1
|
Norte-Muñoz M, Portela-Lomba M, Sobrado-Calvo P, Simón D, Di Pierdomenico J, Gallego-Ortega A, Pérez M, Cabrera-Maqueda JM, Sierra J, Vidal-Sanz M, Moreno-Flores MT, Agudo-Barriuso M. Differential response of injured and healthy retinas to syngeneic and allogeneic transplantation of a clonal cell line of immortalized olfactory ensheathing glia: a double-edged sword. Neural Regen Res 2025; 20:2395-2407. [PMID: 39359096 DOI: 10.4103/nrr.nrr-d-23-01631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 04/19/2024] [Indexed: 10/04/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202508000-00029/figure1/v/2024-09-30T120553Z/r/image-tiff Olfactory ensheathing glia promote axonal regeneration in the mammalian central nervous system, including retinal ganglion cell axonal growth through the injured optic nerve. Still, it is unknown whether olfactory ensheathing glia also have neuroprotective properties. Olfactory ensheathing glia express brain-derived neurotrophic factor, one of the best neuroprotectants for axotomized retinal ganglion cells. Therefore, we aimed to investigate the neuroprotective capacity of olfactory ensheating glia after optic nerve crush. Olfactory ensheathing glia cells from an established rat immortalized clonal cell line, TEG3, were intravitreally injected in intact and axotomized retinas in syngeneic and allogeneic mode with or without microglial inhibition or immunosuppressive treatments. Anatomical and gene expression analyses were performed. Olfactory bulb-derived primary olfactory ensheathing glia and TEG3 express major histocompatibility complex class II molecules. Allogeneically and syngenically transplanted TEG3 cells survived in the vitreous for up to 21 days, forming an epimembrane. In axotomized retinas, only the allogeneic TEG3 transplant rescued retinal ganglion cells at 7 days but not at 21 days. In these retinas, microglial anatomical activation was higher than after optic nerve crush alone. In intact retinas, both transplants activated microglial cells and caused retinal ganglion cell death at 21 days, a loss that was higher after allotransplantation, triggered by pyroptosis and partially rescued by microglial inhibition or immunosuppression. However, neuroprotection of axotomized retinal ganglion cells did not improve with these treatments. The different neuroprotective properties, different toxic effects, and different responses to microglial inhibitory treatments of olfactory ensheathing glia in the retina depending on the type of transplant highlight the importance of thorough preclinical studies to explore these variables.
Collapse
Affiliation(s)
- María Norte-Muñoz
- Grupo de Investigación Oftalmología Experimental, Departamento de Oftalmología, Optometría, Otorrinolaringología y Anatomía Patológica, Facultad de Medicina, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Campus de Ciencias de la Salud, Murcia, Spain
| | - María Portela-Lomba
- Experimental Sciences Faculty, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | - Paloma Sobrado-Calvo
- Grupo de Investigación Oftalmología Experimental, Departamento de Oftalmología, Optometría, Otorrinolaringología y Anatomía Patológica, Facultad de Medicina, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Campus de Ciencias de la Salud, Murcia, Spain
| | - Diana Simón
- Experimental Sciences Faculty, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | - Johnny Di Pierdomenico
- Grupo de Investigación Oftalmología Experimental, Departamento de Oftalmología, Optometría, Otorrinolaringología y Anatomía Patológica, Facultad de Medicina, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Campus de Ciencias de la Salud, Murcia, Spain
| | - Alejandro Gallego-Ortega
- Grupo de Investigación Oftalmología Experimental, Departamento de Oftalmología, Optometría, Otorrinolaringología y Anatomía Patológica, Facultad de Medicina, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Campus de Ciencias de la Salud, Murcia, Spain
| | - Mar Pérez
- Anatomy, Histology and Neuroscience Department, Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
| | - José M Cabrera-Maqueda
- Grupo de Investigación Oftalmología Experimental, Departamento de Oftalmología, Optometría, Otorrinolaringología y Anatomía Patológica, Facultad de Medicina, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Campus de Ciencias de la Salud, Murcia, Spain
- Center of Neuroimmunology, Service of Neurology, Laboratory of Advanced Imaging in Neuroimmunological Diseases, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), and Universitat de Barcelona, Barcelona, Spain
| | - Javier Sierra
- Medicine Faculty, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | - Manuel Vidal-Sanz
- Grupo de Investigación Oftalmología Experimental, Departamento de Oftalmología, Optometría, Otorrinolaringología y Anatomía Patológica, Facultad de Medicina, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Campus de Ciencias de la Salud, Murcia, Spain
| | - María Teresa Moreno-Flores
- Anatomy, Histology and Neuroscience Department, Medicine Faculty, Universidad Autónoma de Madrid, Madrid, Spain
| | - Marta Agudo-Barriuso
- Grupo de Investigación Oftalmología Experimental, Departamento de Oftalmología, Optometría, Otorrinolaringología y Anatomía Patológica, Facultad de Medicina, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Campus de Ciencias de la Salud, Murcia, Spain
| |
Collapse
|
2
|
Brockie S, Zhou C, Fehlings MG. Resident immune responses to spinal cord injury: role of astrocytes and microglia. Neural Regen Res 2024; 19:1678-1685. [PMID: 38103231 PMCID: PMC10960308 DOI: 10.4103/1673-5374.389630] [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: 05/04/2023] [Revised: 09/08/2023] [Accepted: 10/18/2023] [Indexed: 12/18/2023] Open
Abstract
Spinal cord injury can be traumatic or non-traumatic in origin, with the latter rising in incidence and prevalence with the aging demographics of our society. Moreover, as the global population ages, individuals with co-existent degenerative spinal pathology comprise a growing number of traumatic spinal cord injury cases, especially involving the cervical spinal cord. This makes recovery and treatment approaches particularly challenging as age and comorbidities may limit regenerative capacity. For these reasons, it is critical to better understand the complex milieu of spinal cord injury lesion pathobiology and the ensuing inflammatory response. This review discusses microglia-specific purinergic and cytokine signaling pathways, as well as microglial modulation of synaptic stability and plasticity after injury. Further, we evaluate the role of astrocytes in neurotransmission and calcium signaling, as well as their border-forming response to neural lesions. Both the inflammatory and reparative roles of these cells have eluded our complete understanding and remain key therapeutic targets due to their extensive structural and functional roles in the nervous system. Recent advances have shed light on the roles of glia in neurotransmission and reparative injury responses that will change how interventions are directed. Understanding key processes and existing knowledge gaps will allow future research to effectively target these cells and harness their regenerative potential.
Collapse
Affiliation(s)
- Sydney Brockie
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Cindy Zhou
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Michael G. Fehlings
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery and Spine Program, Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
3
|
Church KA, Cardona AE, Hopp SC. Roles in Innate Immunity. ADVANCES IN NEUROBIOLOGY 2024; 37:263-286. [PMID: 39207697 DOI: 10.1007/978-3-031-55529-9_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia are best known as the resident phagocytes of the central nervous system (CNS). As a resident brain immune cell population, microglia play key roles during the initiation, propagation, and resolution of inflammation. The discovery of resident adaptive immune cells in the CNS has unveiled a relationship between microglia and adaptive immune cells for CNS immune-surveillance during health and disease. The interaction of microglia with elements of the peripheral immune system and other CNS resident cells mediates a fine balance between neuroprotection and tissue damage. In this chapter, we highlight the innate immune properties of microglia, with a focus on how pattern recognition receptors, inflammatory signaling cascades, phagocytosis, and the interaction between microglia and adaptive immune cells regulate events that initiate an inflammatory or neuroprotective response within the CNS that modulates immune-mediated disease exacerbation or resolution.
Collapse
Affiliation(s)
- Kaira A Church
- Department of Molecular Microbiology & Immunology, The University of Texas at San Antonio, San Antonio, TX, USA
- South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Astrid E Cardona
- Department of Molecular Microbiology & Immunology, The University of Texas at San Antonio, San Antonio, TX, USA
- South Texas Center for Emerging Infectious Diseases, The University of Texas at San Antonio, San Antonio, TX, USA
| | - Sarah C Hopp
- Department of Pharmacology, Biggs Institute for Alzheimer's and Neurodegenerative Disease, The University of Texas Health Science Center San Antonio, San Antonio, TX, USA.
| |
Collapse
|
4
|
Poppell M, Hammel G, Ren Y. Immune Regulatory Functions of Macrophages and Microglia in Central Nervous System Diseases. Int J Mol Sci 2023; 24:5925. [PMID: 36982999 PMCID: PMC10059890 DOI: 10.3390/ijms24065925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/08/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
Macrophages can be characterized as a very multifunctional cell type with a spectrum of phenotypes and functions being observed spatially and temporally in various disease states. Ample studies have now demonstrated a possible causal link between macrophage activation and the development of autoimmune disorders. How these cells may be contributing to the adaptive immune response and potentially perpetuating the progression of neurodegenerative diseases and neural injuries is not fully understood. Within this review, we hope to illustrate the role that macrophages and microglia play as initiators of adaptive immune response in various CNS diseases by offering evidence of: (1) the types of immune responses and the processes of antigen presentation in each disease, (2) receptors involved in macrophage/microglial phagocytosis of disease-related cell debris or molecules, and, finally, (3) the implications of macrophages/microglia on the pathogenesis of the diseases.
Collapse
Affiliation(s)
| | | | - Yi Ren
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| |
Collapse
|
5
|
Jin M, Alam MM, Liu AYC, Jiang P. Rag2 -/- accelerates lipofuscin accumulation in the brain: Implications for human stem cell brain transplantation studies. Stem Cell Reports 2022; 17:2381-2391. [PMID: 36270284 PMCID: PMC9669406 DOI: 10.1016/j.stemcr.2022.09.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 11/05/2022] Open
Abstract
Immunodeficient mice are widely used in human stem cell transplantation research. Recombination activating gene 1 (Rag1) deletion results in immunodeficiency and leads to accelerated aging in zebrafish with increased cytosolic accumulation of lipofuscin (LF). Unlike zebrafish, mammals have two homologs, Rag1 and Rag2, that regulate adaptive immunity. Currently, little is known if and how Rag1-/- and Rag2-/- may impact aging and LF accumulation in immunodeficient mouse brains and how this may confound results in human neural cell transplantation studies. Here, we demonstrate that in Rag2-/- mouse brains, LF appears early, spreads broadly, emits strong autofluorescence, and accumulates with age. LF is found in various types of glial cells, including xenografted human microglia. Surprisingly, in Rag1-/- mouse brains, LF autofluorescence is seen at much older ages compared with Rag2-/- brains. This study provides direct evidence that Rag2-/- expedites LF occurrence and sets a context for studies using aged immunodeficient mice.
Collapse
Affiliation(s)
- Mengmeng Jin
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Mahabub Maraj Alam
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Alice Y-C Liu
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Peng Jiang
- Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA.
| |
Collapse
|
6
|
Fernández-Albarral JA, de Hoz R, Matamoros JA, Chen L, López-Cuenca I, Salobrar-García E, Sánchez-Puebla L, Ramírez JM, Triviño A, Salazar JJ, Ramírez AI. Retinal Changes in Astrocytes and Müller Glia in a Mouse Model of Laser-Induced Glaucoma: A Time-Course Study. Biomedicines 2022; 10:biomedicines10050939. [PMID: 35625676 PMCID: PMC9138377 DOI: 10.3390/biomedicines10050939] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/15/2022] [Accepted: 04/16/2022] [Indexed: 11/28/2022] Open
Abstract
Macroglia (astrocytes and Müller glia) may play an important role in the pathogenesis of glaucoma. In a glaucoma mouse model, we studied the effects of unilateral laser-induced ocular hypertension (OHT) on macroglia in OHT and contralateral eyes at different time points after laser treatment (1, 3, 5, 8 and 15 days) using anti-GFAP and anti-MHC-II, analyzing the morphological changes, GFAP-labelled retinal area (GFAP-PA), and GFAP and MHC-II immunoreactivity intensities ((GFAP-IRI and MHC-II-IRI)). In OHT and contralateral eyes, with respect to naïve eyes, at all the time points, we found the following: (i) astrocytes with thicker somas and more secondary processes, mainly in the intermediate (IR) and peripheral retina (PR); (ii) astrocytes with low GFAP-IRI and only primary processes near the optic disc (OD); (iii) an increase in total GFAP-RA, which was higher at 3 and 5 days, except for at 15 days; (iv) an increase in GFAP-IRI in the IR and especially in the PR; (v) a decrease in GFAP-IRI near the OD, especially at 1 and 5 days; (vi) a significant increase in MHC-II-IRI, which was higher in the IR and PR; and (vii) the Müller glia were GFAP+ and MHC-II+. In conclusion, in this model of glaucoma, there is a bilateral macroglial activation maintained over time involved in the inflammatory glaucoma process.
Collapse
Affiliation(s)
- Jose A. Fernández-Albarral
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (J.A.M.); (L.C.); (I.L.-C.); (E.S.-G.); (L.S.-P.); (J.M.R.); (A.T.)
| | - Rosa de Hoz
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (J.A.M.); (L.C.); (I.L.-C.); (E.S.-G.); (L.S.-P.); (J.M.R.); (A.T.)
- Departamento de Inmunología, Facultad de Óptica y Optometría, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - José A. Matamoros
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (J.A.M.); (L.C.); (I.L.-C.); (E.S.-G.); (L.S.-P.); (J.M.R.); (A.T.)
| | - Lejing Chen
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (J.A.M.); (L.C.); (I.L.-C.); (E.S.-G.); (L.S.-P.); (J.M.R.); (A.T.)
| | - Inés López-Cuenca
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (J.A.M.); (L.C.); (I.L.-C.); (E.S.-G.); (L.S.-P.); (J.M.R.); (A.T.)
| | - Elena Salobrar-García
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (J.A.M.); (L.C.); (I.L.-C.); (E.S.-G.); (L.S.-P.); (J.M.R.); (A.T.)
