451
|
Pires-Afonso Y, Niclou SP, Michelucci A. Revealing and Harnessing Tumour-Associated Microglia/Macrophage Heterogeneity in Glioblastoma. Int J Mol Sci 2020; 21:E689. [PMID: 31973030 PMCID: PMC7037936 DOI: 10.3390/ijms21030689] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/14/2020] [Accepted: 01/20/2020] [Indexed: 12/14/2022] Open
Abstract
Abstract: Cancer heterogeneity and progression are subject to complex interactions between neoplastic cells and their microenvironment, including the immune system. Although glioblastomas (GBMs) are classified as 'cold tumours' with very little lymphocyte infiltration, they can contain up to 30-40% of tumour-associated macrophages, reported to contribute to a supportive microenvironment that facilitates tumour proliferation, survival and migration. In GBM, tumour-associated macrophages comprise either resident parenchymal microglia, perivascular macrophages or peripheral monocyte-derived cells. They are recruited by GBMs and in turn release growth factors and cytokines that affect the tumour. Notably, tumour-associated microglia/macrophages (TAMs) acquire different expression programs, which shape the tumour microenvironment and contribute to GBM molecular subtyping. Further, emerging evidence highlights that TAM programs may adapt to specific tumour features and landscapes. Here, we review key evidence describing TAM transcriptional and functional heterogeneity in GBM. We propose that unravelling the intricate complexity and diversity of the myeloid compartment as well as understanding how different TAM subsets may affect tumour progression will possibly pave the way to new immune therapeutic avenues for GBM patients.
Collapse
Affiliation(s)
- Yolanda Pires-Afonso
- Neuro-Immunology Group, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg;
- Doctoral School of Science and Technology, University of Luxembourg, L-4365 Esch-sur-Alzette, Luxembourg
| | - Simone P. Niclou
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg;
- Department of Biomedicine, University of Bergen, N-5007 Bergen, Norway
| | - Alessandro Michelucci
- Neuro-Immunology Group, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg;
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4365 Esch-sur-Alzette, Luxembourg
| |
Collapse
|
452
|
Adhikaree J, Moreno-Vicente J, Kaur AP, Jackson AM, Patel PM. Resistance Mechanisms and Barriers to Successful Immunotherapy for Treating Glioblastoma. Cells 2020; 9:E263. [PMID: 31973059 PMCID: PMC7072315 DOI: 10.3390/cells9020263] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 12/22/2022] Open
Abstract
Glioblastoma (GBM) is inevitably refractory to surgery and chemoradiation. The hope for immunotherapy has yet to be realised in the treatment of GBM. Immune checkpoint blockade antibodies, particularly those targeting the Programme death 1 (PD-1)/PD-1 ligand (PD-L1) pathway, have improved the prognosis in a range of cancers. However, its use in combination with chemoradiation or as monotherapy has proved unsuccessful in treating GBM. This review focuses on our current knowledge of barriers to immunotherapy success in treating GBM, such as diminished pre-existing anti-tumour immunity represented by low levels of PD-L1 expression, low tumour mutational burden and a severely exhausted T-cell tumour infiltrate. Likewise, systemic T-cell immunosuppression is seen driven by tumoural factors and corticosteroid use. Furthermore, unique anatomical differences with primary intracranial tumours such as the blood-brain barrier, the type of antigen-presenting cells and lymphatic drainage contribute to differences in treatment success compared to extracranial tumours. There are, however, shared characteristics with those known in other tumours such as the immunosuppressive tumour microenvironment. We conclude with a summary of ongoing and future immune combination strategies in GBM, which are representative of the next wave in immuno-oncology therapeutics.
Collapse
Affiliation(s)
- Jason Adhikaree
- Host-Tumour Interactions Group, Division of Cancer and Stem Cells, BioDiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; (A.P.K.); (A.M.J.); (P.M.P.)
| | - Julia Moreno-Vicente
- Antibody and Vaccine Group, Centre for Cancer Immunology, University of Southampton, Southampton General Hospital, Southampton, Hants SO16 6YD, UK;
| | - Aanchal Preet Kaur
- Host-Tumour Interactions Group, Division of Cancer and Stem Cells, BioDiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; (A.P.K.); (A.M.J.); (P.M.P.)
| | - Andrew Mark Jackson
- Host-Tumour Interactions Group, Division of Cancer and Stem Cells, BioDiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; (A.P.K.); (A.M.J.); (P.M.P.)
| | - Poulam M. Patel
- Host-Tumour Interactions Group, Division of Cancer and Stem Cells, BioDiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; (A.P.K.); (A.M.J.); (P.M.P.)
| |
Collapse
|
453
|
Hirbec H, Déglon N, Foo LC, Goshen I, Grutzendler J, Hangen E, Kreisel T, Linck N, Muffat J, Regio S, Rion S, Escartin C. Emerging technologies to study glial cells. Glia 2020; 68:1692-1728. [PMID: 31958188 DOI: 10.1002/glia.23780] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022]
Abstract
Development, physiological functions, and pathologies of the brain depend on tight interactions between neurons and different types of glial cells, such as astrocytes, microglia, oligodendrocytes, and oligodendrocyte precursor cells. Assessing the relative contribution of different glial cell types is required for the full understanding of brain function and dysfunction. Over the recent years, several technological breakthroughs were achieved, allowing "glio-scientists" to address new challenging biological questions. These technical developments make it possible to study the roles of specific cell types with medium or high-content workflows and perform fine analysis of their mutual interactions in a preserved environment. This review illustrates the potency of several cutting-edge experimental approaches (advanced cell cultures, induced pluripotent stem cell (iPSC)-derived human glial cells, viral vectors, in situ glia imaging, opto- and chemogenetic approaches, and high-content molecular analysis) to unravel the role of glial cells in specific brain functions or diseases. It also illustrates the translation of some techniques to the clinics, to monitor glial cells in patients, through specific brain imaging methods. The advantages, pitfalls, and future developments are discussed for each technique, and selected examples are provided to illustrate how specific "gliobiological" questions can now be tackled.
Collapse
Affiliation(s)
- Hélène Hirbec
- Institute for Functional Genomics (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Nicole Déglon
- Laboratory of Neurotherapies and Neuromodulation, Department of Clinical Neuroscience, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland.,Laboratory of Neurotherapies and Neuromodulation, Neuroscience Research Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Lynette C Foo
- Neuroimmunology and Neurodegeneration Section, The Neuroscience and Rare Diseases Discovery and Translational Area, F. Hoffman-La Roche, Basel, Switzerland
| | - Inbal Goshen
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jaime Grutzendler
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Emilie Hangen
- Commissariat à l'Energie Atomique et aux Energies Alternatives, Département de la Recherche Fondamentale, Institut de Biologie François Jacob, MIRCen, Fontenay-aux-Roses, France.,Centre National de la Recherche Scientifique, Neurodegenerative Diseases Laboratory, Université Paris-Sud, Université Paris-Saclay, UMR 9199, Fontenay-aux-Roses, France
| | - Tirzah Kreisel
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nathalie Linck
- Institute for Functional Genomics (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Julien Muffat
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, and Department of Molecular Genetics, The University of Toronto, Toronto, Canada
| | - Sara Regio
- Laboratory of Neurotherapies and Neuromodulation, Department of Clinical Neuroscience, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland.,Laboratory of Neurotherapies and Neuromodulation, Neuroscience Research Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Sybille Rion
- Neuroimmunology and Neurodegeneration Section, The Neuroscience and Rare Diseases Discovery and Translational Area, F. Hoffman-La Roche, Basel, Switzerland
| | - Carole Escartin
- Commissariat à l'Energie Atomique et aux Energies Alternatives, Département de la Recherche Fondamentale, Institut de Biologie François Jacob, MIRCen, Fontenay-aux-Roses, France.,Centre National de la Recherche Scientifique, Neurodegenerative Diseases Laboratory, Université Paris-Sud, Université Paris-Saclay, UMR 9199, Fontenay-aux-Roses, France
| |
Collapse
|
454
|
Brioschi S, Zhou Y, Colonna M. Brain Parenchymal and Extraparenchymal Macrophages in Development, Homeostasis, and Disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:294-305. [PMID: 31907272 PMCID: PMC7034672 DOI: 10.4049/jimmunol.1900821] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/27/2019] [Indexed: 12/23/2022]
Abstract
Microglia are parenchymal macrophages of the CNS; as professional phagocytes they are important for maintenance of the brain's physiology. These cells are generated through primitive hematopoiesis in the yolk sac and migrate into the brain rudiment after establishment of embryonic circulation. Thereafter, microglia develop in a stepwise fashion, reaching complete maturity after birth. In the CNS, microglia self-renew without input from blood monocytes. Recent RNA-sequencing studies have defined a molecular signature for microglia under homeostasis. However, during disease, microglia undergo remarkable phenotypic changes, which reflect the acquisition of specialized functions tailored to the pathological context. In addition to microglia, the brain-border regions host populations of extraparenchymal macrophages with disparate origins and phenotypes that have recently been delineated. In this review we outline recent findings that provide a deeper understanding of both parenchymal microglia and extraparenchymal brain macrophages in homeostasis and during disease.
Collapse
Affiliation(s)
- Simone Brioschi
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110
| | - Yingyue Zhou
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO 63110
| |
Collapse
|
455
|
Chhatbar C, Detje CN, Grabski E, Borst K, Spanier J, Ghita L, Elliott DA, Jordão MJC, Mueller N, Sutton J, Prajeeth CK, Gudi V, Klein MA, Prinz M, Bradke F, Stangel M, Kalinke U. Type I Interferon Receptor Signaling of Neurons and Astrocytes Regulates Microglia Activation during Viral Encephalitis. Cell Rep 2020; 25:118-129.e4. [PMID: 30282022 PMCID: PMC7103936 DOI: 10.1016/j.celrep.2018.09.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 06/06/2018] [Accepted: 08/30/2018] [Indexed: 12/11/2022] Open
Abstract
In sterile neuroinflammation, a pathological role is proposed for microglia, whereas in viral encephalitis, their function is not entirely clear. Many viruses exploit the odorant system and enter the CNS via the olfactory bulb (OB). Upon intranasal vesicular stomatitis virus instillation, we show an accumulation of activated microglia and monocytes in the OB. Depletion of microglia during encephalitis results in enhanced virus spread and increased lethality. Activation, proliferation, and accumulation of microglia are regulated by type I IFN receptor signaling of neurons and astrocytes, but not of microglia. Morphological analysis of myeloid cells shows that type I IFN receptor signaling of neurons has a stronger impact on the activation of myeloid cells than of astrocytes. Thus, in the infected CNS, the cross talk among neurons, astrocytes, and microglia is critical for full microglia activation and protection from lethal encephalitis.
