peripheral and cerebral immune response and protector of BBB integrity. We studied the immunomodulatory and protective cerebrovascular potential of prenatally administered MAPCs in a preclinical ovine model for antenatal inflammation. Ovine fetuses were intra-amniotically (i.a.) exposed to lipopolysaccharide (LPS) or saline at gestational day 125, followed by the intravenous administration of 1*107 MAPCs or saline at gestational day 127. Circulating inflammation markers were measured. Fetal brains were examined immuno-histochemically post-mortem at gestational day 132. Fetal plasma IL-6 levels were elevated significantly 24 h after LPS administration. In utero systemic MAPC treatment after LPS exposure increased Annexin A1 (ANXA1) expression in the cerebrovascular endothelium, indicating enforcement of BBB integrity, and increased the number of leukocytes at brain barriers throughout the brain. Further characterisation of brain barrier-associated leukocytes showed that monocyte/choroid plexus macrophage (IBA-1+/CD206+) and neutrophil (MPO+) populations predominantly contributed to the LPS-MAPC-induced increase of CD45+cells. In the choroid plexus, the percentage of leukocytes expressing the proresolving mediator ANXA1 tended to be decreased after LPS-induced antenatal inflammation, an effect reversed by systemic MAPC treatment. Accordingly, expression levels of ANXA1 per leukocyte were decreased after LPS and restored after subsequent MAPC treatment. Increased expression of ANXA1 by the cerebrovasculature and immune cells at brain barriers following MAPC treatment in an infectious setting indicate a MAPC driven early defence mechanism to protect the neonatal brain against infection-driven inflammation and potential additional pro-inflammatory insults in the neonatal period. MAPCs administered systemically enhance the brain directed immune response in an inflammation dependent manner in preterm fetuses. Annexin A1 expression is increased in cerebrovasculature and immune cells at brain barriers when MAPCs were i.v. administered in the infectious setting. MAPCs potentially protect the neonatal brain by enforcing the blood brain barrier and modulating inflammation.
Collapse
Affiliation(s)
- Luise Klein
- School for Oncology and Reproduction (GROW), Maastricht University, Maastricht, the Netherlands
- Department of Pediatrics, Maastricht University, Maastricht, the Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, the Netherlands
| | - Daan R.M.G. Ophelders
- School for Oncology and Reproduction (GROW), Maastricht University, Maastricht, the Netherlands
- Department of Pediatrics, Maastricht University, Maastricht, the Netherlands
| | - Daniel van den Hove
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, the Netherlands
- Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany
- Department of Psychiatry and Neuropsychology, European Graduate School of Neuroscience (EURON), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, the Netherlands
| | - Maurits Damoiseaux
- School for Oncology and Reproduction (GROW), Maastricht University, Maastricht, the Netherlands
- Department of Pediatrics, Maastricht University, Maastricht, the Netherlands
| | - Bart P.F. Rutten
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, the Netherlands
- Department of Psychiatry and Neuropsychology, European Graduate School of Neuroscience (EURON), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, the Netherlands
| | - Chris P.M. Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | - Leon J. Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands
| | - Tim G.A.M. Wolfs
- School for Oncology and Reproduction (GROW), Maastricht University, Maastricht, the Netherlands
- Department of Pediatrics, Maastricht University, Maastricht, the Netherlands
- Corresponding author. School for Oncology and Reproduction (GROW), Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
408
|
Huynh H, Upadhyay P, Lopez CH, Miyashiro MK, Van Winkle LS, Thomasy SM, Pinkerton KE. Inhalation of Silver Silicate Nanoparticles Leads to Transient and Differential Microglial Activation in the Rodent Olfactory Bulb. Toxicol Pathol 2022; 50:763-775. [PMID: 35768951 PMCID: PMC9529873 DOI: 10.1177/01926233221107607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Engineered silver nanoparticles (AgNPs), including silver silicate nanoparticles (Ag-SiO2 NPs), are used in a wide variety of medical and consumer applications. Inhaled AgNPs have been found to translocate to the olfactory bulb (OB) after inhalation and intranasal instillation. However, the biological effects of Ag-SiO2 NPs and their potential nose-to-brain transport have not been evaluated. The present study assessed whether inhaled Ag-SiO2 NPs can elicit microglial activation in the OB. Adult Sprague-Dawley rats inhaled aerosolized Ag-SiO2 NPs at a concentration of 1 mg/ml for 6 hours. On day 0, 1, 7, and 21 post-exposure, rats were necropsied and OB were harvested. Immunohistochemistry on OB tissues were performed with anti-ionized calcium-binding adapter molecule 1 and heme oxygenase-1 as markers of microglial activation and oxidative stress, respectively. Aerosol characterization indicated Ag-SiO2 NPs were sufficiently aerosolized with moderate agglomeration and high-efficiency deposition in the nasal cavity and olfactory epithelium. Findings suggested that acute inhalation of Ag-SiO2 NPs elicited transient and differential microglial activation in the OB without significant microglial recruitment or oxidative stress. The delayed and differential pattern of microglial activation in the OB implied that inhaled Ag-SiO2 may have translocated to the central nervous system via intra-neuronal pathways.
Collapse
Affiliation(s)
- Huong Huynh
- William R Pritchard Veterinary Medical Teaching Hospital, University of California-Davis, Davis, CA, USA.,Center for Health and the Environment, University of California – Davis, Davis, CA, USA
| | - Priya Upadhyay
- Center for Health and the Environment, University of California – Davis, Davis, CA, USA
| | - Cora H Lopez
- Center for Health and the Environment, University of California – Davis, Davis, CA, USA
| | - Malia K Miyashiro
- Center for Health and the Environment, University of California – Davis, Davis, CA, USA
| | - Laura S Van Winkle
- Center for Health and the Environment, University of California – Davis, Davis, CA, USA.,Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California – Davis, Davis, CA, USA
| | - Sara M Thomasy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, Davis, CA, USA.,Department of Ophthalmology and Vision Science, School of Medicine, University of California - Davis, Davis, CA, USA
| | - Kent E Pinkerton
- Center for Health and the Environment, University of California – Davis, Davis, CA, USA.,Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California – Davis, Davis, CA, USA
| |
Collapse
|
409
|
Song J, Han K, Wang Y, Qu R, Liu Y, Wang S, Wang Y, An Z, Li J, Wu H, Wu W. Microglial Activation and Oxidative Stress in PM2.5-Induced Neurodegenerative Disorders. Antioxidants (Basel) 2022; 11:antiox11081482. [PMID: 36009201 PMCID: PMC9404971 DOI: 10.3390/antiox11081482] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 07/20/2022] [Accepted: 07/27/2022] [Indexed: 11/22/2022] Open
Abstract
Fine particulate matter (PM2.5) pollution remains a prominent environmental problem worldwide, posing great threats to human health. The adverse effects of PM2.5 on the respiratory and cardiovascular systems have been extensively studied, while its detrimental effects on the central nervous system (CNS), specifically neurodegenerative disorders, are less investigated. Neurodegenerative disorders are characterized by reduced neurogenesis, activated microglia, and neuroinflammation. A variety of studies involving postmortem examinations, epidemiological investigations, animal experiments, and in vitro cell models have shown that PM2.5 exposure results in neuroinflammation, oxidative stress, mitochondrial dysfunction, neuronal apoptosis, and ultimately neurodegenerative disorders, which are strongly associated with the activation of microglia. Microglia are the major innate immune cells of the brain, surveilling and maintaining the homeostasis of CNS. Upon activation by environmental and endogenous insults, such as PM exposure, microglia can enter an overactivated state that is featured by amoeboid morphology, the over-production of reactive oxygen species, and pro-inflammatory mediators. This review summarizes the evidence of microglial activation and oxidative stress and neurodegenerative disorders following PM2.5 exposure. Moreover, the possible mechanisms underlying PM2.5-induced microglial activation and neurodegenerative disorders are discussed. This knowledge provides certain clues for the development of therapies that may slow or halt the progression of neurodegenerative disorders induced by ambient PM.
Collapse
Affiliation(s)
- Jie Song
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.S.); (K.H.); (R.Q.); (Y.L.); (S.W.); (Y.W.); (Z.A.); (J.L.); (H.W.)
| | - Keyang Han
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.S.); (K.H.); (R.Q.); (Y.L.); (S.W.); (Y.W.); (Z.A.); (J.L.); (H.W.)
| | - Ya Wang
- Nursing School, Zhenjiang College, Zhenjiang 212028, China;
| | - Rongrong Qu
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.S.); (K.H.); (R.Q.); (Y.L.); (S.W.); (Y.W.); (Z.A.); (J.L.); (H.W.)
| | - Yuan Liu
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.S.); (K.H.); (R.Q.); (Y.L.); (S.W.); (Y.W.); (Z.A.); (J.L.); (H.W.)
| | - Shaolan Wang
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.S.); (K.H.); (R.Q.); (Y.L.); (S.W.); (Y.W.); (Z.A.); (J.L.); (H.W.)
| | - Yinbiao Wang
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.S.); (K.H.); (R.Q.); (Y.L.); (S.W.); (Y.W.); (Z.A.); (J.L.); (H.W.)
| | - Zhen An
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.S.); (K.H.); (R.Q.); (Y.L.); (S.W.); (Y.W.); (Z.A.); (J.L.); (H.W.)
| | - Juan Li
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.S.); (K.H.); (R.Q.); (Y.L.); (S.W.); (Y.W.); (Z.A.); (J.L.); (H.W.)
| | - Hui Wu
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.S.); (K.H.); (R.Q.); (Y.L.); (S.W.); (Y.W.); (Z.A.); (J.L.); (H.W.)
| | - Weidong Wu
- School of Public Health, Xinxiang Medical University, Xinxiang 453003, China; (J.S.); (K.H.); (R.Q.); (Y.L.); (S.W.); (Y.W.); (Z.A.); (J.L.); (H.W.)
- Correspondence:
| |
Collapse
|
410
|
The Role of Vitamin D in Alzheimer’s Disease: A Transcriptional Regulator of Amyloidopathy and Gliopathy. Biomedicines 2022; 10:biomedicines10081824. [PMID: 36009371 PMCID: PMC9404847 DOI: 10.3390/biomedicines10081824] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by amyloid-beta (Aβ) accumulation and cognitive mental decline. Epidemiological studies have suggested an association between low serum vitamin D levels and an increased risk of AD. Vitamin D regulates gene expression via the vitamin D receptor, a nuclear ligand-dependent transcription factor. However, the molecular mechanism underlying the pathogenic and therapeutic effects of vitamin D on AD is not fully understood yet. To better understand how vitamin D regulates the expression of genes related to AD pathology, first, we induced vitamin D deficiency in 5xFAD mice by providing a vitamin-D-deficient diet and observed the changes in the mRNA level of genes related to Aβ processing, which resulted in an increase in the Aβ load in the brain. The vitamin D-deficient diet also suppressed the expression of genes for microglial Aβ phagocytosis. Interestingly, vitamin D deficiency in the early stage of AD resulted in earlier memory impairment. In addition, we administered vitamin D intraperitoneally to 5xFAD mice with a normal diet and found lower Aβ levels with the suppressed expression of genes for Aβ generation and observed improved memory function, which may be potentially associated with reduced MAO-B expression. These findings strongly suggest the role of vitamin D as a crucial disease-modifying factor that may modulate the amyloid pathology with regard to reducing AD symptoms.
Collapse
|
411
|
Badrah A, Al-Tuwairqi S. Modeling the dynamics of innate immune response to Parkinson disease with therapeutic approach. Phys Biol 2022; 19. [PMID: 35901788 DOI: 10.1088/1478-3975/ac8516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/28/2022] [Indexed: 11/12/2022]
Abstract
This paper aims to mathematically model the dynamics of Parkinson's disease with therapeutic strategies. The constructed model consists of five state variables: healthy neurons, infected neurons, extracellular $\alpha$-syn, active microglia, and resting microglia. The qualitative analysis of the model produced an unstable free equilibrium point and a stable endemic equilibrium point. Moreover, these results are validated by numerical experiments with different initial values. Two therapeutic interventions, reduction of extracellular $\alpha$-syn and reduction of inflammation induced by activated microglia in the central nervous system, are investigated. It is observed that the latter has no apparent effect in delaying the deterioration of neurons. However, treatment to reduce extracellular $\alpha$-syn preserves neurons and delays the onset of Parkinson's disease, whether alone or in combination with another treatment.