- Departamento de Inmunología, Facultad de Óptica y Optometría, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
| | - Lidia Sánchez-Puebla
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (J.A.M.); (L.C.); (I.L.-C.); (E.S.-G.); (L.S.-P.); (J.M.R.); (A.T.)
| | - José M. Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (J.A.M.); (L.C.); (I.L.-C.); (E.S.-G.); (L.S.-P.); (J.M.R.); (A.T.)
- Departamento de Inmunología, Facultad de Medicina, Oftalmología y ORL, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Alberto Triviño
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (J.A.M.); (L.C.); (I.L.-C.); (E.S.-G.); (L.S.-P.); (J.M.R.); (A.T.)
- Departamento de Inmunología, Facultad de Medicina, Oftalmología y ORL, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Juan J. Salazar
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (J.A.M.); (L.C.); (I.L.-C.); (E.S.-G.); (L.S.-P.); (J.M.R.); (A.T.)
- Departamento de Inmunología, Facultad de Óptica y Optometría, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
- Correspondence: (J.J.S.); (A.I.R.)
| | - Ana I. Ramírez
- Instituto de Investigaciones Oftalmológicas Ramón Castroviejo, Grupo UCM 920105, IdISSC, Universidad Complutense de Madrid, 28040 Madrid, Spain; (J.A.F.-A.); (R.d.H.); (J.A.M.); (L.C.); (I.L.-C.); (E.S.-G.); (L.S.-P.); (J.M.R.); (A.T.)
- Departamento de Inmunología, Facultad de Óptica y Optometría, Oftalmología y ORL, Universidad Complutense de Madrid, 28037 Madrid, Spain
- Correspondence: (J.J.S.); (A.I.R.)
| |
Collapse
|
7
|
Zelenka L, Pägelow D, Krüger C, Seele J, Ebner F, Rausch S, Rohde M, Lehnardt S, van Vorst K, Fulde M. Novel protocol for the isolation of highly purified neonatal murine microglia and astrocytes. J Neurosci Methods 2022; 366:109420. [PMID: 34808220 DOI: 10.1016/j.jneumeth.2021.109420] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/02/2021] [Accepted: 11/11/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND The crosstalk and reactivity of the cell type glia, especially microglia and astrocytes, have progressively gathered research attention in understanding proper brain function regulated by the innate immune response. Therefore, methods to isolate highly viable and pure glia for the analysis on a cell-specific level are indispensable. NEW METHOD We modified previously established techniques: Animal numbers were reduced by multiple microglial harvests from the same mixed glial culture, thereby maximizing microglial yields following the principles of the 3Rs (replacement, reduction, and refinement). We optimized Magnetic-activated cell sorting (MACS®) of microglia and astrocytes by applying cultivated primary glial cell suspensions instead of directly sorting dissociated single cell suspension. RESULTS We generated highly viable and pure microglia and astrocytes derived from a single mixed culture with a purity of ~99%, as confirmed by FACS analysis. Field emission scanning electron microscopy (FESEM) demonstrated integrity of the MACS-purified glial cells. Tumor necrosis factor (TNF) and Interleukin-10 (IL-10) ELISA confirmed pro- and anti-inflammatory responses to be functional in purified glia, but significantly weakened compared to non-purified cells, further highlighting the importance of cellular crosstalk for proper immune activation. COMPARISON WITH EXISTING METHOD(S) Unlike previous studies that either isolated a single type of glia or displayed a substantial proportion of contamination with other cell types, we achieved isolation of both microglia and astrocytes at an increased purity (99-100%). CONCLUSIONS We have created an optimized protocol for the efficient purification of both primary microglia and astrocytes. Our results clearly demonstrate the importance of purity in glial cell cultivation in order to examine immune responses, which particularly holds true for astrocytes. We propose the novel protocol as a tool to investigate the cell type-specific crosstalk between microglia and astrocytes in the frame of CNS diseases.
Collapse
Affiliation(s)
- Laura Zelenka
- Institute of Microbiology and Epizootics, Centre of Infection Medicine, Freie Universität Berlin, Robert-von-Ostertag-Straße 7-13, 14163 Berlin, Germany
| | - Dennis Pägelow
- Institute of Microbiology and Epizootics, Centre of Infection Medicine, Freie Universität Berlin, Robert-von-Ostertag-Straße 7-13, 14163 Berlin, Germany
| | - Christina Krüger
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jana Seele
- University Medical Center Göttingen, Institute of Neuropathology, Göttingen, Germany
| | - Friederike Ebner
- Freie Universität Berlin, Institute of Immunology, Berlin, Germany
| | - Sebastian Rausch
- Freie Universität Berlin, Institute of Immunology, Berlin, Germany
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Seija Lehnardt
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Kira van Vorst
- Institute of Microbiology and Epizootics, Centre of Infection Medicine, Freie Universität Berlin, Robert-von-Ostertag-Straße 7-13, 14163 Berlin, Germany
| | - Marcus Fulde
- Institute of Microbiology and Epizootics, Centre of Infection Medicine, Freie Universität Berlin, Robert-von-Ostertag-Straße 7-13, 14163 Berlin, Germany.
| |
Collapse
|
8
|
Dando SJ, Kazanis R, McMenamin PG. Myeloid Cells in the Mouse Retina and Uveal Tract Respond Differently to Systemic Inflammatory Stimuli. Invest Ophthalmol Vis Sci 2021; 62:10. [PMID: 34379096 PMCID: PMC8363776 DOI: 10.1167/iovs.62.10.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 07/03/2021] [Indexed: 01/14/2023] Open
Abstract
Purpose In spite of clear differences in tissue function and significance to ocular disease, little is known about how immune responses differ between the retina and uveal tract. To this end we compared the effects of acute systemic inflammation on myeloid cells within the mouse retina, iris-ciliary body, and choroid. Methods Systemic inflammation was induced in Cx3cr1gfp/gfp and CD11c-eYFP Crb1wt/wtmice by intraperitoneal lipopolysaccharide (LPS). In vivo fundus imaging was performed at two, 24, and 48 hours after LPS, and ocular tissue wholemounts were immunostained and studied by confocal microscopy. Flow cytometry was used to investigate the expression of activation markers (MHC class II, CD80, CD86) on myeloid cell populations at 24 hours. For functional studies, retinal microglia were isolated from LPS-exposed mice and cocultured with naïve OT-II CD4+ T-cells and ovalbumin peptide. T-cell proliferation was measured by flow cytometry and cytokine assays. Results Systemic LPS altered the density and morphology of retinal microglia; however, retinal microglia did not upregulate antigen presentation markers and failed to stimulate naïve CD4+ T-cell proliferation in vitro. In contrast, uveal tract myeloid cells displayed a phenotype consistent with late-activated antigen-presenting cells at 24 hours. Systemic LPS induced remodeling of myeloid populations within the uveal tract, particularly in the choroid, where dendritic cells were partially displaced by macrophages at 24 hours. Conclusions The disparate myeloid cell responses in the retina and uveal tract after systemic LPS highlight differential regulation of innate immunity within these tissue environments, observations that underpin and advance our understanding of ocular immune privilege.
Collapse
Affiliation(s)
- Samantha J. Dando
- Queensland University of Technology, Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Brisbane, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
| | - Renee Kazanis
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
| | - Paul G. McMenamin
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
| |
Collapse
|
9
|
Murdock BJ, Famie JP, Piecuch CE, Pawlowski KD, Mendelson FE, Pieroni CH, Iniguez SD, Zhao L, Goutman SA, Feldman EL. NK cells associate with ALS in a sex- and age-dependent manner. JCI Insight 2021; 6:147129. [PMID: 33974561 PMCID: PMC8262328 DOI: 10.1172/jci.insight.147129] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 05/05/2021] [Indexed: 12/11/2022] Open
Abstract
NK cells are innate immune cells implicated in ALS; whether NK cells impact ALS in a sex- and age-specific manner was investigated. Herein, NK cells were depleted in male and female SOD1G93A ALS mice, survival and neuroinflammation were assessed, and data were stratified by sex. NK cell depletion extended survival in female but not male ALS mice with sex-specific effects on spinal cord microglia. In humans, NK cell numbers, NK cell subpopulations, and NK cell surface markers were examined in prospectively blood collected from subjects with ALS and control subjects; longitudinal changes in these metrics were correlated to revised ALS functional rating scale (ALSFRS-R) slope and stratified by sex and age. Expression of NK cell trafficking and cytotoxicity markers was elevated in subjects with ALS, and changes in CXCR3+ NK cells and 7 trafficking and cytotoxicity markers (CD11a, CD11b, CD38, CX3CR1, NKG2D, NKp30, NKp46) correlated with disease progression. Age affected the associations between ALSFRS-R and markers NKG2D and NKp46, whereas sex impacted the NKp30 association. Collectively, these findings suggest that NK cells contribute to ALS progression in a sex- and age-specific manner and demonstrate that age and sex are critical variables when designing and assessing ALS immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lili Zhao
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | | | | |
Collapse
|
10
|
Abstract
Peptide and dendritic cell vaccines activate the immune system against tumor antigens to combat brain tumors. Vaccines stimulate a systemic immune response by inducing both antitumor T cells as well as humoral immunity through antibody production to cross the blood-brain barrier and combat brain tumors. Recent trials investigating vaccines against peptides (ie, epithelial growth factor receptor variant III, survivin, heat shock proteins, or personalized tumor antigens) and dendritic cells pulsed with known peptides, messenger RNA or unknown tumor lysate targets demonstrate the potential for therapeutic cancer vaccines to become an important therapy for brain tumor treatment.
Collapse
Affiliation(s)
- Justin Lee
- UCLA Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California Los Angeles, 300 Stein Plaza Driveway Suite 420 Los Angeles, CA 90095, USA
| | - Benjamin R Uy
- UCLA Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California Los Angeles, 300 Stein Plaza Driveway Suite 420 Los Angeles, CA 90095, USA
| | - Linda M Liau
- UCLA Department of Neurosurgery, David Geffen School of Medicine at UCLA, University of California Los Angeles, 300 Stein Plaza Driveway Suite 420 Los Angeles, CA 90095, USA.
| |
Collapse
|
11
|
Fernández A, Quintana E, Velasco P, Moreno-Jimenez B, de Andrés B, Gaspar ML, Liste I, Vilar M, Mira H, Cano E. Senescent accelerated prone 8 (SAMP8) mice as a model of age dependent neuroinflammation. J Neuroinflammation 2021; 18:75. [PMID: 33736657 PMCID: PMC7977588 DOI: 10.1186/s12974-021-02104-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 02/03/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Aging and age-related diseases are strong risk factors for the development of neurodegenerative diseases. Neuroinflammation (NIF), as the brain's immune response, plays an important role in aged associated degeneration of central nervous system (CNS). There is a need for well characterized animal models that will allow the scientific community to understand and modulate this process. METHODS We have analyzed aging-phenotypical and inflammatory changes of brain myeloid cells (bMyC) in a senescent accelerated prone aged (SAMP8) mouse model, and compared with their senescence resistant control mice (SAMR1). We have performed morphometric methods to evaluate the architecture of cellular prolongations and determined the appearance of Iba1+ clustered cells with aging. To analyze specific constant brain areas, we have performed stereology measurements of Iba1+ cells in the hippocampal formation. We have isolated bMyC from brain parenchyma (BP) and choroid plexus plus meningeal membranes (m/Ch), and analyzed their response to systemic lipopolysaccharide (LPS)-driven inflammation. RESULTS Aged 10 months old SAMP8 mice present many of the hallmarks of aging-dependent neuroinflammation when compared with their SAMR1 control, i.e., increase of protein aggregates, presence of Iba1+ clusters, but not an increase in the number of Iba1+ cells. We have further observed an increase of main inflammatory mediator IL-1β, and an augment of border MHCII+Iba1+ cells. Isolated CD45+ bMyC from brain parenchyma (BP) and choroid plexus plus meningeal membranes (m/Ch) have been analyzed, showing that there is not a significant increase of CD45+ cells from the periphery. Our data support that aged-driven pro-inflammatory cytokine interleukin 1 beta (IL-1β) transcription is enhanced in CD45+BP cells. Furthermore, LPS-driven systemic inflammation produces inflammatory cytokines mainly in border bMyC, sensed to a lesser extent by the BP bMyC, showing that IL-1β expression is further augmented in aged SAMP8 compared to control SAMR1. CONCLUSION Our data validate the SAMP8 model to study age-associated neuroinflammatory events, but careful controls for age and strain are required. These animals show morphological changes in their bMyC cell repertoires associated to age, corresponding to an increase in the production of pro-inflammatory cytokines such as IL-1β, which predispose the brain to an enhanced inflammatory response after LPS-systemic challenge.