Collapse
Affiliation(s)
- Chintan Chhatbar
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Claudia N Detje
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Elena Grabski
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Katharina Borst
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Julia Spanier
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Luca Ghita
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - David A Elliott
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Disease Research (DZNE), Bonn, Germany
| | - Marta Joana Costa Jordão
- Institute of Neuropathology, Freiburg University Medical Centre, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Nora Mueller
- Institute for Virology and Immunobiology, University of Wuerzburg, Wuerzburg, Germany
| | - James Sutton
- Novartis Institutes for Biomedical Research, Emeryville, CA, USA
| | - Chittappen K Prajeeth
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Viktoria Gudi
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Michael A Klein
- Institute for Virology and Immunobiology, University of Wuerzburg, Wuerzburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Freiburg University Medical Centre, Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Frank Bradke
- Axonal Growth and Regeneration Group, German Center for Neurodegenerative Disease Research (DZNE), Bonn, Germany
| | - Martin Stangel
- Clinical Neuroimmunology and Neurochemistry, Department of Neurology, Hannover Medical School, Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
456
|
Datta D, Leslie SN, Morozov YM, Duque A, Rakic P, van Dyck CH, Nairn AC, Arnsten AFT. Classical complement cascade initiating C1q protein within neurons in the aged rhesus macaque dorsolateral prefrontal cortex. J Neuroinflammation 2020; 17:8. [PMID: 31906973 PMCID: PMC6945481 DOI: 10.1186/s12974-019-1683-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/17/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Cognitive impairment in schizophrenia, aging, and Alzheimer's disease is associated with spine and synapse loss from the dorsolateral prefrontal cortex (dlPFC) layer III. Complement cascade signaling is critical in driving spine loss and disease pathogenesis. Complement signaling is initiated by C1q, which tags synapses for elimination. C1q is thought to be expressed predominately by microglia, but its expression in primate dlPFC has never been examined. The current study assayed C1q levels in aging primate dlPFC and rat medial PFC (mPFC) and used immunoelectron microscopy (immunoEM), immunoblotting, and co-immunoprecipitation (co-IP) to reveal the precise anatomical distribution and interactions of C1q. METHODS Age-related changes in C1q levels in rhesus macaque dlPFC and rat mPFC were examined using immunoblotting. High-spatial resolution immunoEM was used to interrogate the subcellular localization of C1q in aged macaque layer III dlPFC and aged rat layer III mPFC. co-IP techniques quantified protein-protein interactions for C1q and proteins associated with excitatory and inhibitory synapses in macaque dlPFC. RESULTS C1q levels were markedly increased in the aged macaque dlPFC. Ultrastructural localization found the expected C1q localization in glia, including those ensheathing synapses, but also revealed extensive localization within neurons. C1q was found near synapses, within terminals and in spines, but was also observed in dendrites, often near abnormal mitochondria. Similar analyses in aging rat mPFC corroborated the findings in rhesus macaques. C1q protein increasingly associated with PSD95 with age in macaque, consistent with its synaptic localization as evidenced by EM. CONCLUSIONS These findings reveal novel, intra-neuronal distribution patterns for C1q in the aging primate cortex, including evidence of C1q in dendrites. They suggest that age-related changes in the dlPFC may increase C1q expression and synaptic tagging for glial phagocytosis, a possible mechanism for age-related degeneration.
Collapse
Affiliation(s)
- Dibyadeep Datta
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06511, USA.
| | - Shannon N Leslie
- Department of Psychiatry, Yale University School of Medicine, New Haven, USA
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, USA
| | - Yury M Morozov
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06511, USA
| | - Alvaro Duque
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06511, USA
| | - Pasko Rakic
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06511, USA
| | - Christopher H van Dyck
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06511, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, USA
| | - Angus C Nairn
- Department of Psychiatry, Yale University School of Medicine, New Haven, USA
| | - Amy F T Arnsten
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06511, USA.
| |
Collapse
|
457
|
Macke EL, Henningsen E, Jessen E, Zumwalde NA, Landowski M, Western DE, Lee WH, Liu C, Gruenke NP, Doebley AL, Miller S, Pattnaik B, Ikeda S, Gumperz JE, Ikeda A. Loss of Chondroitin Sulfate Modification Causes Inflammation and Neurodegeneration in skt Mice. Genetics 2020; 214:121-134. [PMID: 31754016 PMCID: PMC6944401 DOI: 10.1534/genetics.119.302834] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 11/17/2019] [Indexed: 12/17/2022] Open
Abstract
One major aspect of the aging process is the onset of chronic, low-grade inflammation that is highly associated with age-related diseases. The molecular mechanisms that regulate these processes have not been fully elucidated. We have identified a spontaneous mutant mouse line, small with kinky tail (skt), that exhibits accelerated aging and age-related disease phenotypes including increased inflammation in the brain and retina, enhanced age-dependent retinal abnormalities including photoreceptor cell degeneration, neurodegeneration in the hippocampus, and reduced lifespan. By positional cloning, we identified a deletion in chondroitin sulfate synthase 1 (Chsy1) that is responsible for these phenotypes in skt mice. CHSY1 is a member of the chondroitin N-acetylgalactosaminyltransferase family that plays critical roles in the biosynthesis of chondroitin sulfate, a glycosaminoglycan (GAG) that is attached to the core protein to form the chondroitin sulfate proteoglycan (CSPG). Consistent with this function, the Chsy1 mutation dramatically decreases chondroitin sulfate GAGs in the retina and hippocampus. In addition, macrophage and neutrophil populations appear significantly altered in the bone marrow and spleen of skt mice, suggesting an important role for CHSY1 in the functioning of these immune cell types. Thus, our study reveals a previously unidentified impact of CHSY1 in the retina and hippocampus. Specifically, chondroitin sulfate (CS) modification of proteins by CHSY1 appears critical for proper regulation of immune cells of the myeloid lineage and for maintaining the integrity of neuronal tissues, since a defect in this gene results in increased inflammation and abnormal phenotypes associated with age-related diseases.
Collapse
Affiliation(s)
- Erica L Macke
- Department of Medical Genetics, University of Wisconsin-Madison, Wisconsin 53706
| | - Erika Henningsen
- Department of Medical Genetics, University of Wisconsin-Madison, Wisconsin 53706
| | - Erik Jessen
- Department of Medical Genetics, University of Wisconsin-Madison, Wisconsin 53706
- Department of Biochemistry, University of Wisconsin-Madison, Wisconsin 53706
| | - Nicholas A Zumwalde
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Wisconsin 53706
| | - Michael Landowski
- Department of Medical Genetics, University of Wisconsin-Madison, Wisconsin 53706
| | - Daniel E Western
- Department of Medical Genetics, University of Wisconsin-Madison, Wisconsin 53706
| | - Wei-Hua Lee
- Department of Medical Genetics, University of Wisconsin-Madison, Wisconsin 53706
| | - Che Liu
- Institute for Molecular Virology, University of Wisconsin-Madison, Wisconsin 53706
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Wisconsin 53706
| | - Nathan P Gruenke
- Department of Medical Genetics, University of Wisconsin-Madison, Wisconsin 53706
| | - Anna-Lisa Doebley
- Department of Medical Genetics, University of Wisconsin-Madison, Wisconsin 53706
| | - Samuel Miller
- Department of Medical Genetics, University of Wisconsin-Madison, Wisconsin 53706
| | - Bikash Pattnaik
- Department of Pediatrics, University of Wisconsin-Madison, Wisconsin 53706
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Wisconsin 53706
- McPherson Eye Research Institute, University of Wisconsin-Madison, Wisconsin 53706
| | - Sakae Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison, Wisconsin 53706
- McPherson Eye Research Institute, University of Wisconsin-Madison, Wisconsin 53706
| | - Jenny E Gumperz
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Wisconsin 53706
| | - Akihiro Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison, Wisconsin 53706
- McPherson Eye Research Institute, University of Wisconsin-Madison, Wisconsin 53706
| |
Collapse
|
458
|
Plemel JR, Stratton JA, Michaels NJ, Rawji KS, Zhang E, Sinha S, Baaklini CS, Dong Y, Ho M, Thorburn K, Friedman TN, Jawad S, Silva C, Caprariello AV, Hoghooghi V, Yue J, Jaffer A, Lee K, Kerr BJ, Midha R, Stys PK, Biernaskie J, Yong VW. Microglia response following acute demyelination is heterogeneous and limits infiltrating macrophage dispersion. SCIENCE ADVANCES 2020; 6:eaay6324. [PMID: 31998844 PMCID: PMC6962036 DOI: 10.1126/sciadv.aay6324] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 11/08/2019] [Indexed: 05/22/2023]
Abstract
Microglia and infiltrating macrophages are thought to orchestrate the central nervous system (CNS) response to injury; however, the similarities between these cells make it challenging to distinguish their relative contributions. We genetically labeled microglia and CNS-associated macrophages to distinguish them from infiltrating macrophages. Using single-cell RNA sequencing, we describe multiple microglia activation states, one of which was enriched for interferon associated signaling. Although blood-derived macrophages acutely infiltrated the demyelinated lesion, microglia progressively monopolized the lesion environment where they surrounded infiltrating macrophages. In the microglia-devoid sciatic nerve, the infiltrating macrophage response was sustained. In the CNS, the preferential proliferation of microglia and sparse microglia death contributed to microglia dominating the lesion. Microglia ablation reversed the spatial restriction of macrophages with the demyelinated spinal cord, highlighting an unrealized macrophages-microglia interaction. The restriction of peripheral inflammation by microglia may be a previously unidentified mechanism by which the CNS maintains its "immune privileged" status.
Collapse
Affiliation(s)
- Jason R. Plemel
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, Alberta, Canada
- Corresponding author. (J.R.P.); (J.B.); (V.W.Y.)
| | - Jo Anne Stratton
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Nathan J. Michaels
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Khalil S. Rawji
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Eric Zhang
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sarthak Sinha
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Charbel S. Baaklini
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, Alberta, Canada
| | - Yifei Dong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Madelene Ho
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Kevin Thorburn
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Timothy N. Friedman
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Sana Jawad
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Claudia Silva
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Andrew V. Caprariello
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Vahid Hoghooghi
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Julie Yue
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Arzina Jaffer
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Kelly Lee
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Bradley J. Kerr
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Anesthesiology and Pain Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Raj Midha
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Peter K. Stys
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jeff Biernaskie
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Corresponding author. (J.R.P.); (J.B.); (V.W.Y.)
| | - V. Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Corresponding author. (J.R.P.); (J.B.); (V.W.Y.)
| |
Collapse
|
459
|
Nur S, Sparber F, Lemberg C, Guiducci E, Schweizer TA, Zwicky P, Becher B, LeibundGut-Landmann S. IL-23 supports host defense against systemic Candida albicans infection by ensuring myeloid cell survival. PLoS Pathog 2019; 15:e1008115. [PMID: 31887131 PMCID: PMC6957211 DOI: 10.1371/journal.ppat.1008115] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 01/13/2020] [Accepted: 10/01/2019] [Indexed: 01/21/2023] Open
Abstract
The opportunistic fungal pathogen Candida albicans can cause invasive infections in susceptible hosts and the innate immune system, in particular myeloid cell-mediated immunity, is critical for rapid immune protection and host survival during systemic candidiasis. Using a mouse model of the human disease, we identified a novel role of IL-23 in antifungal defense. IL-23-deficient mice are highly susceptible to systemic infection with C. albicans. We found that this results from a drastic reduction in all subsets of myeloid cells in the infected kidney, which in turn leads to rapid fungal overgrowth and renal tissue injury. The loss in myeloid cells is not due to a defect in emergency myelopoiesis or the recruitment of newly generated cells to the site of infection but, rather, is a consequence of impaired survival of myeloid cells at the site of infection. In fact, the absence of a functional IL-23 pathway causes massive myeloid cell apoptosis upon C. albicans infection. Importantly, IL-23 protects myeloid cells from apoptosis independently of the IL-23-IL-17 immune axis and independently of lymphocytes and innate lymphoid cells. Instead, our results suggest that IL-23 acts in a partially autocrine but not cell-intrinsic manner within the myeloid compartment to promote host protection from systemic candidiasis. Collectively, our data highlight an unprecedented and non-canonical role of IL-23 in securing survival of myeloid cells, which is key for maintaining sufficient numbers of cells at the site of infection to ensure efficient host protection. Linked to advances in medical technology and the resulting increase in the number of intensive care patients, nosocomial infections with Candida albicans are on the rise. In patients suffering from invasive candidiasis the innate immune response is typically severely impaired. Strengthening the innate immune system has become a promising approach complementing the use of antifungal drugs. Our findings identify an unexpected and IL-17-independent role of IL-23 that prevents rapid death of myeloid cells during systemic candidiasis and thereby promotes optimal protection from disease. As such, IL-23 represents an important new piece in the puzzle of the finely tuned network of cytokines that regulates the innate immune response to fungal infection. Our results contribute to a better understanding of myeloid cell regulation during infection and thereby open new perspectives for future immunotherapeutic applications that may improve patient outcome.