Collapse
Affiliation(s)
- Asma Badrah
- Mathematics, King Abdulaziz University, Jeddah, Saudi Arabia, Jeddah, 21589, SAUDI ARABIA
| | - Salma Al-Tuwairqi
- Mathematics, King Abdulaziz University, Jeddah, Saudi Arabia, Jeddah, 21589, SAUDI ARABIA
| |
Collapse
|
412
|
WKYMVm/FPR2 Alleviates Spinal Cord Injury by Attenuating the Inflammatory Response of Microglia. Mediators Inflamm 2022; 2022:4408099. [PMID: 35935810 PMCID: PMC9348919 DOI: 10.1155/2022/4408099] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/11/2022] [Indexed: 11/17/2022] Open
Abstract
Spinal cord injury (SCI) is a common traumatic disease of the nervous system. The pathophysiological process of SCI includes primary injury and secondary injuries. An excessive inflammatory response leads to secondary tissue damage, which in turn exacerbates cellular and organ dysfunction. Due to the irreversibility of primary injury, current research on SCI mainly focuses on secondary injury, and the inflammatory response is considered the primary target. Thus, modulating the inflammatory response has been suggested as a new strategy for the treatment of SCI. In this study, microglial cell lines, primary microglia, and a rat SCI model were used, and we found that WKYMVm/FPR2 plays an anti-inflammatory role and reduces tissue damage after SCI by suppressing the extracellular signal-regulated kinases 1 and 2 (ERK1/2) and nuclear factor-κB (NF-κB) signaling pathways. FPR2 was activated by WKYMVm, suppressing the secretion of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and interleukin-1β (IL-1β) by inhibiting M1 microglial polarization. Moreover, FPR2 activation by WKYMVm could reduce structural disorders and neuronal loss in SCI rats. Overall, this study illustrated that the activation of FPR2 by WKYMVm repressed M1 microglial polarization by suppressing the ERK1/2 and NF-κB signaling pathways to alleviate tissue damage and locomotor decline after SCI. These findings provide further insight into SCI and help identify novel treatment strategies.
Collapse
|
413
|
Myeloperoxidase as a Marker to Differentiate Mouse Monocyte/Macrophage Subsets. Int J Mol Sci 2022; 23:ijms23158246. [PMID: 35897821 PMCID: PMC9330004 DOI: 10.3390/ijms23158246] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/21/2022] [Accepted: 07/23/2022] [Indexed: 02/01/2023] Open
Abstract
Macrophages are present in every tissue in the body and play essential roles in homeostasis and host defense against microorganisms. Some tissue macrophages derive from the yolk sac/fetal liver that populate tissues for life. Other tissue macrophages derive from monocytes that differentiate in the bone marrow and circulate through tissues via the blood and lymphatics. Circulating monocytes are very plastic and differentiate into macrophages with specialized functions upon entering tissues. Specialized monocyte/macrophage subsets have been difficult to differentiate based on cell surface markers. Here, using a combination of "pan" monocyte/macrophage markers and flow cytometry, we asked whether myeloperoxidase (MPO) could be used as a marker of pro-inflammatory monocyte/macrophage subsets. MPO is of interest because of its potent microbicidal activity. In wild-type SPF housed mice, we found that MPO+ monocytes/macrophages were present in peripheral blood, spleen, small and large intestines, and mesenteric lymph nodes, but not the central nervous system. Only monocytes/macrophages that expressed cell surface F4/80 and/or Ly6C co-expressed MPO with the highest expression in F4/80HiLy6CHi subsets regardless of tissue. These cumulative data indicate that MPO expression can be used as an additional marker to differentiate between monocyte/macrophage subsets with pro-inflammatory and microbicidal activity in a variety of tissues.
Collapse
|
414
|
Activation of NLRP3 Is Required for a Functional and Beneficial Microglia Response after Brain Trauma. Pharmaceutics 2022; 14:pharmaceutics14081550. [PMID: 35893807 PMCID: PMC9332196 DOI: 10.3390/pharmaceutics14081550] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 12/04/2022] Open
Abstract
Despite the numerous research studies on traumatic brain injury (TBI), many physiopathologic mechanisms remain unknown. TBI is a complex process, in which neuroinflammation and glial cells play an important role in exerting a functional immune and damage-repair response. The activation of the NLRP3 inflammasome is one of the first steps to initiate neuroinflammation and so its regulation is essential. Using a closed-head injury model and a pharmacological (MCC950; 3 mg/kg, pre- and post-injury) and genetical approach (NLRP3 knockout (KO) mice), we defined the transcriptional and behavioral profiles 24 h after TBI. Wild-type (WT) mice showed a strong pro-inflammatory response, with increased expression of inflammasome components, microglia and astrocytes markers, and cytokines. There was no difference in the IL1β production between WT and KO, nor compensatory mechanisms of other inflammasomes. However, some microglia and astrocyte markers were overexpressed in KO mice, resulting in an exacerbated cytokine expression. Pretreatment with MCC950 replicated the behavioral and blood-brain barrier results observed in KO mice and its administration 1 h after the lesion improved the damage. These findings highlight the importance of NLRP3 time-dependent activation and its role in the fine regulation of glial response.
Collapse
|
415
|
Ceccarelli L, Marchetti L, Rizzo M, Moscardini A, Cappello V, Da Pozzo E, Romano M, Giacomelli C, Bergese P, Martini C. Human Microglia Extracellular Vesicles Derived from Different Microglia Cell Lines: Similarities and Differences. ACS OMEGA 2022; 7:23127-23137. [PMID: 35847267 PMCID: PMC9280972 DOI: 10.1021/acsomega.2c00816] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Microglial cells are a component of the innate immune system in the brain that support cell-to-cell communication via secreted molecules and extracellular vesicles (EVs). EVs can be divided into two major populations: large (LEVs) and small (SEVs) EVs, carrying different mediators, such as proteins, lipids, and miRNAs. The microglia EVs cargo crucially reflects the status of parental cells and can lead to both beneficial and detrimental effects in many physiopathological states. Herein, a workflow for the extraction and characterization of SEVs and LEVs from human C20 and HMC3 microglia cell lines derived, respectively, from adult and embryonic microglia is reported. EVs were gathered from the culture media of the two cell lines by sequential ultracentrifugation steps and their biochemical and biophysical properties were analyzed by Western blot, transmission electron microscopy, and dynamic light scattering. Although the C20- and HMC3-derived EVs shared several common features, C20-derived EVs were slightly lower in number and more polydispersed. Interestingly, C20- but not HMC3-SEVs were able to interfere with the proliferation of U87 glioblastoma cells. This correlated with the different relative levels of eight miRNAs involved in neuroinflammation and tumor progression in the C20- and HMC3-derived EVs, which in turn reflected a different basal activation state of the two cell types. Our data fill a gap in the community of microglia EVs, in which the preparations from human cells have been poorly characterized so far. Furthermore, these results shed light on both the differences and similarities of EVs extracted from different human microglia cell models, underlining the need to better characterize the features and biological effects of EVs for therein useful and correct application.
Collapse
Affiliation(s)
- Lorenzo Ceccarelli
- Department
of Pharmacy, University of Pisa, Pisa 56126, Italy
- Department
of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Laura Marchetti
- Department
of Pharmacy, University of Pisa, Pisa 56126, Italy
| | - Milena Rizzo
- Institute
of Clinical Physiology (IFC), CNR, Pisa 56124, Italy
| | - Aldo Moscardini
- SNS
(Scuola Normale Superiore, NEST laboratories), Pisa 56127, Italy
| | - Valentina Cappello
- Center
for Materials Interfaces, Electron Crystallography, Istituto Italiano di Tecnologia, Pontedera 56025, Italy
| | | | - Miriam Romano
- Department
of Molecular and Translational Medicine, University of Brescia, Brescia 25121, Italy
- Center
for Colloid and Surface Science (CSGI), Firenze 50019, Italy
| | | | - Paolo Bergese
- Department
of Molecular and Translational Medicine, University of Brescia, Brescia 25121, Italy
- Center
for Colloid and Surface Science (CSGI), Firenze 50019, Italy
- Institute
for Research and Biomedical Innovation- IRIB, Consiglio Nazionale delle Ricerche—CNR, Palermo 900146, Italy
| | - Claudia Martini
- Department
of Pharmacy, University of Pisa, Pisa 56126, Italy
| |
Collapse
|
416
|
Zhang L, Zhang X, Liu Y, Wang S, Jia G. Vagus nerve stimulation promotes the M1-to-M2 transition via inhibition of TLR4/NF-κB in microglial to rescue the reperfusion injury. J Stroke Cerebrovasc Dis 2022; 31:106596. [PMID: 35834936 DOI: 10.1016/j.jstrokecerebrovasdis.2022.106596] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE To specify the effect of vagus nerve stimulation (VNS) on microglial polarization following ischemic-reperfusion and further investigate its underlying mechanism. MATERIALS AND METHODS Sprague-Dawley rats were randomly divided into the sham, ischemic reperfusion group (IR), IR+VNS groups. VNS intervention lasting for 1 hour was administered after 30 minutes of occlusion. We analyzed the expression of Arginase 1 (Arg1), the number of M2 microglial in the peri-infarction cortex and assessed the neurological scores at the 1, 3, 7 days after reperfusion to determine the research time point. Then, we assessed polarization status of microglial, the infarct volume, neurological scores, the cellular distribution of Toll-like Receptor 4 (TLR4), the TLR4-associated pathway protein and the p-NF-κB in microglial at 3 days after reperfusion. RESULTS We found that VNS could increase the specific marker of M2 Arg1 and upregulate the M2 microglial after reperfusion, and the increase of Arg1, M2 microglial and the neurological scores was largest at the 3 days after reperfusion. VNS treatment significantly reduced the number and percent of M1, improved the number and percent of M2 and upregulated the M2 to M1 ratio without changing the number of total microglial at the 3 days after reperfusion. Moreover, VNS reduced the infarct volume and neurological deficits. In addition, VNS significantly reduced the microglial-specific TLR4, inhibited the activated TLR4/MyD88/NF-κB pathway following ischemic-reperfusion, and ultimately suppressed the p-NF-κB in microglial. CONCLUSION Our study revealed that VNS can promote the M1-to-M2 phenotype conversion to alleviate inflammatory response and brain injury through inhibition of TLR4/MyD88/NF-κB pathway in microglia following ischemic-reperfusion.
Collapse
Affiliation(s)
- Liping Zhang
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing 400010, China; Chongqing Medical University, Yixueyuan Road, Yuzhong District, Chongqing 40010, China.
| | - Xin Zhang
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing 400010, China; Chongqing Medical University, Yixueyuan Road, Yuzhong District, Chongqing 40010, China.
| | - Yilin Liu
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing 400010, China; Chongqing Medical University, Yixueyuan Road, Yuzhong District, Chongqing 40010, China
| | - Sanrong Wang
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing 400010, China
| | - Gongwei Jia
- Department of Rehabilitation, The Second Affiliated Hospital of Chongqing Medical University, 76 Linjiang Road, Yuzhong District, Chongqing 400010, China.
| |
Collapse
|
417
|
Galoppin M, Kari S, Soldati S, Pal A, Rival M, Engelhardt B, Astier A, Thouvenot E. Full spectrum of vitamin D immunomodulation in multiple sclerosis: mechanisms and therapeutic implications. Brain Commun 2022; 4:fcac171. [PMID: 35813882 PMCID: PMC9260308 DOI: 10.1093/braincomms/fcac171] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/03/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022] Open
Abstract
Vitamin D deficiency has been associated with the risk of multiple sclerosis, disease activity and progression. Results from in vitro experiments, animal models and analysis of human samples from randomized controlled trials provide comprehensive data illustrating the pleiotropic actions of Vitamin D on the immune system. They globally result in immunomodulation by decreasing differentiation of effector T and B cells while promoting regulatory subsets. Vitamin D also modulates innate immune cells such as macrophages, monocytes and dendritic cells, and acts at the level of the blood–brain barrier reducing immune cell trafficking. Vitamin D exerts additional activity within the central nervous system reducing microglial and astrocytic activation. The immunomodulatory role of Vitamin D detected in animal models of multiple sclerosis has suggested its potential therapeutic use for treating multiple sclerosis. In this review, we focus on recent published data describing the biological effects of Vitamin D in animal models of multiple sclerosis on immune cells, blood–brain barrier function, activation of glial cells and its potential neuroprotective effects. Based on the current knowledge, we also discuss optimization of therapeutic interventions with Vitamin D in patients with multiple sclerosis, as well as new technologies allowing in-depth analysis of immune cell regulations by vitamin D.