Collapse
Affiliation(s)
- Andrés Fernández
- Chronic Disease Programme, Neuroinflammation Unit, Instituto de Salud Carlos III, Carretera Majadahonda-Pozuelo, Km.2,2, Majadahonda, 28220, Madrid, Spain
| | - Elena Quintana
- Chronic Disease Programme, Neuroinflammation Unit, Instituto de Salud Carlos III, Carretera Majadahonda-Pozuelo, Km.2,2, Majadahonda, 28220, Madrid, Spain
| | - Patricia Velasco
- Chronic Disease Programme, Neuroinflammation Unit, Instituto de Salud Carlos III, Carretera Majadahonda-Pozuelo, Km.2,2, Majadahonda, 28220, Madrid, Spain
| | - Belén Moreno-Jimenez
- Chronic Disease Programme, Neuroinflammation Unit, Instituto de Salud Carlos III, Carretera Majadahonda-Pozuelo, Km.2,2, Majadahonda, 28220, Madrid, Spain
| | - Belén de Andrés
- Unidad de Inmunobiología, Instituto de Salud Carlos II, Madrid, Spain
| | | | - Isabel Liste
- Chronic Disease Programme, Neuroinflammation Unit, Instituto de Salud Carlos III, Carretera Majadahonda-Pozuelo, Km.2,2, Majadahonda, 28220, Madrid, Spain
| | - Marçal Vilar
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Helena Mira
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Eva Cano
- Chronic Disease Programme, Neuroinflammation Unit, Instituto de Salud Carlos III, Carretera Majadahonda-Pozuelo, Km.2,2, Majadahonda, 28220, Madrid, Spain.
| |
Collapse
|
12
|
Frühauf M, Zeitschel U, Höfling C, Ullm F, Rabiger FV, Alber G, Pompe T, Müller U, Roßner S. Construction of a 3D brain extracellular matrix model to study the interaction between microglia and T cells in co-culture. Eur J Neurosci 2020; 53:4034-4050. [PMID: 32954591 DOI: 10.1111/ejn.14978] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 12/24/2022]
Abstract
Neurodegenerative disorders are characterised by the activation of brain-resident microglia cells and by the infiltration of peripheral T cells. However, their interplay in disease has not been clarified yet. It is difficult to investigate complex cellular dynamics in living animals, and simple two-dimensional (2D) cell culture models do not resemble the soft 3D structure of brain tissue. Therefore, we developed a biomimetic 3D in vitro culture system for co-cultivation of microglia and T cells. As the activation and/or migration of immune cells in the brain might be affected by components of the extracellular matrix, defined 3D fibrillar collagen I-based matrices were constructed and modified with hyaluronan and/or chondroitin sulphate, resembling aspects of brain extracellular matrix. Murine microglia and spleen-derived T cells were cultured alone or in co-culture on the constructed matrices. Microglia exhibited in vivo-like morphology and T cells showed enhanced survival when co-cultured with microglia or to a minor degree in the presence of glia-conditioned medium. The open and porous fibrillar structure of the matrix allowed for cell invasion and direct cell-cell interaction, with stronger invasion of T cells. Both cell types showed no dependence on the matrix modifications. Microglia could be activated on the matrices by lipopolysaccharide resulting in interleukin-6 and tumour necrosis factor-α secretion. The findings herein indicate that biomimetic 3D matrices allow for co-cultivation and activation of primary microglia and T cells and provide useful tools to study their interaction in vitro.
Collapse
Affiliation(s)
- Marie Frühauf
- Paul Flechsig Institute for Brain Research, Medical Faculty, University of Leipzig, Leipzig, Germany.,Institute of Immunology/Molecular Pathogenesis, Centre for Biotechnology and Biomedicine, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany.,Institute of Biochemistry, Faculty of Life Sciences, University of Leipzig, Leipzig, Germany
| | - Ulrike Zeitschel
- Paul Flechsig Institute for Brain Research, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Corinna Höfling
- Paul Flechsig Institute for Brain Research, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Franziska Ullm
- Institute of Biochemistry, Faculty of Life Sciences, University of Leipzig, Leipzig, Germany
| | - Friederike V Rabiger
- Institute of Immunology/Molecular Pathogenesis, Centre for Biotechnology and Biomedicine, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Gottfried Alber
- Institute of Immunology/Molecular Pathogenesis, Centre for Biotechnology and Biomedicine, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Tilo Pompe
- Institute of Biochemistry, Faculty of Life Sciences, University of Leipzig, Leipzig, Germany
| | - Uwe Müller
- Institute of Immunology/Molecular Pathogenesis, Centre for Biotechnology and Biomedicine, Faculty of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Steffen Roßner
- Paul Flechsig Institute for Brain Research, Medical Faculty, University of Leipzig, Leipzig, Germany
| |
Collapse
|
13
|
Rostami J, Fotaki G, Sirois J, Mzezewa R, Bergström J, Essand M, Healy L, Erlandsson A. Astrocytes have the capacity to act as antigen-presenting cells in the Parkinson's disease brain. J Neuroinflammation 2020; 17:119. [PMID: 32299492 PMCID: PMC7164247 DOI: 10.1186/s12974-020-01776-7] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 03/17/2020] [Indexed: 02/10/2023] Open
Abstract
Background Many lines of evidence suggest that accumulation of aggregated alpha-synuclein (αSYN) in the Parkinson’s disease (PD) brain causes infiltration of T cells. However, in which ways the stationary brain cells interact with the T cells remain elusive. Here, we identify astrocytes as potential antigen-presenting cells capable of activating T cells in the PD brain. Astrocytes are a major component of the nervous system, and accumulating data indicate that astrocytes can play a central role during PD progression. Methods To investigate the role of astrocytes in antigen presentation and T-cell activation in the PD brain, we analyzed post mortem brain tissue from PD patients and controls. Moreover, we studied the capacity of cultured human astrocytes and adult human microglia to act as professional antigen-presenting cells following exposure to preformed αSYN fibrils. Results Our analysis of post mortem brain tissue demonstrated that PD patients express high levels of MHC-II, which correlated with the load of pathological, phosphorylated αSYN. Interestingly, a very high proportion of the MHC-II co-localized with astrocytic markers. Importantly, we found both perivascular and infiltrated CD4+ T cells to be surrounded by MHC-II expressing astrocytes, confirming an astrocyte T cell cross-talk in the PD brain. Moreover, we showed that αSYN accumulation in cultured human astrocytes triggered surface expression of co-stimulatory molecules critical for T-cell activation, while cultured human microglia displayed very poor antigen presentation capacity. Notably, intercellular transfer of αSYN/MHC-II deposits occurred between astrocytes via tunneling nanotubes, indicating spreading of inflammation in addition to toxic protein aggregates. Conclusions In conclusion, our data from histology and cell culture studies suggest an important role for astrocytes in antigen presentation and T-cell activation in the PD brain, highlighting astrocytes as a promising therapeutic target in the context of chronic inflammation.
Collapse
Affiliation(s)
- Jinar Rostami
- Molecular Geriatrics, Department of Public Health and Caring Sciences/Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Grammatiki Fotaki
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Julien Sirois
- Neuroimmunology Unit, department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, 3801, Canada
| | - Ropafadzo Mzezewa
- Molecular Geriatrics, Department of Public Health and Caring Sciences/Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Joakim Bergström
- Molecular Geriatrics, Department of Public Health and Caring Sciences/Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Magnus Essand
- Department of Immunology, Genetics and Pathology, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Luke Healy
- Neuroimmunology Unit, department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, 3801, Canada
| | - Anna Erlandsson
- Molecular Geriatrics, Department of Public Health and Caring Sciences/Molecular Geriatrics, Rudbeck Laboratory, Uppsala University, SE-751 85, Uppsala, Sweden.
| |
Collapse
|
14
|
Shukla AK, McIntyre LL, Marsh SE, Schneider CA, Hoover EM, Walsh CM, Lodoen MB, Blurton-Jones M, Inlay MA. CD11a expression distinguishes infiltrating myeloid cells from plaque-associated microglia in Alzheimer's disease. Glia 2018; 67:844-856. [PMID: 30588668 DOI: 10.1002/glia.23575] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/11/2018] [Accepted: 11/15/2018] [Indexed: 11/10/2022]
Abstract
Alzheimer's disease (AD) is the leading cause of age-related neurodegeneration and is characterized neuropathologically by the accumulation of insoluble beta-amyloid (Aβ) peptides. In AD brains, plaque-associated myeloid (PAM) cells cluster around Aβ plaques but fail to effectively clear Aβ by phagocytosis. PAM cells were originally thought to be brain-resident microglia. However, several studies have also suggested that Aβ-induced inflammation causes peripheral monocytes to enter the otherwise immune-privileged brain. The relationship between AD progression and inflammation in the brain remains ambiguous because microglia and monocyte-derived macrophages are extremely difficult to distinguish from one another in an inflamed brain. Whether PAM cells are microglia, peripheral macrophages, or a mixture of both remains unclear. CD11a is a component of the β2 integrin LFA1. We have determined that CD11a is highly expressed on peripheral immune cells, including macrophages, but is not expressed by mouse microglia. These expression patterns remain consistent in LPS-treated inflamed mice, as well as in two mouse models of AD. Thus, CD11a can be used as a marker to distinguish murine microglia from infiltrating peripheral immune cells. Using CD11a, we show that PAM cells in AD transgenic brains are comprised entirely of microglia. We also demonstrate a novel fluorescence-assisted quantification technique (FAQT), which reveals a significant increase in T lymphocytes, especially in the brains of female AD mice. Our findings support the notion that microglia are the lead myeloid players in AD and that rejuvenating their phagocytic potential may be an important therapeutic strategy.
Collapse
Affiliation(s)
- Ankita K Shukla
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, California.,Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, California
| | - Laura L McIntyre
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, California.,Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, California
| | - Samuel E Marsh
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, California.,Department of Neurobiology and Behavior, University of California Irvine, Irvine, California
| | - Christine A Schneider
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, California
| | - Evelyn M Hoover
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, California
| | - Craig M Walsh
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, California.,Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, California
| | - Melissa B Lodoen
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, California
| | - Mathew Blurton-Jones
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, California.,Department of Neurobiology and Behavior, University of California Irvine, Irvine, California
| | - Matthew A Inlay
- Sue and Bill Gross Stem Cell Research Center, University of California Irvine, Irvine, California.,Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, California
| |
Collapse
|
15
|
Antigen-presenting cell diversity for T cell reactivation in central nervous system autoimmunity. J Mol Med (Berl) 2018; 96:1279-1292. [PMID: 30386908 DOI: 10.1007/s00109-018-1709-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/19/2018] [Accepted: 10/24/2018] [Indexed: 12/13/2022]
Abstract
Autoreactive T cells are considered the major culprits in the pathogenesis of many autoimmune diseases like multiple sclerosis (MS). Upon activation in the lymphoid organs, autoreactive T cells migrate towards the central nervous system (CNS) and target the myelin sheath-forming oligodendrocytes, resulting in detrimental neurological symptoms. Despite the availability of extensively studied systems like the experimental autoimmune encephalomyelitis (EAE) model, our understanding of this disease and the underlying pathogenesis is still elusive. One vividly discussed subject represents the T cell reactivation in the CNS. In order to exert their effector functions in the CNS, autoreactive T cells must encounter antigen-presenting cells (APCs). This interaction provides an antigen-restricted stimulus in the context of major histocompatibility complex class II (MHC-II) and other co-stimulatory molecules. Peripherally derived dendritic cells (DCs), B cells, border-associated macrophages (BAM), CNS-resident microglia, and astrocytes have the capacity to express molecules required for antigen presentation under inflammatory conditions. Also, endothelial cells can fulfill these prerequisites in certain situations. Which of these cells in fact act as APCs for T cell reactivation and to which extent they can exert this function has been studied intensively, but unfortunately with no firm conclusion. In this review, we will summarize the findings that support or question the antigen presenting capacities of the mentioned cell types of CNS-localized T cell reactivation.
Collapse
|
16
|
Harms AS, Delic V, Thome AD, Bryant N, Liu Z, Chandra S, Jurkuvenaite A, West AB. α-Synuclein fibrils recruit peripheral immune cells in the rat brain prior to neurodegeneration. Acta Neuropathol Commun 2017; 5:85. [PMID: 29162163 PMCID: PMC5698965 DOI: 10.1186/s40478-017-0494-9] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 11/12/2017] [Indexed: 12/20/2022] Open
Abstract
Genetic variation in a major histocompatibility complex II (MHCII)-encoding gene (HLA-DR) increases risk for Parkinson disease (PD), and the accumulation of MHCII-expressing immune cells in the brain correlates with α-synuclein inclusions. However, the timing of MHCII-cell recruitment with respect to ongoing neurodegeneration, and the types of cells that express MHCII in the PD brain, has been difficult to understand. Recent studies show that the injection of short α-synuclein fibrils into the rat substantia nigra pars compacta (SNpc) induces progressive inclusion formation in SNpc neurons that eventually spread to spiny projection neurons in the striatum. Herein, we find that α-synuclein fibrils rapidly provoke a persistent MHCII response in the brain. In contrast, equivalent amounts of monomeric α-synuclein fail to induce MHCII or persistent microglial activation, consistent with our results in primary microglia. Flow cytometry and immunohistochemical analyses reveal that MHCII-expressing cells are composed of both resident microglia as well as cells from the periphery that include monocytes, macrophages, and lymphocytes. Over time, α-Synuclein fibril exposures in the SNpc causes both axon loss as well as monocyte recruitment in the striatum. While these monocytes in the striatum initially lack MHCII expression, α-synuclein inclusions later form in nearby spiny projection neurons and MHCII expression becomes robust. In summary, in the rat α-synuclein fibril model, peripheral immune cell recruitment occurs prior to neurodegeneration and microglia, monocytes and macrophages all contribute to MHCII expression.
Collapse
Affiliation(s)
- Ashley S Harms
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
- , 1719 6th Ave South, Birmingham, AL, 35233, USA.
| | - Vedad Delic
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Aaron D Thome
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Nicole Bryant
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Zhiyong Liu
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Sidhanth Chandra
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Asta Jurkuvenaite
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Andrew B West
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| |
Collapse
|
17
|
Carriba P, Davies AM. CD40 is a major regulator of dendrite growth from developing excitatory and inhibitory neurons. eLife 2017; 6:30442. [PMID: 29111976 PMCID: PMC5687868 DOI: 10.7554/elife.30442] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 11/06/2017] [Indexed: 12/13/2022] Open
Abstract
Dendrite size and morphology are key determinants of the functional properties of neurons and neural circuits. Here we show that CD40, a member of the TNF receptor superfamily, is a major regulator of dendrite growth and elaboration in the developing brain. The dendrites of hippocampal excitatory neurons were markedly stunted in Cd40-/- mice, whereas those of striatal inhibitory neurons were much more exuberant. These striking and opposite phenotypic changes were also observed in excitatory and inhibitory neurons cultured from Cd40-/- mice and were rescued by soluble CD40. The changes in excitatory and inhibitory neurons cultured from Cd40-/- mice were mimicked in neurons of Cd40+/+ mice by treatment with soluble CD40L and were dependent on PKC-β and PKC-γ, respectively. These results suggest that CD40-activated CD40L reverse signalling has striking and opposite effects on the growth and elaboration of dendrites among major classes of brain neurons by PKC-dependent mechanisms.