Collapse
Affiliation(s)
- Selim Nur
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
| | - Florian Sparber
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
| | - Christina Lemberg
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
| | - Eva Guiducci
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
| | - Tiziano A. Schweizer
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
| | - Pascale Zwicky
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | | |
Collapse
|
460
|
Zhang W, Zhang X, Zhang Y, Qu C, Zhou X, Zhang S. Histamine Induces Microglia Activation and the Release of Proinflammatory Mediators in Rat Brain Via H 1R or H 4R. J Neuroimmune Pharmacol 2019; 15:280-291. [PMID: 31863333 DOI: 10.1007/s11481-019-09887-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 10/01/2019] [Indexed: 12/17/2022]
Abstract
Histamine is a major peripheral inflammatory mediator and a neurotransmitter in the central nervous system. We have reported that histamine induces microglia activation and releases proinflammatory factors in primary cultured microglia. Whether histamine has similar effects in vivo is unknown. In the present study, we aimed to investigate the role of histamine and its receptors in the release of inflammatory mediators and activation of microglia in rat brain. We site-directed injected histamine, histamine receptor agonists or histamine receptor antagonists in the rat lateral ventricle using stereotaxic techniques. Flow cytometry was employed to determine histamine receptor expression in rat microglia. Microglia activation was assessed by Iba1 immunohistochemistry. The levels of tumour necrosis factor-alpha (TNF-α), interleukin-1beta (IL-1β) and interleukin-10 (IL-10) were measured with commercial enzyme-linked immunosorbent assay (ELISA) kits, TNF-α, IL-1β and IL-10 mRNA expressions were determined with Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR). We found that all four types of histamine receptors were expressed in rat brain microglia. Histamine was able to induce microglia activation and subsequent production of the inflammatory factors TNF-α, IL-1β and IL-10, and these effects were partially abolished by H1R and H4R antagonists. However, H2R and H3R antagonists significantly increased production of TNF-α and IL-1β, and decreased IL-10 levels. The H1R or H4R agonists stimulated the production of TNF-α and IL-1β, while the H2R or H3R agonists increased IL-10 release. Our results demonstrate that histamine induces microglia activation and the release of both proinflammatory and anti-inflammatory factors in rat brain, thus contributing to the development of inflammation in the brain. Graphical Abstract Histamine induces microglia activation and the release of both proinflammatory (TNF-α and IL-1β) and anti-inflammatory factors (IL-10) in rat brain, thus contributing to the development of inflammation in the brain.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Laboratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Xiaojun Zhang
- Department of Rheumatology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Yan Zhang
- Department of Anesthesiology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chen Qu
- Department of Geriatrics, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Xiqiao Zhou
- Department of Gastroenterology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, People's Republic of China.
| | - Shu Zhang
- Clinical Research Center, the First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu, 210029, People's Republic of China.
| |
Collapse
|
461
|
Tsai AS, Berry K, Beneyto MM, Gaudilliere D, Ganio EA, Culos A, Ghaemi MS, Choisy B, Djebali K, Einhaus JF, Bertrand B, Tanada A, Stanley N, Fallahzadeh R, Baca Q, Quach LN, Osborn E, Drag L, Lansberg MG, Angst MS, Gaudilliere B, Buckwalter MS, Aghaeepour N. A year-long immune profile of the systemic response in acute stroke survivors. Brain 2019; 142:978-991. [PMID: 30860258 DOI: 10.1093/brain/awz022] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 11/18/2018] [Accepted: 12/14/2018] [Indexed: 02/07/2023] Open
Abstract
Stroke is a leading cause of cognitive impairment and dementia, but the mechanisms that underlie post-stroke cognitive decline are not well understood. Stroke produces profound local and systemic immune responses that engage all major innate and adaptive immune compartments. However, whether the systemic immune response to stroke contributes to long-term disability remains ill-defined. We used a single-cell mass cytometry approach to comprehensively and functionally characterize the systemic immune response to stroke in longitudinal blood samples from 24 patients over the course of 1 year and correlated the immune response with changes in cognitive functioning between 90 and 365 days post-stroke. Using elastic net regularized regression modelling, we identified key elements of a robust and prolonged systemic immune response to ischaemic stroke that occurs in three phases: an acute phase (Day 2) characterized by increased signal transducer and activator of transcription 3 (STAT3) signalling responses in innate immune cell types, an intermediate phase (Day 5) characterized by increased cAMP response element-binding protein (CREB) signalling responses in adaptive immune cell types, and a late phase (Day 90) by persistent elevation of neutrophils, and immunoglobulin M+ (IgM+) B cells. By Day 365 there was no detectable difference between these samples and those from an age- and gender-matched patient cohort without stroke. When regressed against the change in the Montreal Cognitive Assessment scores between Days 90 and 365 after stroke, the acute inflammatory phase Elastic Net model correlated with post-stroke cognitive trajectories (r = -0.692, Bonferroni-corrected P = 0.039). The results demonstrate the utility of a deep immune profiling approach with mass cytometry for the identification of clinically relevant immune correlates of long-term cognitive trajectories.
Collapse
Affiliation(s)
- Amy S Tsai
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine, CA, USA
| | - Kacey Berry
- Stanford Stroke Center, Stanford School of Medicine, CA, USA.,Department of Neurology and Neurological Sciences, Stanford School of Medicine, CA, USA
| | - Maxime M Beneyto
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine, CA, USA
| | - Dyani Gaudilliere
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, CA, USA
| | - Edward A Ganio
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine, CA, USA
| | - Anthony Culos
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine, CA, USA
| | - Mohammad S Ghaemi
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine, CA, USA
| | - Benjamin Choisy
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine, CA, USA
| | - Karim Djebali
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine, CA, USA
| | - Jakob F Einhaus
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine, CA, USA
| | - Basile Bertrand
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine, CA, USA
| | - Athena Tanada
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine, CA, USA
| | - Natalie Stanley
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine, CA, USA
| | - Ramin Fallahzadeh
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine, CA, USA
| | - Quentin Baca
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine, CA, USA
| | - Lisa N Quach
- Stanford Stroke Center, Stanford School of Medicine, CA, USA.,Department of Neurology and Neurological Sciences, Stanford School of Medicine, CA, USA
| | - Elizabeth Osborn
- Stanford Stroke Center, Stanford School of Medicine, CA, USA.,Department of Neurology and Neurological Sciences, Stanford School of Medicine, CA, USA
| | - Lauren Drag
- Department of Neurology and Neurological Sciences, Stanford School of Medicine, CA, USA
| | - Maarten G Lansberg
- Stanford Stroke Center, Stanford School of Medicine, CA, USA.,Department of Neurology and Neurological Sciences, Stanford School of Medicine, CA, USA
| | - Martin S Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine, CA, USA
| | - Brice Gaudilliere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine, CA, USA
| | - Marion S Buckwalter
- Stanford Stroke Center, Stanford School of Medicine, CA, USA.,Department of Neurology and Neurological Sciences, Stanford School of Medicine, CA, USA.,Department of Neurosurgery, Stanford School of Medicine, CA, USA
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford School of Medicine, CA, USA
| |
Collapse
|
462
|
Schonhoff AM, Williams GP, Wallen ZD, Standaert DG, Harms AS. Innate and adaptive immune responses in Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2019; 252:169-216. [PMID: 32247364 DOI: 10.1016/bs.pbr.2019.10.006] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Parkinson's disease (PD) has classically been defined as a movement disorder, in which motor symptoms are explained by the aggregation of alpha-synuclein (α-syn) and subsequent death of dopaminergic neurons of the substantia nigra pars compacta (SNpc). More recently, the multisystem effects of the disease have been investigated, with the immune system being implicated in a number of these processes in the brain, the blood, and the gut. In this review, we highlight the dysfunctional immune system found in both human PD and animal models of the disease, and discuss how genetic risk factors and risk modifiers are associated with pro-inflammatory immune responses. Finally, we emphasize evidence that the immune response drives the pathogenesis and progression of PD, and discuss key questions that remain to be investigated in order to identify immunomodulatory therapies in PD.
Collapse
Affiliation(s)
- Aubrey M Schonhoff
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Gregory P Williams
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Zachary D Wallen
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - David G Standaert
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ashley S Harms
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, The University of Alabama at Birmingham, Birmingham, AL, United States.
| |
Collapse
|
463
|
Koizumi T, Kerkhofs D, Mizuno T, Steinbusch HWM, Foulquier S. Vessel-Associated Immune Cells in Cerebrovascular Diseases: From Perivascular Macrophages to Vessel-Associated Microglia. Front Neurosci 2019; 13:1291. [PMID: 31866808 PMCID: PMC6904330 DOI: 10.3389/fnins.2019.01291] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 11/14/2019] [Indexed: 01/04/2023] Open
Abstract
Cerebral small vessels feed and protect the brain parenchyma thanks to the unique features of the blood-brain barrier. Cerebrovascular dysfunction is therefore seen as a detrimental factor for the initiation of several central nervous system (CNS) disorders, such as stroke, cerebral small vessel disease (cSVD), and Alzheimer's disease. The main working hypothesis linking cerebrovascular dysfunction to brain disorders includes the contribution of neuroinflammation. While our knowledge on microglia cells - the brain-resident immune cells - has been increasing in the last decades, the specific populations of microglia and macrophages surrounding brain vessels, vessel-associated microglia (VAM), and perivascular macrophages (PVMs), respectively, have been overlooked. This review aims to summarize the knowledge gathered on VAM and PVMs, to discuss existing knowledge gaps of importance for later studies and to summarize evidences for their contribution to cerebrovascular dysfunction.
Collapse
Affiliation(s)
- Takashi Koizumi
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Pharmacology and Toxicology, School for Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Danielle Kerkhofs
- Department of Neurology, School for Cardiovascular Diseases, Maastricht University Medical Center+, Maastricht, Netherlands
- Department of Pathology, School for Cardiovascular Diseases, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Toshiki Mizuno
- Department of Neurology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Harry W. M. Steinbusch
- Department of Translational Neuroscience, School for Mental Health and Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Sébastien Foulquier
- Department of Pharmacology and Toxicology, School for Mental Health and Neuroscience, Maastricht University Medical Center+, Maastricht, Netherlands
| |
Collapse
|
464
|
Sankowski R, Böttcher C, Masuda T, Geirsdottir L, Sagar, Sindram E, Seredenina T, Muhs A, Scheiwe C, Shah MJ, Heiland DH, Schnell O, Grün D, Priller J, Prinz M. Mapping microglia states in the human brain through the integration of high-dimensional techniques. Nat Neurosci 2019; 22:2098-2110. [PMID: 31740814 DOI: 10.1038/s41593-019-0532-y] [Citation(s) in RCA: 277] [Impact Index Per Article: 55.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 10/07/2019] [Indexed: 01/01/2023]
Abstract
Microglia are tissue-resident macrophages of the CNS that orchestrate local immune responses and contribute to several neurological and psychiatric diseases. Little is known about human microglia and how they orchestrate their highly plastic, context-specific adaptive responses during pathology. Here we combined two high-dimensional technologies, single-cell RNA-sequencing and time-of-flight mass cytometry, to identify microglia states in the human brain during homeostasis and disease. This approach enabled us to identify and characterize a previously unappreciated spectrum of transcriptional states in human microglia. These transcriptional states are determined by their spatial distribution, and they further change with aging and brain tumor pathology. This description of multiple microglia phenotypes in the human CNS may open promising new avenues for subset-specific therapeutic interventions.
