351
|
Ago T. [Why are pericytes important for brain functions?]. Rinsho Shinkeigaku 2019; 59:707-715. [PMID: 31656270 DOI: 10.5692/clinicalneurol.cn-001357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Pericytes are mural cells embedded in the basal membrane surrounding endothelial cells in capillary and small vessels (from precapillary arterioles to postcapillary venules). They exist with a high coverage ratio to endothelial cells in the brain and play crucial roles in the formation and maintenance of the blood-brain barrier and the control of blood flow through a close interaction with endothelial cells. Thus, intactness of pericyte is absolutely needed for neuronal/brain functions. Ageing, life-style diseases, hypoperfusion/ischemia, drugs, and genetic factors can primarily cause pericyte dysfunctions, thereby leading to the development or progression of various brain disorders, including cerebrovascular diseases. Because pericytes also play an important role in tissue repair after brain injuries, they have received much attention as a therapeutic target even from the standpoint of functional recovery.
Collapse
Affiliation(s)
- Tetsuro Ago
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University
| |
Collapse
|
352
|
Neumann B, Baror R, Zhao C, Segel M, Dietmann S, Rawji KS, Foerster S, McClain CR, Chalut K, van Wijngaarden P, Franklin RJM. Metformin Restores CNS Remyelination Capacity by Rejuvenating Aged Stem Cells. Cell Stem Cell 2019; 25:473-485.e8. [PMID: 31585093 PMCID: PMC6863391 DOI: 10.1016/j.stem.2019.08.015] [Citation(s) in RCA: 291] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 06/27/2019] [Accepted: 08/23/2019] [Indexed: 01/06/2023]
Abstract
The age-related failure to produce oligodendrocytes from oligodendrocyte progenitor cells (OPCs) is associated with irreversible neurodegeneration in multiple sclerosis (MS). Consequently, regenerative approaches have significant potential for treating chronic demyelinating diseases. Here, we show that the differentiation potential of adult rodent OPCs decreases with age. Aged OPCs become unresponsive to pro-differentiation signals, suggesting intrinsic constraints on therapeutic approaches aimed at enhancing OPC differentiation. This decline in functional capacity is associated with hallmarks of cellular aging, including decreased metabolic function and increased DNA damage. Fasting or treatment with metformin can reverse these changes and restore the regenerative capacity of aged OPCs, improving remyelination in aged animals following focal demyelination. Aged OPCs treated with metformin regain responsiveness to pro-differentiation signals, suggesting synergistic effects of rejuvenation and pro-differentiation therapies. These findings provide insight into aging-associated remyelination failure and suggest therapeutic interventions for reversing such declines in chronic disease.
Collapse
Affiliation(s)
- Björn Neumann
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Roey Baror
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Chao Zhao
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Michael Segel
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Sabine Dietmann
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Khalil S Rawji
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Sarah Foerster
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Crystal R McClain
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Kevin Chalut
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK; Department of Physics, University of Cambridge, Cambridge CB3 0HE, UK
| | - Peter van Wijngaarden
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK; Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia; Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Australia.
| | - Robin J M Franklin
- Wellcome - MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0AW, UK.
| |
Collapse
|
353
|
Wang A, Wang R, Cui D, Huang X, Yuan L, Liu H, Fu Y, Liang L, Wang W, He Q, Shi C, Guan X, Teng Z, Zhao G, Li Y, Gao Y, Han H. The Drainage of Interstitial Fluid in the Deep Brain is Controlled by the Integrity of Myelination. Aging Dis 2019; 10:937-948. [PMID: 31595193 PMCID: PMC6764732 DOI: 10.14336/ad.2018.1206] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/26/2018] [Indexed: 12/17/2022] Open
Abstract
In searching for the drainage route of the interstitial fluid (ISF) in the deep brain, we discovered a regionalized ISF drainage system as well as a new function of myelin in regulating the drainage. The traced ISF from the caudate nucleus drained to the ipsilateral cortex along myelin fiber tracts, while in the opposite direction, its movement to the adjacent thalamus was completely impeded by a barrier structure, which was identified as the converged, compact myelin fascicle. The regulating and the barrier effects of myelin were unchanged in AQP4-knockout rats but were impaired as the integrity of boundary structure of drainage system was destroyed in a demyelinated rat model. We thus proposed that the brain homeostasis was maintained within each ISF drainage division locally, rather than across the brain as a whole. A new brain division system and a new pathogenic mechanism of demyelination are therefore proposed.
Collapse
Affiliation(s)
- Aibo Wang
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Rui Wang
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Dehua Cui
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Xinrui Huang
- Department of Biophysics, School of Basic Medical Sciences, Peking University, Beijing, China.
| | - Lan Yuan
- Peking University Medical and Health Analysis Center, Peking University Health Science Center, Beijing, China.
| | - Huipo Liu
- Institute of Applied Physics and Computational Mathematics, Beijing, China.
| | - Yu Fu
- Department of Neurology, Peking University Third Hospital, Beijing, China.
| | - Lei Liang
- Department of Medical Chemistry, School of Pharmaceutical Sciences, Peking University, Beijing, China.
| | - Wei Wang
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Qingyuan He
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Chunyan Shi
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Xiangping Guan
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Ze Teng
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Guomei Zhao
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Yuanyuan Li
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Yajuan Gao
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| | - Hongbin Han
- Department of Radiology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Magnetic Resonance Imaging Equipment and Technique, Beijing, China.
| |
Collapse
|
354
|
Blumenfeld Kan S, Staun-Ram E, Golan D, Miller A. HDL-cholesterol elevation associated with fingolimod and dimethyl fumarate therapies in multiple sclerosis. Mult Scler J Exp Transl Clin 2019; 5:2055217319882720. [PMID: 31662882 PMCID: PMC6794658 DOI: 10.1177/2055217319882720] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/01/2019] [Accepted: 09/16/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Patients with Multiple Sclerosis (PwMS) display altered lipoproteins levels and function, which seem to affect disease risk and progress. Whether disease-modifying therapies affect the lipoprotein profile in PwMS has scarcely been studied. OBJECTIVE The study aims to assess whether fingolimod and dimethyl fumarate (DMF) affect lipoproteins in PwMS. METHODS We compared retrospectively the blood lipoprotein levels of 29 fingolimod-treated and 41 DMF-treated patients before and after 3 and 12 months of therapy. Patients treated with cholesterol-reducing medications were not included. Data on weight change and disease activity during 1-year follow-up were obtained. RESULTS HDL level, HDL/LDL ratio and HDL/total cholesterol ratio were increased in both treatment groups after 3 months' therapy and sustained, with no change in LDL or triglycerides. While at baseline only 26% of patients met the recommended minimum of HDL 60 mg/dl, after 3 months' therapy, 43% of fingolimod-treated and 47% of DMF-treated patients reached the recommended level. The majority of patients had no weight reduction. CONCLUSIONS Fingolimod and DMF therapies are associated with a specific increase in HDL in PwMS. Further studies are required to validate these findings and their potential implication as biomarker of reduced inflammatory state and/or reduced risk of neurodegeneration or cardiovascular comorbidity.
Collapse
Affiliation(s)
- S Blumenfeld Kan
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - E Staun-Ram
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Neuroimmunology Unit & Multiple Sclerosis Center, Department of Neurology, Carmel Medical Center, Haifa, Israel
| | - D Golan
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Neuroimmunology Unit & Multiple Sclerosis Center, Department of Neurology, Carmel Medical Center, Haifa, Israel
| | - A Miller
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Neuroimmunology Unit & Multiple Sclerosis Center, Department of Neurology, Carmel Medical Center, Haifa, Israel
| |
Collapse
|
355
|
CPT1a downregulation protects against cholesterol-induced fibrosis in tubular epithelial cells by downregulating TGFβ-1 and inflammasome. Biochem Biophys Res Commun 2019; 517:715-721. [DOI: 10.1016/j.bbrc.2019.07.121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 07/30/2019] [Indexed: 01/10/2023]
|
356
|
Stadelmann C, Timmler S, Barrantes-Freer A, Simons M. Myelin in the Central Nervous System: Structure, Function, and Pathology. Physiol Rev 2019; 99:1381-1431. [PMID: 31066630 DOI: 10.1152/physrev.00031.2018] [Citation(s) in RCA: 400] [Impact Index Per Article: 66.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Oligodendrocytes generate multiple layers of myelin membrane around axons of the central nervous system to enable fast and efficient nerve conduction. Until recently, saltatory nerve conduction was considered the only purpose of myelin, but it is now clear that myelin has more functions. In fact, myelinating oligodendrocytes are embedded in a vast network of interconnected glial and neuronal cells, and increasing evidence supports an active role of oligodendrocytes within this assembly, for example, by providing metabolic support to neurons, by regulating ion and water homeostasis, and by adapting to activity-dependent neuronal signals. The molecular complexity governing these interactions requires an in-depth molecular understanding of how oligodendrocytes and axons interact and how they generate, maintain, and remodel their myelin sheaths. This review deals with the biology of myelin, the expanded relationship of myelin with its underlying axons and the neighboring cells, and its disturbances in various diseases such as multiple sclerosis, acute disseminated encephalomyelitis, and neuromyelitis optica spectrum disorders. Furthermore, we will highlight how specific interactions between astrocytes, oligodendrocytes, and microglia contribute to demyelination in hereditary white matter pathologies.