Collapse
Affiliation(s)
- Paulina Carriba
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Alun M Davies
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
18
|
Menzel F, Kaiser N, Haehnel S, Rapp F, Patties I, Schöneberg N, Haimon Z, Immig K, Bechmann I. Impact of X-irradiation on microglia. Glia 2017; 66:15-33. [DOI: 10.1002/glia.23239] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 09/05/2017] [Accepted: 09/18/2017] [Indexed: 12/20/2022]
Affiliation(s)
| | - Nicole Kaiser
- Institute of Anatomy, Leipzig University; Leipzig Germany
| | - Susann Haehnel
- Institute of Anatomy, Leipzig University; Leipzig Germany
| | - Felicitas Rapp
- Institute of Anatomy, Leipzig University; Leipzig Germany
| | - Ina Patties
- Department of Radiation Therapy; Leipzig University; Leipzig Germany
| | | | - Zhana Haimon
- Department of Immunology; Weizmann Institute of Science; Rehovot Israel
| | - Kerstin Immig
- Institute of Anatomy, Leipzig University; Leipzig Germany
| | - Ingo Bechmann
- Institute of Anatomy, Leipzig University; Leipzig Germany
| |
Collapse
|
19
|
Sun BL, Wang LH, Yang T, Sun JY, Mao LL, Yang MF, Yuan H, Colvin RA, Yang XY. Lymphatic drainage system of the brain: A novel target for intervention of neurological diseases. Prog Neurobiol 2017; 163-164:118-143. [PMID: 28903061 DOI: 10.1016/j.pneurobio.2017.08.007] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 08/11/2017] [Accepted: 08/31/2017] [Indexed: 12/20/2022]
Abstract
The belief that the vertebrate brain functions normally without classical lymphatic drainage vessels has been held for many decades. On the contrary, new findings show that functional lymphatic drainage does exist in the brain. The brain lymphatic drainage system is composed of basement membrane-based perivascular pathway, a brain-wide glymphatic pathway, and cerebrospinal fluid (CSF) drainage routes including sinus-associated meningeal lymphatic vessels and olfactory/cervical lymphatic routes. The brain lymphatic systems function physiological as a route of drainage for interstitial fluid (ISF) from brain parenchyma to nearby lymph nodes. Brain lymphatic drainage helps maintain water and ion balance of the ISF, waste clearance, and reabsorption of macromolecular solutes. A second physiological function includes communication with the immune system modulating immune surveillance and responses of the brain. These physiological functions are influenced by aging, genetic phenotypes, sleep-wake cycle, and body posture. The impairment and dysfunction of the brain lymphatic system has crucial roles in age-related changes of brain function and the pathogenesis of neurovascular, neurodegenerative, and neuroinflammatory diseases, as well as brain injury and tumors. In this review, we summarize the key component elements (regions, cells, and water transporters) of the brain lymphatic system and their regulators as potential therapeutic targets in the treatment of neurologic diseases and their resulting complications. Finally, we highlight the clinical importance of ependymal route-based targeted gene therapy and intranasal drug administration in the brain by taking advantage of the unique role played by brain lymphatic pathways in the regulation of CSF flow and ISF/CSF exchange.
Collapse
Affiliation(s)
- Bao-Liang Sun
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong (Taishan Medical University), Department of Neurology, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, China.
| | - Li-Hua Wang
- Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261031, China
| | - Tuo Yang
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jing-Yi Sun
- Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, Gangwon 220-701, Republic of Korea
| | - Lei-Lei Mao
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong (Taishan Medical University), Department of Neurology, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, China
| | - Ming-Feng Yang
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong (Taishan Medical University), Department of Neurology, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, China
| | - Hui Yuan
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong (Taishan Medical University), Department of Neurology, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, China
| | - Robert A Colvin
- Department of Biological Sciences, Interdisciplinary Graduate Program in Molecular and Cellular Biology, Neuroscience Program, Ohio University, Athens, OH 45701, USA
| | - Xiao-Yi Yang
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong (Taishan Medical University), Department of Neurology, Affiliated Hospital of Taishan Medical University, Tai'an, Shandong 271000, China.
| |
Collapse
|
20
|
Wong EL, Stowell RD, Majewska AK. What the Spectrum of Microglial Functions Can Teach us About Fetal Alcohol Spectrum Disorder. Front Synaptic Neurosci 2017; 9:11. [PMID: 28674490 PMCID: PMC5474469 DOI: 10.3389/fnsyn.2017.00011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 05/29/2017] [Indexed: 12/18/2022] Open
Abstract
Alcohol exposure during gestation can lead to severe defects in brain development and lifelong physical, behavioral and learning deficits that are classified under the umbrella term fetal alcohol spectrum disorder (FASD). Sadly, FASD is diagnosed at an alarmingly high rate, affecting 2%–5% of live births in the United States, making it the most common non-heritable cause of mental disability. Currently, no standard therapies exist that are effective at battling FASD symptoms, highlighting a pressing need to better understand the underlying mechanisms by which alcohol affects the developing brain. While it is clear that sensory and cognitive deficits are driven by inappropriate development and remodeling of the neural circuits that mediate these processes, alcohol’s actions acutely and long-term on the brain milieu are diverse and complex. Microglia, the brain’s immune cells, have been thought to be a target for alcohol during development because of their exquisite ability to rapidly detect and respond to perturbations affecting the brain. Additionally, our view of these immune cells is rapidly changing, and recent studies have revealed a myriad of microglial physiological functions critical for normal brain development and long-term function. A clear and complete understanding of how microglial roles on this end of the spectrum may be altered in FASD is currently lacking. Such information could provide important insights toward novel therapeutic targets for FASD treatment. Here we review the literature that links microglia to neural circuit remodeling and provide a discussion of the current understanding of how developmental alcohol exposure affects microglial behavior in the context of developing brain circuits.
Collapse
Affiliation(s)
- Elissa L Wong
- Department of Environmental Medicine, University of Rochester Medical CenterRochester, NY, United States
| | - Rianne D Stowell
- Department of Neuroscience, University of Rochester Medical CenterRochester, NY, United States
| | - Ania K Majewska
- Department of Neuroscience, University of Rochester Medical CenterRochester, NY, United States
| |
Collapse
|
21
|
Schiefenhövel F, Immig K, Prodinger C, Bechmann I. Indications for cellular migration from the central nervous system to its draining lymph nodes in CD11c-GFP + bone-marrow chimeras following EAE. Exp Brain Res 2017; 235:2151-2166. [PMID: 28421248 DOI: 10.1007/s00221-017-4956-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Accepted: 04/07/2017] [Indexed: 12/25/2022]
Abstract
The concept as to how the brain maintains its immune privilege has initially been based on observations that it is lacking classical lymph vessels and later, the absence of dendritic cells (DC). This view has been challenged by several groups demonstrating drainage/migration of injected tracers and cells into cervical lymph nodes (CLNs) and the presence of brain antigens in CLNs in the course of various brain pathologies. Using CD11c-diphtheria toxin receptor (DTR)-green fluorescent protein (GFP) transgenic (tg) mice, we have shown the existence of CD11c+ cells, a main DC marker, within the brain parenchyma. Since injecting tracers or cells may cause barrier artefacts, we have now transplanted wild type (wt)-bone marrow (BM) to lethally irradiated CD11c-DTR-GFP tg mice to restrict the CD11c-DTR-GFP+ population to the brain and induced experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). We observed ramified GFP+ cells in the olfactory bulb, the cribriform plate, the nasal mucosa and superficial CLNs. We measured a significant increase of host gfp genomic DNA (gDNA) levels in lymph nodes (LNs) previously described as draining stations for the central nervous system (CNS). Using flow cytometry analysis, we observed an increase of the percentage of CD11c-GFP+ cells in brain parenchyma in the course of EAE which is most likely due to an up-regulation of CD11c of resident microglial cells since levels of gfp gDNA did not increase. Our data supports the hypothesis that brain-resident antigen presenting cells (APC) are capable of migrating to CNS-draining LNs to present myelin-associated epitopes.
Collapse
Affiliation(s)
- Fridtjof Schiefenhövel
- Medical Faculty, Institute of Anatomy, Leipzig University, Oststraße 25, 04317, Leipzig, Germany
| | - Kerstin Immig
- Medical Faculty, Institute of Anatomy, Leipzig University, Oststraße 25, 04317, Leipzig, Germany.
| | - Carolin Prodinger
- Medical Faculty, Institute of Anatomy, Leipzig University, Oststraße 25, 04317, Leipzig, Germany
| | - Ingo Bechmann
- Medical Faculty, Institute of Anatomy, Leipzig University, Oststraße 25, 04317, Leipzig, Germany
| |
Collapse
|
22
|
Warden A, Erickson E, Robinson G, Harris RA, Mayfield RD. The neuroimmune transcriptome and alcohol dependence: potential for targeted therapies. Pharmacogenomics 2016; 17:2081-2096. [PMID: 27918243 DOI: 10.2217/pgs-2016-0062] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Transcriptome profiling enables discovery of gene networks that are altered in alcoholic brains. This technique has revealed involvement of the brain's neuroimmune system in regulating alcohol abuse and dependence, and has provided potential therapeutic targets. In this review, we discuss Toll-like-receptor pathways, hypothesized to be key players in many stages of the alcohol addiction cycle. The growing appreciation of the neuroimmune system's involvement in alcoholism has also led to consideration of crucial roles for glial cells, including astrocytes and microglia, in the brain's response to alcohol abuse. We discuss current knowledge and hypotheses on the roles that specific neuroimmune cell types may play in addiction. Current strategies for repurposing US FDA-approved drugs for the treatment of alcohol use disorders are also discussed.
Collapse
Affiliation(s)
- Anna Warden
- The University of Texas at Austin, Waggoner Center for Alcohol & Addiction Research, Austin, TX, USA
| | - Emma Erickson
- The University of Texas at Austin, Waggoner Center for Alcohol & Addiction Research, Austin, TX, USA
| | - Gizelle Robinson
- The University of Texas at Austin, Waggoner Center for Alcohol & Addiction Research, Austin, TX, USA
| | - R Adron Harris
- The University of Texas at Austin, Waggoner Center for Alcohol & Addiction Research, Austin, TX, USA
| | - R Dayne Mayfield
- The University of Texas at Austin, Waggoner Center for Alcohol & Addiction Research, Austin, TX, USA
| |
Collapse
|
23
|
Hernandez A, Donovan V, Grinberg YY, Obenaus A, Carson MJ. Differential detection of impact site versus rotational site injury by magnetic resonance imaging and microglial morphology in an unrestrained mild closed head injury model. J Neurochem 2016; 136 Suppl 1:18-28. [PMID: 26806371 PMCID: PMC5047732 DOI: 10.1111/jnc.13402] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/05/2015] [Accepted: 10/06/2015] [Indexed: 01/31/2023]
Abstract
Seventy‐five percent of all traumatic brain injuries are mild and do not cause readily visible abnormalities on routine medical imaging making it difficult to predict which individuals will develop unwanted clinical sequelae. Microglia are brain‐resident macrophages and early responders to brain insults. Their activation is associated with changes in morphology or expression of phenotypic markers including P2Y12 and major histocompatibility complex class II. Using a murine model of unrestrained mild closed head injury (mCHI), we used microglia as reporters of acute brain injury at sites of impact versus sites experiencing rotational stress 24 h post‐mCHI. Consistent with mild injury, a modest 20% reduction in P2Y12 expression was detected by quantitative real‐time PCR (qPCR) analysis but only in the impacted region of the cortex. Furthermore, neither an influx of blood‐derived immune cells nor changes in microglial expression of CD45, TREM1, TREM2, major histocompatibility complex class II or CD40 were detected. Using magnetic resonance imaging (MRI), small reductions in T2 weighted values were observed but only near the area of impact and without overt tissue damage (blood deposition, edema). Microglial morphology was quantified without cryosectioning artifacts using ScaleA2 clarified brains from CX3CR1‐green fluorescence protein (GFP) mice. The cortex rostral to the mCHI impact site receives greater rotational stress but neither MRI nor molecular markers of microglial activation showed significant changes from shams in this region. However, microglia in this rostral region did display signs of morphologic activation equivalent to that observed in severe CHI. Thus, mCHI‐triggered rotational stress is sufficient to cause injuries undetectable by routine MRI that could result in altered microglial surveillance of brain homeostasis.
Acute changes in microglial morphology reveal brain responses to unrestrained mild traumatic brain injury
In areas subjected to rotational stress distant from impact site In the absence of detectable changes in standard molecular indicators of brain damage, inflammation or microglial activation. That might result in decreased surveillance of brain function and increased susceptibility to subsequent brain insults.