Collapse
Affiliation(s)
- Roman Sankowski
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Berta-Ottenstein-Programme for Clinician Scientists, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Chotima Böttcher
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Takahiro Masuda
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Laufey Geirsdottir
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sagar
- Max-Planck-Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Elena Sindram
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | | | - Christian Scheiwe
- Department of Neurosurgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Mukesch Johannes Shah
- Department of Neurosurgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dieter Henrik Heiland
- Department of Neurosurgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Oliver Schnell
- Department of Neurosurgery, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dominic Grün
- Max-Planck-Institute of Immunobiology and Epigenetics, Freiburg, Germany.
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg im Breisgau, Germany.
| | - Josef Priller
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin, Berlin, Germany.
- DZNE and BIH, Berlin, Germany.
- University of Edinburgh and UK DRI, Edinburgh, UK.
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg im Breisgau, Germany.
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany.
| |
Collapse
|
465
|
Olcum M, Tastan B, Kiser C, Genc S, Genc K. Microglial NLRP3 inflammasome activation in multiple sclerosis. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2019; 119:247-308. [PMID: 31997770 DOI: 10.1016/bs.apcsb.2019.08.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Multiple sclerosis (MS) is a chronic, autoimmune and neuroinflammatory disease of the central nervous system (CNS) mediated by autoreactive T cells directed against myelin antigens. Although the crucial role of adaptive immunity is well established in MS, the contribution of innate immunity has only recently been appreciated. Microglia are the main innate immune cells of the CNS. Similar to other myeloid cells, microglia recognize both exogenous and host-derived endogenous danger signals through pattern recognition receptors (PRRs) localized on their cell surface such as Toll Like receptor 4, or in the cytosol such as NLRP3. The second one is the sensor protein of the multi-molecular NLRP3 inflammasome complex in activated microglia that promotes the maturation and secretion of proinflammatory cytokines, interleukin-1β and interleukin-18. Overactivation of microglia and aberrant activation of the NLRP3 inflammasome have been implicated in the pathogenesis of MS. Indeed, experimental data, together with post-mortem and clinical studies have revealed an increased expression of NLRP3 inflammasome complex elements in microglia and other immune cells. In this review, we focus on microglial NLRP3 inflammasome activation in MS. First, we overview the basic knowledge about MS, microglia and the NLRP3 inflammasome. Then, we summarize studies about microglial NLRP3 inflammasome activation in MS and its animal models. We also highlight experimental therapeutic approaches that target different steps of NLRP inflammasome activation. Finally, we discuss future research avenues and new methods in this rapidly evolving area.
Collapse
Affiliation(s)
- Melis Olcum
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus Balcova, Izmir, Turkey
| | - Bora Tastan
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus Balcova, Izmir, Turkey; Izmir International Biomedicine and Genome Institute (iBG-Izmir), Dokuz Eylul University Health Campus, Balcova, Izmir, Turkey
| | - Cagla Kiser
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus Balcova, Izmir, Turkey; Izmir International Biomedicine and Genome Institute (iBG-Izmir), Dokuz Eylul University Health Campus, Balcova, Izmir, Turkey
| | - Sermin Genc
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus Balcova, Izmir, Turkey; Izmir International Biomedicine and Genome Institute (iBG-Izmir), Dokuz Eylul University Health Campus, Balcova, Izmir, Turkey; Department of Neuroscience, Institute of Health and Science, Dokuz Eylul University Health Campus, Balcova, Izmir, Turkey
| | - Kursad Genc
- Department of Neuroscience, Institute of Health and Science, Dokuz Eylul University Health Campus, Balcova, Izmir, Turkey
| |
Collapse
|
466
|
Yang T, Guo R, Zhang F. Brain perivascular macrophages: Recent advances and implications in health and diseases. CNS Neurosci Ther 2019; 25:1318-1328. [PMID: 31749316 PMCID: PMC7154594 DOI: 10.1111/cns.13263] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/24/2019] [Accepted: 10/25/2019] [Indexed: 12/20/2022] Open
Abstract
Brain perivascular macrophages (PVMs) belong to a distinct population of brain‐resident myeloid cells located within the perivascular space surrounding arterioles and venules. Their characterization depends on the combination of anatomical localization, phagocytic capacity, and molecular markers. Under physiological status, they provide structural and functional support for maintaining brain homeostasis, including facilitation of blood‐brain barrier integrity and lymphatic drainage, and exertion of immune functions such as phagocytosis and antigen presentation. Increasing evidence also implicates their specific roles in diseased brain, ranging from cerebrovascular diseases, Aβ pathologies, infections, and autoimmunity. Collectively, PVMs are key components of the brain‐resident immune system, actively participate in a broad‐spectrum of processes in normal and diseased status. Details of the processes are largely underexplored. Targeting PVMs would lead to new insights and be a promising strategy for a broad array of human diseases.
Collapse
Affiliation(s)
- Tuo Yang
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ruiming Guo
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Feng Zhang
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
467
|
Bordt EA, Ceasrine AM, Bilbo SD. Microglia and sexual differentiation of the developing brain: A focus on ontogeny and intrinsic factors. Glia 2019; 68:1085-1099. [PMID: 31743527 DOI: 10.1002/glia.23753] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/24/2019] [Accepted: 10/29/2019] [Indexed: 12/15/2022]
Abstract
Sexual differentiation of the brain during early development likely underlies the strong sex biases prevalent in many neurological conditions. Mounting evidence indicates that microglia, the innate immune cells of the central nervous system, are intricately involved in these sex-specific processes of differentiation. In this review, we synthesize literature demonstrating sex differences in microglial number, morphology, transcriptional state, and functionality throughout spatiotemporal development as well as highlight current literature regarding ontogeny of microglia. Along with vanRyzin et al. in this issue, we explore the idea that differences in microglia imparted by chromosomal or ontogeny-related programming can influence microglial-driven sexual differentiation of the brain, as well as the idea that extrinsic differences in the male and female brain microenvironment may in turn impart sex differences in microglia.
Collapse
Affiliation(s)
- Evan A Bordt
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital for Children, Harvard Medical School, Boston, Massachusetts
| | - Alexis M Ceasrine
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina
| | - Staci D Bilbo
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital for Children, Harvard Medical School, Boston, Massachusetts.,Department of Psychology and Neuroscience, Duke University, Durham, North Carolina
| |
Collapse
|
468
|
Hatton CF, Duncan CJA. Microglia Are Essential to Protective Antiviral Immunity: Lessons From Mouse Models of Viral Encephalitis. Front Immunol 2019; 10:2656. [PMID: 31798586 PMCID: PMC6863772 DOI: 10.3389/fimmu.2019.02656] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 10/28/2019] [Indexed: 12/22/2022] Open
Abstract
Viral encephalitis is a rare but clinically serious consequence of viral invasion of the brain and insight into its pathogenesis is urgently needed. Important research questions concern the involvement of the host innate immune response in pathogenesis, key to which is the role played by microglia, resident macrophages of the brain parenchyma. Do microglia have a protective function, by coordinating the innate immune response to viral infection, or do they drive pathogenic neuroinflammation? Here we synthesize recent data from mouse models of acute viral encephalitis, which reveal an unambiguously protective role for microglia. Depletion of microglia, via blockade of colony-stimulating factor 1 receptor (CSF1R) signaling, led to increased viral replication accompanied by more severe neurological disease and heightened mortality. Whilst the underlying mechanism(s) remain to be defined, microglial interactions with T cells and phagocytosis of infected neurones appear to play a role. Paradoxically, the production of inflammatory cytokines was increased in several instances following viral infection in microglia-depleted brains, suggesting that: (i) cells other than microglia mediate inflammatory responses and/or (ii) microglia may exert a regulatory function. Under certain circumstances the microglial antiviral response might contribute negatively to longer-term neurological sequelae, although fewer studies have focused on this aspect in encephalitis models. Understanding regulation of the microglial response, and how it contributes to disease is therefore a priority for future studies. Collectively, these findings demonstrate the central role of microglia in pathogenesis, suggesting the exciting possibility that defects of microglial function might contribute to encephalitis susceptibility and/or outcome in humans.
Collapse
Affiliation(s)
- Catherine F Hatton
- Immunity and Inflammation Theme, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Christopher J A Duncan
- Immunity and Inflammation Theme, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,Department of Infection and Tropical Medicine, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
469
|
Mrdjen D, Fox EJ, Bukhari SA, Montine KS, Bendall SC, Montine TJ. The basis of cellular and regional vulnerability in Alzheimer's disease. Acta Neuropathol 2019; 138:729-749. [PMID: 31392412 PMCID: PMC6802290 DOI: 10.1007/s00401-019-02054-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 07/24/2019] [Accepted: 07/31/2019] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) differentially and specifically affects brain regions and neuronal cell types in a predictable pattern. Damage to the brain appears to spread and worsens with time, taking over more regions and activating multiple stressors that can converge to promote vulnerability of certain cell types. At the same time, other cell types and brain regions remain intact in the face of this onslaught of neuropathology. Although neuropathologic descriptions of AD have been extensively expanded and mapped over the last several decades, our understanding of the mechanisms underlying how certain regions and cell populations are specifically vulnerable or resistant has lagged behind. In this review, we detail what is known about the selectivity of local initiation of AD pathology in the hippocampus, its proposed spread via synaptic connections, and the diversity of clinical phenotypes and brain atrophy patterns that may arise from different fibrillar strains of pathologic proteins or genetic predispositions. We summarize accumulated and emerging knowledge of the cellular and molecular basis for neuroanatomic selectivity, consider potential disease-relevant differences between vulnerable and resistant neuronal cell types and isolate molecular markers to identify them.
Collapse
Affiliation(s)
- Dunja Mrdjen
- Department of Pathology, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Edward J Fox
- Department of Pathology, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Syed A Bukhari
- Department of Pathology, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Kathleen S Montine
- Department of Pathology, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Sean C Bendall
- Department of Pathology, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Thomas J Montine
- Department of Pathology, School of Medicine, Stanford University, Palo Alto, CA, USA.
| |
Collapse
|
470
|
Perus LJM, Walsh LA. Microenvironmental Heterogeneity in Brain Malignancies. Front Immunol 2019; 10:2294. [PMID: 31632393 PMCID: PMC6779728 DOI: 10.3389/fimmu.2019.02294] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 09/11/2019] [Indexed: 12/26/2022] Open
Abstract
Brain tumors are among the deadliest malignancies. The brain tumor microenvironment (TME) hosts a unique collection of cells, soluble factors, and extracellular matrix components that regulate disease evolution of both primary and metastatic brain malignancies. It is established that macrophages and other myeloid cells are abundant in the brain TME and strongly correlate with aggressive phenotypes and distinct genetic signatures, while lymphoid cells are less frequent but are now known to have a pronounced effect on disease progression. Different types of brain tumors vary widely in their microenvironmental contexture, and the proportion of various stromal components impacts tumor biology. Indeed, emerging evidence suggests an intimate link between the molecular signature of tumor cells and the composition of the TME, shedding light on the mechanisms which underlie microenvironmental heterogeneity in brain cancer. In this review, we discuss the association between TME composition and the diverse molecular profiles of primary gliomas and brain metastases. We also discuss the implications of these associations on the efficacy of immunotherapy in brain malignancies. An appreciation for the causes and functional consequences of microenvironmental heterogeneity in brain cancer will be of crucial importance to the rational design of microenvironment-targeted therapies for these deadly diseases.