Collapse
Affiliation(s)
- Manon Galoppin
- IGF, University Montpellier, CNRS, INSERM , Montpellier , France
| | - Saniya Kari
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291 – CNRS UMR5051 – Université Toulouse III , 31024 Toulouse cedex 3 , France
| | - Sasha Soldati
- Theodor Kocher Institute, University of Bern , Bern , Switzerland
| | - Arindam Pal
- Theodor Kocher Institute, University of Bern , Bern , Switzerland
| | - Manon Rival
- IGF, University Montpellier, CNRS, INSERM , Montpellier , France
- Department of Neurology, Nîmes University Hospital, University Montpellier , Nîmes , France
| | | | - Anne Astier
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291 – CNRS UMR5051 – Université Toulouse III , 31024 Toulouse cedex 3 , France
| | - Eric Thouvenot
- IGF, University Montpellier, CNRS, INSERM , Montpellier , France
- Department of Neurology, Nîmes University Hospital, University Montpellier , Nîmes , France
| |
Collapse
|
418
|
Childers E, Bowen EFW, Rhodes CH, Granger R. Immune-Related Genomic Schizophrenic Subtyping Identified in DLPFC Transcriptome. Genes (Basel) 2022; 13:1200. [PMID: 35885983 PMCID: PMC9319783 DOI: 10.3390/genes13071200] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/26/2022] [Accepted: 06/29/2022] [Indexed: 02/04/2023] Open
Abstract
Well-documented evidence of the physiologic, genetic, and behavioral heterogeneity of schizophrenia suggests that diagnostic subtyping may clarify the underlying pathobiology of the disorder. Recent studies have demonstrated that increased inflammation may be a prominent feature of a subset of schizophrenics. However, these findings are inconsistent, possibly due to evaluating schizophrenics as a single group. In this study, we segregated schizophrenic patients into two groups ("Type 1", "Type 2") by their gene expression in the dorsolateral prefrontal cortex and explored biological differences between the subgroups. The study included post-mortem tissue samples that were sequenced in multiple, publicly available gene datasets using different sequencing methods. To evaluate the role of inflammation, the expression of genes in multiple components of neuroinflammation were examined: complement cascade activation, glial cell activation, pro-inflammatory mediator secretion, blood-brain barrier (BBB) breakdown, chemokine production and peripheral immune cell infiltration. The Type 2 schizophrenics showed widespread abnormal gene expression across all the neuroinflammation components that was not observed in Type 1 schizophrenics. Our results demonstrate the importance of separating schizophrenic patients into their molecularly defined subgroups and provide supporting evidence for the involvement of the immune-related pathways in a schizophrenic subset.
Collapse
Affiliation(s)
- Eva Childers
- Dartmouth College, Hanover, NH 03755, USA; (E.C.); (E.F.W.B.)
| | | | | | - Richard Granger
- Dartmouth College, Hanover, NH 03755, USA; (E.C.); (E.F.W.B.)
| |
Collapse
|
419
|
Ye F, Alvarez-Carbonell D, Nguyen K, Leskov K, Garcia-Mesa Y, Sreeram S, Valadkhan S, Karn J. Recruitment of the CoREST transcription repressor complexes by Nerve Growth factor IB-like receptor (Nurr1/NR4A2) mediates silencing of HIV in microglial cells. PLoS Pathog 2022; 18:e1010110. [PMID: 35797416 PMCID: PMC9295971 DOI: 10.1371/journal.ppat.1010110] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 07/19/2022] [Accepted: 06/15/2022] [Indexed: 12/13/2022] Open
Abstract
Human immune deficiency virus (HIV) infection in the brain leads to chronic neuroinflammation due to the production of pro-inflammatory cytokines, which in turn promotes HIV transcription in infected microglial cells. However, powerful counteracting silencing mechanisms in microglial cells result in the rapid shutdown of HIV expression after viral reactivation to limit neuronal damage. Here we investigated whether the Nerve Growth Factor IB-like nuclear receptor Nurr1 (NR4A2), which is a repressor of inflammation in the brain, acts directly to restrict HIV expression. HIV silencing following activation by TNF-α, or a variety of toll-like receptor (TLR) agonists, in both immortalized human microglial cells (hμglia) and induced pluripotent stem cells (iPSC)-derived human microglial cells (iMG) was enhanced by Nurr1 agonists. Similarly, overexpression of Nurr1 led to viral suppression, while conversely, knock down (KD) of endogenous Nurr1 blocked HIV silencing. The effect of Nurr1 on HIV silencing is direct: Nurr1 binds directly to the specific consensus binding sites in the U3 region of the HIV LTR and mutation of the Nurr1 DNA binding domain blocked its ability to suppress HIV-1 transcription. Chromatin immunoprecipitation (ChIP) assays also showed that after Nurr1 binding to the LTR, the CoREST/HDAC1/G9a/EZH2 transcription repressor complex is recruited to the HIV provirus. Finally, transcriptomic studies demonstrated that in addition to repressing HIV transcription, Nurr1 also downregulated numerous cellular genes involved in inflammation, cell cycle, and metabolism, further promoting HIV latency and microglial homoeostasis. Nurr1 therefore plays a pivotal role in modulating the cycles of proviral reactivation by potentiating the subsequent proviral transcriptional shutdown. These data highlight the therapeutic potential of Nurr1 agonists for inducing HIV silencing and microglial homeostasis and ultimately for the amelioration of the neuroinflammation associated with HIV-associated neurocognitive disorders (HAND).
Collapse
Affiliation(s)
- Fengchun Ye
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - David Alvarez-Carbonell
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Kien Nguyen
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Konstantin Leskov
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Yoelvis Garcia-Mesa
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Sheetal Sreeram
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Saba Valadkhan
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Jonathan Karn
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio, United States of America
| |
Collapse
|
420
|
Mineur YS, Garcia-Rivas V, Thomas MA, Soares AR, McKee SA, Picciotto MR. Sex differences in stress-induced alcohol intake: a review of preclinical studies focused on amygdala and inflammatory pathways. Psychopharmacology (Berl) 2022; 239:2041-2061. [PMID: 35359158 PMCID: PMC9704113 DOI: 10.1007/s00213-022-06120-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/14/2022] [Indexed: 02/06/2023]
Abstract
Clinical studies suggest that women are more likely than men to relapse to alcohol drinking in response to stress; however, the mechanisms underlying this sex difference are not well understood. A number of preclinical behavioral models have been used to study stress-induced alcohol intake. Here, we review paradigms used to study effects of stress on alcohol intake in rodents, focusing on findings relevant to sex differences. To date, studies of sex differences in stress-induced alcohol drinking have been somewhat limited; however, there is evidence that amygdala-centered circuits contribute to effects of stress on alcohol seeking. In addition, we present an overview of inflammatory pathways leading to microglial activation that may contribute to alcohol-dependent behaviors. We propose that sex differences in neuronal function and inflammatory signaling in circuits centered on the amygdala are involved in sex-dependent effects on stress-induced alcohol seeking and suggest that this is an important area for future studies.
Collapse
Affiliation(s)
- Yann S Mineur
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA
| | - Vernon Garcia-Rivas
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA
| | - Merrilee A Thomas
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA
| | - Alexa R Soares
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA
- Yale Interdepartmental Neuroscience Program, New Haven, CT, USA
| | - Sherry A McKee
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA
| | - Marina R Picciotto
- Department of Psychiatry, Yale University, 34 Park Street, 3Rd Floor Research, New Haven, CT, 06508, USA.
- Yale Interdepartmental Neuroscience Program, New Haven, CT, USA.
| |
Collapse
|
421
|
Wendimu MY, Hooks SB. Microglia Phenotypes in Aging and Neurodegenerative Diseases. Cells 2022; 11:2091. [PMID: 35805174 PMCID: PMC9266143 DOI: 10.3390/cells11132091] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 02/08/2023] Open
Abstract
Neuroinflammation is a hallmark of many neurodegenerative diseases (NDs) and plays a fundamental role in mediating the onset and progression of disease. Microglia, which function as first-line immune guardians of the central nervous system (CNS), are the central drivers of neuroinflammation. Numerous human postmortem studies and in vivo imaging analyses have shown chronically activated microglia in patients with various acute and chronic neuropathological diseases. While microglial activation is a common feature of many NDs, the exact role of microglia in various pathological states is complex and often contradictory. However, there is a consensus that microglia play a biphasic role in pathological conditions, with detrimental and protective phenotypes, and the overall response of microglia and the activation of different phenotypes depends on the nature and duration of the inflammatory insult, as well as the stage of disease development. This review provides a comprehensive overview of current research on the various microglia phenotypes and inflammatory responses in health, aging, and NDs, with a special emphasis on the heterogeneous phenotypic response of microglia in acute and chronic diseases such as hemorrhagic stroke (HS), Alzheimer's disease (AD), and Parkinson's disease (PD). The primary focus is translational research in preclinical animal models and bulk/single-cell transcriptome studies in human postmortem samples. Additionally, this review covers key microglial receptors and signaling pathways that are potential therapeutic targets to regulate microglial inflammatory responses during aging and in NDs. Additionally, age-, sex-, and species-specific microglial differences will be briefly reviewed.
Collapse
Affiliation(s)
| | - Shelley B. Hooks
- Hooks Lab, Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, USA;
| |
Collapse
|
422
|
Near-infrared light reduces glia activation and modulates neuroinflammation in the brains of diet-induced obese mice. Sci Rep 2022; 12:10848. [PMID: 35761012 PMCID: PMC9237037 DOI: 10.1038/s41598-022-14812-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 06/13/2022] [Indexed: 12/06/2022] Open
Abstract
Neuroinflammation is a key event in neurodegenerative conditions such as Alzheimer's disease (AD) and characterizes metabolic pathologies like obesity and type 2 diabetes (T2D). Growing evidence in humans shows that obesity increases the risk of developing AD by threefold. Hippocampal neuroinflammation in rodents correlates with poor memory performance, suggesting that it contributes to cognitive decline. Here we propose that reducing obesity/T2D-driven neuroinflammation may prevent the progression of cognitive decline associated with AD-like neurodegenerative states. Near-infrared light (NIR) has attracted increasing attention as it was shown to improve learning and memory in both humans and animal models. We previously reported that transcranial NIR delivery reduced amyloid beta and Tau pathology and improved memory function in mouse models of AD. Here, we report the effects of NIR in preventing obesity-induced neuroinflammation in a diet-induced obese mouse model. Five-week-old wild-type mice were fed a high-fat diet (HFD) for 13 weeks to induce obesity prior to transcranial delivery of NIR for 4 weeks during 90-s sessions given 5 days a week. After sacrifice, brain slices were subjected to free-floating immunofluorescence for microglia and astrocyte markers to evaluate glial activation and quantitative real-time polymerase chain reaction (PCR) to evaluate expression levels of inflammatory cytokines and brain-derived neurotrophic factor (BDNF). The hippocampal and cortical regions of the HFD group had increased expression of the activated microglial marker CD68 and the astrocytic marker glial fibrillary acidic protein. NIR-treated HFD groups showed decreased levels of these markers. PCR revealed that hippocampal tissue from the HFD group had increased levels of pro-inflammatory interleukin (IL)-1β and tumor necrosis factor-α. Interestingly, the same samples showed increased levels of the anti-inflammatory IL-10. All these changes were attenuated by NIR treatment. Lastly, hippocampal levels of the neurotrophic factor BDNF were increased in NIR-treated HFD mice, compared to untreated HFD mice. The marked reductions in glial activation and pro-inflammatory cytokines along with elevated BDNF provide insights into how NIR could reduce neuroinflammation. These results support the use of NIR as a potential non-invasive and preventive therapeutic approach against chronic obesity-induced deficits that are known to occur with AD neuropathology.