Collapse
Affiliation(s)
- Alfredo Hernandez
- Center for Glial-Neuronal Interactions, University of California Riverside, School of Medicine, Riverside, California, USA.,MarcU Program, University of California Riverside, Riverside, California, USA.,Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, California, USA
| | - Virgina Donovan
- Center for Glial-Neuronal Interactions, University of California Riverside, School of Medicine, Riverside, California, USA.,Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, California, USA.,Cell Molecular and Developmental Biology Program, University of California Riverside, Riverside, California, USA.,Loma Linda University School of Medicine, Loma Linda California, Loma Linda, CA, USA
| | - Yelena Y Grinberg
- Center for Glial-Neuronal Interactions, University of California Riverside, School of Medicine, Riverside, California, USA.,Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, California, USA
| | - Andre Obenaus
- Center for Glial-Neuronal Interactions, University of California Riverside, School of Medicine, Riverside, California, USA.,Cell Molecular and Developmental Biology Program, University of California Riverside, Riverside, California, USA.,Loma Linda University School of Medicine, Loma Linda California, Loma Linda, CA, USA
| | - Monica J Carson
- Center for Glial-Neuronal Interactions, University of California Riverside, School of Medicine, Riverside, California, USA.,Division of Biomedical Sciences, University of California Riverside, School of Medicine, Riverside, California, USA.,Cell Molecular and Developmental Biology Program, University of California Riverside, Riverside, California, USA
| |
Collapse
|
24
|
Ferretti MT, Merlini M, Späni C, Gericke C, Schweizer N, Enzmann G, Engelhardt B, Kulic L, Suter T, Nitsch RM. T-cell brain infiltration and immature antigen-presenting cells in transgenic models of Alzheimer's disease-like cerebral amyloidosis. Brain Behav Immun 2016; 54:211-225. [PMID: 26872418 DOI: 10.1016/j.bbi.2016.02.009] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 01/26/2016] [Accepted: 02/09/2016] [Indexed: 11/18/2022] Open
Abstract
Cerebral beta-amyloidosis, one of the pathological hallmarks of Alzheimer's disease (AD), elicits a well-characterised, microglia-mediated local innate immune response. In contrast, it is not clear whether cells of the adaptive immune system, in particular T-cells, react to cerebral amyloidosis in AD. Even though parenchymal T-cells have been described in post-mortem brains of AD patients, it is not known whether infiltrating T-cells are specifically recruited to the extracellular deposits of beta-amyloid, and whether they are locally activated into proliferating, effector cells upon interaction with antigen-presenting cells (APCs). To address these issues we have analysed by confocal microscopy and flow-cytometry the localisation and activation status of both T-cells and APCs in transgenic (tg) mice models of AD-like cerebral amyloidosis. Increased numbers of infiltrating T-cells were found in amyloid-burdened brain regions of tg mice, with concomitant up-regulation of endothelial adhesion molecules ICAM-1 and VCAM-1, compared to non-tg littermates. The infiltrating T-cells in tg brains did not co-localise with amyloid plaques, produced less interferon-gamma than those in controls and did not proliferate locally. Bona-fide dendritic cells were virtually absent from the brain parenchyma of both non-tg and tg mice, and APCs from tg brains showed an immature phenotype, with accumulation of MHC-II in intracellular compartments. These results indicate that cerebral amyloidosis promotes T-cell infiltration but interferes with local antigen presentation and T-cell activation. The inability of the brain immune surveillance to orchestrate a protective immune response to amyloid-beta peptide might contribute to the accumulation of amyloid in the progression of the disease.
Collapse
Affiliation(s)
- M T Ferretti
- Institute for Regenerative Medicine (IREM), University of Zurich, Schlieren, Wagistrasse 12, 8952, Switzerland.
| | - M Merlini
- Institute for Regenerative Medicine (IREM), University of Zurich, Schlieren, Wagistrasse 12, 8952, Switzerland
| | - C Späni
- Institute for Regenerative Medicine (IREM), University of Zurich, Schlieren, Wagistrasse 12, 8952, Switzerland
| | - C Gericke
- Institute for Regenerative Medicine (IREM), University of Zurich, Schlieren, Wagistrasse 12, 8952, Switzerland
| | - N Schweizer
- Neurology, Neuroimmunology and Multiple Sclerosis Research, University Hospital Zurich, Sternwartstrasse 14, 8006 Zurich, Switzerland
| | - G Enzmann
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012 Bern, Switzerland
| | - B Engelhardt
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012 Bern, Switzerland
| | - L Kulic
- Institute for Regenerative Medicine (IREM), University of Zurich, Schlieren, Wagistrasse 12, 8952, Switzerland
| | - T Suter
- Neurology, Neuroimmunology and Multiple Sclerosis Research, University Hospital Zurich, Sternwartstrasse 14, 8006 Zurich, Switzerland
| | - R M Nitsch
- Institute for Regenerative Medicine (IREM), University of Zurich, Schlieren, Wagistrasse 12, 8952, Switzerland
| |
Collapse
|
25
|
Abstract
UNLABELLED Ischemic preconditioning (IPC) is a robust neuroprotective phenomenon whereby brief ischemic exposure confers tolerance to a subsequent ischemic challenge. IPC has not been studied selectively in CNS white matter (WM), although stroke frequently involves WM. We determined whether IPC is present in WM and, if so, its mechanism. We delivered a brief in vivo preconditioning ischemic insult (unilateral common carotid artery ligation) to 12- to 14-week-old mice and determined WM ischemic vulnerability [oxygen-glucose deprivation (OGD)] 72 h later, using acutely isolated optic nerves (CNS WM tracts) from the preconditioned (ipsilateral) and control (contralateral) hemispheres. Functional and structural recovery was assessed by quantitative measurement of compound action potentials (CAPs) and immunofluorescent microscopy. Preconditioned mouse optic nerves (MONs) showed better functional recovery after OGD than the non-preconditioned MONs (31 ± 3 vs 17 ± 3% normalized CAP area, p < 0.01). Preconditioned MONs also showed improved axon integrity and reduced oligodendrocyte injury compared with non-preconditioned MONs. Toll-like receptor-4 (TLR4) and type 1 interferon receptor (IFNAR1), key receptors in innate immune response, are implicated in gray matter preconditioning. Strikingly, IPC-mediated WM protection was abolished in both TLR4(-/-) and IFNAR1(-/-) mice. In addition, IPC-mediated protection in WM was also abolished in IFNAR1(fl/fl) LysM(cre), but not in IFNAR1(fl/fl) control, mice. These findings demonstrated for the first time that IPC was robust in WM, the phenomenon being intrinsic to WM itself. Furthermore, WM IPC was dependent on innate immune cell signaling pathways. Finally, these data demonstrated that microglial-specific expression of IFNAR1 plays an indispensable role in WM IPC. SIGNIFICANCE STATEMENT Ischemic preconditioning (IPC) has been studied predominantly in gray matter, but stroke in humans frequently involves white matter (WM) as well. Here we describe a novel, combined in vivo/ex vivo mouse model to determine whether IPC occurs in WM. It does. Using genetically altered mice, we identified two innate immune cell receptors, Toll-like receptor 4 and type 1 interferon receptor (IFNAR1), that are required for IPC-mediated protection in WM. Furthermore, using microglia-targeted IFNAR1 knockdown, we demonstrate that interferon signaling specifically in microglia is essential for this protection. The discovery of IPC as an intrinsic capability of WM is novel and important. This is also the first in vivo demonstration that cell-type-specific expression of an individual gene plays an indispensable role in IPC-mediated protection.
Collapse
|
26
|
Su W, Aloi MS, Garden GA. MicroRNAs mediating CNS inflammation: Small regulators with powerful potential. Brain Behav Immun 2016; 52:1-8. [PMID: 26148445 PMCID: PMC5030842 DOI: 10.1016/j.bbi.2015.07.003] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 07/02/2015] [Accepted: 07/02/2015] [Indexed: 01/17/2023] Open
Abstract
MicroRNAs (miRNAs) are a family of small non-coding RNAs (~22 nucleotides) that fine-tune protein expression by either silencing mRNA translation or directly targeting gene transcripts for degradation. In the central nervous system (CNS), neuroinflammation plays a critical role in brain injury and neurodegeneration. Increasing evidence supports the involvement of miRNAs as key regulators of neuroinflammation. Altered expression or function of particular miRNAs has been identified in various CNS pathological conditions, including neuroinflammation, neurodegeneration, and autoimmune diseases. Several miRNAs have been shown to play a critical role in the microglia-mediated inflammatory response including miR-155 and miR-146a. In this review, we summarize recent advances in the field of miRNAs associated with CNS inflammation, including our studies of unique inflammatory pathways involving miR-155 and miR-146a. We discuss how specific miRNAs influence microglia activation states in response to inflammatory stimuli, and describe the potential of miRNAs as both biomarkers of inflammation and therapeutic tools for the modulation of microglia behavior.
Collapse
Affiliation(s)
- Wei Su
- Department of Neurology, University of Washington, Seattle, WA, USA
| | - Macarena S Aloi
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Gwenn A Garden
- Department of Neurology, University of Washington, Seattle, WA, USA; Department of Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
27
|
Liu Z, Chen HQ, Huang Y, Qiu YH, Peng YP. Transforming growth factor-β1 acts via TβR-I on microglia to protect against MPP(+)-induced dopaminergic neuronal loss. Brain Behav Immun 2016; 51:131-143. [PMID: 26254549 DOI: 10.1016/j.bbi.2015.08.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 07/07/2015] [Accepted: 08/04/2015] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation is associated with pathogenesis of Parkinson's disease (PD), a neurodegenerative disorder characterized by a progressive loss of dopaminergic (DAergic) neurons within the substantia nigra. Transforming growth factor (TGF)-β1 exerts anti-inflammatory and neuroprotective properties. However, it is unclear if microglia are required for TGF-β1 neuroprotection in PD. Here we used both shRNA and pharmacologic inhibition to determine the role of microglial TGF-β receptor (TβR)-I and its downstream signaling pathways in 1-methyl-4-phenylpyridinium (MPP(+))-induced DAergic neuronal toxicity. As expected, MPP(+) reduced the number of tyrosine hydroxylase (TH)-immunoreactive cells in ventral mesencephalic cell cultures. We found that MPP(+) activated microglia as determined by an upregulation in expression of CD11b and inducible nitric oxide synthase (iNOS), an increase in expression and secretion of tumor necrosis factor (TNF)-α and interleukin (IL)-1β, and a decrease in expression and secretion of the neurotrophic factor, insulin-like growth factor (IGF)-1. Pretreatment with TGF-β1 significantly inhibited all these changes caused by MPP(+). Expression of microglial TβR-I was upregulated by TGF-β1. Silencing of the TβR-I gene in microglia abolished both the neuroprotective and anti-inflammatory properties of TGF-β1. TGF-β1 increased microglial p38 MAPK and Akt phosphorylation, both of which were blocked by the p38 inhibitor SB203580 and the PI3K inhibitor LY294002, respectively. Pretreatment of microglia with either SB203580 or LY294002 impaired the ability of TGF-β1 to inhibit MPP(+)-induced DAergic neuronal loss and microglial activation. These findings establish that TGF-β1 activates TβR-I and its downstream p38 MAPK and PI3K-Akt signaling pathways in microglia to protect against DAergic neuronal loss that characterizes in PD.
Collapse
Affiliation(s)
- Zhan Liu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province 226001, China
| | - Hui-Qiao Chen
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province 226001, China
| | - Yan Huang
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province 226001, China
| | - Yi-Hua Qiu
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province 226001, China.
| | - Yu-Ping Peng
- Department of Physiology, School of Medicine, and Co-innovation Center of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, Jiangsu Province 226001, China.
| |
Collapse
|
28
|
Su W, Kang J, Sopher B, Gillespie J, Aloi MS, Odom GL, Hopkins S, Case A, Wang DB, Chamberlain JS, Garden GA. Recombinant adeno-associated viral (rAAV) vectors mediate efficient gene transduction in cultured neonatal and adult microglia. J Neurochem 2016; 136 Suppl 1:49-62. [PMID: 25708596 PMCID: PMC4547919 DOI: 10.1111/jnc.13081] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 02/13/2015] [Accepted: 02/18/2015] [Indexed: 12/20/2022]
Abstract
Microglia are a specialized population of myeloid cells that mediate CNS innate immune responses. Efforts to identify the cellular and molecular mechanisms that regulate microglia behaviors have been hampered by the lack of effective tools for manipulating gene expression. Cultured microglia are refractory to most chemical and electrical transfection methods, yielding little or no gene delivery and causing toxicity and/or inflammatory activation. Recombinant adeno-associated viral (rAAVs) vectors are non-enveloped, single-stranded DNA vectors commonly used to transduce many primary cell types and tissues. In this study, we evaluated the feasibility and efficiency of utilizing rAAV serotype 2 (rAAV2) to modulate gene expression in cultured microglia. rAAV2 yields high transduction and causes minimal toxicity or inflammatory response in both neonatal and adult microglia. To demonstrate that rAAV transduction can induce functional protein expression, we used rAAV2 expressing Cre recombinase to successfully excise a LoxP-flanked miR155 gene in cultured microglia. We further evaluated rAAV serotypes 5, 6, 8, and 9, and observed that all efficiently transduced cultured microglia to varying degrees of success and caused little or no alteration in inflammatory gene expression. These results provide strong encouragement for the application of rAAV-mediated gene expression in microglia for mechanistic and therapeutic purposes. Neonatal microglia are functionally distinct from adult microglia, although the majority of in vitro studies utilize rodent neonatal microglia cultures because of difficulties of culturing adult cells. In addition, cultured microglia are refractory to most methods for modifying gene expression. Here, we developed a novel protocol for culturing adult microglia and evaluated the feasibility and efficiency of utilizing Recombinant Adeno-Associated Virus (rAAV) to modulate gene expression in cultured microglia.
Collapse
Affiliation(s)
- Wei Su
- Department of Neurology, University of Washington, Seattle, Washington, USA
| | - John Kang
- Department of Neurology, University of Washington, Seattle, Washington, USA
| | - Bryce Sopher
- Department of Neurology, University of Washington, Seattle, Washington, USA
| | - James Gillespie
- Department of Neurology, University of Washington, Seattle, Washington, USA
| | - Macarena S. Aloi
- Department of Pathology, University of Washington, Seattle, Washington, USA
| | - Guy L. Odom
- Department of Neurology, University of Washington, Seattle, Washington, USA
| | - Stephanie Hopkins
- Department of Neurology, University of Washington, Seattle, Washington, USA
| | - Amanda Case
- Department of Neurology, University of Washington, Seattle, Washington, USA
| | - David B. Wang
- Department of Neurological Surgery, University of Washington, Seattle, Washington, USA
| | | | - Gwenn A. Garden
- Department of Neurology, University of Washington, Seattle, Washington, USA
- Department of Pathology, University of Washington, Seattle, Washington, USA
- Center on Human Development and Disability
| |
Collapse
|
29
|
|
30
|
Kielczewski JL, Horai R, Jittayasothorn Y, Chan CC, Caspi RR. Tertiary Lymphoid Tissue Forms in Retinas of Mice with Spontaneous Autoimmune Uveitis and Has Consequences on Visual Function. THE JOURNAL OF IMMUNOLOGY 2015; 196:1013-25. [PMID: 26712943 DOI: 10.4049/jimmunol.1501570] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 11/21/2015] [Indexed: 12/19/2022]
Abstract
During chronic inflammation, tertiary lymphoid tissue (TLT) can form within an inflamed organ, including the CNS. However, little is known about TLT formation in the neuroretina. In a novel spontaneous autoimmune mouse model of uveitis (R161H), we identified well-organized lymphoid aggregates in the retina and examined them for TLT characteristics. Presence of immune cells, tissue-specific markers, and gene expression patterns typically associated with germinal centers and T follicular helper cells were examined using immunohistochemistry and gene analysis of laser capture microdissected retina. Our data revealed the retinal lymphoid structures contained CD4(+) T cells and B cells in well-defined zonal areas that expressed classic germinal center markers, peanut lectin (agglutinin) and GL-7. Gene expression analysis showed upregulation of T follicular helper cell markers, most notably CXCR5 and its ligand CXCL13, and immunohistochemical analysis confirmed CXCR5 expression, typically associated with CD4(+) T follicular helper cells. Highly organized stromal cell networks, a hallmark of organized lymphoid tissue, were also present. Positive staining for phospho-Zap70 in retina-specific T cells indicated CD4(+) T cells were being activated within these lymphoid structures. CD138(+)/B220(+) plasma cells were detected, suggesting the retinal lymphoid aggregates give rise to functional germinal centers, which produce Abs. Interestingly, eyes with lymphoid aggregates exhibited lower inflammatory scores by fundus examination and a slower initial rate of loss of visual function by electroretinography, compared with eyes without these structures. Our findings suggest that the lymphoid aggregates in the retina of R161H mice represent organized TLT, which impact the course of chronic uveitis.