Collapse
Affiliation(s)
- Christine Stadelmann
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| | - Sebastian Timmler
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| | - Alonso Barrantes-Freer
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| | - Mikael Simons
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| |
Collapse
|
357
|
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: 57] [Impact Index Per Article: 9.5] [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
|
358
|
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: 133] [Impact Index Per Article: 22.2] [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
|
359
|
Galloway DA, Gowing E, Setayeshgar S, Kothary R. Inhibitory milieu at the multiple sclerosis lesion site and the challenges for remyelination. Glia 2019; 68:859-877. [PMID: 31441132 DOI: 10.1002/glia.23711] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 06/26/2019] [Accepted: 07/26/2019] [Indexed: 12/14/2022]
Abstract
Regeneration of myelin, following injury, can occur within the central nervous system to reinstate proper axonal conductance and provide trophic support. Failure to do so renders the axons vulnerable, leading to eventual degeneration, and neuronal loss. Thus, it is essential to understand the mechanisms by which remyelination or failure to remyelinate occur, particularly in the context of demyelinating and neurodegenerative disorders. In multiple sclerosis, oligodendrocyte progenitor cells (OPCs) migrate to lesion sites to repair myelin. However, during disease progression, the ability of OPCs to participate in remyelination diminishes coincident with worsening of the symptoms. Remyelination is affected by a broad range of cues from intrinsic programming of OPCs and extrinsic local factors to the immune system and other systemic elements including diet and exercise. Here we review the literature on these diverse inhibitory factors and the challenges they pose to remyelination. Results spanning several disciplines from fundamental preclinical studies to knowledge gained in the clinic will be discussed.
Collapse
Affiliation(s)
- Dylan A Galloway
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Canada
| | - Elizabeth Gowing
- Neurosciences Department, Faculty of Medicine, Centre de recherche du CHUM, Université de Montreal, Montreal, Quebec, Canada
| | - Solmaz Setayeshgar
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rashmi Kothary
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Medicine, Department of Biochemistry, Microbiology and Immunology, and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
360
|
Papadopoulos C, Kravic B, Meyer H. Repair or Lysophagy: Dealing with Damaged Lysosomes. J Mol Biol 2019; 432:231-239. [PMID: 31449799 DOI: 10.1016/j.jmb.2019.08.010] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Revised: 08/12/2019] [Accepted: 08/14/2019] [Indexed: 12/21/2022]
Abstract
Lysosomal membrane permeabilization or full rupture of lysosomes is a common and severe stress condition that is relevant for degenerative disease, infection and cancer. If damage is limited, cells can repair lysosomes by means of the endosomal sorting complex required for transport (ESCRT) machinery. Presumably, if repair fails, lysosomes are tagged with ubiquitin to initiate clearance by selective macroautophagy, termed lysophagy. Accumulating evidence suggests damage-induced exposure of luminal glycans to the cytosol as the key trigger for ubiquitination. In this review, we discuss recent data on cellular damage sensing, the underlying ubiquitination and autophagy machinery as well as additional layers of regulation such as processing of ubiquitinated proteins by the AAA-ATPase VCP/p97. We conclude with thoughts on how these mechanisms may regulate decision making between lysosome repair and lysophagy.
Collapse
Affiliation(s)
| | - Bojana Kravic
- Centre for Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Hemmo Meyer
- Centre for Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
361
|
Zhou Y, Miles JR, Tavori H, Lin M, Khoshbouei H, Borchelt DR, Bazick H, Landreth GE, Lee S, Fazio S, Notterpek L. PMP22 Regulates Cholesterol Trafficking and ABCA1-Mediated Cholesterol Efflux. J Neurosci 2019; 39:5404-5418. [PMID: 31061090 PMCID: PMC6607759 DOI: 10.1523/jneurosci.2942-18.2019] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 04/22/2019] [Accepted: 04/26/2019] [Indexed: 12/21/2022] Open
Abstract
The absence of functional peripheral myelin protein 22 (PMP22) is associated with shortened lifespan in rodents and severe peripheral nerve myelin abnormalities in several species including humans. Schwann cells and nerves from PMP22 knock-out (KO) mice show deranged cholesterol distribution and aberrant lipid raft morphology, supporting an unrecognized role for PMP22 in cellular lipid metabolism. To examine the mechanisms underlying these abnormalities, we studied Schwann cells and nerves from male and female PMP22 KO mice. Whole-cell current-clamp recordings in cultured Schwann cells revealed increased membrane capacitance and decreased membrane resistance in the absence of PMP22, which was consistent with a reduction in membrane cholesterol. Nerves from PMP22-deficient mice contained abnormal lipid droplets, with both mRNA and protein levels of apolipoprotein E (apoE) and ATP-binding cassette transporter A1 (ABCA1) being highly upregulated. Despite the upregulation of ABCA1 and apoE, the absence of PMP22 resulted in reduced localization of the transporter to the cell membrane and diminished secretion of apoE. The absence of PMP22 also impaired ABCA1-mediated cholesterol efflux capacity. In nerves from ABCA1 KO mice, the expression of PMP22 was significantly elevated and the subcellular processing of the overproduced protein was aberrant. In wild-type samples, double immunolabeling identified overlapping distribution of PMP22 and ABCA1 at the Schwann cell plasma membrane and the two proteins were coimmunoprecipitated from Schwann cell and nerve lysates. Together, these results reveal a novel role for PMP22 in regulating lipid metabolism and cholesterol trafficking through functional interaction with the cholesterol efflux regulatory protein ABCA1.SIGNIFICANCE STATEMENT Understanding the subcellular events that underlie abnormal myelin formation in hereditary neuropathies is critical for advancing therapy development. Peripheral myelin protein 22 (PMP22) is an essential peripheral myelin protein because its genetic abnormalities account for ∼80% of hereditary neuropathies. Here, we demonstrate that in the absence of PMP22, the cellular and electrophysiological properties of the Schwann cells' plasma membrane are altered and cholesterol trafficking and lipid homeostasis are perturbed. The molecular mechanisms for these abnormalities involve a functional interplay among PMP22, cholesterol, apolipoprotein E, and the major cholesterol-efflux transporter protein ATP-binding cassette transporter A1 (ABCA1). These findings establish a critical role for PMP22 in the maintenance of cholesterol homeostasis in Schwann cells.
Collapse
Affiliation(s)
| | - Joshua R Miles
- Department of Medicine
- Department of Physiology and Pharmacology, Knight Cardiovascular Institute, Center of Preventive Cardiology, Oregon Health & Science University, Portland, Oregon 27332, and
| | - Hagai Tavori
- Department of Medicine
- Department of Physiology and Pharmacology, Knight Cardiovascular Institute, Center of Preventive Cardiology, Oregon Health & Science University, Portland, Oregon 27332, and
| | | | | | | | | | - Gary E Landreth
- Department of Neurosciences, Indiana University, Indianapolis, Indiana 46202
| | | | - Sergio Fazio
- Department of Medicine
- Department of Physiology and Pharmacology, Knight Cardiovascular Institute, Center of Preventive Cardiology, Oregon Health & Science University, Portland, Oregon 27332, and
| | - Lucia Notterpek
- Department of Neuroscience,
- Department of Neurology, College of Medicine, McKnight Brain Institute, University of Florida, Gainesville, Florida 32610
| |
Collapse
|
362
|
Baror R, Neumann B, Segel M, Chalut KJ, Fancy SPJ, Schafer DP, Franklin RJM. Transforming growth factor-beta renders ageing microglia inhibitory to oligodendrocyte generation by CNS progenitors. Glia 2019; 67:1374-1384. [PMID: 30861188 PMCID: PMC6563458 DOI: 10.1002/glia.23612] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 02/17/2019] [Accepted: 02/22/2019] [Indexed: 01/26/2023]
Abstract
It is now well-established that the macrophage and microglial response to CNS demyelination influences remyelination by removing myelin debris and secreting a variety of signaling molecules that influence the behaviour of oligodendrocyte progenitor cells (OPCs). Previous studies have shown that changes in microglia contribute to the age-related decline in the efficiency of remyelination. In this study, we show that microglia increase their expression of the proteoglycan NG2 with age, and that this is associated with an altered micro-niche generated by aged, but not young, microglia that can divert the differentiation OPCs from oligodendrocytes into astrocytes in vitro. We further show that these changes in ageing microglia are generated by exposure to high levels of TGFβ. Thus, our findings suggest that the rising levels of circulating TGFβ known to occur with ageing contribute to the age-related decline in remyelination by impairing the ability of microglia to promote oligodendrocyte differentiation from OPCs, and therefore could be a potential therapeutic target to promote remyelination.