Collapse
|
423
|
Single-Cell RNA-Sequencing: Astrocyte and Microglial Heterogeneity in Health and Disease. Cells 2022; 11:cells11132021. [PMID: 35805105 PMCID: PMC9265979 DOI: 10.3390/cells11132021] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 12/13/2022] Open
Abstract
Astrocytes and microglia are non-neuronal cells that maintain homeostasis within the central nervous system via their capacity to regulate neuronal transmission and prune synapses. Both astrocytes and microglia can undergo morphological and transcriptomic changes in response to infection with human immunodeficiency virus (HIV). While both astrocytes and microglia can be infected with HIV, HIV viral proteins in the local environment can interact with and activate these cells. Given that both astrocytes and microglia play critical roles in maintaining neuronal function, it will be critical to have an understanding of their heterogeneity and to identify genes and mechanisms that modulate their responses to HIV. Heterogeneity may include a depletion or increase in one or more astrocyte or microglial subtypes in different regions of the brain or spine as well as the gain or loss of a specific function. Single-cell RNA sequencing (scRNA-seq) has emerged as a powerful tool that can be used to characterise these changes within a given population. The use of this method facilitates the identification of subtypes and changes in cellular transcriptomes that develop in response to activation and various disease processes. In this review, we will examine recent studies that have used scRNA-seq to explore astrocyte and microglial heterogeneity in neurodegenerative diseases including Alzheimer’s disease and amyotrophic lateral sclerosis as well as in response to HIV infection. A careful review of these studies will expand our current understanding of cellular heterogeneity at homeostasis and in response to specific disease states.
Collapse
|
424
|
Lin YJ, Wu CYJ, Wu JY, Lim M. The Role of Myeloid Cells in GBM Immunosuppression. Front Immunol 2022; 13:887781. [PMID: 35711434 PMCID: PMC9192945 DOI: 10.3389/fimmu.2022.887781] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/25/2022] [Indexed: 12/12/2022] Open
Abstract
Gliomas are intrinsic brain tumors that originate from glial cells. Glioblastoma (GBM) is the most aggressive glioma type and resistant to immunotherapy, mainly due to its unique immune environment. Dimensional data analysis reveals that the intra-tumoral heterogeneity of immune cell populations in the glioma microenvironment is largely made up of cells of myeloid lineage. Conventional therapies of combined surgery, chemotherapy and radiotherapy have achieved limited improvements in the prognosis of glioma patients, as myeloid cells are prominent mediators of immune and therapeutic responses—like immunotherapy resistance—in glioma. Myeloid cells are frequently seen in the tumor microenvironment (TME), and they are polarized to promote tumorigenesis and immunosuppression. Reprogramming myeloid cells has emerged as revolutionary, new types of immunotherapies for glioma treatment. Here we detail the current advances in classifying epigenetic, metabolic, and phenotypic characteristics and functions of different populations of myeloid cells in glioma TME, including myeloid-derived suppressor cells (MDSCs), glioma-associated microglia/macrophages (GAMs), glioma-associated neutrophils (GANs), and glioma-associated dendritic cells (GADCs), as well as the mechanisms underlying promotion of tumorigenesis. The final goal of this review will be to provide new insights into novel therapeutic approaches for specific targeting of myeloid cells to improve the efficacy of current treatments in glioma patients.
Collapse
Affiliation(s)
- Ya-Jui Lin
- Department of Neurosurgery, Chang Gung Medical Foundation, Linkou Medical Center, Taoyuan, Taiwan.,Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Caren Yu-Ju Wu
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States.,Department of Neurosurgery, Chang Gung Medical Foundation, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Janet Yuling Wu
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Michael Lim
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
425
|
Wicks EE, Ran KR, Kim JE, Xu R, Lee RP, Jackson CM. The Translational Potential of Microglia and Monocyte-Derived Macrophages in Ischemic Stroke. Front Immunol 2022; 13:897022. [PMID: 35795678 PMCID: PMC9251541 DOI: 10.3389/fimmu.2022.897022] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
The immune response to ischemic stroke is an area of study that is at the forefront of stroke research and presents promising new avenues for treatment development. Upon cerebral vessel occlusion, the innate immune system is activated by danger-associated molecular signals from stressed and dying neurons. Microglia, an immune cell population within the central nervous system which phagocytose cell debris and modulate the immune response via cytokine signaling, are the first cell population to become activated. Soon after, monocytes arrive from the peripheral immune system, differentiate into macrophages, and further aid in the immune response. Upon activation, both microglia and monocyte-derived macrophages are capable of polarizing into phenotypes which can either promote or attenuate the inflammatory response. Phenotypes which promote the inflammatory response are hypothesized to increase neuronal damage and impair recovery of neuronal function during the later phases of ischemic stroke. Therefore, modulating neuroimmune cells to adopt an anti-inflammatory response post ischemic stroke is an area of current research interest and potential treatment development. In this review, we outline the biology of microglia and monocyte-derived macrophages, further explain their roles in the acute, subacute, and chronic stages of ischemic stroke, and highlight current treatment development efforts which target these cells in the context of ischemic stroke.
Collapse
|
426
|
Wang H, He Y, Sun Z, Ren S, Liu M, Wang G, Yang J. Microglia in depression: an overview of microglia in the pathogenesis and treatment of depression. J Neuroinflammation 2022; 19:132. [PMID: 35668399 PMCID: PMC9168645 DOI: 10.1186/s12974-022-02492-0] [Citation(s) in RCA: 159] [Impact Index Per Article: 79.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/18/2022] [Indexed: 02/07/2023] Open
Abstract
Major depressive disorder is a highly debilitating psychiatric disorder involving the dysfunction of different cell types in the brain. Microglia are the predominant resident immune cells in the brain and exhibit a critical role in depression. Recent studies have suggested that depression can be regarded as a microglial disease. Microglia regulate inflammation, synaptic plasticity, and the formation of neural networks, all of which affect depression. In this review, we highlighted the role of microglia in the pathology of depression. First, we described microglial activation in animal models and clinically depressed patients. Second, we emphasized the possible mechanisms by which microglia recognize depression-associated stress and regulate conditions. Third, we described how antidepressants (clinical medicines and natural products) affect microglial activation. Thus, this review aimed to objectively analyze the role of microglia in depression and focus on potential antidepressants. These data suggested that regulation of microglial actions might be a novel therapeutic strategy to counteract the adverse effects of devastating mental disorders.
Collapse
Affiliation(s)
- Haixia Wang
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, 5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, 10 Xi tou tiao, You An Men Wai, Fengtai District, Beijing, 100069, China
| | - Yi He
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, 5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, 10 Xi tou tiao, You An Men Wai, Fengtai District, Beijing, 100069, China
| | - Zuoli Sun
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, 5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, 10 Xi tou tiao, You An Men Wai, Fengtai District, Beijing, 100069, China
| | - Siyu Ren
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, 5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, 10 Xi tou tiao, You An Men Wai, Fengtai District, Beijing, 100069, China
| | - Mingxia Liu
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, 5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, 10 Xi tou tiao, You An Men Wai, Fengtai District, Beijing, 100069, China
| | - Gang Wang
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, 5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088, China. .,Advanced Innovation Center for Human Brain Protection, Capital Medical University, 10 Xi tou tiao, You An Men Wai, Fengtai District, Beijing, 100069, China.
| | - Jian Yang
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, 5 Ankang Lane, Dewai Avenue, Xicheng District, Beijing, 100088, China. .,Advanced Innovation Center for Human Brain Protection, Capital Medical University, 10 Xi tou tiao, You An Men Wai, Fengtai District, Beijing, 100069, China.
| |
Collapse
|
427
|
Venugopal D, Vishwakarma S, Kaur I, Samavedi S. Electrospun fiber-based strategies for controlling early innate immune cell responses: Towards immunomodulatory mesh designs that facilitate robust tissue repair. Acta Biomater 2022; 163:228-247. [PMID: 35675893 DOI: 10.1016/j.actbio.2022.06.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 05/16/2022] [Accepted: 06/01/2022] [Indexed: 12/16/2022]
Abstract
Electrospun fibrous meshes are widely used for tissue repair due to their ability to guide a host of cell responses including phenotypic differentiation and tissue maturation. A critical factor determining the eventual biological outcomes of mesh-based regeneration strategies is the early innate immune response following implantation. The natural healing process involves a sequence of tightly regulated, temporally varying and delicately balanced pro-/anti-inflammatory events which together promote mesh integration with host tissue. Matrix designs that do not account for the immune milieu can result in dysregulation, chronic inflammation and fibrous capsule formation, thus obliterating potential therapeutic outcomes. In this review, we provide systematic insights into the effects of specific fiber/mesh properties and mechanical stimulation on the responses of early innate immune modulators viz., neutrophils, monocytes and macrophages. We identify matrix characteristics that promote anti-inflammatory immune phenotypes, and we correlate such responses with pro-regenerative in vivo outcomes. We also discuss recent advances in 3D fabrication technologies, bioactive functionalization approaches and biomimetic/bioinspired immunomodulatory mesh design strategies for tissue repair and wound healing. The mechanobiological insights and immunoregulatory strategies discussed herein can help improve the translational outcomes of fiber-based regeneration and may also be leveraged for intervention in degenerative diseases associated with dysfunctional immune responses. STATEMENT OF SIGNIFICANCE: The crucial role played by immune cells in promoting biomaterial-based tissue regeneration is being increasingly recognized. In this review focusing on the interactions of innate immune cells (primarily neutrophils, monocytes and macrophages) with electrospun fibrous meshes, we systematically elucidate the effects of the fiber microenvironment and mechanical stimulation on biological responses, and build upon these insights to inform the rational design of immunomodulatory meshes for effective tissue repair. We discuss state-of-the-art fabrication methods and mechanobiological advances that permit the orchestration of temporally controlled phenotypic switches in immune cells during different phases of healing. The design strategies discussed herein can also be leveraged to target several complex autoimmune and inflammatory diseases.
Collapse
|
428
|
Opioid receptor activation suppresses the neuroinflammatory response by promoting microglial M2 polarization. Mol Cell Neurosci 2022; 121:103744. [PMID: 35660086 DOI: 10.1016/j.mcn.2022.103744] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 05/02/2022] [Accepted: 05/29/2022] [Indexed: 11/20/2022] Open
Abstract
Activation of microglia is considered the most important component of neuroinflammation. Microglia can adopt a pro-inflammatory (M1) or anti-inflammatory (M2) phenotype. Opioid receptors (ORs) have been shown to control neurotransmission of various peptidergic neurons, but their potential role in regulating microglial function is largely unknown. Here, we aimed to investigate the effect of the OR agonists DAMGO, DADLE and U-50488 on the polarization of C8-B4 microglial cells. We observed that opioids suppressed lipopolysaccharide (LPS)-triggered M1 polarization and promoted M2 polarization. This was reflected in lower phagocytic activity, lower production of NO, lower expression of TNF-α, IL-1β, IL-6, IL-86 and IL-12 beta p40 together with higher migration rate, and increased expression of IL-4, IL-10, arginase 1 and CD 206 in microglia, compared to cells affected by LPS. We demonstrated that the effect of opioids on microglial polarization is mediated by the TREM2/NF-κB signaling pathway. These results provide new insights into the anti-inflammatory and neuroprotective effects of opioids and highlight their potential in combating neurodegenerative diseases.
