1
|
Festa LK, Jordan-Sciutto KL, Grinspan JB. Neuroinflammation: An Oligodendrocentric View. Glia 2025; 73:1113-1129. [PMID: 40059542 PMCID: PMC12014387 DOI: 10.1002/glia.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/16/2025]
Abstract
Chronic neuroinflammation, driven by central nervous system (CNS)-resident astrocytes and microglia, as well as infiltration of the peripheral immune system, is an important pathologic mechanism across a range of neurologic diseases. For decades, research focused almost exclusively on how neuroinflammation impacted neuronal function; however, there is accumulating evidence that injury to the oligodendrocyte lineage is an important component for both pathologic and clinical outcomes. While oligodendrocytes are able to undergo an endogenous repair process known as remyelination, this process becomes inefficient and usually fails in the presence of sustained inflammation. The present review focuses on our current knowledge regarding activation of the innate and adaptive immune systems in the chronic demyelinating disease, multiple sclerosis, and provides evidence that sustained neuroinflammation in other neurologic conditions, such as perinatal white matter injury, traumatic brain injury, and viral infections, converges on oligodendrocyte injury. Lastly, the therapeutic potential of targeting the impact of inflammation on the oligodendrocyte lineage in these diseases is discussed.
Collapse
Affiliation(s)
- Lindsay K Festa
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Kelly L Jordan-Sciutto
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Judith B Grinspan
- Department of Neurology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
2
|
Liran M, Fischer I, Elboim M, Rahamim N, Gordon T, Urshansky N, Assaf Y, Barak B, Barak S. Long-Term Excessive Alcohol Consumption Enhances Myelination in the Mouse Nucleus Accumbens. J Neurosci 2025; 45:e0280242025. [PMID: 39909566 PMCID: PMC11968546 DOI: 10.1523/jneurosci.0280-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 12/15/2024] [Accepted: 01/08/2025] [Indexed: 02/07/2025] Open
Abstract
Chronic excessive alcohol (ethanol) consumption induces neuroadaptations in the brain's reward system, including biochemical and structural abnormalities in white matter that are implicated in addiction phenotypes. Here, we demonstrate that long-term (12 week) voluntary ethanol consumption enhances myelination in the nucleus accumbens (NAc) of female and male adult mice, as evidenced by molecular, ultrastructural, and cellular alterations. Specifically, transmission electron microscopy analysis showed increased myelin thickness in the NAc following long-term ethanol consumption, while axon diameter remained unaffected. These changes were paralleled by increased mRNA transcript levels of key transcription factors essential for oligodendrocyte (OL) differentiation, along with elevated expression of critical myelination-related genes. In addition, diffusion tensor imaging revealed increased connectivity between the NAc and the prefrontal cortex, reflected by a higher number of tracts connecting these regions. We also observed ethanol-induced effects on OL lineage cells, with a reduction in the number of mature OLs after 3 weeks of ethanol consumption, followed by an increase after 6 weeks. These findings suggest that ethanol alters OL development prior to increasing myelination in the NAc. Finally, chronic administration of the promyelination drug clemastine to mice with a history of heavy ethanol consumption further elevated ethanol intake and preference, suggesting that increased myelination may contribute to escalated drinking behavior. Together, these findings suggest that heavy ethanol consumption disrupts OL development, induces enhanced myelination in the NAc, and may drive further ethanol intake, reinforcing addictive behaviors.