Collapse
Affiliation(s)
- Roey Baror
- Wellcome‐MRC Stem Cell InstituteUniversity of CambridgeCambridgeUnited Kingdom
- Department of PaediatricsUniversity California San FranciscoSan FranciscoCalifornia
| | - Björn Neumann
- Wellcome‐MRC Stem Cell InstituteUniversity of CambridgeCambridgeUnited Kingdom
| | - Michael Segel
- Wellcome‐MRC Stem Cell InstituteUniversity of CambridgeCambridgeUnited Kingdom
| | - Kevin J. Chalut
- Wellcome‐MRC Stem Cell InstituteUniversity of CambridgeCambridgeUnited Kingdom
| | - Stephen P. J. Fancy
- Department of PaediatricsUniversity California San FranciscoSan FranciscoCalifornia
| | - Dorothy P. Schafer
- Department of Neurobiology and the Brudnik Neuropsychiatric InstituteUniversity of Massachusetts Medical SchoolWorcesterMassachusetts
| | | |
Collapse
|
363
|
Leyrolle Q, Layé S, Nadjar A. Direct and indirect effects of lipids on microglia function. Neurosci Lett 2019; 708:134348. [PMID: 31238131 DOI: 10.1016/j.neulet.2019.134348] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 06/05/2019] [Accepted: 06/21/2019] [Indexed: 02/07/2023]
Abstract
Microglia are key players in brain function by maintaining brain homeostasis across lifetime. They participate to brain development and maturation through their ability to release neurotrophic factors, to remove immature synapses or unnecessary neural progenitors. They modulate neuronal activity in healthy adult brains and they also orchestrate the neuroinflammatory response in various pathophysiological contexts such as aging and neurodegenerative diseases. One of the main features of microglia is their high sensitivity to environmental factors, partly via the expression of a wide range of receptors. Recent data pinpoint that dietary fatty acids modulate microglia function. Both the quantity and the type of fatty acid are potent modulators of microglia physiology. The present review aims at dissecting the current knowledge on the direct and indirect mechanisms (focus on gut microbiota and hormones) through which fatty acids influence microglial physiology. We summarize main discoveries from in vitro and in vivo models on fatty acid-mediated microglial modulation. All these studies represent a promising field of research that could promote using nutrition as a novel therapeutic or preventive tool in diseases involving microglia dysfunctions.
Collapse
Affiliation(s)
- Q Leyrolle
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France
| | - S Layé
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France
| | - A Nadjar
- INRA, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France; Univ. Bordeaux, Nutrition et Neurobiologie Intégrée, UMR 1286, 33076 Bordeaux, France.
| |
Collapse
|
364
|
Liu C, Jiang W, Tian X, Yang P, Xiao L, Li J, Qiu L, Tu H, Tan W. Identification of Vigilin as a Potential Ischemia Biomarker by Brain Slice-Based Systematic Evolution of Ligands by Exponential Enrichment. Anal Chem 2019; 91:6675-6681. [PMID: 30993971 PMCID: PMC6625766 DOI: 10.1021/acs.analchem.9b00609] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Stroke is one of the leading causes of disability and death among adults worldwide and results in numerous biochemical alterations. However, few efficient biomarkers are clinically available to diagnose stroke because of the limitations of biomarkers and their probes. In this work, we utilized frozen brain slices of middle cerebral artery occlusion (MCAO) in a mouse model of ischemia to select a specific binding aptamer, termed LCW17, by tissue-based SELEX (systematic evolution of ligands by exponential enrichment). LCW17 was enhanced in binding in ischemic brain slices compared to sham control. We identified the binding target of LCW17 as vigilin. Vigilin is increased in ischemia brain slices and exhibits enhanced release from cultured hippocampal neurons after oxygen glucose deprivation in vitro. Taken together, ischemic brain slice-based aptamer selection will enable identification of more probes and potential target molecules for diagnosis and therapy of ischemic stroke. Aptamer LCW17 and vigilin may potentially be applied to define the molecular mechanism underlying ischemic stroke, as well as its diagnosis.
Collapse
Affiliation(s)
- Chao Liu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University Changsha, Hunan, 410082, China
| | - Wei Jiang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University Changsha, Hunan, 410082, China
| | - Xibin Tian
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University Changsha, Hunan, 410082, China
| | - Peng Yang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University Changsha, Hunan, 410082, China
| | - Le Xiao
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University Changsha, Hunan, 410082, China
| | - Jianglin Li
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University Changsha, Hunan, 410082, China
| | - Liping Qiu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University Changsha, Hunan, 410082, China
| | - Haijun Tu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University Changsha, Hunan, 410082, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University Changsha, Hunan, 410082, China
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Department of Chemistry, Department of Physiology and Functional Genomics, Center for Research at the Bio/Nano Interface, UF Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|
365
|
Targeting Apolipoprotein E for Alzheimer's Disease: An Industry Perspective. Int J Mol Sci 2019; 20:ijms20092161. [PMID: 31052389 PMCID: PMC6539182 DOI: 10.3390/ijms20092161] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 04/27/2019] [Accepted: 04/28/2019] [Indexed: 02/08/2023] Open
Abstract
Apolipoprotein E (apoE), a key lipid transport protein in the brain, is predominantly produced by astrocytes. Astrocytes are the most numerous cell type in the brain and are the main support network for neurons. They play a critical role in the synthesis and delivery of cholesterol in the brain. Humans have three common apoE isoforms, apoE2, apoE3 and apoE4, that show a strong genotype effect on the risk and age of onset for sporadic and late onset forms of Alzheimer’s disease (AD). Carriers of an ε4 allele have an increased risk of developing AD, while those with an ε2 allele are protected. Investigations into the contribution of apoE to the development of AD has yielded conflicting results and there is still much speculation about the role of this protein in disease. Here, we review the opposing hypotheses currently described in the literature and the approaches that have been considered for targeting apoE as a novel therapeutic strategy for AD. Additionally, we provide our perspective on the rationale for targeting apoE and the challenges that arise with respect to “drug-ability” of this target.
Collapse
|
366
|
Hill RA, Grutzendler J. Uncovering the biology of myelin with optical imaging of the live brain. Glia 2019; 67:2008-2019. [PMID: 31033062 DOI: 10.1002/glia.23635] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 03/26/2019] [Accepted: 04/11/2019] [Indexed: 12/31/2022]
Abstract
Myelin has traditionally been considered a static structure that is produced and assembled during early developmental stages. While this characterization is accurate in some contexts, recent studies have revealed that oligodendrocyte generation and patterns of myelination are dynamic and potentially modifiable throughout life. Unique structural and biochemical properties of the myelin sheath provide opportunities for the development and implementation of multimodal label-free and fluorescence optical imaging approaches. When combined with genetically encoded fluorescent tags targeted to distinct cells and subcellular structures, these techniques offer a powerful methodological toolbox for uncovering mechanisms of myelin generation and plasticity in the live brain. Here, we discuss recent advances in these approaches that have allowed the discovery of several forms of myelin plasticity in developing and adult nervous systems. Using these techniques, long-standing questions related to myelin generation, remodeling, and degeneration can now be addressed.
Collapse
Affiliation(s)
- Robert A Hill
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire
| | - Jaime Grutzendler
- Departments of Neurology and Neuroscience, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
367
|
Zabala A, Vazquez-Villoldo N, Rissiek B, Gejo J, Martin A, Palomino A, Perez-Samartín A, Pulagam KR, Lukowiak M, Capetillo-Zarate E, Llop J, Magnus T, Koch-Nolte F, Rassendren F, Matute C, Domercq M. P2X4 receptor controls microglia activation and favors remyelination in autoimmune encephalitis. EMBO Mol Med 2019; 10:emmm.201708743. [PMID: 29973381 PMCID: PMC6079537 DOI: 10.15252/emmm.201708743] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Microglia survey the brain microenvironment for signals of injury or infection and are essential for the initiation and resolution of pathogen‐ or tissue damage‐induced inflammation. Understanding the mechanism of microglia responses during pathology is hence vital to promote regenerative responses. Here, we analyzed the role of purinergic receptor P2X4 (P2X4R) in microglia/macrophages during autoimmune inflammation. Blockade of P2X4R signaling exacerbated clinical signs in the experimental autoimmune encephalomyelitis (EAE) model and also favored microglia activation to a pro‐inflammatory phenotype and inhibited myelin phagocytosis. Moreover, P2X4R blockade in microglia halted oligodendrocyte differentiation in vitro and remyelination after lysolecithin‐induced demyelination. Conversely, potentiation of P2X4R signaling by the allosteric modulator ivermectin (IVM) favored a switch in microglia to an anti‐inflammatory phenotype, potentiated myelin phagocytosis, promoted the remyelination response, and ameliorated clinical signs of EAE. Our results provide evidence that P2X4Rs modulate microglia/macrophage inflammatory responses and identify IVM as a potential candidate among currently used drugs to promote the repair of myelin damage.