Collapse
|
429
|
Tabaa MME, Aboalazm HM, Shaalan M, Khedr NF. Silymarin constrains diacetyl-prompted oxidative stress and neuroinflammation in rats: involvements of Dyn/GDNF and MAPK signaling pathway. Inflammopharmacology 2022; 30:961-980. [PMID: 35366745 PMCID: PMC9135832 DOI: 10.1007/s10787-022-00961-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/25/2022] [Indexed: 11/26/2022]
Abstract
Neuroinflammation, a major component of many CNS disorders, has been suggested to be associated with diacetyl (DA) exposure. DA is commonly used as a food flavoring additive and condiment. Lately, silymarin (Sily) has shown protective and therapeutic effects on neuronal inflammation. The study aimed to explore the role of Sily in protecting and/or treating DA-induced neuroinflammation. Neuroinflammation was induced in rats by administering DA (25 mg/kg) orally. Results revealed that Sily (50 mg/kg) obviously maintained cognitive and behavioral functions, alleviated brain antioxidant status, and inhibited microglial activation. Sily enhanced IL-10, GDNF and Dyn levels, reduced IFN-γ, TNFα, and IL-1β levels, and down-regulated the MAPK pathway. Immunohistochemical investigation of EGFR and GFAP declared that Sily could conserve neurons from inflammatory damage. However, with continuing DA exposure during Sily treatment, oxidative stress and neuroinflammation were less mitigated. These findings point to a novel mechanism involving the Dyn/GDNF and MAPK pathway through which Sily might prevent and treat DA-induced neuroinflammation.
Collapse
Affiliation(s)
- Manar Mohammed El Tabaa
- Pharmacology & Environmental Toxicology, Environmental Studies & Research Institute (ESRI), University of Sadat City, Minofia Governorate, Sadat city, Egypt
| | - Hamdi M. Aboalazm
- Biochemistry, Environmental Studies & Research Institute (ESRI), University of Sadat City, Sadat City, Egypt
| | - Mohamed Shaalan
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | | |
Collapse
|
430
|
Liu X, Vigorito M, Huang W, Khan MAS, Chang SL. The Impact of Alcohol-Induced Dysbiosis on Diseases and Disorders of the Central Nervous System. J Neuroimmune Pharmacol 2022; 17:131-151. [PMID: 34843074 DOI: 10.1007/s11481-021-10033-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/11/2021] [Indexed: 12/29/2022]
Abstract
The human digestive tract contains a diverse and abundant microbiota that is important for health. Excessive alcohol use can disrupt the balance of these microbes (known as dysbiosis), leading to elevated blood endotoxin levels and systemic inflammation. Using QIAGEN Ingenuity Pathway Analysis (IPA) bioinformatics tool, we have confirmed that peripheral endotoxin (lipopolysaccharide) mediates various cytokines to enhance the neuroinflammation signaling pathway. The literature has identified alcohol-mediated neuroinflammation as a possible risk factor for the onset and progression of neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD), and psychiatric disorders such as addiction to alcohol and other drugs. In this review, we discuss alcohol-use-induced dysbiosis in the gut and other body parts as a causal factor in the progression of Central Nervous System (CNS) diseases including neurodegenerative disease and possibly alcohol use disorder.
Collapse
Affiliation(s)
- Xiangqian Liu
- Institute of Neuroimmune Pharmacology, Seton Hall University, South Orange, NJ, 07079, USA
- Department of Histology and Embryology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, P.R. China
| | - Michael Vigorito
- Institute of Neuroimmune Pharmacology, Seton Hall University, South Orange, NJ, 07079, USA
- Department of Psychology, Seton Hall University, South Orange, NJ, 07079, USA
| | - Wenfei Huang
- Institute of Neuroimmune Pharmacology, Seton Hall University, South Orange, NJ, 07079, USA
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, 07079, USA
| | - Mohammed A S Khan
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children, Boston, MA, 02114, USA.
| | - Sulie L Chang
- Institute of Neuroimmune Pharmacology, Seton Hall University, South Orange, NJ, 07079, USA.
- Department of Biological Sciences, Seton Hall University, South Orange, NJ, 07079, USA.
| |
Collapse
|
431
|
Characterization of Ex Vivo and In Vitro Wnt Transcriptome Induced by Spinal Cord Injury in Rat Microglial Cells. Brain Sci 2022; 12:brainsci12060708. [PMID: 35741593 PMCID: PMC9221341 DOI: 10.3390/brainsci12060708] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/18/2022] [Accepted: 05/27/2022] [Indexed: 12/10/2022] Open
Abstract
It is well known that inflammation is crucial in the onset and progression of neurodegenerative diseases and traumatic central nervous system (CNS) injuries, and that microglia and monocyte-derived macrophages (MDMs) play a pivotal role in neuroinflammation. Therefore, the exploration of molecular signaling pathways that are involved in the microglia/macrophage response might help us to shed light on their eventual therapeutic modulation. Interestingly, there is growing evidence showing that the Wnt family of proteins is involved in different neuropathologies that are characterized by a dysregulated neuroinflammatory response, including spinal cord injury (SCI). Here, we aimed to validate a methodology with competence to assess the physiologically relevant Wnt expression patterns of active microglia and MDMs in a rat model of SCI. For that purpose, we have selected and adapted an in vitro system of primary microglia culture that were stimulated with a lesioned spinal cord extract (SCE), together with an ex vivo protocol of flow cytometry sorting of rat microglia/MDMs at different time-points after contusive SCI. Our study demonstrates that the expression profile of Wnt-related genes in microglia/MDM cells exhibit important differences between these particular scenarios which would be in line with previous studies where similar discrepancies have been described for other molecules. Moreover, our results provide for a first experimental report of the Wnt transcriptome in rat microglia and MDMs after SCI which, together with the research platform that was used in the study, and considering its limitations, we expect might contribute to foster the research on Wnt-driven immunomodulatory therapies.
Collapse
|
432
|
Jackson A, Engen PA, Forsyth CB, Shaikh M, Naqib A, Wilber S, Frausto DM, Raeisi S, Green SJ, Bradaric BD, Persons AL, Voigt RM, Keshavarzian A. Intestinal Barrier Dysfunction in the Absence of Systemic Inflammation Fails to Exacerbate Motor Dysfunction and Brain Pathology in a Mouse Model of Parkinson's Disease. Front Neurol 2022; 13:882628. [PMID: 35665034 PMCID: PMC9159909 DOI: 10.3389/fneur.2022.882628] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/14/2022] [Indexed: 01/01/2023] Open
Abstract
Introduction Parkinson's disease (PD) is the second most common neurodegenerative disease associated with aging. PD patients have systemic and neuroinflammation which is hypothesized to contribute to neurodegeneration. Recent studies highlight the importance of the gut-brain axis in PD pathogenesis and suggest that gut-derived inflammation can trigger and/or promote neuroinflammation and neurodegeneration in PD. However, it is not clear whether microbiota dysbiosis, intestinal barrier dysfunction, or intestinal inflammation (common features in PD patients) are primary drivers of disrupted gut-brain axis in PD that promote neuroinflammation and neurodegeneration. Objective To determine the role of microbiota dysbiosis, intestinal barrier dysfunction, and colonic inflammation in neuroinflammation and neurodegeneration in a genetic rodent model of PD [α-synuclein overexpressing (ASO) mice]. Methods To distinguish the role of intestinal barrier dysfunction separate from inflammation, low dose (1%) dextran sodium sulfate (DSS) was administered in cycles for 52 days to ASO and control mice. The outcomes assessed included intestinal barrier integrity, intestinal inflammation, stool microbiome community, systemic inflammation, motor function, microglial activation, and dopaminergic neurons. Results Low dose DSS treatment caused intestinal barrier dysfunction (sugar test, histological analysis), intestinal microbiota dysbiosis, mild intestinal inflammation (colon shortening, elevated MPO), but it did not increase systemic inflammation (serum cytokines). However, DSS did not exacerbate motor dysfunction, neuroinflammation (microglial activation), or dopaminergic neuron loss in ASO mice. Conclusion Disruption of the intestinal barrier without overt intestinal inflammation is not associated with worsening of PD-like behavior and pathology in ASO mice.
Collapse
Affiliation(s)
- Aeja Jackson
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Phillip A. Engen
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Christopher B. Forsyth
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, United States
| | - Maliha Shaikh
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Ankur Naqib
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Sherry Wilber
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Dulce M. Frausto
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Shohreh Raeisi
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Stefan J. Green
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
- Genomics and Microbiome Core Facility, Rush University Medical Center, Chicago, IL, United States
| | - Brinda Desai Bradaric
- Bachelor of Science in Health Sciences Program, College of Health Sciences, Rush University Medical Center, Chicago, IL, United States
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, United States
| | - Amanda L. Persons
- Center for Compulsive Behavior and Addiction, Rush University Medical Center, Chicago, IL, United States
- Department of Physician Assistant Studies, Rush University Medical Center, Chicago, IL, United States
| | - Robin M. Voigt
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, United States
| | - Ali Keshavarzian
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, United States
- Department of Physiology, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|
433
|
Slota JA, Medina SJ, Frost KL, Booth SA. Neurons and Astrocytes Elicit Brain Region Specific Transcriptional Responses to Prion Disease in the Murine CA1 and Thalamus. Front Neurosci 2022; 16:918811. [PMID: 35651626 PMCID: PMC9149297 DOI: 10.3389/fnins.2022.918811] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 04/29/2022] [Indexed: 01/14/2023] Open
Abstract
Progressive dysfunction and loss of neurons ultimately culminates in the symptoms and eventual fatality of prion disease, yet the pathways and mechanisms that lead to neuronal degeneration remain elusive. Here, we used RNAseq to profile transcriptional changes in microdissected CA1 and thalamus brain tissues from prion infected mice. Numerous transcripts were altered during clinical disease, whereas very few transcripts were reliably altered at pre-clinical time points. Prion altered transcripts were assigned to broadly defined brain cell types and we noted a strong transcriptional signature that was affiliated with reactive microglia and astrocytes. While very few neuronal transcripts were common between the CA1 and thalamus, we described transcriptional changes in both regions that were related to synaptic dysfunction. Using transcriptional profiling to compare how different neuronal populations respond during prion disease may help decipher mechanisms that lead to neuronal demise and should be investigated with greater detail.
Collapse
Affiliation(s)
- Jessy A. Slota
- One Health Division, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Sarah J. Medina
- One Health Division, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Kathy L. Frost
- One Health Division, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Stephanie A. Booth
- One Health Division, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
- Department of Medical Microbiology and Infectious Diseases, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
434
|
Taddei RN, Sanchez-Mico MV, Bonnar O, Connors T, Gaona A, Denbow D, Frosch MP, Gómez-Isla T. Changes in glial cell phenotypes precede overt neurofibrillary tangle formation, correlate with markers of cortical cell damage, and predict cognitive status of individuals at Braak III-IV stages. Acta Neuropathol Commun 2022; 10:72. [PMID: 35534858 PMCID: PMC9082857 DOI: 10.1186/s40478-022-01370-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 04/18/2022] [Indexed: 11/10/2022] Open
Abstract
Clinico-pathological correlation studies show that some otherwise healthy elderly individuals who never developed cognitive impairment harbor a burden of Alzheimer's disease lesions (plaques and tangles) that would be expected to result in dementia. In the absence of comorbidities explaining such discrepancies, there is a need to identify other brain changes that meaningfully contribute to the cognitive status of an individual in the face of such burdens of plaques and tangles. Glial inflammatory responses, a universal phenomenon in symptomatic AD, show robust association with degree of cognitive impairment, but their significance in early tau pathology stages and contribution to the trajectory of cognitive decline at an individual level remain widely unexplored. We studied 55 brains from individuals at intermediate stages of tau tangle pathology (Braak III-IV) with diverging antemortem cognition (demented vs. non-demented, here termed `resilient'), and age-matched cognitively normal controls (Braak 0-II). We conducted quantitative assessments of amyloid and tau lesions, cellular vulnerability markers, and glial phenotypes in temporal pole (Braak III-IV region) and visual cortex (Braak V-VI region) using artificial-intelligence based semiautomated quantifications. We found distinct glial responses with increased proinflammatory and decreased homeostatic markers, both in regions with tau tangles (temporal pole) and without overt tau deposits (visual cortex) in demented but not in resilient. These changes were significantly associated with markers of cortical cell damage. Similar phenotypic glial changes were detected in the white matter of demented but not resilient and were associated with higher burden of overlying cortical cellular damage in regions with and without tangles. Our data suggest that changes in glial phenotypes in cortical and subcortical regions represent an early phenomenon that precedes overt tau deposition and likely contributes to cell damage and loss of brain function predicting the cognitive status of individuals at intermediate stages of tau aggregate burden (Braak III-IV).