Collapse
Affiliation(s)
- Jennifer L Kielczewski
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892
| | - Reiko Horai
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892
| | - Yingyos Jittayasothorn
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892
| | - Chi-Chao Chan
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892
| | - Rachel R Caspi
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
31
|
Kokiko-Cochran O, Ransohoff L, Veenstra M, Lee S, Saber M, Sikora M, Teknipp R, Xu G, Bemiller S, Wilson G, Crish S, Bhaskar K, Lee YS, Ransohoff RM, Lamb BT. Altered Neuroinflammation and Behavior after Traumatic Brain Injury in a Mouse Model of Alzheimer's Disease. J Neurotrauma 2015; 33:625-40. [PMID: 26414955 DOI: 10.1089/neu.2015.3970] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Traumatic brain injury (TBI) has acute and chronic sequelae, including an increased risk for the development of Alzheimer's disease (AD). TBI-associated neuroinflammation is characterized by activation of brain-resident microglia and infiltration of monocytes; however, recent studies have implicated beta-amyloid as a major manipulator of the inflammatory response. To examine neuroinflammation after TBI and development of AD-like features, these studies examined the effects of TBI in the presence and absence of beta-amyloid. The R1.40 mouse model of cerebral amyloidosis was used, with a focus on time points well before robust AD pathologies. Unexpectedly, in R1.40 mice, the acute neuroinflammatory response to TBI was strikingly muted, with reduced numbers of CNS myeloid cells acquiring a macrophage phenotype and decreased expression of inflammatory cytokines. At chronic time points, macrophage activation substantially declined in non-Tg TBI mice; however, it was relatively unchanged in R1.40 TBI mice. The persistent inflammatory response coincided with significant tissue loss between 3 and 120 days post-injury in R1.40 TBI mice, which was not observed in non-Tg TBI mice. Surprisingly, inflammatory cytokine expression was enhanced in R1.40 mice compared with non-Tg mice, regardless of injury group. Although R1.40 TBI mice demonstrated task-specific deficits in cognition, overall functional recovery was similar to non-Tg TBI mice. These findings suggest that accumulating beta-amyloid leads to an altered post-injury macrophage response at acute and chronic time points. Together, these studies emphasize the role of post-injury neuroinflammation in regulating long-term sequelae after TBI and also support recent studies implicating beta-amyloid as an immunomodulator.
Collapse
Affiliation(s)
| | - Lena Ransohoff
- 1 Department of Neurosciences, Cleveland Clinic , Cleveland, Ohio
| | - Mike Veenstra
- 1 Department of Neurosciences, Cleveland Clinic , Cleveland, Ohio
| | - Sungho Lee
- 1 Department of Neurosciences, Cleveland Clinic , Cleveland, Ohio
| | - Maha Saber
- 1 Department of Neurosciences, Cleveland Clinic , Cleveland, Ohio
| | - Matt Sikora
- 1 Department of Neurosciences, Cleveland Clinic , Cleveland, Ohio
| | - Ryan Teknipp
- 1 Department of Neurosciences, Cleveland Clinic , Cleveland, Ohio
| | - Guixiang Xu
- 1 Department of Neurosciences, Cleveland Clinic , Cleveland, Ohio
| | - Shane Bemiller
- 1 Department of Neurosciences, Cleveland Clinic , Cleveland, Ohio
| | - Gina Wilson
- 2 Department of Pharmaceutical Science, Northeast Ohio Medical University , Rootstown, Ohio
| | - Samuel Crish
- 2 Department of Pharmaceutical Science, Northeast Ohio Medical University , Rootstown, Ohio
| | - Kiran Bhaskar
- 3 Department of Molecular Genetics Microbiology and Neurology, University of New Mexico , Albuquerque New Mexico
| | - Yu-Shang Lee
- 1 Department of Neurosciences, Cleveland Clinic , Cleveland, Ohio
| | | | - Bruce T Lamb
- 1 Department of Neurosciences, Cleveland Clinic , Cleveland, Ohio.,5 Department of Genetics and Department of Neurosciences, Case Western Reserve University , Cleveland, Ohio
| |
Collapse
|
32
|
Weinstein JR, Quan Y, Hanson JF, Colonna L, Iorga M, Honda SI, Shibuya K, Shibuya A, Elkon KB, Möller T. IgM-Dependent Phagocytosis in Microglia Is Mediated by Complement Receptor 3, Not Fcα/μ Receptor. THE JOURNAL OF IMMUNOLOGY 2015; 195:5309-17. [PMID: 26500348 DOI: 10.4049/jimmunol.1401195] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Accepted: 09/29/2015] [Indexed: 12/11/2022]
Abstract
Microglia play an important role in receptor-mediated phagocytosis in the CNS. In brain abscess and other CNS infections, invading bacteria undergo opsonization with Igs or complement. Microglia recognize these opsonized pathogens by Fc or complement receptors triggering phagocytosis. In this study, we investigated the role of Fcα/μR, the less-studied receptor for IgM and IgA, in microglial phagocytosis. We showed that primary microglia, as well as N9 microglial cells, express Fcα/μR. We also showed that anti-Staphylococcus aureus IgM markedly increased the rate of microglial S. aureus phagocytosis. To unequivocally test the role of Fcα/μR in IgM-mediated phagocytosis, we performed experiments in microglia from Fcα/μR(-/-) mice. Surprisingly, we found that IgM-dependent phagocytosis of S. aureus was similar in microglia derived from wild-type or Fcα/μR(-/-) mice. We hypothesized that IgM-dependent activation of complement receptors might contribute to the IgM-mediated increase in phagocytosis. To test this, we used immunologic and genetic inactivation of complement receptor 3 components (CD11b and CD18) as well as C3. IgM-, but not IgG-mediated phagocytosis of S. aureus was reduced in wild-type microglia and macrophages following preincubation with an anti-CD11b blocking Ab. IgM-dependent phagocytosis of S. aureus was also reduced in microglia derived from CD18(-/-) and C3(-/-) mice. Taken together, our findings implicate complement receptor 3 and C3, but not Fcα/μR, in IgM-mediated phagocytosis of S. aureus by microglia.
Collapse
Affiliation(s)
- Jonathan R Weinstein
- Department of Neurology, School of Medicine, University of Washington, Seattle, WA 98195;
| | - Yi Quan
- Department of Neurology, School of Medicine, University of Washington, Seattle, WA 98195
| | - Josiah F Hanson
- Department of Neurology, School of Medicine, University of Washington, Seattle, WA 98195
| | - Lucrezia Colonna
- Division of Rheumatology, Department of Medicine, School of Medicine, University of Washington, Seattle, WA 98195; and
| | - Michael Iorga
- Department of Neurology, School of Medicine, University of Washington, Seattle, WA 98195
| | - Shin-ichiro Honda
- Department of Immunology, Institute of Basic Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Kazuko Shibuya
- Department of Immunology, Institute of Basic Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Akira Shibuya
- Department of Immunology, Institute of Basic Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki 305-8575, Japan
| | - Keith B Elkon
- Division of Rheumatology, Department of Medicine, School of Medicine, University of Washington, Seattle, WA 98195; and
| | - Thomas Möller
- Department of Neurology, School of Medicine, University of Washington, Seattle, WA 98195
| |
Collapse
|
33
|
Quintana E, Fernández A, Velasco P, de Andrés B, Liste I, Sancho D, Gaspar ML, Cano E. DNGR-1(+) dendritic cells are located in meningeal membrane and choroid plexus of the noninjured brain. Glia 2015; 63:2231-48. [PMID: 26184558 DOI: 10.1002/glia.22889] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 06/17/2015] [Accepted: 06/24/2015] [Indexed: 12/14/2022]
Abstract
The role and different origin of brain myeloid cells in the brain is central to understanding how the central nervous system (CNS) responds to injury. C-type lectin receptor family 9, member A (DNGR-1/CLEC9A) is a marker of specific DC subsets that share functional similarities, such as CD8α(+) DCs in lymphoid tissues and CD103(+) CD11b(low) DCs in peripheral tissues. Here, we analyzed the presence of DNGR-1 in DCs present in the mouse brain (bDCs). Dngr-1/Clec9a mRNA is expressed mainly in the meningeal membranes and choroid plexus (m/Ch), and its expression is enhanced by fms-like tyrosine kinase 3 ligand (Flt3L), a cytokine involved in DC homeostasis. Using Clec9a(egfp/egfp) mice, we show that Flt3L induces accumulation of DNGR-1-EGFP(+) cells in the brain m/Ch. Most of these cells also express major histocompatibility complex class II (MHCII) molecules. We also observed an increase in specific markers of cDC CD8α+ cells such as Batf-3 and Irf-8, but not of costimulatory molecules such as Cd80 and Cd86, indicating an immature phenotype for these bDCs in the noninjured brain. The presence of DNGR-1 in the brain provides a potential marker for the study of this specific brain cell subset. Knowledge and targeting of brain antigen presenting cells (APCs) has implications for the fight against brain diseases such as neuroinflammation-based neurodegenerative diseases, microbe-induced encephalitis, and brain tumors such as gliomas.
Collapse
Affiliation(s)
- Elena Quintana
- Neuroinflammation Unit. Unidad Funcional De Investigación De Enfermedades Crónicas, Instituto De Salud Carlos III, Majadahonda, Madrid, Spain
| | - Andrés Fernández
- Neuroinflammation Unit. Unidad Funcional De Investigación De Enfermedades Crónicas, Instituto De Salud Carlos III, Majadahonda, Madrid, Spain
| | - Patricia Velasco
- Neuroinflammation Unit. Unidad Funcional De Investigación De Enfermedades Crónicas, Instituto De Salud Carlos III, Majadahonda, Madrid, Spain
| | - Belén de Andrés
- Department of Immunology, Centro Nacional De Microbiología, Instituto De Salud Carlos III, Madrid, Spain
| | - Isabel Liste
- Instituto De Salud Carlos III, Neural Regeneration Unit. Unidad Funcional De Investigación De Enfermedades Crónicas, Majadahonda, Madrid, Spain
| | - David Sancho
- Centro Nacional De Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - María Luisa Gaspar
- Department of Immunology, Centro Nacional De Microbiología, Instituto De Salud Carlos III, Madrid, Spain
| | - Eva Cano
- Neuroinflammation Unit. Unidad Funcional De Investigación De Enfermedades Crónicas, Instituto De Salud Carlos III, Majadahonda, Madrid, Spain
| |
Collapse
|
34
|
Ji R, Meng L, Li Q, Lu Q. TAM receptor deficiency affects adult hippocampal neurogenesis. Metab Brain Dis 2015; 30:633-44. [PMID: 25487541 PMCID: PMC4414696 DOI: 10.1007/s11011-014-9636-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 11/17/2014] [Indexed: 10/24/2022]
Abstract
The Tyro3, Axl and Mertk (TAM) subfamily of receptor protein tyrosine kinases functions in cell growth, differentiation, survival, and most recently found, in the regulation of immune responses and phagocytosis. All three receptors and their ligands, Gas6 (growth arrest-specific gene 6) and protein S, are expressed in the central nervous system (CNS). TAM receptors play pivotal roles in adult hippocampal neurogenesis. Loss of these receptors causes a comprised neurogenesis in the dentate gyrus of adult hippocampus. TAM receptors have a negative regulatory effect on microglia and peripheral antigen-presenting cells, and play a critical role in preventing overproduction of pro-inflammatory cytokines detrimental to the proliferation, differentiation, and survival of adult neuronal stem cells (NSCs). Besides, these receptors also play an intrinsic trophic function in supporting NSC survival, proliferation, and differentiation into immature neurons. All these events collectively ensure a sustained neurogenesis in adult hippocampus.
Collapse
Affiliation(s)
- Rui Ji
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Lingbin Meng
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Qiutang Li
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Qingxian Lu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, KY 40202, USA
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
35
|
Legroux L, Pittet CL, Beauseigle D, Deblois G, Prat A, Arbour N. An optimized method to process mouse CNS to simultaneously analyze neural cells and leukocytes by flow cytometry. J Neurosci Methods 2015; 247:23-31. [DOI: 10.1016/j.jneumeth.2015.03.021] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 03/03/2015] [Accepted: 03/18/2015] [Indexed: 11/24/2022]
|
36
|
Ritzel RM, Patel AR, Pan S, Crapser J, Hammond M, Jellison E, McCullough LD. Age- and location-related changes in microglial function. Neurobiol Aging 2015; 36:2153-63. [PMID: 25816747 DOI: 10.1016/j.neurobiolaging.2015.02.016] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Revised: 02/08/2015] [Accepted: 02/13/2015] [Indexed: 11/26/2022]
Abstract
Inflammation in the central nervous system (CNS) is primarily regulated by microglia. No longer considered a homogenous population, microglia display a high degree of heterogeneity, immunological diversity and regional variability in function. Given their low rate of self-renewal, the microenvironment in which microglia reside may play an important role in microglial senescence. This study examines age-related changes in microglia in the brain and spinal cord. Using ex-vivo flow cytometry analyses, functional assays were performed to assess changes in microglial morphology, oxidative stress, cytokine production, and phagocytic activity with age in both the brain and spinal cord. The regional CNS environment had a significant effect on microglial activity with age. Blood-CNS barrier permeability was greater in the aging spinal cord compared with aging brain; this was associated with increased tissue cytokine levels. Aged microglia had deficits in phagocytosis at baseline and after stimulus-induced activation. The identification of age-specific, high scatter microglia together with the use of ex-vivo functional analyses provides the first functional characterization of senescent microglia. Age and regional-specificity of CNS disease should be taken into consideration when developing immune-modulatory treatments.