Collapse
Affiliation(s)
- Alazne Zabala
- Achucarro Basque Center for Neurosciences, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco, Leioa, Spain
| | - Nuria Vazquez-Villoldo
- Achucarro Basque Center for Neurosciences, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco, Leioa, Spain
| | - Björn Rissiek
- Department of Neurology, University Medical Center, Hamburg, Germany
| | - Jon Gejo
- Achucarro Basque Center for Neurosciences, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco, Leioa, Spain
| | - Abraham Martin
- Molecular Imaging Unit, CIC biomaGUNE, San Sebastian, Spain
| | - Aitor Palomino
- Achucarro Basque Center for Neurosciences, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco, Leioa, Spain
| | - Alberto Perez-Samartín
- Achucarro Basque Center for Neurosciences, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco, Leioa, Spain
| | | | - Marco Lukowiak
- Department of Neurology, University Medical Center, Hamburg, Germany
| | - Estibaliz Capetillo-Zarate
- Achucarro Basque Center for Neurosciences, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco, Leioa, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Jordi Llop
- Molecular Imaging Unit, CIC biomaGUNE, San Sebastian, Spain
| | - Tim Magnus
- Department of Neurology, University Medical Center, Hamburg, Germany
| | | | | | - Carlos Matute
- Achucarro Basque Center for Neurosciences, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco, Leioa, Spain
| | - María Domercq
- Achucarro Basque Center for Neurosciences, CIBERNED and Departamento de Neurociencias, Universidad del País Vasco, Leioa, Spain
| |
Collapse
|
368
|
Wu F, Xu K, Liu L, Zhang K, Xia L, Zhang M, Teng C, Tong H, He Y, Xue Y, Zhang H, Chen D, Hu A. Vitamin B 12 Enhances Nerve Repair and Improves Functional Recovery After Traumatic Brain Injury by Inhibiting ER Stress-Induced Neuron Injury. Front Pharmacol 2019; 10:406. [PMID: 31105562 PMCID: PMC6491933 DOI: 10.3389/fphar.2019.00406] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 04/01/2019] [Indexed: 12/31/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the most common causes of neurological damage in young human populations. Vitamin B12 has been reported to promote axon growth of neuronal cells after peripheral nerve injury, which is currently used for the treatment of peripheral nerve damage in the clinical trial. Thus, we hypothesized that TBI can be attenuated by vitaminB12 treatment through its beneficial role on axon regeneration after nerve injury. To confirm it, the biological function of vitaminB12 was characterized using hematoxylin and eosin (H&E) staining, Luxol fast blue (LFB) staining, western blot analysis, and immunohistochemistry staining. The results showed that the neurological functional recovery was improved in the VitaminB12-treated group after TBI, which may be due to downregulation of the endoplasmic reticulum stress-related apoptosis signaling pathway. Moreover, the microtubule stabilization, remyelination and myelin reparation were rescued by vitamin B12, which was consistent with the treatment of 4-phenylbutyric acid (4-PBA), an endoplasmic reticulum stress inhibitor. The study suggests that vitamin B12 may be useful as a novel neuroprotective drug for TBI.
Collapse
Affiliation(s)
- Fangfang Wu
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ke Xu
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
| | - Lei Liu
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Kairui Zhang
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Leilei Xia
- Department of Emergency, Wenzhou People's Hospital, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, China
| | - Man Zhang
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Chenhuai Teng
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Heyan Tong
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yifang He
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yujie Xue
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hongyu Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Daqing Chen
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Aiping Hu
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
369
|
Gabandé‐Rodríguez E, Keane L, Capasso M. Microglial phagocytosis in aging and Alzheimer's disease. J Neurosci Res 2019; 98:284-298. [DOI: 10.1002/jnr.24419] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/20/2019] [Accepted: 03/08/2019] [Indexed: 01/24/2023]
Affiliation(s)
- Enrique Gabandé‐Rodríguez
- Department of Molecular Neuropathology Centro de Biología Molecular “Severo Ochoa” (CSIC‐UAM) Madrid Spain
| | - Lily Keane
- German Center for Neurodegenerative Diseases (DZNE) Bonn Germany
| | - Melania Capasso
- German Center for Neurodegenerative Diseases (DZNE) Bonn Germany
| |
Collapse
|
370
|
Tall AR, Westerterp M. Inflammasomes, neutrophil extracellular traps, and cholesterol. J Lipid Res 2019; 60:721-727. [PMID: 30782961 PMCID: PMC6446695 DOI: 10.1194/jlr.s091280] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 02/09/2019] [Indexed: 02/07/2023] Open
Abstract
Activation of macrophage inflammasomes leads to interleukin (IL)-1β and IL-18 secretion and promotes atherosclerosis and its complications in mice and humans. However, the specific role and underlying mechanisms of the inflammasome in atherogenesis are topics of active research. Several studies in hyperlipidemic mouse models found that the NOD-like receptor protein 3 (NLRP3) inflammasome contributes to atherosclerosis, but recent work suggests that a second hit, such as defective cholesterol efflux or accumulation of oxidized mitochondrial DNA, may be required for significant inflammasome activation. Cholesterol crystal uptake or formation in lysosomes may damage membranes and activate NLRP3 inflammasomes. Alternatively, plasma or ER membrane cholesterol accumulation may condition macrophages for inflammasome activation in the presence of danger-associated molecular patterns, such as oxidized LDL. Inflammasome activation in macrophages or neutrophils leads to gasdermin-D cleavage that induces membrane pore formation, releasing IL-1β and IL-18, and eventuating in pyroptosis or neutrophil extracellular trap formation (NETosis). In humans, inflammasome activation and NETosis may contribute to atherosclerotic plaque erosion and thrombosis, especially in patients with type 2 diabetes, chronic kidney disease, or clonal hematopoiesis. Suppression of the inflammasome by activation of cholesterol efflux or by direct inhibition of inflammasome components may benefit patients with CVD and underlying susceptibility to inflammasome activation.
Collapse
Affiliation(s)
- Alan R Tall
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY 10032.
| | - Marit Westerterp
- Department of Pediatrics, Section Molecular Genetics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
371
|
Hudry E, Klickstein J, Cannavo C, Jackson R, Muzikansky A, Gandhi S, Urick D, Sargent T, Wrobleski L, Roe AD, Hou SS, Kuchibhotla KV, Betensky RA, Spires-Jones T, Hyman BT. Opposing Roles of apolipoprotein E in aging and neurodegeneration. Life Sci Alliance 2019; 2:2/1/e201900325. [PMID: 30760557 PMCID: PMC6374993 DOI: 10.26508/lsa.201900325] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/05/2019] [Accepted: 02/06/2019] [Indexed: 11/24/2022] Open
Abstract
Apolipoprotein E (APOE) effects on brain function remain controversial. Removal of APOE not only impairs cognitive functions but also reduces neuritic amyloid plaques in mouse models of Alzheimer's disease (AD). Can APOE simultaneously protect and impair neural circuits? Here, we dissociated the role of APOE in AD versus aging to determine its effects on neuronal function and synaptic integrity. Using two-photon calcium imaging in awake mice to record visually evoked responses, we found that genetic removal of APOE improved neuronal responses in adult APP/PSEN1 mice (8-10 mo). These animals also exhibited fewer neuritic plaques with less surrounding synapse loss, fewer neuritic dystrophies, and reactive glia. Surprisingly, the lack of APOE in aged mice (18-20 mo), even in the absence of amyloid, disrupted visually evoked responses. These results suggest a dissociation in APOE's role in AD versus aging: APOE may be neurotoxic during early stages of amyloid deposition, although being neuroprotective in latter stages of aging.
Collapse
Affiliation(s)
- Eloise Hudry
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Jacob Klickstein
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Claudia Cannavo
- Centre for Discovery Brain Sciences, UK Dementia Research Institute, and Edinburgh Neuroscience, The University of Edinburgh, Edinburgh, UK
| | - Rosemary Jackson
- Centre for Discovery Brain Sciences, UK Dementia Research Institute, and Edinburgh Neuroscience, The University of Edinburgh, Edinburgh, UK
| | - Alona Muzikansky
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Sheetal Gandhi
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - David Urick
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Taylie Sargent
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Lauren Wrobleski
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Allyson D Roe
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Steven S Hou
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | | | - Rebecca A Betensky
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Tara Spires-Jones
- Centre for Discovery Brain Sciences, UK Dementia Research Institute, and Edinburgh Neuroscience, The University of Edinburgh, Edinburgh, UK
| | - Bradley T Hyman
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
372
|
Elbaz B, Popko B. Molecular Control of Oligodendrocyte Development. Trends Neurosci 2019; 42:263-277. [PMID: 30770136 DOI: 10.1016/j.tins.2019.01.002] [Citation(s) in RCA: 207] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/07/2019] [Accepted: 01/15/2019] [Indexed: 12/27/2022]
Abstract
Myelin is a multilayer lipid membrane structure that wraps and insulates axons, allowing for the efficient propagation of action potentials. During developmental myelination of the central nervous system (CNS), oligodendrocyte progenitor cells (OPCs) proliferate and migrate to their final destination, where they terminally differentiate into mature oligodendrocytes and myelinate axons. Lineage progression and terminal differentiation of oligodendrocyte lineage cells are under tight transcriptional and post-transcriptional control. The characterization of several recently identified regulatory factors that govern these processes, which are the focus of this review, has greatly increased our understanding of oligodendrocyte development and function. These insights are critical to facilitate efforts to enhance OPC differentiation in neurological disorders that disrupt CNS myelin.