Collapse
Affiliation(s)
- Raquel N Taddei
- Department of Neurology, Massachusetts General Hospital, 15th Parkman St, Boston, MA, 02114, USA
- Massachusetts Alzheimer's Disease Research Center, Boston, MA, USA
- Department of Neurology, Dementia Research Institute, University College London, London, UK
| | - Maria V Sanchez-Mico
- Department of Neurology, Massachusetts General Hospital, 15th Parkman St, Boston, MA, 02114, USA
| | - Orla Bonnar
- Department of Neurology, Massachusetts General Hospital, 15th Parkman St, Boston, MA, 02114, USA
| | - Theresa Connors
- Massachusetts Alzheimer's Disease Research Center, Boston, MA, USA
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, MA, USA
| | - Angelica Gaona
- Massachusetts Alzheimer's Disease Research Center, Boston, MA, USA
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, MA, USA
| | - Dominique Denbow
- Department of Neurology, Massachusetts General Hospital, 15th Parkman St, Boston, MA, 02114, USA
| | - Matthew P Frosch
- Massachusetts Alzheimer's Disease Research Center, Boston, MA, USA
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, MA, USA
| | - Teresa Gómez-Isla
- Department of Neurology, Massachusetts General Hospital, 15th Parkman St, Boston, MA, 02114, USA.
- Massachusetts Alzheimer's Disease Research Center, Boston, MA, USA.
| |
Collapse
|
435
|
Zileuton, a 5-Lipoxygenase Inhibitor, Attenuates Haemolysate-Induced BV-2 Cell Activation by Suppressing the MyD88/NF-κB Pathway. Int J Mol Sci 2022; 23:ijms23094910. [PMID: 35563304 PMCID: PMC9104905 DOI: 10.3390/ijms23094910] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 02/04/2023] Open
Abstract
M1 microglia induce neuroinflammation-related neuronal death in animal models of spontaneous subarachnoid haemorrhage. Zileuton is a 5-lipoxygenase inhibitor that reduces the levels of downstream pro-inflammatory cytokines. This study aimed to investigate whether zileuton inhibits microglial activation and describe its underlying mechanisms. BV-2 cells were exposed to 1 mg/mL haemolysate for 30 min, followed by treatment with different concentrations (5, 10, 15, or 20 μM) of zileuton for 24 h. The cells were then assessed for viability, polarisation, and protein expression levels. Haemolysate increases the viability of BV-2 cells and induces M1 polarisation. Subsequent exposure to high concentrations of zileuton decreased the viability of BV-2 cells, shifted the polarisation to the M2 phenotype, suppressed the expression of 5-lipoxygenase, decreased tumour necrosis factor α levels, and increased interleukin-10 levels. Furthermore, high concentrations of zileuton suppressed the expression of myeloid differentiation primary response protein 88 and reduced the phosphorylated-nuclear factor-kappa B (NF-kB)/NF-kB ratio. Therefore, phenotype reversal from M1 to M2 is a possible mechanism by which zileuton attenuates haemolysate-induced neuroinflammation after spontaneous subarachnoid haemorrhage.
Collapse
|
436
|
Zhou J, Ni W, Ling Y, Lv X, Niu D, Zeng Y, Qiu Y, Si Y, Wang Z, Hu J. Human neural stem cells secretome inhibits lipopolysaccharide-induced neuroinflammation through modulating microglia polarization by activating PPAR-γ. Stem Cells Dev 2022; 31:369-382. [PMID: 35481777 DOI: 10.1089/scd.2022.0081] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Neuroinflammation is one of the typical events in multiple neurodegenerative diseases, whereas microglia are the critical participants in the pathogenesis of neuroinflammation. Several studies suggest that neural stem cells (NSCs) present immunomodulatory benefits due to their paracrine products, which contain mounting trophic factors. In the current study, the anti-inflammatory effects of neural stem cells secretome (NSC-S) on lipopolysaccharide (LPS)-induced neuroinflammatory models were evaluated in vivo and the underlying mechanism was further investigated in vitro. It was revealed that NSC-S significantly attenuated the severity of LPS-induced behaviour disorders and inflammatory response in mice. In vitro studies found that NSC-S significantly promoted the polarization of microglia from proinflammatory M1 to anti-inflammatory M2 phenotype, and reduced the production of proinflammatory cytokines while elevated anti-inflammatory cytokines in BV2 cells. NSC-S promoted peroxisome proliferator-activated receptor gamma (PPAR-γ) pathway activation. However, these effects of NSC-S were abrogated by PPAR-γ inhibitor GW9662. Notably, the fatty acid binding protein 5 (FABP5) in NSC-S may mediate PPAR-γ activation and inflammation remission. In summary, NSC-S promotes the regression of LPS-induced microglia-mediated inflammation through the PPAR-γ pathway. This function might be achieved via FABP5.
Collapse
Affiliation(s)
- Jiqin Zhou
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, , Jiangsu, China;
| | - Wei Ni
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, , Jiangsu, China;
| | - Yating Ling
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, , Jiangsu, China;
| | - Xiaorui Lv
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, , Jiangsu, China;
| | - Dongdong Niu
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, , Jiangsu, China;
| | - Yu Zeng
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, , Jiangsu, China;
| | - Yun Qiu
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, , Jiangsu, China;
| | - Yu Si
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, , Jiangsu, China;
| | - Ziyu Wang
- Health Clinical Laboratories, Health BioMed Co.,Ltd, Ningbo, Zhejiang, China;
| | - Jiabo Hu
- Jinagsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, 301, , Jiangsu, China, 212013;
| |
Collapse
|
437
|
Guselnikova VV, Razenkova VA, Sufieva DA, Korzhevskii DE. Microglia and putative macrophages of the subfornical organ: structural and functional features. BULLETIN OF RUSSIAN STATE MEDICAL UNIVERSITY 2022. [DOI: 10.24075/brsmu.2022.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The subfornical organ is an important regulator of water-salt metabolism and energy balance of the body, involved in the control of the cardiovascular system and immune regulation. The organ comprises several cell populations, among which microglia and macrophages remain uncharacterized. This study aimed at structural, cytochemical, and functional characterization of microglia and macrophages of the subfornical organ in rats. Brain specimens were collected from mature male Wistar rats (n = 8). Microglia and macrophages were revealed by immunostaining with poly- and monoclonal antibodies against calcium-binding protein Iba1 and lysosomal protein CD68; the slides were examined by light and confocal laser microscopy. The study provides a comprehensive morphological characterization of microglial cells and macrophages of the subfornical organ. We demonstrate that the majority of Iba1-expressing cells in this area of the brain are microglial cells, not macrophages. Pre-activated state of the subfornical organ microglia may reflect structural and functional features of this organ and specific functions of local microglia. Subependymal microglial cells, the processes of which penetrate into the third ventricle of the brain, constitute a distinct subpopulation among the Iba1-expressing cells of the subfornical organ. Apart from microglial elements, the subfornical organ contains few tissue macrophages with characteristic strong expression of CD68 accompanied by undetectable or weak expression of Iba1.
Collapse
Affiliation(s)
- VV Guselnikova
- Institute of Experimental Medicine, St Petersburg, Russia
| | - VA Razenkova
- Institute of Experimental Medicine, St Petersburg, Russia
| | - DA Sufieva
- Institute of Experimental Medicine, St Petersburg, Russia
| | - DE Korzhevskii
- Institute of Experimental Medicine, St Petersburg, Russia
| |
Collapse
|
438
|
Advances in Visualizing Microglial Cells in Human Central Nervous System Tissue. Biomolecules 2022; 12:biom12050603. [PMID: 35625531 PMCID: PMC9138569 DOI: 10.3390/biom12050603] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 02/06/2023] Open
Abstract
Neuroinflammation has recently been identified as a fundamentally important pathological process in most, if not all, CNS diseases. The main contributor to neuroinflammation is the microglia, which constitute the innate immune response system. Accurate identification of microglia and their reactivity state is therefore essential to further our understanding of CNS pathophysiology. Many staining techniques have been used to visualise microglia in rodent and human tissue, and immunostaining is currently the most frequently used. Historically, identification of microglia was predominantly based on morphological structure, however, recently there has been a reliance on selective antigen expression, and microglia-specific markers have been identified providing increased certainty that the cells observed are in fact microglia, rather than the similar yet distinct macrophages. To date, the most microglia-specific markers are P2Y12 and TMEM119. However, other microglia-related markers can also be useful for demonstrating activation state, phagocytic state, and for neuroimaging purposes in longitudinal studies. Overall, it is important to be aware of the microglia-selectivity issues of the various stains and immunomarkers used by researchers to distinguish microglia in CNS tissue to avoid misinterpretation.
Collapse
|
439
|
Scipioni L, Ciaramellano F, Carnicelli V, Leuti A, Lizzi AR, De Dominicis N, Oddi S, Maccarrone M. Microglial Endocannabinoid Signalling in AD. Cells 2022; 11:1237. [PMID: 35406803 PMCID: PMC8997504 DOI: 10.3390/cells11071237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 03/30/2022] [Accepted: 04/04/2022] [Indexed: 02/01/2023] Open
Abstract
Chronic inflammation in Alzheimer's disease (AD) has been recently identified as a major contributor to disease pathogenesis. Once activated, microglial cells, which are brain-resident immune cells, exert several key actions, including phagocytosis, chemotaxis, and the release of pro- or anti-inflammatory mediators, which could have opposite effects on brain homeostasis, depending on the stage of disease and the particular phenotype of microglial cells. The endocannabinoids (eCBs) are pleiotropic bioactive lipids increasingly recognized for their essential roles in regulating microglial activity both under normal and AD-driven pathological conditions. Here, we review the current literature regarding the involvement of this signalling system in modulating microglial phenotypes and activity in the context of homeostasis and AD-related neurodegeneration.
Collapse
Affiliation(s)
- Lucia Scipioni
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, Via Vetoio Snc, 67100 L’Aquila, Italy; (L.S.); (V.C.); (A.R.L.); (N.D.D.)
- European Center for Brain Research/IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy; (F.C.); (A.L.)
| | - Francesca Ciaramellano
- European Center for Brain Research/IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy; (F.C.); (A.L.)
- Faculty of Veterinary Medicine, University of Teramo, Via R. Balzarini 1, 64100 Teramo, Italy
| | - Veronica Carnicelli
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, Via Vetoio Snc, 67100 L’Aquila, Italy; (L.S.); (V.C.); (A.R.L.); (N.D.D.)
| | - Alessandro Leuti
- European Center for Brain Research/IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy; (F.C.); (A.L.)
- Department of Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Anna Rita Lizzi
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, Via Vetoio Snc, 67100 L’Aquila, Italy; (L.S.); (V.C.); (A.R.L.); (N.D.D.)
| | - Noemi De Dominicis
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, Via Vetoio Snc, 67100 L’Aquila, Italy; (L.S.); (V.C.); (A.R.L.); (N.D.D.)
- Department of Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy
| | - Sergio Oddi
- European Center for Brain Research/IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy; (F.C.); (A.L.)