Collapse
Affiliation(s)
- Lucas J. M. Perus
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Logan A. Walsh
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
- Department of Human Genetics, Faculty of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
471
|
Mundt S, Greter M, Flügel A, Becher B. The CNS Immune Landscape from the Viewpoint of a T Cell. Trends Neurosci 2019; 42:667-679. [DOI: 10.1016/j.tins.2019.07.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 02/07/2023]
|
472
|
Single-cell transcriptomic profiling of the aging mouse brain. Nat Neurosci 2019; 22:1696-1708. [PMID: 31551601 DOI: 10.1038/s41593-019-0491-3] [Citation(s) in RCA: 373] [Impact Index Per Article: 74.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 08/09/2019] [Indexed: 01/09/2023]
Abstract
The mammalian brain is complex, with multiple cell types performing a variety of diverse functions, but exactly how each cell type is affected in aging remains largely unknown. Here we performed a single-cell transcriptomic analysis of young and old mouse brains. We provide comprehensive datasets of aging-related genes, pathways and ligand-receptor interactions in nearly all brain cell types. Our analysis identified gene signatures that vary in a coordinated manner across cell types and gene sets that are regulated in a cell-type specific manner, even at times in opposite directions. These data reveal that aging, rather than inducing a universal program, drives a distinct transcriptional course in each cell population, and they highlight key molecular processes, including ribosome biogenesis, underlying brain aging. Overall, these large-scale datasets (accessible online at https://portals.broadinstitute.org/single_cell/study/aging-mouse-brain ) provide a resource for the neuroscience community that will facilitate additional discoveries directed towards understanding and modifying the aging process.
Collapse
|
473
|
Baaklini CS, Rawji KS, Duncan GJ, Ho MFS, Plemel JR. Central Nervous System Remyelination: Roles of Glia and Innate Immune Cells. Front Mol Neurosci 2019; 12:225. [PMID: 31616249 PMCID: PMC6764409 DOI: 10.3389/fnmol.2019.00225] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 09/04/2019] [Indexed: 12/31/2022] Open
Abstract
In diseases such as multiple sclerosis (MS), inflammation can injure the myelin sheath that surrounds axons, a process known as demyelination. The spontaneous regeneration of myelin, called remyelination, is associated with restoration of function and prevention of axonal degeneration. Boosting remyelination with therapeutic intervention is a promising new approach that is currently being tested in several clinical trials. The endogenous regulation of remyelination is highly dependent on the immune response. In this review article, we highlight the cell biology of remyelination and its regulation by innate immune cells. For the purpose of this review, we discuss the roles of microglia, and also astrocytes and oligodendrocyte progenitor cells (OPCs) as they are being increasingly recognized to have immune cell functions.
Collapse
Affiliation(s)
- Charbel S. Baaklini
- Department of Medicine, Division of Neurology, Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Khalil S. Rawji
- Wellcome Trust-Medical Research Council, Cambridge Stem Cell Institute, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Greg J. Duncan
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, OR, United States
| | - Madelene F. S. Ho
- Department of Medicine, Division of Neurology, Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| | | |
Collapse
|
474
|
When encephalitogenic T cells collaborate with microglia in multiple sclerosis. Nat Rev Neurol 2019; 15:704-717. [PMID: 31527807 DOI: 10.1038/s41582-019-0253-6] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2019] [Indexed: 01/07/2023]
Abstract
Immune cells mediate critical inflammatory and neurodegenerative processes in the CNS in individuals with multiple sclerosis (MS). In MS, activated microglia, border-associated macrophages and monocyte-derived macrophages in the CNS can encounter T cells that have infiltrated the brain parenchyma from the circulation. Although microglia and T cells both contribute to normal CNS development and homeostasis, evidence suggests that the meeting of activated microglia and macrophages with encephalitogenic T cells exacerbates their capacity to inflict injury. This crosstalk involves many cell-surface molecules, cytokines and neurotoxic factors. In this Review, we summarize the mechanisms and consequences of T cell-microglia interactions as identified with in vitro experiments and animal models, and discuss the challenges that arise when translating this preclinical knowledge to MS in humans. We also consider therapeutic approaches to MS of which the mechanisms involve prevention or modulation of T cell and microglia responses and their interactions.
Collapse
|
475
|
Bohlen CJ, Friedman BA, Dejanovic B, Sheng M. Microglia in Brain Development, Homeostasis, and Neurodegeneration. Annu Rev Genet 2019; 53:263-288. [PMID: 31518519 DOI: 10.1146/annurev-genet-112618-043515] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Advances in human genetics have implicated a growing number of genes in neurodegenerative diseases, providing insight into pathological processes. For Alzheimer disease in particular, genome-wide association studies and gene expression studies have emphasized the pathogenic contributions from microglial cells and motivated studies of microglial function/dysfunction. Here, we summarize recent genetic evidence for microglial involvement in neurodegenerative disease with a focus on Alzheimer disease, for which the evidence is most compelling. To provide context for these genetic discoveries, we discuss how microglia influence brain development and homeostasis, how microglial characteristics change in disease, and which microglial activities likely influence the course of neurodegeneration. In all, we aim to synthesize varied aspects of microglial biology and highlight microglia as possible targets for therapeutic interventions in neurodegenerative disease.
Collapse
Affiliation(s)
- Christopher J Bohlen
- Department of Neuroscience, Genentech, South San Francisco, California 94080, USA; ,
| | - Brad A Friedman
- Department of Bioinformatics, Genentech, South San Francisco, California 94080, USA
| | - Borislav Dejanovic
- Department of Neuroscience, Genentech, South San Francisco, California 94080, USA; ,
| | - Morgan Sheng
- Department of Neuroscience, Genentech, South San Francisco, California 94080, USA; ,
| |
Collapse
|
476
|
Murakami Y, Ishikawa K, Nakao S, Sonoda KH. Innate immune response in retinal homeostasis and inflammatory disorders. Prog Retin Eye Res 2019; 74:100778. [PMID: 31505218 DOI: 10.1016/j.preteyeres.2019.100778] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 08/12/2019] [Accepted: 09/02/2019] [Indexed: 01/03/2023]
Abstract
Innate immune cells such as neutrophils, monocyte-macrophages and microglial cells are pivotal for the health and disease of the retina. For the maintenance of retinal homeostasis, these cells and immunosuppressive molecules in the eye actively regulate the induction and the expression of inflammation in order to prevent excessive activation and subsequent tissue damage. In the disease context, these regulatory mechanisms are modulated genetically and/or by environmental stimuli such as damage-associated molecular patterns (DAMPs), and a chronic innate immune response regulates or contributes to the formation of diverse retinal disorders such as uveitis, retinitis pigmentosa, retinal vascular diseases and retinal fibrosis. Here we summarize the recent knowledge regarding the innate immune response in both ocular immune regulation and inflammatory retinal diseases, and we describe the potential of the innate immune response as a biomarker and therapeutic target.
Collapse
Affiliation(s)
- Yusuke Murakami
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka, 812-8582, Japan
| | - Keijiro Ishikawa
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka, 812-8582, Japan
| | - Shintaro Nakao
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka, 812-8582, Japan
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Graduate School of Medical Science, Kyushu University, Fukuoka, 812-8582, Japan.
| |
Collapse
|
477
|
Sodium oligomannate therapeutically remodels gut microbiota and suppresses gut bacterial amino acids-shaped neuroinflammation to inhibit Alzheimer's disease progression. Cell Res 2019; 29:787-803. [PMID: 31488882 PMCID: PMC6796854 DOI: 10.1038/s41422-019-0216-x] [Citation(s) in RCA: 661] [Impact Index Per Article: 132.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 07/29/2019] [Indexed: 01/01/2023] Open
Abstract
Recently, increasing evidence has suggested the association between gut dysbiosis and Alzheimer’s disease (AD) progression, yet the role of gut microbiota in AD pathogenesis remains obscure. Herein, we provide a potential mechanistic link between gut microbiota dysbiosis and neuroinflammation in AD progression. Using AD mouse models, we discovered that, during AD progression, the alteration of gut microbiota composition leads to the peripheral accumulation of phenylalanine and isoleucine, which stimulates the differentiation and proliferation of pro-inflammatory T helper 1 (Th1) cells. The brain-infiltrated peripheral Th1 immune cells are associated with the M1 microglia activation, contributing to AD-associated neuroinflammation. Importantly, the elevation of phenylalanine and isoleucine concentrations and the increase of Th1 cell frequency in the blood were also observed in two small independent cohorts of patients with mild cognitive impairment (MCI) due to AD. Furthermore, GV-971, a sodium oligomannate that has demonstrated solid and consistent cognition improvement in a phase 3 clinical trial in China, suppresses gut dysbiosis and the associated phenylalanine/isoleucine accumulation, harnesses neuroinflammation and reverses the cognition impairment. Together, our findings highlight the role of gut dysbiosis-promoted neuroinflammation in AD progression and suggest a novel strategy for AD therapy by remodelling the gut microbiota.
Collapse
|
478
|
Stratoulias V, Venero JL, Tremblay M, Joseph B. Microglial subtypes: diversity within the microglial community. EMBO J 2019; 38:e101997. [PMID: 31373067 PMCID: PMC6717890 DOI: 10.15252/embj.2019101997] [Citation(s) in RCA: 340] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/29/2019] [Accepted: 05/03/2019] [Indexed: 12/03/2022] Open
Abstract
Microglia are brain-resident macrophages forming the first active immune barrier in the central nervous system. They fulfill multiple functions across development and adulthood and under disease conditions. Current understanding revolves around microglia acquiring distinct phenotypes upon exposure to extrinsic cues in their environment. However, emerging evidence suggests that microglia display differences in their functions that are not exclusively driven by their milieu, rather by the unique properties these cells possess. This microglial intrinsic heterogeneity has been largely overlooked, favoring the prevailing view that microglia are a single-cell type endowed with spectacular plasticity, allowing them to acquire multiple phenotypes and thereby fulfill their numerous functions in health and disease. Here, we review the evidence that microglia might form a community of cells in which each member (or "subtype") displays intrinsic properties and performs unique functions. Distinctive features and functional implications of several microglial subtypes are considered, across contexts of health and disease. Finally, we suggest that microglial subtype categorization shall be based on function and we propose ways for studying them. Hence, we advocate that plasticity (reaction states) and diversity (subtypes) should both be considered when studying the multitasking microglia.
Collapse
Affiliation(s)
- Vassilis Stratoulias
- Toxicology UnitInstitute of Environmental MedicineKarolinska InstitutetStockholmSweden
| | - Jose Luis Venero
- Departamento de Bioquímica y Biología MolecularFacultad de FarmaciaUniversidad de SevillaSevillaSpain
- Instituto de Biomedicina de Sevilla‐Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevillaSpain
| | - Marie‐Ève Tremblay
- Department of Molecular MedicineUniversité LavalQuebecQCCanada
- Axe NeurosciencesCentre de Recherche du CHU de Québec‐Université LavalQuebecQCCanada
| | - Bertrand Joseph
- Toxicology UnitInstitute of Environmental MedicineKarolinska InstitutetStockholmSweden
| |
Collapse
|
479
|
Abstract
Microglia are the primary innate immune cells in the CNS. In the healthy brain, they exhibit a unique molecular homeostatic 'signature', consisting of a specific transcriptional profile and surface protein expression pattern, which differs from that of tissue macrophages. In recent years, there have been a number of important advances in our understanding of the molecular signatures of homeostatic microglia and disease-associated microglia that have provided insight into how these cells are regulated in health and disease and how they contribute to the maintenance of the neural environment.