Collapse
Affiliation(s)
- Mirit Liran
- Department of Neurobiology, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Inbar Fischer
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - May Elboim
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Nofar Rahamim
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Tamar Gordon
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Nataly Urshansky
- School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yaniv Assaf
- Department of Neurobiology, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Boaz Barak
- Department of Neurobiology, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
- School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Segev Barak
- Department of Neurobiology, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
- School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
3
|
Wang Q, Huang T, Zheng Z, Su Y, Wu Z, Zeng C, Yu G, Liu Y, Wang X, Li H, Chen X, Jiang Z, Zhang J, Zhuang Y, Tian Y, Yang Q, Verkhratsky A, Wan Y, Yi C, Niu J. Oligodendroglial precursor cells modulate immune response and early demyelination in a murine model of multiple sclerosis. Sci Transl Med 2025; 17:eadn9980. [PMID: 40173259 DOI: 10.1126/scitranslmed.adn9980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 06/23/2024] [Accepted: 03/12/2025] [Indexed: 04/04/2025]
Abstract
Reproducing the pathophysiology of human multiple sclerosis (MS) in animal models is critical to identifying mechanisms triggering demyelination and to developing early intervention strategies. Here, we aimed to model overactivated Wnt (wingless-related integration site) signaling previously shown in postmortem brain tissues of patients with MS by inducing experimental autoimmune encephalomyelitis (EAE) in PdgfraCreER;Apcfl/fl and Olig2Cre;Apcfl/fl mice. These mice have overactivated Wnt signaling in oligodendrocyte precursor cells (OPCs) because of a conditional knockout of the pathway repressor adenomatous polyposis coli (APC). PdgfraCreER;Apcfl/fl EAE mice exhibited increased expression of markers for Wnt activation such as Axis inhibition protein 2 (AXIN2) and Wnt inhibitory factor 1 (WIF1) in OPCs and showed exacerbated EAE progression in both the spinal cord and the brain. Genetic or antibody-mediated ablation of CC-chemokine ligand 4 (CCL4) prevented infiltration of CD4+ T cells and arrested disease progression in these mice. A characterization of CNS (central nervous system) immune cell clusters identified an augmented subpopulation of NK1.1+CD11b+Gr-1+ cytotoxic macrophages in PdgfraCreER;Apcfl/fl EAE mice. Microinjection of this subpopulation of macrophages into the brains of wild-type C57/B6J mice was sufficient to induce demyelination. Ablation of CD4+ T cells prevented the effects of Wnt overactivation on demyelination and immune cell infiltration. Antagonizing chemokine receptor 5 (CCR5) using a European Medicines Agency-approved drug, maraviroc, reduced immune cell infiltration, alleviated demyelination, and attenuated EAE progression. We found an OPC-orchestrated immune cellular network that instigates early demyelination, provides insight into MS pathophysiology, and suggests avenues for early interventions.
Collapse
Affiliation(s)
- Qi Wang
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
- Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China
| | - Taida Huang
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Zihan Zheng
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China
| | - Yixun Su
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
- Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China
| | - Zhonghao Wu
- Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China
| | - Cong Zeng
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China
| | - Guangdan Yu
- China Astronaut Research and Training Center, Beijing 100094, China
| | - Yang Liu
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China
| | - Xiaorui Wang
- Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China
| | - Hui Li
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
- Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China
| | - Xiaoying Chen
- Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China
| | - Zhuoxu Jiang
- Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China
| | - Jinyu Zhang
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Yuan Zhuang
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Yi Tian
- Institute of Immunology, Third Military Medical University, Chongqing 400038, China
| | - Qingwu Yang
- Department of Neurology, Second Affiliated Hospital, Third Military Medical University, Chongqing 400038, China
- Chongqing Institute for Brain and Intelligence, Chongqing 400037, China
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M139PL, UK
- Department of Neurosciences, University of the Basque Country, Leioa 48940, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
- International Joint Research Centre on Purinergic Signalling of Sichuan Province, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, China
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang 110122, China
- Celica Biomedical, Technology Park 24, 1000 Ljubljana, Slovenia
| | - Ying Wan
- Biomedical Analysis Center, Third Military Medical University, Chongqing 400038, China
- Institute for Translational Immunology, Chongqing 400038, China
| | - Chenju Yi
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou 510080, China
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen 518107, China
| | - Jianqin Niu
- Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China
- Chongqing Key Laboratory of Neurobiology, Chongqing 400038, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing 400042, China
| |
Collapse
|
4
|
Kornberg MD, Calabresi PA. Multiple Sclerosis and Other Acquired Demyelinating Diseases of the Central Nervous System. Cold Spring Harb Perspect Biol 2025; 17:a041374. [PMID: 38806240 PMCID: PMC11875095 DOI: 10.1101/cshperspect.a041374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Acquired demyelinating diseases of the central nervous system (CNS) comprise inflammatory conditions, including multiple sclerosis (MS) and related diseases, as well as noninflammatory conditions caused by toxic, metabolic, infectious, traumatic, and neurodegenerative insults. Here, we review the spectrum of diseases producing acquired CNS demyelination before focusing on the prototypical example of MS, exploring the pathologic mechanisms leading to myelin injury in relapsing and progressive MS and summarizing the mechanisms and modulators of remyelination. We highlight the complex interplay between the immune system, oligodendrocytes and oligodendrocyte progenitor cells (OPCs), and other CNS glia cells such as microglia and astrocytes in the pathogenesis and clinical course of MS. Finally, we review emerging therapeutic strategies that exploit our growing understanding of disease mechanisms to limit progression and promote remyelination.