- Faculty of Veterinary Medicine, University of Teramo, Via R. Balzarini 1, 64100 Teramo, Italy
| | - Mauro Maccarrone
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, Via Vetoio Snc, 67100 L’Aquila, Italy; (L.S.); (V.C.); (A.R.L.); (N.D.D.)
- European Center for Brain Research/IRCCS Santa Lucia Foundation, Via del Fosso di Fiorano 64, 00143 Rome, Italy; (F.C.); (A.L.)
| |
Collapse
|
440
|
Ton ST, Laghi JR, Tsai SY, Blackwell AA, Adamczyk NS, Oltmanns JRO, Britten RA, Wallace DG, Kartje GL. Exposure to 5 cGy 28Si Particles Induces Long-Term Microglial Activation in the Striatum and Subventricular Zone and Concomitant Neurogenic Suppression. Radiat Res 2022; 198:28-39. [DOI: 10.1667/rade-21-00021.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/17/2022] [Indexed: 11/03/2022]
Abstract
The proposed mission to Mars will expose astronauts to space radiation that is known to adversely affect cognition and tasks that rely on fine sensorimotor function. Space radiation has also been shown to affect the microglial and neurogenic responses in the center nervous system (CNS). We recently reported that a low dose of 5 cGy 600 MeV/n 28Si results in impaired cognition and skilled motor behavior in adult rats. Since these tasks rely at least in part on the proper functioning of the striatum, we examined striatal microglial cells in these same subjects. Using morphometric analysis, we found that 28Si exposure increased activated microglial cells in the striatum. The majority of these striatal Iba1+ microglia were ED1–, indicating that they were in an alternatively activated state, where microglia do not have phagocytic activity but may be releasing cytokines that could negatively impact neuronal function. In the other areas studied, Iba1+ microglial cells were increased in the subventricular zone (SVZ), but not in the dentate gyrus (DG). Additionally, we examined the relationship between the microglial response and neurogenesis. An analysis of new neurons in the DG revealed an increase in doublecortin-positive (DCX+) hilar ectopic granule cells (hEGC) which correlated with Iba1+ cells, suggesting that microglial cells contributed to this aberrant distribution which may adversely affect hippocampal function. Taken together, these results indicate that a single dose of 28Si radiation results in persistent cellular effects in the CNS that may impact astronauts both in the short and long-term following deep space missions.
Collapse
Affiliation(s)
- Son T. Ton
- Research Service, Edward Hines Jr. VA Hospital, Hines, Illinois
| | - Julia R. Laghi
- Research Service, Edward Hines Jr. VA Hospital, Hines, Illinois
| | - Shih-Yen Tsai
- Research Service, Edward Hines Jr. VA Hospital, Hines, Illinois
| | | | | | | | - Richard A. Britten
- Department of Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia
| | - Douglas G. Wallace
- Department of Psychology, Northern Illinois University, DeKalb, Illinois
| | - Gwendolyn L. Kartje
- Research Service, Edward Hines Jr. VA Hospital, Hines, Illinois
- Department of Molecular Pharmacology and Neuroscience, Loyola University Chicago Health Sciences Division, Maywood, Illinois
| |
Collapse
|
441
|
Takahashi K, Nelvagal HR, Lange J, Cooper JD. Glial Dysfunction and Its Contribution to the Pathogenesis of the Neuronal Ceroid Lipofuscinoses. Front Neurol 2022; 13:886567. [PMID: 35444603 PMCID: PMC9013902 DOI: 10.3389/fneur.2022.886567] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/16/2022] [Indexed: 01/05/2023] Open
Abstract
While significant efforts have been made in developing pre-clinical treatments for the neuronal ceroid lipofuscinoses (NCLs), many challenges still remain to bring children with NCLs a cure. Devising effective therapeutic strategies for the NCLs will require a better understanding of pathophysiology, but little is known about the mechanisms by which loss of lysosomal proteins causes such devastating neurodegeneration. Research into glial cells including astrocytes, microglia, and oligodendrocytes have revealed many of their critical functions in brain homeostasis and potential contributions to neurodegenerative diseases. Genetically modified mouse models have served as a useful platform to define the disease progression in the central nervous system across NCL subtypes, revealing a wide range of glial responses to disease. The emerging evidence of glial dysfunction questions the traditional “neuron-centric” view of NCLs, and would suggest that directly targeting glia in addition to neurons could lead to better therapeutic outcomes. This review summarizes the most up-to-date understanding of glial pathologies and their contribution to the pathogenesis of NCLs, and highlights some of the associated challenges that require further research.
Collapse
Affiliation(s)
- Keigo Takahashi
- Pediatric Storage Disorders Laboratory, Department of Pediatrics, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
| | - Hemanth R. Nelvagal
- Department of Pharmacology, School of Pharmacy, University College London, London, United Kingdom
| | - Jenny Lange
- Zayed Centre for Research into Rare Disease in Children, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Jonathan D. Cooper
- Pediatric Storage Disorders Laboratory, Department of Pediatrics, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Genetics, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- Department of Neurology, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States
- *Correspondence: Jonathan D. Cooper
| |
Collapse
|
442
|
LONG-TERM EFFECTS OF SHAM SURGERY ON PHAGOCYTE FUNCTIONS IN RATS. BIOTECHNOLOGIA ACTA 2022. [DOI: 10.15407/biotech15.02.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Animal models of inflammatory disorders, including those of the nervous system are commonly used to explore the pathophysiological role of immune cell response in disease triggering and course and to develop biotechnology products for therapeutic use. Modeling some of these disorders, particularly neurodegenerative diseases, implies surgical manipulations for the intracerebral introduction of disease-initiating substances (toxins, amyloids etc.). Design of these experiments involves the use of sham-operated animals as a control of non-specific intrinsic side-effects elicited by surgical manipulations per se, including local and systemic inflammation, where phagocytic cells are key participants. Short-term post-surgical immunomodulatory effects are widely reported. However, no study thus far has examined the long term effects of sham-surgery on phagocyte functions. The purpose of this study was to evaluate the effect of sham-surgery, commonly used for modeling neurodegenerative diseases, on phagocyte functions in the far terms after the surgical manipulations. Materials and Methods. Adult male Wistar rats were used in the study. Sham surgery consisted of stereotactic unilateral injection of saline solution into the median forebrain bundle (sham-operated 1, SO1) or directly into the substantia nigra (sham-operated 2, SO2). Before the placebo surgery, animals were anaesthetized using nembutal and ketamine/xylazine correspondingly. Functional characteristics (phagocytic activity, oxidative metabolism, CD80/86 and CD206 expression) of phagocytes (microglia, peritoneal macrophages, circulating monocytes and granulocytes) were examined by flow cytometry. Differential leukocyte count was conducted using hematological analyzer. Results. Phagocytes from animals underwent of different protocols of placebo surgery, demonstrated various patterns of functional changes on day 29 after the manipulations. In animals from SO1 group, we observed signs of residual neuroinflammation (pro-inflammatory shift of microglia functional profile) along with ongoing resolution of systemic inflammation (anti-inflammatory metabolic shift of circulating phagocytes and peritoneal macrophages). In rats from SO2 group, pro-inflammatory polarized activation of peritoneal phagocytes was registered along with anti-inflammatory shift in microglia and circulating phagocytes. Conclusions. Sham surgery influences functions of phagocytic cells of different locations even in the far terms after the manipulations. These effects can be considered as combined long-term consequences of surgical brain injury and the use of anesthetics. Our observations evidences, that sham associated non-specific immunomodulatory effects should always be taken into consideration in animal models of inflammatory central nervous system diseases.
Collapse
|
443
|
Wolf J, Boneva S, Rosmus DD, Agostini H, Schlunck G, Wieghofer P, Schlecht A, Lange C. In-Depth Molecular Profiling Specifies Human Retinal Microglia Identity. Front Immunol 2022; 13:863158. [PMID: 35371110 PMCID: PMC8971200 DOI: 10.3389/fimmu.2022.863158] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 02/22/2022] [Indexed: 12/20/2022] Open
Abstract
Microglia are the tissue-resident macrophages of the retina and brain, being critically involved in organ development, tissue homeostasis, and response to cellular damage. Until now, little is known about the molecular signature of human retinal microglia and how it differs from the one of brain microglia and peripheral monocytes. In addition, it is not yet clear to what extent murine retinal microglia resemble those of humans, which represents an important prerequisite for translational research. The present study applies fluorescence-activated cell sorting to isolate human retinal microglia from enucleated eyes and compares their transcriptional profile with the one of whole retinal tissue, human brain microglia as well as classical, intermediate and non-classical monocytes. Finally, human retinal microglia are compared to murine retinal microglia, isolated from Cx3cr1GFP/+ mice. Whereas human retinal microglia exhibited a high grade of similarity in comparison to their counterparts in the brain, several enriched genes were identified in retinal microglia when compared to whole retinal tissue, as well as classical, intermediate, and non-classical monocytes. In relation to whole retina sequencing, several risk genes associated with age-related macular degeneration (AMD) and diabetic retinopathy (DR) were preferentially expressed in retinal microglia, indicating their potential pathophysiological involvement. Although a high degree of similarity was observed between human and murine retinal microglia, several species-specific genes were identified, which should be kept in mind when employing mouse models to investigate retinal microglia biology. In summary, this study provides detailed insights into the molecular profile of human retinal microglia, identifies a plethora of tissue-specific and species-specific genes in comparison to human brain microglia and murine retinal microglia, and thus highlights the significance of retinal microglia in human retinal diseases and for translational research approaches.
Collapse
Affiliation(s)
- Julian Wolf
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefaniya Boneva
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Hansjürgen Agostini
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Günther Schlunck
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter Wieghofer
- Institute of Anatomy, Leipzig University, Leipzig, Germany
- Cellular Neuroanatomy, Institute of Theoretical Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Anja Schlecht
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Wuerzburg, Wuerzburg, Germany
| | - Clemens Lange
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Ophtha-Lab, Department of Ophthalmology at St. Franziskus Hospital, Muenster, Germany
- *Correspondence: Clemens Lange,
| |
Collapse
|
444
|
Markert F, Storch A. Hyperoxygenation During Mid-Neurogenesis Accelerates Cortical Development in the Fetal Mouse Brain. Front Cell Dev Biol 2022; 10:732682. [PMID: 35372333 PMCID: PMC8969024 DOI: 10.3389/fcell.2022.732682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 01/20/2022] [Indexed: 11/13/2022] Open
Abstract
Oxygen tension is well-known to affect cortical development. Fetal brain hyperoxygenation during mid-neurogenesis in mice (embryonic stage E14.5. to E16.5) increases brain size evoked through an increase of neuroprecursor cells. Nevertheless, it is unknown whether these effects can lead to persistent morphological changes within the highly orchestrated brain development. To shed light on this, we used our model of controlled fetal brain hyperoxygenation in time-pregnant C57BL/6J mice housed in a chamber with 75% atmospheric oxygen from E14.5 to E16.5 and analyzed the brains from E14.5, E16.5, P0.5, and P3.5 mouse embryos and pups via immunofluorescence staining. Mid-neurogenesis hyperoxygenation led to an acceleration of cortical development by temporal expansion of the cortical plate with increased NeuN+ neuron counts in hyperoxic brains only until birth. More specifically, the number of Ctip2+ cortical layer 5 (L5) neurons was increased at E16.5 and at birth in hyperoxic brains but normalized in the early postnatal stage (P3.5). The absence of cleaved caspase 3 within the extended Ctip2+ L5 cell population largely excluded apoptosis as a major compensatory mechanism. Timed BrdU/EdU analyses likewise rule out a feedback mechanism. The normalization was, on the contrary, accompanied by an increase of active microglia within L5 targeting Ctip2+ neurons without any signs of apoptosis. Together, hyperoxygenation during mid-neurogenesis phase of fetal brain development provoked a specific transient overshoot of cortical L5 neurons leading to an accelerated cortical development without detectable persistent changes. These observations provide insight into cortical and L5 brain development.