Collapse
|
480
|
Rustenhoven J, Herz J, Kipnis J. Old T Cells Interfer(on) with Neurogenesis. Trends Immunol 2019; 40:783-785. [PMID: 31422900 DOI: 10.1016/j.it.2019.07.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 07/31/2019] [Indexed: 10/26/2022]
Abstract
Adult neurogenesis plays an important role in brain function and declines with aging. A recent report demonstrates clonal T cell expansion within neurogenic niches of the aged brain, impairing neurogenesis through IFNγ signaling (Dulken et al.,Nature, 2019). These results highlight T cells as important contributors to and potential therapeutic targets for age-related brain dysfunction.
Collapse
Affiliation(s)
- Justin Rustenhoven
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA 22908, USA; Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| | - Jasmin Herz
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA 22908, USA; Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA 22908, USA; Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
481
|
Park C, Ponath G, Levine-Ritterman M, Bull E, Swanson EC, De Jager PL, Segal BM, Pitt D. The landscape of myeloid and astrocyte phenotypes in acute multiple sclerosis lesions. Acta Neuropathol Commun 2019; 7:130. [PMID: 31405387 PMCID: PMC6689891 DOI: 10.1186/s40478-019-0779-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 07/23/2019] [Indexed: 02/06/2023] Open
Abstract
Activated myeloid cells and astrocytes are the predominant cell types in active multiple sclerosis (MS) lesions. Both cell types can adopt diverse functional states that play critical roles in lesion formation and resolution. In order to identify phenotypic subsets of myeloid cells and astrocytes, we profiled two active MS lesions with thirteen glial activation markers using imaging mass cytometry (IMC), a method for multiplexed labeling of histological sections. In the acutely demyelinating lesion, we found multiple distinct myeloid and astrocyte phenotypes that populated separate lesion zones. In the post-demyelinating lesion, phenotypes were less distinct and more uniformly distributed. In both lesions cell-to-cell interactions were not random, but occurred between specific glial subpopulations and lymphocytes. Finally, we demonstrated that myeloid, but not astrocyte phenotypes were activated along a lesion rim-to-center gradient, and that marker expression in glial cells at the lesion rim was driven more by cell-extrinsic factors than in cells at the center. This proof-of-concept study demonstrates that highly multiplexed tissue imaging, combined with the appropriate computational tools, is a powerful approach to study heterogeneity, spatial distribution and cellular interactions in the context of MS lesions. Identifying glial phenotypes and their interactions at different lesion stages may provide novel therapeutic targets for inhibiting acute demyelination and low-grade, chronic inflammation.
Collapse
Affiliation(s)
- Calvin Park
- Department of Neurology, Yale School of Medicine, 300 George Street, Suite 353I, New Haven, CT 06511 USA
| | - Gerald Ponath
- Department of Neurology, Yale School of Medicine, 300 George Street, Suite 353I, New Haven, CT 06511 USA
| | - Maya Levine-Ritterman
- Department of Neurology, Yale School of Medicine, 300 George Street, Suite 353I, New Haven, CT 06511 USA
| | - Edward Bull
- Department of Neurology, Yale School of Medicine, 300 George Street, Suite 353I, New Haven, CT 06511 USA
| | | | - Philip L. De Jager
- Department of Neurology, Columbia University Medical Center, New York, NY USA
| | | | - David Pitt
- Department of Neurology, Yale School of Medicine, 300 George Street, Suite 353I, New Haven, CT 06511 USA
| |
Collapse
|
482
|
Sie C, Perez LG, Kreutzfeldt M, Potthast M, Ohnmacht C, Merkler D, Huber S, Krug A, Korn T. Dendritic Cell Accumulation in the Gut and Central Nervous System Is Differentially Dependent on α4 Integrins. THE JOURNAL OF IMMUNOLOGY 2019; 203:1417-1427. [PMID: 31399516 DOI: 10.4049/jimmunol.1900468] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/13/2019] [Indexed: 12/11/2022]
Abstract
Homing of pathogenic CD4+ T cells to the CNS is dependent on α4 integrins. However, it is uncertain whether α4 integrins are also required for the migration of dendritic cell (DC) subsets, which sample Ags from nonlymphoid tissues to present it to T cells. In this study, after genetic ablation of Itga4 in DCs and monocytes in mice via the promoters of Cd11c and Lyz2 (also known as LysM), respectively, the recruitment of α4 integrin-deficient conventional and plasmacytoid DCs to the CNS was unaffected, whereas α4 integrin-deficient, monocyte-derived DCs accumulated less efficiently in the CNS during experimental autoimmune encephalomyelitis in a competitive setting than their wild-type counterparts. In a noncompetitive setting, α4 integrin deficiency on monocyte-derived DCs was fully compensated. In contrast, in small intestine and colon, the fraction of α4 integrin-deficient CD11b+CD103+ DCs was selectively reduced in steady-state. Yet, T cell-mediated inflammation and host defense against Citrobacter rodentium were not impaired in the absence of α4 integrins on DCs. Thus, inflammatory conditions can promote an environment that is indifferent to α4 integrin expression by DCs.
Collapse
Affiliation(s)
- Christopher Sie
- Abteilung für Experimentelle Neuroimmunologie, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany.,Klinik für Neurologie, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Laura Garcia Perez
- I. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Mario Kreutzfeldt
- Division of Clinical Pathology, Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Maria Potthast
- Center of Allergy and Environment, Helmholtz Center and Technical University of Munich, 80802 Munich, Germany
| | - Caspar Ohnmacht
- Center of Allergy and Environment, Helmholtz Center and Technical University of Munich, 80802 Munich, Germany
| | - Doron Merkler
- Division of Clinical Pathology, Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland
| | - Samuel Huber
- I. Medizinische Klinik und Poliklinik, Universitätsklinikum Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Anne Krug
- Institute for Immunology, Biomedical Center, Ludwig Maximilians University of Munich, 82152 Planegg-Martinsried, Germany; and
| | - Thomas Korn
- Abteilung für Experimentelle Neuroimmunologie, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany; .,Klinik für Neurologie, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany.,Munich Cluster for Systems Neurology, SyNergy, 81377 Munich, Germany
| |
Collapse
|
483
|
Chen G, Zhang YQ, Qadri YJ, Serhan CN, Ji RR. Microglia in Pain: Detrimental and Protective Roles in Pathogenesis and Resolution of Pain. Neuron 2019; 100:1292-1311. [PMID: 30571942 DOI: 10.1016/j.neuron.2018.11.009] [Citation(s) in RCA: 481] [Impact Index Per Article: 96.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 10/22/2018] [Accepted: 11/02/2018] [Indexed: 12/18/2022]
Abstract
The previous decade has seen a rapid increase in microglial studies on pain, with a unique focus on microgliosis in the spinal cord after nerve injury and neuropathic pain. Numerous signaling molecules are altered in microglia and contribute to the pathogenesis of pain. Here, we discuss how microglial signaling regulates spinal cord synaptic plasticity in acute and chronic pain conditions with different degrees and variations of microgliosis. We highlight that microglial mediators such as pro- and anti-inflammatory cytokines are powerful neuromodulators that regulate synaptic transmission and pain via neuron-glial interactions. We also reveal an emerging role of microglia in the resolution of pain, in part via specialized pro-resolving mediators including resolvins, protectins, and maresins. We also discuss a possible role of microglia in chronic itch.
Collapse
Affiliation(s)
- Gang Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Yu-Qiu Zhang
- Institute of Neurobiology, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Yawar J Qadri
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Charles N Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Hale Transformative Medicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
484
|
Li T, Zhu J. Entanglement of CCR5 and Alzheimer's Disease. Front Aging Neurosci 2019; 11:209. [PMID: 31447666 PMCID: PMC6692443 DOI: 10.3389/fnagi.2019.00209] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 07/24/2019] [Indexed: 12/22/2022] Open
Abstract
Although the mechanisms of Alzheimer's disease are diverse and unclear, the past 20 years have witnessed the unprecedented development of the AD inflammation theory. As a key inflammatory receptor family, the C-C chemokine receptor family is a remarkable participant in the cause of Alzheimer's disease; of this family, CCR5 is the most widely studied. CCR5 is an essential entrance when HIV infects immune cells and is also involved in other inflammatory and immune activities. New evidence on the inevitably intertwined link between Alzheimer's disease and CCR5 indicates that CCR5 accelerates the development of Alzheimer's disease, and few studies disputed it. The role of CCR5 in Alzheimer's disease remains elusive. However, as the research progresses, this intricate relationship will gradually be uncovered.
Collapse
Affiliation(s)
- Tianwen Li
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, Shanghai, China
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianhong Zhu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, Shanghai, China
- Institutes of Brain Science, Shanghai, China
| |
Collapse
|
485
|
Eyo UB, Wu LJ. Microglia: Lifelong patrolling immune cells of the brain. Prog Neurobiol 2019; 179:101614. [PMID: 31075285 PMCID: PMC6599472 DOI: 10.1016/j.pneurobio.2019.04.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/11/2019] [Accepted: 04/19/2019] [Indexed: 02/02/2023]
Abstract
Microglial cells are the predominant parenchymal immune cell of the brain. Recent evidence suggests that like peripheral immune cells, microglia patrol the brain in health and disease. Reviewing these data, we first examine the evidence that microglia invade the brain mesenchyme early in embryonic development, establish residence therein, proliferate and subsequently maintain their numbers throughout life. We, then, summarize established and novel evidence for microglial process surveillance in the healthy and injured brain. Finally, we discuss emerging evidence for microglial cell body dynamics that challenge existing assumptions of their sessile nature. We conclude that microglia are long-lived immune cells that patrol the brain through both cell body and process movements. This recognition has significant implications for neuroimmune interactions throughout the animal lifespan.
Collapse
Affiliation(s)
- Ukpong B Eyo
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA; Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
486
|
Fani Maleki A, Rivest S. Innate Immune Cells: Monocytes, Monocyte-Derived Macrophages and Microglia as Therapeutic Targets for Alzheimer's Disease and Multiple Sclerosis. Front Cell Neurosci 2019; 13:355. [PMID: 31427930 PMCID: PMC6690269 DOI: 10.3389/fncel.2019.00355] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 07/17/2019] [Indexed: 12/20/2022] Open
Abstract
The immune system provides protection in the CNS via resident microglial cells and those that traffic into it in the course of pathological challenges. These populations of cells are key players in modulating immune functions that are involved in disease outcomes. In this review, we briefly summarize and highlight the current state of knowledge of the differential contributions of microglia and monocytes in Alzheimer’s disease and multiple sclerosis. The role of innate immunity is frequently seen as a Yin and Yang in both diseases, but this depends on the environment, pre-clinical disease models and the type of cells involved.