Collapse
Affiliation(s)
- Benayahu Elbaz
- The Center for Peripheral Neuropathy, The Department of Neurology, University of Chicago, Chicago, IL, USA
| | - Brian Popko
- The Center for Peripheral Neuropathy, The Department of Neurology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
373
|
Bergner CG, van der Meer F, Winkler A, Wrzos C, Türkmen M, Valizada E, Fitzner D, Hametner S, Hartmann C, Pfeifenbring S, Stoltenburg-Didinger G, Brück W, Nessler S, Stadelmann C. Microglia damage precedes major myelin breakdown in X-linked adrenoleukodystrophy and metachromatic leukodystrophy. Glia 2019; 67:1196-1209. [PMID: 30980503 PMCID: PMC6594046 DOI: 10.1002/glia.23598] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 01/20/2019] [Accepted: 01/21/2019] [Indexed: 12/28/2022]
Abstract
X-linked adrenoleukodystrophy (X-ALD) and metachromatic leukodystrophy (MLD) are two relatively common examples of hereditary demyelinating diseases caused by a dysfunction of peroxisomal or lysosomal lipid degradation. In both conditions, accumulation of nondegraded lipids leads to the destruction of cerebral white matter. Because of their high lipid content, oligodendrocytes are considered key to the pathophysiology of these leukodystrophies. However, the response to allogeneic stem cell transplantation points to the relevance of cells related to the hematopoietic lineage. In the present study, we aimed to better characterize the pathogenetic role of microglia in the above-mentioned diseases. Applying recently established microglia markers to human autopsy cases of X-ALD and MLD we were able to delineate distinct lesion stages in evolving demyelinating lesions. The immune-phenotype of microglia was altered already early in lesion evolution, and microglia loss preceded full-blown myelin degeneration both in X-ALD and MLD. DNA fragmentation indicating phagocyte death was observed in areas showing microglia loss. The morphology and dynamics of phagocyte decay differed between the diseases and between lesion stages, hinting at distinct pathways of programmed cell death. In summary, the present study shows an early and severe damage to microglia in the pathogenesis of X-ALD and MLD. This hints at a central pathophysiologic role of these cells in the diseases and provides evidence for an ongoing transfer of toxic substrates primarily enriched in myelinating cells to microglia.
Collapse
Affiliation(s)
- Caroline G Bergner
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Anne Winkler
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Claudia Wrzos
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Mevlude Türkmen
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany.,Department of Cardiology, University Medical Center Göttingen, Göttingen, Germany
| | - Emil Valizada
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Dirk Fitzner
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Simon Hametner
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany.,Institute of Neurology, Medical University Vienna, Vienna, Austria
| | - Christian Hartmann
- Institute of Pathology, Section of Neuropathology, Hannover Medical School, Hannover, Germany
| | - Sabine Pfeifenbring
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Wolfgang Brück
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Stefan Nessler
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Christine Stadelmann
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
374
|
Filippi M, Brück W, Chard D, Fazekas F, Geurts JJG, Enzinger C, Hametner S, Kuhlmann T, Preziosa P, Rovira À, Schmierer K, Stadelmann C, Rocca MA. Association between pathological and MRI findings in multiple sclerosis. Lancet Neurol 2019; 18:198-210. [DOI: 10.1016/s1474-4422(18)30451-4] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/22/2018] [Accepted: 11/12/2018] [Indexed: 12/12/2022]
|
375
|
Gabandé-Rodríguez E, Pérez-Cañamás A, Soto-Huelin B, Mitroi DN, Sánchez-Redondo S, Martínez-Sáez E, Venero C, Peinado H, Ledesma MD. Lipid-induced lysosomal damage after demyelination corrupts microglia protective function in lysosomal storage disorders. EMBO J 2018; 38:embj.201899553. [PMID: 30530526 PMCID: PMC6331723 DOI: 10.15252/embj.201899553] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 10/12/2018] [Accepted: 10/25/2018] [Indexed: 12/20/2022] Open
Abstract
Neuropathic lysosomal storage disorders (LSDs) present with activated pro‐inflammatory microglia. However, anti‐inflammatory treatment failed to improve disease pathology. We characterise the mechanisms underlying microglia activation in Niemann–Pick disease type A (NPA). We establish that an NPA patient and the acid sphingomyelinase knockout (ASMko) mouse model show amoeboid microglia in neurodegeneration‐prone areas. In vivo microglia ablation worsens disease progression in ASMko mice. We demonstrate the coexistence of different microglia phenotypes in ASMko brains that produce cytokines or counteract neuronal death by clearing myelin debris. Overloading microglial lysosomes through myelin debris accumulation and sphingomyelin build‐up induces lysosomal damage and cathepsin B extracellular release by lysosomal exocytosis. Inhibition of cathepsin B prevents neuronal death and behavioural anomalies in ASMko mice. Similar microglia phenotypes occur in a Niemann–Pick disease type C mouse model and patient. Our results show a protective function for microglia in LSDs and how this is corrupted by lipid lysosomal overload. Data indicate cathepsin B as a key molecule mediating neurodegeneration, opening research pathways for therapeutic targeting of LSDs and other demyelinating diseases.
Collapse
Affiliation(s)
- Enrique Gabandé-Rodríguez
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Madrid, Spain .,Barts Cancer Institute, Centre for Cancer & Inflammation, Queen Mary University of London, London, UK
| | - Azucena Pérez-Cañamás
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Madrid, Spain
| | - Beatriz Soto-Huelin
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Madrid, Spain
| | - Daniel N Mitroi
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Madrid, Spain
| | - Sara Sánchez-Redondo
- Microenvironment and Metastasis Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Elena Martínez-Sáez
- Department of Pathology, Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | - César Venero
- Department of Psychobiology, Universidad Nacional de Educación a Distancia, Madrid, Spain
| | - Héctor Peinado
- Microenvironment and Metastasis Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain.,Department of Pediatrics, Drukier Institute for Children's Health and Meyer Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - María Dolores Ledesma
- Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Madrid, Spain
| |
Collapse
|
376
|
Zarei S, Eggert J, Franqui-Dominguez L, Carl Y, Boria F, Stukova M, Avila A, Rubi C, Chinea A. Comprehensive review of neuromyelitis optica and clinical characteristics of neuromyelitis optica patients in Puerto Rico. Surg Neurol Int 2018; 9:242. [PMID: 30603227 PMCID: PMC6293609 DOI: 10.4103/sni.sni_224_18] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 08/21/2018] [Indexed: 12/14/2022] Open
Abstract
Neuromyelitis optica (NMO) is an immune-mediated inflammatory disorder of the central nervous system. It is characterized by concurrent inflammation and demyelination of the optic nerve (optic neuritis [ON]) and the spinal cord (myelitis). Multiple studies show variations in prevalence, clinical, and demographic features of NMO among different populations. In addition, ethnicity and race are known as important factors on disease phenotype and clinical outcomes. There are little data on information about NMO patients in underserved groups, including Puerto Rico (PR). In this research, we will provide a comprehensive overview of all aspects of NMO, including epidemiology, environmental risk factors, genetic factors, molecular mechanism, symptoms, comorbidities and clinical differentiation, diagnosis, treatment, its management, and prognosis. We will also evaluate the demographic features and clinical phenotype of NMO patients in PR. This will provide a better understanding of NMO and establish a basis of knowledge that can be used to improve care. Furthermore, this type of population-based study can distinguish the clinical features variation among NMO patients and will provide insight into the potential mechanisms that cause these variations.
Collapse
Affiliation(s)
- Sara Zarei
- San Juan Bautista School of Medicine, Caguas, Puerto Rico, USA
| | - James Eggert
- San Juan Bautista School of Medicine, Caguas, Puerto Rico, USA
| | | | - Yonatan Carl
- San Juan Bautista School of Medicine, Caguas, Puerto Rico, USA
| | - Fernando Boria
- San Juan Bautista School of Medicine, Caguas, Puerto Rico, USA
| | - Marina Stukova
- San Juan Bautista School of Medicine, Caguas, Puerto Rico, USA
| | | | - Cristina Rubi
- Caribbean Neurological Center, Guaynabo, Puerto Rico, USA
| | - Angel Chinea
- Caribbean Neurological Center, Guaynabo, Puerto Rico, USA
| |
Collapse
|
377
|
Groh J, Klein D, Berve K, West BL, Martini R. Targeting microglia attenuates neuroinflammation-related neural damage in mice carrying human PLP1
mutations. Glia 2018; 67:277-290. [DOI: 10.1002/glia.23539] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 08/28/2018] [Accepted: 09/11/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Janos Groh
- Department of Neurology, Section of Developmental Neurobiology; University Hospital Wuerzburg; Wuerzburg Germany
| | - Dennis Klein
- Department of Neurology, Section of Developmental Neurobiology; University Hospital Wuerzburg; Wuerzburg Germany
| | - Kristina Berve
- Department of Neurology, Section of Developmental Neurobiology; University Hospital Wuerzburg; Wuerzburg Germany
| | | | - Rudolf Martini
- Department of Neurology, Section of Developmental Neurobiology; University Hospital Wuerzburg; Wuerzburg Germany
| |
Collapse
|
378
|
Grajchen E, Hendriks JJA, Bogie JFJ. The physiology of foamy phagocytes in multiple sclerosis. Acta Neuropathol Commun 2018; 6:124. [PMID: 30454040 PMCID: PMC6240956 DOI: 10.1186/s40478-018-0628-8] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 11/02/2018] [Indexed: 12/15/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic disease of the central nervous system characterized by massive infiltration of immune cells, demyelination, and axonal loss. Active MS lesions mainly consist of macrophages and microglia containing abundant intracellular myelin remnants. Initial studies showed that these foamy phagocytes primarily promote MS disease progression by internalizing myelin debris, presenting brain-derived autoantigens, and adopting an inflammatory phenotype. However, more recent studies indicate that phagocytes can also adopt a beneficial phenotype upon myelin internalization. In this review, we summarize and discuss the current knowledge on the spatiotemporal physiology of foamy phagocytes in MS lesions, and elaborate on extrinsic and intrinsic factors regulating their behavior. In addition, we discuss and link the physiology of myelin-containing phagocytes to that of foamy macrophages in other disorders such atherosclerosis.