Collapse
Affiliation(s)
- Rodney M Ritzel
- Department of Neurology, University of Connecticut Health Center, Farmington, CT, USA
| | - Anita R Patel
- Department of Neurology, University of Connecticut Health Center, Farmington, CT, USA
| | - Sarah Pan
- Department of Neurology, University of Connecticut Health Center, Farmington, CT, USA
| | - Joshua Crapser
- Department of Neurology, University of Connecticut Health Center, Farmington, CT, USA
| | - Matt Hammond
- Department of Neurology, University of Connecticut Health Center, Farmington, CT, USA
| | - Evan Jellison
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, USA
| | - Louise D McCullough
- Department of Neurology, University of Connecticut Health Center, Farmington, CT, USA.
| |
Collapse
|
37
|
Zager A, Peron JP, Mennecier G, Rodrigues SC, Aloia TP, Palermo-Neto J. Maternal immune activation in late gestation increases neuroinflammation and aggravates experimental autoimmune encephalomyelitis in the offspring. Brain Behav Immun 2015; 43:159-71. [PMID: 25108214 DOI: 10.1016/j.bbi.2014.07.021] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 07/24/2014] [Accepted: 07/29/2014] [Indexed: 01/09/2023] Open
Abstract
Multiple sclerosis (MS) is characterized by an autoimmune response against myelin antigens driven by autoreactive T cells. Several lines of evidence indicate that environmental factors, such as previous infection, can influence and trigger autoimmune responses. However, the importance of the gestational period, particularly under inflammatory conditions, on the modulation of MS and related neuroinflammation by the offspring is unknown. This study aimed to evaluate the impact of prenatal exposure to lipopolysaccharide (LPS) during late gestation on the neuroinflammatory response in primary mixed glial cultures and on the progression of experimental autoimmune encephalomyelitis (EAE, an animal model of MS) in the offspring. LPS (Escherichia coli 0127:B8, 120μg/kg) was administered intraperitoneally to pregnant C57BL/6J mice on gestational day 17, and the offspring were assigned to two experiments: (1) mixed glial cultures generated using the brain of neonates, stimulated in vitro with LPS, and (2) adult offspring immunized with MOG35-55. The EAE clinical symptoms were followed for 30days. Different sets of animals were sacrificed either during the onset (7days post-immunization [p.i.]), when spleen and lymph nodes were collected, or the peak of disease (20days p.i.), when CNS were collected for flow cytometry, cytokine production, and protein/mRNA-expression analysis. The primary CNS cultures from the LPS-treated group produced exaggerated amounts of IL-6, IL-1β and nitrites after in vitro stimulus, while IL-10 production was lowered compared to the data of the control group. Prenatal exposure to LPS worsened EAE disease severity in adult offspring, and this worsening was linked to increased CNS-infiltrating macrophages, Th1 cells and Th17 cells at the peak of EAE severity; additionally, exacerbated gliosis was evidenced in microglia (MHC II) and astrocytes (GFAP protein level and immunoreactivity). The IL-2, IL-6 and IL-17 levels in the spleen and lymph nodes were increased in the offspring of the LPS-exposed dams. Our results indicate that maternal immune activation during late gestation predispose the offspring to increased neuroinflammation and potentiate the autoimmune response and clinical manifestation of EAE.
Collapse
Affiliation(s)
- Adriano Zager
- Neuroimmunomodulation Research Group, Department of Pathology, School of Veterinary Medicine, University of São Paulo, São Paulo, Brazil.
| | - Jean Pierre Peron
- Neuroimmune Interactions Laboratory, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Gregory Mennecier
- Neuroimmunomodulation Research Group, Department of Pathology, School of Veterinary Medicine, University of São Paulo, São Paulo, Brazil
| | - Sandra C Rodrigues
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Thiago P Aloia
- Neuroimmunomodulation Research Group, Department of Pathology, School of Veterinary Medicine, University of São Paulo, São Paulo, Brazil
| | - João Palermo-Neto
- Neuroimmunomodulation Research Group, Department of Pathology, School of Veterinary Medicine, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
38
|
Chen CC, Hung TH, Lee CY, Wang LF, Wu CH, Ke CH, Chen SF. Berberine protects against neuronal damage via suppression of glia-mediated inflammation in traumatic brain injury. PLoS One 2014; 9:e115694. [PMID: 25546475 PMCID: PMC4278716 DOI: 10.1371/journal.pone.0115694] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Accepted: 11/26/2014] [Indexed: 01/26/2023] Open
Abstract
Traumatic brain injury (TBI) triggers a series of neuroinflammatory processes that contribute to evolution of neuronal injury. The present study investigated the neuroprotective effects and anti-inflammatory actions of berberine, an isoquinoline alkaloid, in both in vitro and in vivo TBI models. Mice subjected to controlled cortical impact injury were injected with berberine (10 mg·kg−1) or vehicle 10 min after injury. In addition to behavioral studies and histology analysis, blood-brain barrier (BBB) permeability and brain water content were determined. Expression of PI3K/Akt and Erk signaling and inflammatory mediators were also analyzed. The protective effect of berberine was also investigated in cultured neurons either subjected to stretch injury or exposed to conditioned media with activated microglia. Berberine significantly attenuated functional deficits and brain damage associated with TBI up to day 28 post-injury. Berberine also reduced neuronal death, apoptosis, BBB permeability, and brain edema at day 1 post-injury. These changes coincided with a marked reduction in leukocyte infiltration, microglial activation, matrix metalloproteinase-9 activity, and expression of inflammatory mediators. Berberine had no effect on Akt or Erk 1/2 phosphorylation. In mixed glial cultures, berberine reduced TLR4/MyD88/NF-κB signaling. Berberine also attenuated neuronal death induced by microglial conditioned media; however, it did not directly protect cultured neurons subjected to stretch injury. Moreover, administration of berberine at 3 h post-injury also reduced TBI-induced neuronal damage, apoptosis and inflammation in vivo. Berberine reduces TBI-induced brain damage by limiting the production of inflammatory mediators by glial cells, rather than by a direct neuroprotective effect.
Collapse
Affiliation(s)
- Chien-Cheng Chen
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
| | - Tai-Ho Hung
- Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Taipei and College of Medicine, Chang Gung University, Taipei, Taiwan, Republic of China
| | - Chao Yu Lee
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
| | - Liang-Fei Wang
- Departments of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, Republic of China
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Chun-Hu Wu
- Departments of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, Republic of China
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Chia-Hua Ke
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
- Departments of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Szu-Fu Chen
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan, Republic of China
- Departments of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan, Republic of China
- * E-mail:
| |
Collapse
|
39
|
Ballesteros I, Cuartero MI, Moraga A, de la Parra J, Lizasoain I, Moro MÁ. Stereological and flow cytometry characterization of leukocyte subpopulations in models of transient or permanent cerebral ischemia. J Vis Exp 2014:52031. [PMID: 25590380 PMCID: PMC4354492 DOI: 10.3791/52031] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Microglia activation, as well as extravasation of haematogenous macrophages and neutrophils, is believed to play a pivotal role in brain injury after stroke. These myeloid cell subpopulations can display different phenotypes and functions and need to be distinguished and characterized to study their regulation and contribution to tissue damage. This protocol provides two different methodologies for brain immune cell characterization: a precise stereological approach and a flow cytometric analysis. The stereological approach is based on the optical fractionator method, which calculates the total number of cells in an area of interest (infarcted brain) estimated by a systematic random sampling. The second characterization approach provides a simple way to isolate brain leukocyte suspensions and to characterize them by flow cytometry, allowing for the characterization of microglia, infiltrated monocytes and neutrophils of the ischemic tissue. In addition, it also details a cerebral ischemia model in mice that exclusively affects brain cortex, generating highly reproducible infarcts with a low rate of mortality, and the procedure for histological brain processing to characterize infarct volume by the Cavalieri method.
Collapse
Affiliation(s)
- Iván Ballesteros
- Unidad de Investigacón Neurovascular, Departmento de Farmacología, Falcultad de Medicina, Universidad Complutense de Madrid y Instituto de Investigación Hospital 12 de Octubre, Madrid
| | - María Isabel Cuartero
- Unidad de Investigacón Neurovascular, Departmento de Farmacología, Falcultad de Medicina, Universidad Complutense de Madrid y Instituto de Investigación Hospital 12 de Octubre, Madrid;
| | - Ana Moraga
- Unidad de Investigacón Neurovascular, Departmento de Farmacología, Falcultad de Medicina, Universidad Complutense de Madrid y Instituto de Investigación Hospital 12 de Octubre, Madrid
| | - Juan de la Parra
- Unidad de Investigacón Neurovascular, Departmento de Farmacología, Falcultad de Medicina, Universidad Complutense de Madrid y Instituto de Investigación Hospital 12 de Octubre, Madrid
| | - Ignacio Lizasoain
- Unidad de Investigacón Neurovascular, Departmento de Farmacología, Falcultad de Medicina, Universidad Complutense de Madrid y Instituto de Investigación Hospital 12 de Octubre, Madrid
| | - María Ángeles Moro
- Unidad de Investigacón Neurovascular, Departmento de Farmacología, Falcultad de Medicina, Universidad Complutense de Madrid y Instituto de Investigación Hospital 12 de Octubre, Madrid
| |
Collapse
|
40
|
Brain APCs including microglia are only differential and positional polymorphs. Ann Neurosci 2014; 17:191-9. [PMID: 25205905 PMCID: PMC4117011 DOI: 10.5214/ans.0972.7531.1017410] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 09/22/2010] [Accepted: 10/04/2010] [Indexed: 12/23/2022] Open
Abstract
The antigen presentation to lymphocytes in brain occurs in two steps. Initially it happens at perivascular spaces by perivascular microglia/macrophage population and finally at the site of inflammation deep into brain parenchyma by the resident microglia. But recent evidence challanges the existing notion of involvement of distinct and different cells at these sites. Studies have shown that many of these microglial cells show dendritic cell phenotype in pathogenic and cytokine driven environment. Different subsets of the cell show wide range of myeloid lineage functions indicating a pre-differentiated status of the cell. Monocytic CD34(+)/B220(+) precursor cells have been transformed to microglial cells in vitro and transplantation of these cells show Iba-1 or F4/80 positivity with microglial phenotypes in vivo in adults. Even they can be converted into dendritic cell like forms. The interconvertability among macrophage-microglia-dendritic cells and final effector maturation according to the microenvironmental cues indicates existence of a pre-mature myeloid cell population concerned with antigen presentation and related functions in brain. With the substantial recent observation this article sketches the idea that brain APCs appearing as macrophage/microglia/DC like forms are derivatives of the same stock in response to their position and microenvironment. And also microglia is never any distinct cells, both in neonatal stage and adults.
Collapse
|
41
|
Immig K, Gericke M, Menzel F, Merz F, Krueger M, Schiefenhövel F, Lösche A, Jäger K, Hanisch UK, Biber K, Bechmann I. CD11c-positive cells from brain, spleen, lung, and liver exhibit site-specific immune phenotypes and plastically adapt to new environments. Glia 2014; 63:611-25. [DOI: 10.1002/glia.22771] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 11/06/2014] [Indexed: 12/23/2022]
Affiliation(s)
- Kerstin Immig
- Institute of Anatomy, Leipzig University; Leipzig Germany
| | - Martin Gericke
- Institute of Anatomy, Leipzig University; Leipzig Germany
| | | | - Felicitas Merz
- Institute of Anatomy, Leipzig University; Leipzig Germany
| | - Martin Krueger
- Institute of Anatomy, Leipzig University; Leipzig Germany
| | | | - Andreas Lösche
- IZKF-FACS-Core Unit; Leipzig University; Leipzig Germany
| | - Kathrin Jäger
- IZKF-FACS-Core Unit; Leipzig University; Leipzig Germany
| | | | - Knut Biber
- Department of Psychiatry and Psychotherapy; Section of Molecular Psychiatry, University of Freiburg; Freiburg Germany
| | - Ingo Bechmann
- Institute of Anatomy, Leipzig University; Leipzig Germany
| |
Collapse
|
42
|
Alvarado R, O'Brien B, Tanaka A, Dalton JP, Donnelly S. A parasitic helminth-derived peptide that targets the macrophage lysosome is a novel therapeutic option for autoimmune disease. Immunobiology 2014; 220:262-9. [PMID: 25466586 DOI: 10.1016/j.imbio.2014.11.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 11/06/2014] [Accepted: 11/07/2014] [Indexed: 12/24/2022]
Abstract
Parasitic worms (helminths) reside in their mammalian hosts for many years. This is attributable, in part, to their ability to skew the host's immune system away from pro-inflammatory responses and towards anti-inflammatory or regulatory responses. This immune modulatory ability ensures helminth longevity within the host, while simultaneously minimises tissue destruction for the host. The molecules that the parasite releases clearly exert potent immune-modulatory actions, which could be exploited clinically, for example in the prophylactic and therapeutic treatment of pro-inflammatory and autoimmune diseases. We have identified a novel family of immune-modulatory proteins, termed helminth defence molecules (HDMs), which are secreted by several medically important helminth parasites. These HDMs share biochemical and structural characteristics with mammalian cathelicidin-like host defence peptides (HDPs), which are significant components of the innate immune system. Like their mammalian counterparts, parasite HDMs block the activation of macrophages via toll like receptor (TLR) 4 signalling, however HDMs are significantly less cytotoxic than HDPs. HDMs can traverse the cell membrane of macrophages and enter the endolysosomal system where they reduce the acidification of lysosomal compartments by inhibiting vacuolar (v)-ATPase activity. In doing this, HDMs can modulate critical cellular functions, such as cytokine secretion and antigen processing/presentation. Here, we review the role of macrophages, specifically their lysosomal mediated activities, in the initiation and perpetuation of pro-inflammatory immune responses. We also discuss the potential of helminth defence molecules (HDMs) as therapeutics to counteract the pro-inflammatory responses underlying autoimmune disease. Given the current lack of effective, non-cytotoxic treatment options to limit the progression of autoimmune pathologies, HDMs open novel treatment avenues.