Collapse
Affiliation(s)
- Adham Fani Maleki
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec City, QC, Canada
| | - Serge Rivest
- Neuroscience Laboratory, CHU de Québec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec City, QC, Canada
| |
Collapse
|
487
|
Kierdorf K, Masuda T, Jordão MJC, Prinz M. Macrophages at CNS interfaces: ontogeny and function in health and disease. Nat Rev Neurosci 2019; 20:547-562. [PMID: 31358892 DOI: 10.1038/s41583-019-0201-x] [Citation(s) in RCA: 225] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2019] [Indexed: 12/16/2022]
Abstract
The segregation and limited regenerative capacity of the CNS necessitate a specialized and tightly regulated resident immune system that continuously guards the CNS against invading pathogens and injury. Immunity in the CNS has generally been attributed to neuron-associated microglia in the parenchyma, whose origin and functions have recently been elucidated. However, there are several other specialized macrophage populations at the CNS borders, including dural, leptomeningeal, perivascular and choroid plexus macrophages (collectively known as CNS-associated macrophages (CAMs)), whose origins and roles in health and disease have remained largely uncharted. CAMs are thought to be involved in regulating the fine balance between the proper segregation of the CNS, on the one hand, and the essential exchange between the CNS parenchyma and the periphery, on the other. Recent studies that have been empowered by major technological advances have shed new light on these cells and suggest central roles for CAMs in CNS physiology and in the pathogenesis of diseases.
Collapse
Affiliation(s)
- Katrin Kierdorf
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Takahiro Masuda
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany. .,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,Centre for Biological Signalling Studies (BIOSS), University of Freiburg, Freiburg, Germany.
| |
Collapse
|
488
|
Schulz M, Salamero-Boix A, Niesel K, Alekseeva T, Sevenich L. Microenvironmental Regulation of Tumor Progression and Therapeutic Response in Brain Metastasis. Front Immunol 2019; 10:1713. [PMID: 31396225 PMCID: PMC6667643 DOI: 10.3389/fimmu.2019.01713] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/09/2019] [Indexed: 12/14/2022] Open
Abstract
Cellular and non-cellular components of the tumor microenvironment (TME) are emerging as key regulators of primary tumor progression, organ-specific metastasis, and therapeutic response. In the era of TME-targeted- and immunotherapies, cancer-associated inflammation has gained increasing attention. In this regard, the brain represents a unique and highly specialized organ. It has long been regarded as an immunological sanctuary site where the presence of the blood brain barrier (BBB) and blood cerebrospinal fluid barrier (BCB) restricts the entry of immune cells from the periphery. Consequently, tumor cells that metastasize to the brain were thought to be shielded from systemic immune surveillance and destruction. However, the detailed characterization of the immune landscape within border-associated areas of the central nervous system (CNS), such as the meninges and the choroid plexus, as well as the discovery of lymphatics and channels that connect the CNS with the periphery, have recently challenged the dogma of the immune privileged status of the brain. Moreover, the presence of brain metastases (BrM) disrupts the integrity of the BBB and BCB. Indeed, BrM induce the recruitment of different immune cells from the myeloid and lymphoid lineage to the CNS. Blood-borne immune cells together with brain-resident cell-types, such as astrocytes, microglia, and neurons, form a highly complex and dynamic TME that affects tumor cell survival and modulates the mode of immune responses that are elicited by brain metastatic tumor cells. In this review, we will summarize recent findings on heterotypic interactions within the brain metastatic TME and highlight specific functions of brain-resident and recruited cells at different rate-limiting steps of the metastatic cascade. Based on the insight from recent studies, we will discuss new opportunities and challenges for TME-targeted and immunotherapies for BrM.
Collapse
Affiliation(s)
- Michael Schulz
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany.,Biological Sciences, Faculty 15, Goethe University, Frankfurt, Germany
| | - Anna Salamero-Boix
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany
| | - Katja Niesel
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany
| | - Tijna Alekseeva
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany
| | - Lisa Sevenich
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany.,Frankfurt Cancer Institute, Goethe University, Frankfurt, Germany.,German Cancer Consortium (DKTK, Partner Site Frankfurt/Mainz) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
489
|
Naseri NN, Wang H, Guo J, Sharma M, Luo W. The complexity of tau in Alzheimer's disease. Neurosci Lett 2019; 705:183-194. [PMID: 31028844 PMCID: PMC7060758 DOI: 10.1016/j.neulet.2019.04.022] [Citation(s) in RCA: 183] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 03/14/2019] [Accepted: 04/08/2019] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is characterized by two major pathological lesions in the brain, amyloid plaques and neurofibrillary tangles (NFTs) composed mainly of amyloid-β (Aβ) peptides and hyperphosphorylated tau, respectively. Although accumulation of toxic Aβ species in the brain has been proposed as one of the important early events in AD, continued lack of success of clinical trials based on Aβ-targeting drugs has triggered the field to seek out alternative disease mechanisms and related therapeutic strategies. One of the new approaches is to uncover novel roles of pathological tau during disease progression. This review will primarily focus on recent advances in understanding the contributions of tau to AD.
Collapse
Affiliation(s)
- Nima N Naseri
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, USA.
| | - Hong Wang
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, USA
| | - Jennifer Guo
- The University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - Manu Sharma
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, USA
| | - Wenjie Luo
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, USA.
| |
Collapse
|
490
|
Single-cell analysis reveals T cell infiltration in old neurogenic niches. Nature 2019; 571:205-210. [PMID: 31270459 PMCID: PMC7111535 DOI: 10.1038/s41586-019-1362-5] [Citation(s) in RCA: 317] [Impact Index Per Article: 63.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 06/04/2019] [Indexed: 02/06/2023]
Abstract
The mammalian brain contains neurogenic niches comprising neural stem cells (NSCs) and other cell types. Neurogenic niches become less functional with age, but how they change during aging remains unclear. Here we perform single cell RNA-sequencing of young and old neurogenic niches in mice. Analysis of 14,685 single cell transcriptomes reveals a decrease in activated NSCs, changes in endothelial cells and microglia, and infiltration of T cells in old neurogenic niches. Surprisingly, T cells in old brains are clonally expanded and generally distinct from those in old blood, suggesting they may experience specific antigens. T cells from old brains express interferon γ, and the subset of NSCs with a high interferon response shows decreased proliferation in vivo. Interestingly, T cells can inhibit NSC proliferation in co-cultures and in vivo, in part by secreting interferon. Our study reveals an interaction between T cells and NSCs in old brains, opening potential avenues to counter age-related decline in brain function.
Collapse
|
491
|
Lanz TV, Pröbstel AK, Mildenberger I, Platten M, Schirmer L. Single-Cell High-Throughput Technologies in Cerebrospinal Fluid Research and Diagnostics. Front Immunol 2019; 10:1302. [PMID: 31244848 PMCID: PMC6579921 DOI: 10.3389/fimmu.2019.01302] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 05/22/2019] [Indexed: 01/08/2023] Open
Abstract
High-throughput single-cell technologies have recently emerged as essential tools in biomedical research with great potential for clinical pathology when studying liquid and solid biopsies. We provide an update on current single-cell methods in cerebrospinal fluid research and diagnostics, focusing on high-throughput cell-type specific proteomic and genomic technologies. Proteomic methods comprising flow cytometry and mass cytometry as well as genomic approaches including immune cell repertoire and single-cell transcriptomic studies are critically reviewed and future directions discussed.
Collapse
Affiliation(s)
- Tobias V. Lanz
- Department of Neurology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Anne-Katrin Pröbstel
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, United States
- Departments of Medicine and Biomedicine, Neurologic Clinic and Policlinic, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Iris Mildenberger
- Department of Neurology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Michael Platten
- Department of Neurology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- DKTK Clinical Cooperation Unit Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lucas Schirmer
- Department of Neurology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
492
|
Prionisti I, Bühler LH, Walker PR, Jolivet RB. Harnessing Microglia and Macrophages for the Treatment of Glioblastoma. Front Pharmacol 2019; 10:506. [PMID: 31231208 PMCID: PMC6560150 DOI: 10.3389/fphar.2019.00506] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/23/2019] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most malignant form of brain tumors, with a dismal prognosis. During the course of the disease, microglia and macrophages both infiltrate the tumor microenvironment and contribute considerably in glioma development. Thus, tumor-associated microglia and macrophages have recently emerged as potentially key therapeutic targets. Here, we review the physiology of microglia and their responses in brain cancer. We further discuss current treatment options for GBM using radiotherapy, and novel advances in our knowledge of microglia physiology, with emphasis on the recently discovered pathway that controls the baseline motility of microglia processes. We argue that the latter pathway is an interesting therapeutic avenue to pursue for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Ioanna Prionisti
- Division of Digestive and Transplantation Surgery, Geneva University Hospitals, Geneva, Switzerland
- Lemanic Neuroscience Doctoral School, Geneva, Switzerland
| | - Léo H. Bühler
- Division of Digestive and Transplantation Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Paul R. Walker
- Center for Translational Research in Onco-Hematology, Division of Oncology, Geneva University Hospitals – University of Geneva, Geneva, Switzerland
| | - Renaud B. Jolivet
- Département de Physique Nucléaire et Corpusculaire (DPNC), University of Geneva, Geneva, Switzerland
- European Organization for Nuclear Research (CERN), Geneva, Switzerland
| |
Collapse
|
493
|
Bonham LW, Sirkis DW, Yokoyama JS. The Transcriptional Landscape of Microglial Genes in Aging and Neurodegenerative Disease. Front Immunol 2019; 10:1170. [PMID: 31214167 PMCID: PMC6557985 DOI: 10.3389/fimmu.2019.01170] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 05/08/2019] [Indexed: 12/21/2022] Open
Abstract
Microglia, the brain-resident myeloid cells, are strongly implicated in Alzheimer's disease (AD) pathogenesis by human genetics. However, the mechanisms by which microglial gene expression is regulated in a region-specific manner over the course of normal aging and in neurodegenerative disease are only beginning to be deciphered. Herein, we used a specific marker of microglia (TMEM119) and a cell-type expression profiling tool (CellMapper) to identify a human microglial gene expression module. Surprisingly, we found that microglial module genes are robustly expressed in several healthy human brain regions known to be vulnerable in AD, in addition to other regions affected only later in disease or spared in AD. Surveying the microglial gene set for differential expression over the lifespan in mouse models of AD and a related tauopathy revealed that the majority of microglial module genes were significantly upregulated in cortex and hippocampus as a function of age and transgene status. Extending these results, we also observed significant upregulation of microglial module genes in several AD-affected brain regions in addition to other regions using postmortem brain tissue from human AD samples. In pathologically confirmed AD cases, we found preliminary evidence that microglial genes may be dysregulated in a sex-specific manner. Finally, we identified specific and significant overlap between the described microglial gene set—identified by unbiased co-expression analysis—and genes known to impart risk for AD. Our findings suggest that microglial genes show enriched expression in AD-vulnerable brain regions, are upregulated during aging and neurodegeneration in mice, and are upregulated in pathologically affected brain regions in AD. Taken together, our data-driven findings from multiple publicly accessible datasets reemphasize the importance of microglial gene expression alterations in AD and, more importantly, suggest that regional and sex-specific variation in microglial gene expression may be implicated in risk for and progression of neurodegenerative disease.