Collapse
Affiliation(s)
- Elien Grajchen
- Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium/School of Life Sciences, Transnationale Universiteit Limburg, Diepenbeek, Belgium
| | - Jerome J A Hendriks
- Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium/School of Life Sciences, Transnationale Universiteit Limburg, Diepenbeek, Belgium
| | - Jeroen F J Bogie
- Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium/School of Life Sciences, Transnationale Universiteit Limburg, Diepenbeek, Belgium.
| |
Collapse
|
379
|
Liquefaction of the Brain following Stroke Shares a Similar Molecular and Morphological Profile with Atherosclerosis and Mediates Secondary Neurodegeneration in an Osteopontin-Dependent Mechanism. eNeuro 2018; 5:eN-CFN-0076-18. [PMID: 30417081 PMCID: PMC6223114 DOI: 10.1523/eneuro.0076-18.2018] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 10/01/2018] [Accepted: 10/05/2018] [Indexed: 12/23/2022] Open
Abstract
Here we used mouse models of heart and brain ischemia to compare the inflammatory response to ischemia in the heart, a protein rich organ, to the inflammatory response to ischemia in the brain, a lipid rich organ. We report that ischemia-induced inflammation resolves between one and four weeks in the heart compared to between eight and 24 weeks in the brain. Importantly, we discovered that a second burst of inflammation occurs in the brain between four and eight weeks following ischemia, which coincided with the appearance of cholesterol crystals within the infarct. This second wave shares a similar cellular and molecular profile with atherosclerosis and is characterized by high levels of osteopontin (OPN) and matrix metalloproteinases (MMPs). In order to test the role of OPN in areas of liquefactive necrosis, OPN-/- mice were subjected to brain ischemia. We found that at seven weeks following stroke, the expression of pro-inflammatory proteins and MMPs was profoundly reduced in the infarct of the OPN-/- mice, although the number of cholesterol crystals was increased. OPN-/- mice exhibited faster recovery of motor function and a higher number of neuronal nuclei (NeuN) positive cells in the peri-infarct area at seven weeks following stroke. Based on these findings we propose that the brain liquefies after stroke because phagocytic cells in the infarct are unable to efficiently clear cholesterol rich myelin debris, and that this leads to the perpetuation of an OPN-dependent inflammatory response characterized by high levels of degradative enzymes.
Collapse
|
380
|
Prajeeth CK, Dittrich-Breiholz O, Talbot SR, Robert PA, Huehn J, Stangel M. IFN-γ Producing Th1 Cells Induce Different Transcriptional Profiles in Microglia and Astrocytes. Front Cell Neurosci 2018; 12:352. [PMID: 30364000 PMCID: PMC6191492 DOI: 10.3389/fncel.2018.00352] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 09/19/2018] [Indexed: 12/16/2022] Open
Abstract
Autoreactive T cells that infiltrate into the central nervous system (CNS) are believed to have a significant role in mediating the pathology of neuroinflammatory diseases like multiple sclerosis. Their interaction with microglia and astrocytes in the CNS is crucial for the regulation of neuroinflammatory processes. Our previous work demonstrated that effectors secreted by Th1 and Th17 cells have different capacities to influence the phenotype and function of glial cells. We have shown that Th1-derived effectors altered the phenotype and function of both microglia and astrocytes whereas Th17-derived effectors induced direct effects only on astrocytes but not on microglia. Here we investigated if effector molecules associated with IFN-γ producing Th1 cells induced different gene expression profiles in microglia and astrocytes. We performed a microarray analysis of RNA isolated from microglia and astrocytes treated with medium and Th-derived culture supernatants and compared the gene expression data. By using the criteria of 2-fold change and a false discovery rate of 0.01 (corrected p < 0.01), we demonstrated that a total of 2,106 and 1,594 genes were differentially regulated in microglia and astrocytes, respectively, in response to Th1-derived factors. We observed that Th1-derived effectors induce distinct transcriptional changes in microglia and astrocytes in addition to commonly regulated transcripts. These distinct transcriptional changes regulate peculiar physiological functions, and this knowledge can help to better understand T cell mediated neuropathologies.
Collapse
Affiliation(s)
- Chittappen K Prajeeth
- Department of Neurology, Clinical Neuroimmunology and Neurochemistry, Hannover Medical School, Hannover, Germany
| | | | - Steven R Talbot
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Philippe A Robert
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Department of Systems Immunology, Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Martin Stangel
- Department of Neurology, Clinical Neuroimmunology and Neurochemistry, Hannover Medical School, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
381
|
NLRP3 inflammasome activation in inflammaging. Semin Immunol 2018; 40:61-73. [PMID: 30268598 DOI: 10.1016/j.smim.2018.09.001] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 02/06/2023]
Abstract
The process of aging is associated with the appearance of low-grade subclinical inflammation, termed inflammaging, that can accelerate age-related diseases. In Western societies the age-related inflammatory response can additionally be aggravated by an inflammatory response related to modern lifestyles and excess calorie consumption, a pathophysiologic inflammatory response that was coined metaflammation. Here, we summarize the current knowledge of mechanisms that drive both of these processes and focus our discussion the emerging concept that a key innate immune pathway, the NLRP3 inflammasome, is centrally involved in the recognition of triggers that appear during physiological aging and during metabolic stress. We further discuss how these processes are involved in the pathogenesis of common age-related pathologies and highlight potential strategies by which the detrimental inflammatory responses could be pharmacologically addressed.
Collapse
|
382
|
Oost W, Talma N, Meilof JF, Laman JD. Targeting senescence to delay progression of multiple sclerosis. J Mol Med (Berl) 2018; 96:1153-1166. [PMID: 30229272 PMCID: PMC6208951 DOI: 10.1007/s00109-018-1686-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/18/2018] [Accepted: 08/09/2018] [Indexed: 12/11/2022]
Abstract
Multiple sclerosis (MS) is a chronic and often progressive, demyelinating disease of the central nervous system (CNS) white and gray matter and the single most common cause of disability in young adults. Age is one of the factors most strongly influencing the course of progression in MS. One of the hallmarks of aging is cellular senescence. The elimination of senescent cells with senolytics has very recently been shown to delay age-related dysfunction in animal models for other neurological diseases. In this review, the possible link between cellular senescence and the progression of MS is discussed, and the potential use of senolytics as a treatment for progressive MS is explored. Currently, there is no cure for MS and there are limited treatment options to slow the progression of MS. Current treatment is based on immunomodulatory approaches. Various cell types present in the CNS can become senescent and thus potentially contribute to MS disease progression. We propose that, after cellular senescence has indeed been shown to be directly implicated in disease progression, administration of senolytics should be tested as a potential therapeutic approach for the treatment of progressive MS.
Collapse
Affiliation(s)
- Wendy Oost
- University of Groningen, Groningen, The Netherlands
| | - Nynke Talma
- European Institute for the Biology of Ageing (ERIBA), University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Neuroscience, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jan F Meilof
- Department of Neurology, Martini Hospital, Groningen, The Netherlands.,MS Center Noord Nederland (MSCNN), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jon D Laman
- Department of Neuroscience, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands. .,MS Center Noord Nederland (MSCNN), University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
383
|
Mangale V, McIntyre LL, Walsh CM, Loring JF, Lane TE. Promoting remyelination through cell transplantation therapies in a model of viral-induced neurodegenerative disease. Dev Dyn 2018; 248:43-52. [PMID: 30067309 DOI: 10.1002/dvdy.24658] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/05/2018] [Accepted: 07/05/2018] [Indexed: 12/17/2022] Open
Abstract
Multiple sclerosis (MS) is a central nervous system (CNS) disease characterized by chronic neuroinflammation, demyelination, and axonal damage. Infiltration of activated lymphocytes and myeloid cells are thought to be primarily responsible for white matter damage and axonopathy. Several United States Food and Drug Administration-approved therapies exist that impede activated lymphocytes from entering the CNS thereby limiting new lesion formation in patients with relapse-remitting forms of MS. However, a significant challenge within the field of MS research is to develop effective and sustained therapies that allow for axonal protection and remyelination. In recent years, there has been increasing evidence that some kinds of stem cells and their derivatives seem to be able to mute neuroinflammation as well as promote remyelination and axonal integrity. Intracranial infection of mice with the neurotropic JHM strain of mouse hepatitis virus (JHMV) results in immune-mediated demyelination and axonopathy, making this an excellent model to interrogate the therapeutic potential of stem cell derivatives in evoking remyelination. This review provides a succinct overview of our recent findings using intraspinal injection of mouse CNS neural progenitor cells and human neural precursors into JHMV-infected mice. JHMV-infected mice receiving these cells display extensive remyelination associated with axonal sparing. In addition, we discuss possible mechanisms associated with sustained clinical recovery. Developmental Dynamics 248:43-52, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Vrushali Mangale
- Division of Microbiology & Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah
| | - Laura L McIntyre
- Department of Molecular Biology & Biochemistry, Sue & Bill Gross Stem Cell Center, University of California, Irvine, California
| | - Craig M Walsh
- Department of Molecular Biology & Biochemistry, Sue & Bill Gross Stem Cell Center, University of California, Irvine, California
| | - Jeanne F Loring
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California
| | - Thomas E Lane
- Division of Microbiology & Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah.,Department of Bioengineering, University of Utah, Salt Lake City, Utah.,Immunology, Inflammation, and Infectious Disease Initiative, University of Utah, Salt Lake City, Utah
| |
Collapse
|
384
|
Dang EV, Cyster JG. Loss of sterol metabolic homeostasis triggers inflammasomes - how and why. Curr Opin Immunol 2018; 56:1-9. [PMID: 30172069 DOI: 10.1016/j.coi.2018.08.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/01/2018] [Accepted: 08/01/2018] [Indexed: 10/28/2022]
Abstract
Proper regulation of sterol biosynthesis is critical for eukaryotic cellular homeostasis. Cholesterol and isoprenoids serve key roles in eukaryotic cells by regulating membrane fluidity and correct localization of proteins. It is becoming increasingly appreciated that dysregulated sterol metabolism engages pathways that lead to inflammation. Of particular importance are inflammasomes, which are multiplatform protein complexes that activate caspase-1 in order to process the pro-inflammatory and pyrogenic cytokines IL-1β and IL-18. In this review, we highlight recent research that links altered sterol biosynthetic pathway activity to inflammasome activation. We discuss how clues from human genetics have led to new insights into how alterations in isoprenoid biosynthesis connect to inflammation. We also discuss new mechanisms that show how macrophage cholesterol buildup can lead to inflammasome activation.