Collapse
Affiliation(s)
- Franz Markert
- Department of Neurology, University of Rostock, Rostock, Germany
| | - Alexander Storch
- Department of Neurology, University of Rostock, Rostock, Germany
- German Center for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Rostock, Germany
- *Correspondence: Alexander Storch,
| |
Collapse
|
445
|
Abstract
Chandelier cells (ChCs) are a unique type of GABAergic interneuron that form axo-axonic synapses exclusively on the axon initial segment (AIS) of neocortical pyramidal neurons (PyNs), allowing them to exert powerful yet precise control over PyN firing and population output. The importance of proper ChC function is further underscored by the association of ChC connectivity defects with various neurological conditions. Despite this, the cellular mechanisms governing ChC axo-axonic synapse formation remain poorly understood. Here, we identify microglia as key regulators of ChC axonal morphogenesis and AIS synaptogenesis, and show that disease-induced aberrant microglial activation perturbs proper ChC synaptic development/connectivity in the neocortex. In doing so, such findings highlight the therapeutic potential of manipulating microglia to ensure proper brain wiring. Microglia have emerged as critical regulators of synapse development and circuit formation in the healthy brain. To date, examination of microglia in such processes has largely been focused on excitatory synapses. Their roles, however, in the modulation of GABAergic interneuron synapses—particularly those targeting the axon initial segment (AIS)—during development remain enigmatic. Here, we identify a synaptogenic/growth-promoting role for microglia in regulating pyramidal neuron (PyN) AIS synapse formation by chandelier cells (ChCs), a unique interneuron subtype whose axonal terminals, called cartridges, selectively target the AIS. We show that a subset of microglia contacts PyN AISs and ChC cartridges and that such tripartite interactions, which rely on the unique AIS cytoskeleton and microglial GABAB1 receptors, are associated with increased ChC cartridge length and bouton number and AIS synaptogenesis. Conversely, microglia depletion or disease-induced aberrant microglia activation impairs the proper development and maintenance of ChC cartridges and boutons, as well as AIS synaptogenesis. These findings unveil key roles for homeostatic, AIS-associated microglia in regulating proper ChC axonal morphogenesis and synaptic connectivity in the neocortex.
Collapse
|
446
|
Abstract
Macrophages are cells of the innate immune system and represent an important component of the first-line defense against pathogens and tumor cells. Here, their diverse functions in inflammation and tumor defense are described, and the mechanisms, tools, and activation pathways and states applied are presented. The main focus is on the role and origin of reactive oxygen species (ROS), the important signal pathways TLR/NF-κB, and the M1/M2 polarization of macrophages.
Collapse
Affiliation(s)
- Uwe Lendeckel
- Institut für Medizinische Biochemie und Molekularbiologie, Universitätsmedizin Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| | - Simone Venz
- Institut für Medizinische Biochemie und Molekularbiologie, Universitätsmedizin Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| | - Carmen Wolke
- Institut für Medizinische Biochemie und Molekularbiologie, Universitätsmedizin Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| |
Collapse
|
447
|
Beckman D, Seelke AMH, Bennett J, Dougherty P, Van Rompay KKA, Keesler R, Pesavento PA, Coffey LLA, Morrison JH, Bliss-Moreau E. Neuroanatomical abnormalities in a nonhuman primate model of congenital Zika virus infection. eLife 2022; 11:e64734. [PMID: 35261339 PMCID: PMC8906804 DOI: 10.7554/elife.64734] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 12/14/2021] [Indexed: 11/18/2022] Open
Abstract
We evaluated neuropathological consequences of fetal ZIKV exposure in rhesus monkeys, a translatable animal model for human neural development, by carrying out quantitative neuroanatomical analyses of the nearly full-term brains of fetuses infected with ZIKV and procedure-matched controls. For each animal, a complete cerebral hemisphere was evaluated using immunohistochemical (IHC) and neuroanatomical techniques to detect virus, identify affected cell types, and evaluate gross neuroanatomical abnormalities. IHC staining revealed the presence of ZIKV in the frontal lobe, which contained activated microglia and showed increased apoptosis of immature neurons. ZIKV-infected animals exhibited macrostructural changes within the visual pathway. Regional differences tracked with the developmental timing of the brain, suggesting inflammatory processes related to viral infiltration swept through the cortex, followed by a wave of cell death resulting in morphological changes. These findings may help explain why some infants born with normal sized heads during the ZIKV epidemic manifest developmental challenges as they age.
Collapse
Affiliation(s)
- Danielle Beckman
- California National Primate Research Center, UC DavisDavisUnited States
| | - Adele MH Seelke
- California National Primate Research Center, UC DavisDavisUnited States
- Department of Psychology, UC DavisDavisUnited States
| | - Jeffrey Bennett
- California National Primate Research Center, UC DavisDavisUnited States
- Department of Psychology, UC DavisDavisUnited States
| | - Paige Dougherty
- California National Primate Research Center, UC DavisDavisUnited States
- Department of Psychology, UC DavisDavisUnited States
| | - Koen KA Van Rompay
- California National Primate Research Center, UC DavisDavisUnited States
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, UC DavisDavisUnited States
| | - Rebekah Keesler
- California National Primate Research Center, UC DavisDavisUnited States
| | - Patricia A Pesavento
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, UC DavisDavisUnited States
| | - Lark LA Coffey
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, UC DavisDavisUnited States
| | - John H Morrison
- California National Primate Research Center, UC DavisDavisUnited States
- Department of Neurology, School of Medicine, UC DavisDavisUnited States
| | - Eliza Bliss-Moreau
- California National Primate Research Center, UC DavisDavisUnited States
- Department of Psychology, UC DavisDavisUnited States
| |
Collapse
|
448
|
Nociceptor-derived Reg3γ prevents endotoxic death by targeting kynurenine pathway in microglia. Cell Rep 2022; 38:110462. [PMID: 35263589 DOI: 10.1016/j.celrep.2022.110462] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 01/11/2022] [Accepted: 02/09/2022] [Indexed: 11/21/2022] Open
Abstract
Nociceptors can fine-tune local or systemic immunity, but the mechanisms of nociceptive modulation in endotoxic death remain largely unknown. Here, we identified C-type lectin Reg3γ as a nociceptor-enriched hormone that protects the host from endotoxic death. During endotoxemia, nociceptor-derived Reg3γ penetrates the brain and suppresses the expression of microglial indoleamine dioxygenase 1, a critical enzyme of the kynurenine pathway, via the Extl3-Bcl10 axis. Endotoxin-administered nociceptor-null mice and nociceptor-specific Reg3γ-deficient mice exhibit a high mortality rate accompanied by decreased brain HK1 phosphorylation and ATP production despite normal peripheral inflammation. Such metabolic arrest is only observed in the brain, and aberrant production of brain quinolinic acid, a neurotoxic metabolite of the kynurenine pathway, causes HK1 suppression. Strikingly, the central administration of Reg3γ protects mice from endotoxic death by enhancing brain ATP production. By identifying nociceptor-derived Reg3γ as a microglia-targeted hormone, this study provides insights into the understanding of tolerance to endotoxic death.
Collapse
|
449
|
Tumor-Associated Macrophages in Gliomas—Basic Insights and Treatment Opportunities. Cancers (Basel) 2022; 14:cancers14051319. [PMID: 35267626 PMCID: PMC8909866 DOI: 10.3390/cancers14051319] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Macrophages are a specialized immune cell type found in both invertebrates and vertebrates. Versatile in functionality, macrophages carry out important tasks such as cleaning cellular debris in healthy tissues and mounting immune responses during infection. In many cancer types, macrophages make up a significant portion of tumor tissue, and these are aptly called tumor-associated macrophages. In gliomas, a group of primary brain tumors, these macrophages are found in very high frequency. Tumor-associated macrophages can promote glioma development and influence the outcome of various therapeutic regimens. At the same time, these cells provide various potential points of intervention for therapeutic approaches in glioma patients. The significance of tumor-associated macrophages in the glioma microenvironment and potential therapeutic targets are the focus of this review. Abstract Glioma refers to a group of primary brain tumors which includes glioblastoma (GBM), astrocytoma and oligodendroglioma as major entities. Among these, GBM is the most frequent and most malignant one. The highly infiltrative nature of gliomas, and their intrinsic intra- and intertumoral heterogeneity, pose challenges towards developing effective treatments. The glioma microenvironment, in addition, is also thought to play a critical role during tumor development and treatment course. Unlike most other solid tumors, the glioma microenvironment is dominated by macrophages and microglia—collectively known as tumor-associated macrophages (TAMs). TAMs, like their homeostatic counterparts, are plastic in nature and can polarize to either pro-inflammatory or immunosuppressive states. Many lines of evidence suggest that immunosuppressive TAMs dominate the glioma microenvironment, which fosters tumor development, contributes to tumor aggressiveness and recurrence and, very importantly, impedes the therapeutic effect of various treatment regimens. However, through the development of new therapeutic strategies, TAMs can potentially be shifted towards a proinflammatory state which is of great therapeutic interest. In this review, we will discuss various aspects of TAMs in the context of glioma. The focus will be on the basic biology of TAMs in the central nervous system (CNS), potential biomarkers, critical evaluation of model systems for studying TAMs and finally, special attention will be given to the potential targeted therapeutic options that involve the TAM compartment in gliomas.
Collapse
|
450
|
Au NPB, Ma CHE. Neuroinflammation, Microglia and Implications for Retinal Ganglion Cell Survival and Axon Regeneration in Traumatic Optic Neuropathy. Front Immunol 2022; 13:860070. [PMID: 35309305 PMCID: PMC8931466 DOI: 10.3389/fimmu.2022.860070] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 02/14/2022] [Indexed: 12/12/2022] Open
Abstract
Traumatic optic neuropathy (TON) refers to a pathological condition caused by a direct or indirect insult to the optic nerves, which often leads to a partial or permanent vision deficit due to the massive loss of retinal ganglion cells (RGCs) and their axonal fibers. Retinal microglia are immune-competent cells residing in the retina. In rodent models of optic nerve crush (ONC) injury, resident retinal microglia gradually become activated, form end-to-end alignments in the vicinity of degenerating RGC axons, and actively internalized them. Some activated microglia adopt an amoeboid morphology that engulf dying RGCs after ONC. In the injured optic nerve, the activated microglia contribute to the myelin debris clearance at the lesion site. However, phagocytic capacity of resident retinal microglia is extremely poor and therefore the clearance of cellular and myelin debris is largely ineffective. The presence of growth-inhibitory myelin debris and glial scar formed by reactive astrocytes inhibit the regeneration of RGC axons, which accounts for the poor visual function recovery in patients with TON. In this Review, we summarize the current understanding of resident retinal microglia in RGC survival and axon regeneration after ONC. Resident retinal microglia play a key role in facilitating Wallerian degeneration and the subsequent axon regeneration after ONC. However, they are also responsible for producing pro-inflammatory cytokines, chemokines, and reactive oxygen species that possess neurotoxic effects on RGCs. Intraocular inflammation triggers a massive influx of blood-borne myeloid cells which produce oncomodulin to promote RGC survival and axon regeneration. However, intraocular inflammation induces chronic neuroinflammation which exacerbates secondary tissue damages and limits visual function recovery after ONC. Activated retinal microglia is required for the proliferation of oligodendrocyte precursor cells (OPCs); however, sustained activation of retinal microglia suppress the differentiation of OPCs into mature oligodendrocytes for remyelination after injury. Collectively, controlled activation of retinal microglia and infiltrating myeloid cells facilitate axon regeneration and nerve repair. Recent advance in single-cell RNA-sequencing and identification of microglia-specific markers could improve our understanding on microglial biology and to facilitate the development of novel therapeutic strategies aiming to switch resident retinal microglia’s phenotype to foster neuroprotection.
Collapse
Affiliation(s)
- Ngan Pan Bennett Au
- Department of Neuroscience, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Chi Him Eddie Ma
- Department of Neuroscience, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- *Correspondence: Chi Him Eddie Ma,
| |
Collapse
|