Collapse
Affiliation(s)
- Luke W Bonham
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, United States
| | - Daniel W Sirkis
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, United States
| | - Jennifer S Yokoyama
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
494
|
Brummelman J, Haftmann C, Núñez NG, Alvisi G, Mazza EMC, Becher B, Lugli E. Development, application and computational analysis of high-dimensional fluorescent antibody panels for single-cell flow cytometry. Nat Protoc 2019; 14:1946-1969. [PMID: 31160786 DOI: 10.1038/s41596-019-0166-2] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 03/12/2019] [Indexed: 02/07/2023]
Abstract
The interrogation of single cells is revolutionizing biology, especially our understanding of the immune system. Flow cytometry is still one of the most versatile and high-throughput approaches for single-cell analysis, and its capability has been recently extended to detect up to 28 colors, thus approaching the utility of cytometry by time of flight (CyTOF). However, flow cytometry suffers from autofluorescence and spreading error (SE) generated by errors in the measurement of photons mainly at red and far-red wavelengths, which limit barcoding and the detection of dim markers. Consequently, development of 28-color fluorescent antibody panels for flow cytometry is laborious and time consuming. Here, we describe the steps that are required to successfully achieve 28-color measurement capability. To do this, we provide a reference map of the fluorescence spreading errors in the 28-color space to simplify panel design and predict the success of fluorescent antibody combinations. Finally, we provide detailed instructions for the computational analysis of such complex data by existing, popular algorithms (PhenoGraph and FlowSOM). We exemplify our approach by designing a high-dimensional panel to characterize the immune system, but we anticipate that our approach can be used to design any high-dimensional flow cytometry panel of choice. The full protocol takes a few days to complete, depending on the time spent on panel design and data analysis.
Collapse
Affiliation(s)
- Jolanda Brummelman
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Claudia Haftmann
- Laboratory of Inflammation Research, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Nicolás Gonzalo Núñez
- Laboratory of Inflammation Research, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Giorgia Alvisi
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Emilia M C Mazza
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Burkhard Becher
- Laboratory of Inflammation Research, Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Enrico Lugli
- Laboratory of Translational Immunology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy. .,Flow Cytometry Core, Humanitas Clinical and Research Center, Rozzano, Milan, Italy.
| |
Collapse
|
495
|
Lipopolysaccharide-Induced Neuroinflammation as a Bridge to Understand Neurodegeneration. Int J Mol Sci 2019; 20:ijms20092293. [PMID: 31075861 PMCID: PMC6539529 DOI: 10.3390/ijms20092293] [Citation(s) in RCA: 299] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/03/2019] [Accepted: 05/05/2019] [Indexed: 12/19/2022] Open
Abstract
A large body of experimental evidence suggests that neuroinflammation is a key pathological event triggering and perpetuating the neurodegenerative process associated with many neurological diseases. Therefore, different stimuli, such as lipopolysaccharide (LPS), are used to model neuroinflammation associated with neurodegeneration. By acting at its receptors, LPS activates various intracellular molecules, which alter the expression of a plethora of inflammatory mediators. These factors, in turn, initiate or contribute to the development of neurodegenerative processes. Therefore, LPS is an important tool for the study of neuroinflammation associated with neurodegenerative diseases. However, the serotype, route of administration, and number of injections of this toxin induce varied pathological responses. Thus, here, we review the use of LPS in various models of neurodegeneration as well as discuss the neuroinflammatory mechanisms induced by this toxin that could underpin the pathological events linked to the neurodegenerative process.
Collapse
|
496
|
|
497
|
A single-cell atlas of mouse brain macrophages reveals unique transcriptional identities shaped by ontogeny and tissue environment. Nat Neurosci 2019; 22:1021-1035. [PMID: 31061494 DOI: 10.1038/s41593-019-0393-4] [Citation(s) in RCA: 556] [Impact Index Per Article: 111.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 03/21/2019] [Indexed: 12/24/2022]
Abstract
While the roles of parenchymal microglia in brain homeostasis and disease are fairly clear, other brain-resident myeloid cells remain less well understood. By dissecting border regions and combining single-cell RNA-sequencing with high-dimensional cytometry, bulk RNA-sequencing, fate-mapping and microscopy, we reveal the diversity of non-parenchymal brain macrophages. Border-associated macrophages (BAMs) residing in the dura mater, subdural meninges and choroid plexus consisted of distinct subsets with tissue-specific transcriptional signatures, and their cellular composition changed during postnatal development. BAMs exhibited a mixed ontogeny, and subsets displayed distinct self-renewal capacity following depletion and repopulation. Single-cell and fate-mapping analysis both suggested that there is a unique microglial subset residing on the apical surface of the choroid plexus epithelium. Finally, gene network analysis and conditional deletion revealed IRF8 as a master regulator that drives the maturation and diversity of brain macrophages. Our results provide a framework for understanding host-macrophage interactions in both the healthy and diseased brain.
Collapse
|
498
|
Sarntivijai S, He Y, Diehl AD. Cells in ExperimentaL Life Sciences (CELLS-2018): capturing the knowledge of normal and diseased cells with ontologies. BMC Bioinformatics 2019; 20:183. [PMID: 31272374 PMCID: PMC6509796 DOI: 10.1186/s12859-019-2721-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Cell cultures and cell lines are widely used in life science experiments. In conjunction with the 2018 International Conference on Biomedical Ontology (ICBO-2018), the 2nd International Workshop on Cells in ExperimentaL Life Science (CELLS-2018) focused on two themes of knowledge representation, for newly-discovered cell types and for cells in disease states. This workshop included five oral presentations and a general discussion session. Two new ontologies, including the Cancer Cell Ontology (CCL) and the Ontology for Stem Cell Investigations (OSCI), were reported in the workshop. In another representation, the Cell Line Ontology (CLO) framework was applied and extended to represent cell line cells used in China and their Chinese representation. Other presentations included a report on the application of ontologies to cross-compare cell types and marker patterns used in flow cytometry studies, and a presentation on new experimental findings about novel cell types based on single cell RNA sequencing assay and their corresponding ontological representation. The general discussion session focused on the ontology design patterns in representing newly-discovered cell types and cells in disease states.
Collapse
Affiliation(s)
| | - Yongqun He
- University of Michigan Medical School, Ann Arbor, MI USA
| | - Alexander D. Diehl
- Department of Biomedical Informatics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY USA
| |
Collapse
|
499
|
Sala Frigerio C, Wolfs L, Fattorelli N, Thrupp N, Voytyuk I, Schmidt I, Mancuso R, Chen WT, Woodbury ME, Srivastava G, Möller T, Hudry E, Das S, Saido T, Karran E, Hyman B, Perry VH, Fiers M, De Strooper B. The Major Risk Factors for Alzheimer's Disease: Age, Sex, and Genes Modulate the Microglia Response to Aβ Plaques. Cell Rep 2019; 27:1293-1306.e6. [PMID: 31018141 PMCID: PMC7340153 DOI: 10.1016/j.celrep.2019.03.099] [Citation(s) in RCA: 484] [Impact Index Per Article: 96.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/05/2019] [Accepted: 03/26/2019] [Indexed: 12/21/2022] Open
Abstract
Gene expression profiles of more than 10,000 individual microglial cells isolated from cortex and hippocampus of male and female AppNL-G-F mice over time demonstrate that progressive amyloid-β accumulation accelerates two main activated microglia states that are also present during normal aging. Activated response microglia (ARMs) are composed of specialized subgroups overexpressing MHC type II and putative tissue repair genes (Dkk2, Gpnmb, and Spp1) and are strongly enriched with Alzheimer's disease (AD) risk genes. Microglia from female mice progress faster in this activation trajectory. Similar activated states are also found in a second AD model and in human brain. Apoe, the major genetic risk factor for AD, regulates the ARMs but not the interferon response microglia (IRMs). Thus, the ARMs response is the converging point for aging, sex, and genetic AD risk factors.
Collapse
Affiliation(s)
- Carlo Sala Frigerio
- VIB Centre for Brain Disease Research, Leuven, Belgium; University of Leuven, Department of Neurosciences and Leuven Brain Institute, Leuven, Belgium; UK Dementia Research Institute, University College London, London, UK.
| | - Leen Wolfs
- VIB Centre for Brain Disease Research, Leuven, Belgium; University of Leuven, Department of Neurosciences and Leuven Brain Institute, Leuven, Belgium
| | - Nicola Fattorelli
- VIB Centre for Brain Disease Research, Leuven, Belgium; University of Leuven, Department of Neurosciences and Leuven Brain Institute, Leuven, Belgium
| | - Nicola Thrupp
- VIB Centre for Brain Disease Research, Leuven, Belgium; University of Leuven, Department of Neurosciences and Leuven Brain Institute, Leuven, Belgium
| | - Iryna Voytyuk
- VIB Centre for Brain Disease Research, Leuven, Belgium; University of Leuven, Department of Neurosciences and Leuven Brain Institute, Leuven, Belgium
| | - Inga Schmidt
- VIB Centre for Brain Disease Research, Leuven, Belgium; University of Leuven, Department of Neurosciences and Leuven Brain Institute, Leuven, Belgium
| | - Renzo Mancuso
- VIB Centre for Brain Disease Research, Leuven, Belgium; University of Leuven, Department of Neurosciences and Leuven Brain Institute, Leuven, Belgium
| | - Wei-Ting Chen
- VIB Centre for Brain Disease Research, Leuven, Belgium; University of Leuven, Department of Neurosciences and Leuven Brain Institute, Leuven, Belgium
| | - Maya E Woodbury
- Foundational Neuroscience Center, AbbVie, Inc., Cambridge, MA, USA
| | - Gyan Srivastava
- Foundational Neuroscience Center, AbbVie, Inc., Cambridge, MA, USA
| | - Thomas Möller
- Foundational Neuroscience Center, AbbVie, Inc., Cambridge, MA, USA
| | - Eloise Hudry
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Sudeshna Das
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Takaomi Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako-shi, Saitama, Japan
| | - Eric Karran
- Foundational Neuroscience Center, AbbVie, Inc., Cambridge, MA, USA
| | - Bradley Hyman
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - V Hugh Perry
- UK Dementia Research Institute, University College London, London, UK; Centre for Biological Sciences, University of Southampton, Southampton, UK
| | - Mark Fiers
- VIB Centre for Brain Disease Research, Leuven, Belgium; University of Leuven, Department of Neurosciences and Leuven Brain Institute, Leuven, Belgium
| | - Bart De Strooper
- VIB Centre for Brain Disease Research, Leuven, Belgium; University of Leuven, Department of Neurosciences and Leuven Brain Institute, Leuven, Belgium; UK Dementia Research Institute, University College London, London, UK.
| |
Collapse
|
500
|
Distinct metabolic patterns during microglial remodeling by oleate and palmitate. Biosci Rep 2019; 39:BSR20190072. [PMID: 30867255 PMCID: PMC6449521 DOI: 10.1042/bsr20190072] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/20/2019] [Accepted: 03/06/2019] [Indexed: 12/13/2022] Open
Abstract
Microglial activation by oleate and palmitate differentially modulates brain inflammatory status. However, the metabolic reprogramming supporting these reactive phenotypes remains unknown. Employing real-time metabolic measurements and lipidomic analysis, we show that both fatty acids promote microglial oxidative metabolism, while lipopolysaccharide (LPS) enhances glycolytic rates. Interestingly, oleate treatment was followed by enrichment in storage lipids bound to polyunsaturated fatty acids (PUFA), in parallel with protection against oxidative imbalance. Palmitate, in turn, induced a distinct lipid distribution defined by PUFA linked to membrane phospholipids, which are more susceptible to lipid peroxidation and inflammatory signaling cascades. This distribution was mirrored by LPS treatment, which led to a strong pro-inflammatory phenotype in microglia. Thus, although both oleate and palmitate preserve mitochondrial function, a contrasting lipid distribution supports differences in fatty acid-induced neuroinflammation. These data reinforce the concept that reactive microglial profiles are achieved by stimulus-evoked remodeling in cell metabolism.
Collapse
|