Collapse
Affiliation(s)
- Eric V Dang
- Department of Biophysics and Biochemistry, University of California, San Francisco, CA 94158, USA.
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, CA 94143, USA.
| |
Collapse
|
385
|
Westerterp M, Fotakis P, Ouimet M, Bochem AE, Zhang H, Molusky MM, Wang W, Abramowicz S, la Bastide-van Gemert S, Wang N, Welch CL, Reilly MP, Stroes ES, Moore KJ, Tall AR. Cholesterol Efflux Pathways Suppress Inflammasome Activation, NETosis, and Atherogenesis. Circulation 2018; 138:898-912. [PMID: 29588315 PMCID: PMC6160368 DOI: 10.1161/circulationaha.117.032636] [Citation(s) in RCA: 245] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 03/09/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND The CANTOS trial (Canakinumab Antiinflammatory Thrombosis Outcome Study) showed that antagonism of interleukin (IL)-1β reduces coronary heart disease in patients with a previous myocardial infarction and evidence of systemic inflammation, indicating that pathways required for IL-1β secretion increase cardiovascular risk. IL-1β and IL-18 are produced via the NLRP3 inflammasome in myeloid cells in response to cholesterol accumulation, but mechanisms linking NLRP3 inflammasome activation to atherogenesis are unclear. The cholesterol transporters ATP binding cassette A1 and G1 (ABCA1/G1) mediate cholesterol efflux to high-density lipoprotein, and Abca1/g1 deficiency in myeloid cells leads to cholesterol accumulation. METHODS To interrogate mechanisms connecting inflammasome activation with atherogenesis, we used mice with myeloid Abca1/g1 deficiency and concomitant deficiency of the inflammasome components Nlrp3 or Caspase-1/11. Bone marrow from these mice was transplanted into Ldlr-/- recipients, which were fed a Western-type diet. RESULTS Myeloid Abca1/g1 deficiency increased plasma IL-18 levels in Ldlr-/- mice and induced IL-1β and IL-18 secretion in splenocytes, which was reversed by Nlrp3 or Caspase-1/11 deficiency, indicating activation of the NLRP3 inflammasome. Nlrp3 or Caspase-1/11 deficiency decreased atherosclerotic lesion size in myeloid Abca1/g1-deficient Ldlr-/- mice. Myeloid Abca1/g1 deficiency enhanced caspase-1 cleavage not only in splenic monocytes and macrophages, but also in neutrophils, and dramatically enhanced neutrophil accumulation and neutrophil extracellular trap formation in atherosclerotic plaques, with reversal by Nlrp3 or Caspase-1/11 deficiency, suggesting that inflammasome activation promotes neutrophil recruitment and neutrophil extracellular trap formation in atherosclerotic plaques. These effects appeared to be indirectly mediated by systemic inflammation leading to activation and accumulation of neutrophils in plaques. Myeloid Abca1/g1 deficiency also activated the noncanonical inflammasome, causing increased susceptibility to lipopolysaccharide-induced mortality. Patients with Tangier disease, who carry loss-of-function mutations in ABCA1 and have increased myeloid cholesterol content, showed a marked increase in plasma IL-1β and IL-18 levels. CONCLUSIONS Cholesterol accumulation in myeloid cells activates the NLRP3 inflammasome, which enhances neutrophil accumulation and neutrophil extracellular trap formation in atherosclerotic plaques. Patients with Tangier disease, who have increased myeloid cholesterol content, showed markers of inflammasome activation, suggesting human relevance.
Collapse
MESH Headings
- ATP Binding Cassette Transporter 1/deficiency
- ATP Binding Cassette Transporter 1/genetics
- ATP Binding Cassette Transporter 1/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 1/deficiency
- ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism
- Animals
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/prevention & control
- Case-Control Studies
- Caspase 1/genetics
- Caspase 1/metabolism
- Caspases/genetics
- Caspases/metabolism
- Caspases, Initiator
- Cholesterol/metabolism
- Cytokines/blood
- Disease Models, Animal
- Extracellular Traps/metabolism
- Humans
- Inflammasomes/deficiency
- Inflammasomes/genetics
- Inflammasomes/metabolism
- Inflammation/genetics
- Inflammation/metabolism
- Inflammation/pathology
- Inflammation/prevention & control
- Mice, Knockout
- Myeloid Cells/metabolism
- Myeloid Cells/pathology
- NLR Family, Pyrin Domain-Containing 3 Protein/deficiency
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- Plaque, Atherosclerotic
- Receptors, LDL/genetics
- Receptors, LDL/metabolism
- Spleen/metabolism
- Tangier Disease/blood
- Tangier Disease/genetics
Collapse
Affiliation(s)
- Marit Westerterp
- Department of Medicine, Division of Molecular Medicine, Columbia
University, New York NY
- Department of Pediatrics, Section Molecular Genetics, University of
Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Panagiotis Fotakis
- Department of Medicine, Division of Molecular Medicine, Columbia
University, New York NY
| | - Mireille Ouimet
- Department of Medicine, Division of Cardiology, New York University
Medical Center, New York NY
- University of Ottawa Heart Institute, Ontario, Canada &
Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine,
University of Ottawa, Ontario, Canada
| | - Andrea E. Bochem
- Department of Medicine, Division of Molecular Medicine, Columbia
University, New York NY
- Department of Vascular Medicine, Academic Medical Center, University
of Amsterdam, Amsterdam, The Netherlands
| | - Hanrui Zhang
- Department of Medicine, Division of Cardiology, Columbia University,
New York NY
| | - Matthew M. Molusky
- Department of Medicine, Division of Molecular Medicine, Columbia
University, New York NY
| | - Wei Wang
- Department of Medicine, Division of Molecular Medicine, Columbia
University, New York NY
| | - Sandra Abramowicz
- Department of Medicine, Division of Molecular Medicine, Columbia
University, New York NY
| | - Sacha la Bastide-van Gemert
- Department of Epidemiology, University of Groningen, University
Medical Center Groningen, Groningen, The Netherlands
| | - Nan Wang
- Department of Medicine, Division of Molecular Medicine, Columbia
University, New York NY
| | - Carrie L. Welch
- Department of Medicine, Division of Molecular Medicine, Columbia
University, New York NY
| | - Muredach P. Reilly
- Department of Medicine, Division of Cardiology, Columbia University,
New York NY
| | - Erik S. Stroes
- Department of Vascular Medicine, Academic Medical Center, University
of Amsterdam, Amsterdam, The Netherlands
| | - Kathryn J. Moore
- Department of Medicine, Division of Cardiology, New York University
Medical Center, New York NY
| | | |
Collapse
|
386
|
Mononuclear phagocytes locally specify and adapt their phenotype in a multiple sclerosis model. Nat Neurosci 2018; 21:1196-1208. [PMID: 30127427 DOI: 10.1038/s41593-018-0212-3] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 06/29/2018] [Indexed: 02/07/2023]
Abstract
Mononuclear phagocytes are key regulators of both tissue damage and repair in neuroinflammatory conditions such as multiple sclerosis. To examine divergent phagocyte phenotypes in the inflamed CNS, we introduce an in vivo imaging approach that allows us to temporally and spatially resolve the evolution of phagocyte polarization in a murine model of multiple sclerosis. We show that the initial proinflammatory polarization of phagocytes is established after spinal cord entry and critically depends on the compartment they enter. Guided by signals from the CNS environment, individual phagocytes then switch their phenotype as lesions move from expansion to resolution. Our study thus provides a real-time analysis of the temporospatial determinants and regulatory principles of phagocyte specification in the inflamed CNS.