Collapse
Affiliation(s)
- Raquel Alvarado
- School of Medical and Molecular Biosciences, University of Technology, Sydney, Sydney, NSW, Australia
| | - Bronwyn O'Brien
- School of Medical and Molecular Biosciences, University of Technology, Sydney, Sydney, NSW, Australia
| | - Akane Tanaka
- School of Medical and Molecular Biosciences, University of Technology, Sydney, Sydney, NSW, Australia
| | - John P Dalton
- School of Biological Sciences, Medical Biology Centre, Queen's University, Belfast, Belfast, Northern Ireland, UK
| | - Sheila Donnelly
- School of Medical and Molecular Biosciences, University of Technology, Sydney, Sydney, NSW, Australia; The i3 Institute, University of Technology, Sydney, Sydney, NSW, Australia.
| |
Collapse
|
43
|
The contribution of immune and glial cell types in experimental autoimmune encephalomyelitis and multiple sclerosis. Mult Scler Int 2014; 2014:285245. [PMID: 25374694 PMCID: PMC4211315 DOI: 10.1155/2014/285245] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 09/27/2014] [Accepted: 09/27/2014] [Indexed: 12/19/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system characterised by widespread areas of focal demyelination. Its aetiology and pathogenesis remain unclear despite substantial insights gained through studies of animal models, most notably experimental autoimmune encephalomyelitis (EAE). MS is widely believed to be immune-mediated and pathologically attributable to myelin-specific autoreactive CD4+ T cells. In recent years, MS research has expanded beyond its focus on CD4+ T cells to recognise the contributions of multiple immune and glial cell types to the development, progression, and amelioration of the disease. This review summarises evidence of T and B lymphocyte, natural killer cell, macrophage/microglial, astrocytic, and oligodendroglial involvement in both EAE and MS and the intercommunication and influence of each cell subset in the inflammatory process. Despite important advances in the understanding of the involvement of these cell types in MS, many questions still remain regarding the various subsets within each cell population and their exact contribution to different stages of the disease.
Collapse
|
44
|
Virgili N, Mancera P, Chanvillard C, Wegner A, Wappenhans B, Rodríguez MJ, Infante-Duarte C, Espinosa-Parrilla JF, Pugliese M. Diazoxide attenuates autoimmune encephalomyelitis and modulates lymphocyte proliferation and dendritic cell functionality. J Neuroimmune Pharmacol 2014; 9:558-68. [PMID: 24939091 DOI: 10.1007/s11481-014-9551-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 06/09/2014] [Indexed: 12/14/2022]
Abstract
Activation of mitochondrial ATP-sensitive potassium (KATP) channels is postulated as an effective mechanism to confer cardio and neuroprotection, especially in situations associated to oxidative stress. Pharmacological activation of these channels inhibits glia-mediated neuroinflammation. In this way, diazoxide, an old-known mitochondrial KATP channel opener, has been proposed as an effective and safe treatment for different neurodegenerative diseases, demonstrating efficacy in different animal models, including the experimental autoimmune encephalomyelitis (EAE), an animal model for Multiple Sclerosis. Although neuroprotection and modulation of glial reactivity could alone explain the positive effects of diazoxide administration in EAE mice, little is known of its effects on the immune system and the autoimmune reaction that triggers the EAE pathology. The aim of the present work was to study the effects of diazoxide in autoimmune key processes related with EAE, such as antigen presentation and lymphocyte activation and proliferation. Results show that, although diazoxide treatment inhibited in vitro and ex-vivo lymphocyte proliferation from whole splenocytes it had no effect in isolated CD4(+) T cells. In any case, treatment had no impact in lymphocyte activation. Diazoxide can also slightly decrease CD83, CD80, CD86 and major histocompatibility complex class II expression in cultured dendritic cells, demonstrating a possible role in modulating antigen presentation. Taken together, our results indicate that diazoxide treatment attenuates autoimmune encephalomyelitis pathology without immunosuppressive effect.
Collapse
Affiliation(s)
- N Virgili
- Neurotec Pharma S.L., Bioincubadora PCB-Santander, Parc Científic de Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Treatment with a hybrid between the synapsin ABC domains and the B subunit of E. coli heat-labile toxin reduces frequency of proinflammatory cells and cytokines in the central nervous system of rats with EAE. Neuroscience 2014; 277:217-28. [DOI: 10.1016/j.neuroscience.2014.06.070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 06/27/2014] [Accepted: 06/28/2014] [Indexed: 01/07/2023]
|
46
|
Ferrazzano P, Chanana V, Uluc K, Fidan E, Akture E, Kintner DB, Cengiz P, Sun D. Age-dependent microglial activation in immature brains after hypoxia- ischemia. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2014; 12:338-49. [PMID: 23469850 DOI: 10.2174/1871527311312030007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 09/13/2012] [Accepted: 09/22/2012] [Indexed: 12/20/2022]
Abstract
In the present study, we tested whether the ongoing differentiation of microglia in the immature brain results in more robust microglial activation and pro-inflammatory responses than juvenile brains following hypoxia-ischemia (HI). Under normoxic conditions, microglial activation profiles were assessed in postnatal day 9 and postnatal day 30 mice (P9 and P30) by analyzing relative expression levels of CD45 in CD11b+/CD45+ microglia/macrophages. Flow cytometry analysis revealed that the hippocampi of P9 and P30 brains exhibited higher levels of CD45 expression in CD11b+/CD45+ cells than in the cortex and striatum. In response to HI, there was an early increase in number of CD11b+/CD45+ microglia/macrophages in the ipsilateral hippocampus of P9 mice. These cells transformed from a "ramified" to an "amoeboid" morphology in the CA1 region, which was accompanied by a loss of microtubule-associated protein 2 immunostaining in this brain region. The peak response of microglial activation in the ipsilateral hippocampus of P9 mice occurred on day 2 post-HI, which was in contrast to a delayed and persistent microglial activation in the cortex and striatum (peak on day 9 post-HI). P9 brains demonstrated a 2-3 fold greater increase in microglia counts than P30 brains in each region (hippocampus, cortex, and striatum) during day 1-17 post-HI. P9 brains also showed more robust expression of pro-inflammatory cytokines (tumor necrosis factor-alpha, interleukin-1β) than P30 brains. Taken together, compared to P30 mice, P9 mice demonstrated differences in microglial activation and pro-inflammatory responses after HI, which may be important in brain damage and tissue repair.
Collapse
Affiliation(s)
- Peter Ferrazzano
- Department of Pediatrics, University of Wisconsin Medical School, 1500 Highland Ave., Madison, WI 53705, USA.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Caldeira C, Oliveira AF, Cunha C, Vaz AR, Falcão AS, Fernandes A, Brites D. Microglia change from a reactive to an age-like phenotype with the time in culture. Front Cell Neurosci 2014; 8:152. [PMID: 24917789 PMCID: PMC4040822 DOI: 10.3389/fncel.2014.00152] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 05/13/2014] [Indexed: 01/25/2023] Open
Abstract
Age-related neurodegenerative diseases have been associated with chronic neuroinflammation and microglia activation. However, cumulative evidence supports that inflammation only occurs at an early stage once microglia change the endogenous characteristics with aging and switch to irresponsive/senescent and dystrophic phenotypes with disease progression. Thus, it will be important to have the means to assess the role of reactive and aged microglia when studying advanced brain neurodegeneration processes and age-associated related disorders. Yet, most studies are done with microglia from neonates since there are no adequate means to isolate degenerating microglia for experimentation. Indeed, only a few studies report microglia isolation from aged animals, using either short-term cultures or high concentrations of mitogens in the medium, which trigger microglia reactivity. The purpose of this study was to develop an experimental process to naturally age microglia after isolation from neonatal mice and to characterize the cultured cells at 2 days in vitro (DIV), 10 DIV, and 16 DIV. We found that 2 DIV (young) microglia had predominant amoeboid morphology and markers of stressed/reactive phenotype. In contrast, 16 DIV (aged) microglia evidenced ramified morphology and increased matrix metalloproteinase (MMP)-2 activation, as well as reduced MMP-9, glutamate release and nuclear factor kappa-B activation, in parallel with decreased expression of Toll-like receptor (TLR)-2 and TLR-4, capacity to migrate and phagocytose. These findings together with the reduced expression of microRNA (miR)-124, and miR-155, decreased autophagy, enhanced senescence associated beta-galactosidase activity and elevated miR-146a expression, are suggestive that 16 DIV cells mainly correspond to irresponsive/senescent microglia. Data indicate that the model represent an opportunity to understand and control microglial aging, as well as to explore strategies to recover microglia surveillance function.
Collapse
Affiliation(s)
- Cláudia Caldeira
- Research Institute for Medicines - iMed.ULisboa, Faculdade de Farmácia, Universidade de Lisboa Lisboa, Portugal ; Centro de Investigação Interdisciplinar Egas Moniz, Egas Moniz - Cooperativa de Ensino Superior, CRL, Campus Universitário Monte de Caparica, Portugal
| | - Ana F Oliveira
- Research Institute for Medicines - iMed.ULisboa, Faculdade de Farmácia, Universidade de Lisboa Lisboa, Portugal
| | - Carolina Cunha
- Research Institute for Medicines - iMed.ULisboa, Faculdade de Farmácia, Universidade de Lisboa Lisboa, Portugal
| | - Ana R Vaz
- Research Institute for Medicines - iMed.ULisboa, Faculdade de Farmácia, Universidade de Lisboa Lisboa, Portugal ; Department of Biochemistry and Human Biology, Faculdade de Farmácia, Universidade de Lisboa Lisboa, Portugal
| | - Ana S Falcão
- Research Institute for Medicines - iMed.ULisboa, Faculdade de Farmácia, Universidade de Lisboa Lisboa, Portugal ; Department of Biochemistry and Human Biology, Faculdade de Farmácia, Universidade de Lisboa Lisboa, Portugal
| | - Adelaide Fernandes
- Research Institute for Medicines - iMed.ULisboa, Faculdade de Farmácia, Universidade de Lisboa Lisboa, Portugal ; Department of Biochemistry and Human Biology, Faculdade de Farmácia, Universidade de Lisboa Lisboa, Portugal
| | - Dora Brites
- Research Institute for Medicines - iMed.ULisboa, Faculdade de Farmácia, Universidade de Lisboa Lisboa, Portugal ; Department of Biochemistry and Human Biology, Faculdade de Farmácia, Universidade de Lisboa Lisboa, Portugal
| |
Collapse
|
48
|
Mendez-Huergo SP, Maller SM, Farez MF, Mariño K, Correale J, Rabinovich GA. Integration of lectin–glycan recognition systems and immune cell networks in CNS inflammation. Cytokine Growth Factor Rev 2014; 25:247-55. [DOI: 10.1016/j.cytogfr.2014.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 02/24/2014] [Indexed: 12/26/2022]
|
49
|
|
50
|
Wlodarczyk A, Løbner M, Cédile O, Owens T. Comparison of microglia and infiltrating CD11c⁺ cells as antigen presenting cells for T cell proliferation and cytokine response. J Neuroinflammation 2014; 11:57. [PMID: 24666681 PMCID: PMC3987647 DOI: 10.1186/1742-2094-11-57] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 03/09/2014] [Indexed: 12/25/2022] Open
Abstract
Background Tissue-resident antigen-presenting cells (APC) exert a major influence on the local immune environment. Microglia are resident myeloid cells in the central nervous system (CNS), deriving from early post-embryonic precursors, distinct from adult hematopoietic lineages. Dendritic cells (DC) and macrophages infiltrate the CNS during experimental autoimmune encephalomyelitis (EAE). Microglia are not considered to be as effective APC as DC or macrophages. Methods In this work we compared the antigen presenting capacity of CD11c+ and CD11c− microglia subsets with infiltrating CD11c+ APC, which include DC. The microglial subpopulations (CD11c− CD45dim CD11b+ and CD11c+ CD45dim CD11b+) as well as infiltrating CD11c+ CD45high cells were sorted from CNS of C57BL/6 mice with EAE. Sorted cells were characterised by flow cytometry for surface phenotype and by quantitative real-time PCR for cytokine expression. They were co-cultured with primed T cells to measure induction of T cell proliferation and cytokine response. Results The number of CD11c+ microglia cells increased dramatically in EAE. They expressed equivalent levels of major histocompatibility complex and co-stimulatory ligands CD80 and CD86 as those expressed by CD11c+ cells infiltrating from blood. CD11c+ microglia differed significantly from blood-derived CD11c+ cells in their cytokine profile, expressing no detectable IL-6, IL-12 or IL-23, and low levels of IL-1β. By contrast, CD11c− microglia expressed low but detectable levels of all these cytokines. Transforming growth factor β expression was similar in all three populations. Although CNS-resident and blood-derived CD11c+ cells showed equivalent ability to induce proliferation of myelin oligodendrocyte glycoprotein-immunised CD4+ T cells, CD11c+ microglia induced lower levels of T helper (Th)1 and Th17 cytokines, and did not induce Th2 cytokines. Conclusions Our findings show distinct subtypes of APC in the inflamed CNS, with a hierarchy of functional competence for induction of CD4+ T cell responses.
Collapse
Affiliation(s)
| | | | | | - Trevor Owens
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, J,B, Winsløwsvej 25, Odense, DK 5000, Denmark.
| |
Collapse
|