Collapse
|
387
|
Ban N, Lee TJ, Sene A, Dong Z, Santeford A, Lin JB, Ory DS, Apte RS. Disrupted cholesterol metabolism promotes age-related photoreceptor neurodegeneration. J Lipid Res 2018; 59:1414-1423. [PMID: 29946056 PMCID: PMC6071770 DOI: 10.1194/jlr.m084442] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 06/12/2018] [Indexed: 12/17/2022] Open
Abstract
Photoreceptors have high intrinsic metabolic demand and are exquisitely sensitive to metabolic perturbation. In addition, they shed a large portion of their outer segment lipid membranes in a circadian manner, increasing the metabolic burden on the outer retina associated with the resynthesis of cell membranes and disposal of the cellular cargo. Here, we demonstrate that deletion of both ABCA1 and ABCG1 in rod photoreceptors leads to age-related accumulation of cholesterol metabolites in the outer retina, photoreceptor dysfunction, degeneration of rod outer segments, and ultimately blindness. A high-fat diet significantly accelerates rod neurodegeneration and vision loss, further highlighting the role of lipid homeostasis in regulating photoreceptor neurodegeneration and vision.
Collapse
Affiliation(s)
- Norimitsu Ban
- Departments of Ophthalmology and Visual Sciences Washington University School of Medicine in St. Louis, St. Louis, MO 63110
| | - Tae Jun Lee
- Departments of Ophthalmology and Visual Sciences Washington University School of Medicine in St. Louis, St. Louis, MO 63110
| | - Abdoulaye Sene
- Departments of Ophthalmology and Visual Sciences Washington University School of Medicine in St. Louis, St. Louis, MO 63110
| | - Zhenyu Dong
- Departments of Ophthalmology and Visual Sciences Washington University School of Medicine in St. Louis, St. Louis, MO 63110
| | - Andrea Santeford
- Departments of Ophthalmology and Visual Sciences Washington University School of Medicine in St. Louis, St. Louis, MO 63110
| | - Jonathan B Lin
- Departments of Ophthalmology and Visual Sciences Washington University School of Medicine in St. Louis, St. Louis, MO 63110
- Neuroscience Graduate Program, Division of Biology and Biomedical Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO 63110
| | - Daniel S Ory
- Diabetic Cardiovascular Disease Center, Washington University School of Medicine in St. Louis, St. Louis, MO 63110
| | - Rajendra S Apte
- Departments of Ophthalmology and Visual Sciences Washington University School of Medicine in St. Louis, St. Louis, MO 63110
- Neuroscience Graduate Program, Division of Biology and Biomedical Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO 63110
- Medicine and Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110
| |
Collapse
|
388
|
Macrophage Depletion Ameliorates Peripheral Neuropathy in Aging Mice. J Neurosci 2018; 38:4610-4620. [PMID: 29712789 DOI: 10.1523/jneurosci.3030-17.2018] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 02/06/2018] [Accepted: 03/09/2018] [Indexed: 01/02/2023] Open
Abstract
Aging is known as a major risk factor for the structure and function of the nervous system. There is urgent need to overcome such deleterious effects of age-related neurodegeneration. Here we show that peripheral nerves of 24-month-old aging C57BL/6 mice of either sex show similar pathological alterations as nerves from aging human individuals, whereas 12-month-old adult mice lack such alterations. Specifically, nerve fibers showed demyelination, remyelination and axonal lesion. Moreover, in the aging mice, neuromuscular junctions showed features typical for dying-back neuropathies, as revealed by a decline of presynaptic markers, associated with α-bungarotoxin-positive postsynapses. In line with these observations were reduced muscle strengths. These alterations were accompanied by elevated numbers of endoneurial macrophages, partially comprising the features of phagocytosing macrophages. Comparable profiles of macrophages could be identified in peripheral nerve biopsies of aging persons. To determine the pathological impact of macrophages in aging mice, we selectively targeted the cells by applying an orally administered CSF-1R specific kinase (c-FMS) inhibitor. The 6-month-lasting treatment started before development of degenerative changes at 18 months and reduced macrophage numbers in mice by ∼70%, without side effects. Strikingly, nerve structure was ameliorated and muscle strength preserved. We show, for the first time, that age-related degenerative changes in peripheral nerves are driven by macrophages. These findings may pave the way for treating degeneration in the aging peripheral nervous system by targeting macrophages, leading to reduced weakness, improved mobility, and eventually increased quality of life in the elderly.SIGNIFICANCE STATEMENT Aging is a major risk factor for the structure and function of the nervous system. Here we show that peripheral nerves of 24-month-old aging mice show similar degenerative alterations as nerves from aging human individuals. Both in mice and humans, these alterations were accompanied by endoneurial macrophages. To determine the pathological impact of macrophages in aging mice, we selectively targeted the cells by blocking a cytokine receptor, essential for macrophage survival. The treatment strongly reduced macrophage numbers and substantially improved nerve structure and muscle strength. We show, for the first time, that age-related degenerative changes in peripheral nerves are driven by macrophages. These findings may be helpful for treatment weakness and reduced mobility in the elderly.
Collapse
|
389
|
Caprariello AV, Stys PK. Two steps forward for myelin repair in multiple sclerosis. Lancet Neurol 2018; 17:297-298. [DOI: 10.1016/s1474-4422(18)30070-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 02/02/2018] [Indexed: 11/30/2022]
|
390
|
Dresselhaus E, Duerr JM, Vincent F, Sylvain EK, Beyna M, Lanyon LF, LaChapelle E, Pettersson M, Bales KR, Ramaswamy G. Class I HDAC inhibition is a novel pathway for regulating astrocytic apoE secretion. PLoS One 2018; 13:e0194661. [PMID: 29579087 PMCID: PMC5868809 DOI: 10.1371/journal.pone.0194661] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 03/07/2018] [Indexed: 11/19/2022] Open
Abstract
Despite the important role of apolipoprotein E (apoE) secretion from astrocytes in brain lipid metabolism and the strong association of apoE4, one of the human apoE isoforms, with sporadic and late onset forms of Alzheimer's disease (AD) little is known about the regulation of astrocytic apoE. Utilizing annotated chemical libraries and a phenotypic screening strategy that measured apoE secretion from a human astrocytoma cell line, inhibition of pan class I histone deacetylases (HDACs) was identified as a mechanism to increase apoE secretion. Knocking down select HDAC family members alone or in combination revealed that inhibition of the class I HDAC family was responsible for enhancing apoE secretion. Knocking down LXRα and LXRβ genes revealed that the increase in astrocytic apoE in response to HDAC inhibition occurred via an LXR-independent pathway. Collectively, these data suggest that pan class I HDAC inhibition is a novel pathway for regulating astrocytic apoE secretion.
Collapse
Affiliation(s)
- Erica Dresselhaus
- Internal Medicine Research Unit, Pfizer, Cambridge, Massachusetts, United States of America
| | - James M. Duerr
- Internal Medicine Research Unit, Pfizer, Cambridge, Massachusetts, United States of America
| | - Fabien Vincent
- Hit Discovery and Lead Profiling, Pfizer, Groton, Connecticut, United States of America
| | - Emily K. Sylvain
- Internal Medicine Research Unit, Pfizer, Cambridge, Massachusetts, United States of America
| | - Mercedes Beyna
- Internal Medicine Research Unit, Pfizer, Cambridge, Massachusetts, United States of America
| | - Lorraine F. Lanyon
- Hit Discovery and Lead Profiling, Pfizer, Groton, Connecticut, United States of America
| | - Erik LaChapelle
- Medicinal Chemistry, Pfizer, Groton, Connecticut, United States of America
| | - Martin Pettersson
- Medicinal Chemistry, Pfizer, Cambridge, Massachusetts, United States of America
| | - Kelly R. Bales
- Internal Medicine Research Unit, Pfizer, Cambridge, Massachusetts, United States of America
- * E-mail: (GR); (KRB)
| | - Gayathri Ramaswamy
- Internal Medicine Research Unit, Pfizer, Cambridge, Massachusetts, United States of America
- * E-mail: (GR); (KRB)
| |
Collapse
|
391
|
Abstract
Phagolysosome membrane rupture can trigger a maladaptive immune response that promotes tissue damage. In Science, Cantuti-Castelvetri et al. (2018) report that cholesterol-rich myelin debris overwhelms reverse cholesterol transport in aged phagocytes, leading to cholesterol crystal formation, damaged phagolysosomes, and limited tissue repair.
Collapse
Affiliation(s)
- Andrew P Lieberman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| | - Joel A Swanson
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
392
|
Ridler C. Cholesterol crystals thwart repair in old CNS. Nat Rev Neurol 2018; 14:126. [DOI: 10.1038/nrneurol.2018.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
393
|
Affiliation(s)
- Yanan Chen
- Department of Neurology, Center for Peripheral Neuropathy, University of Chicago, Chicago, IL 60637, USA
| | - Brian Popko
- Department of Neurology, Center for Peripheral Neuropathy, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
394
|
Kingwell K. Cholesterol clearance restores remyelination. Nat Rev Drug Discov 2018; 17:94. [DOI: 10.1038/nrd.2018.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|