Collapse
Affiliation(s)
- Michael D Kornberg
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21287, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21287, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland 21205, USA
| |
Collapse
|
5
|
Milstein JL, Kulas JA, Kamal A, Lo AB, Ferris HA. Regulation of glial ApoE secretion by the mevalonate pathway is independent of ApoE isoform. J Alzheimers Dis 2025; 104:473-487. [PMID: 39994996 DOI: 10.1177/13872877251317732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
BackgroundLipids synthesized in astrocytes are distributed to other brain cells in high-density lipoprotein-like ApoE particles. ApoE, which is a powerful genetic risk factor for developing Alzheimer's disease, is secreted differently depending on genotype. Secretion of ApoE from mouse astrocytes is regulated by the mevalonate pathway.ObjectiveWe aimed to understand if the regulation of ApoE secretion from astrocytes by the mevalonate pathway was the same between mouse ApoE and ApoE from humanized mice, and if this is impacted by ApoE isoform.MethodsAstrocyte-enriched glial cultures from wild-type and humanized ApoE targeted-replacement mice were treated with pharmacological inhibitors of various steps along the mevalonate pathway and ApoE in the conditioned media was measured.ResultsWe show that statins and prenylation inhibitors, but not specific cholesterol inhibitors, reduce extracellular ApoE lipoparticle levels in astrocyte-enriched glial cultures, and that this occurs in cells harboring either the mouse ApoE or any of the three human ApoE genotypes to a similar extent. We find that geranylgeranylation modulates ApoE release from astrocytes, and it does so independent of ApoE genotype.ConclusionsOur results suggest that prenylation broadly regulates ApoE secretion from astrocytes regardless of ApoE genotype, and that this is mediated specifically by geranylgeranylation. Therefore, our data implicates geranylgeranylation as a general mechanism modulating ApoE release from astrocytes, but likely is not responsible for the reported baseline differences in ApoE secretion seen in vivo and in vitro across genotypes.
Collapse
Affiliation(s)
- Joshua L Milstein
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Joshua A Kulas
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Aria Kamal
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - An B Lo
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Heather A Ferris
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
6
|
Brown RM, Le HH, Babcock IW, Harris TH, Gaultier A. Functional analysis of antigen presentation by enteric glial cells during intestinal inflammation. Glia 2025; 73:291-308. [PMID: 39495092 DOI: 10.1002/glia.24632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 09/09/2024] [Accepted: 10/10/2024] [Indexed: 11/05/2024]
Abstract
The Enteric Nervous System is composed of a vastly interconnected network of neurons and glial cells that coordinate to regulate homeostatic gut function including intestinal motility, nutrient sensing, and mucosal barrier immunity. Enteric Glial Cells (EGCs) are a heterogeneous cell population located throughout the gastrointestinal tract and have well described roles in regulating intestinal immune responses. Enteric Glial Cells have been suggested to act as nonconventional antigen presenting cells via the Major Histocompatibility Complex II (MHC II), though this has not been confirmed functionally. Here, we investigate the capability of EGCs to present antigen on MHC I and MHC II using in vitro antigen presentation assays performed with primary murine EGC cultures. We found that EGCs are capable of functional antigen presentation on MHC I, including antigen cross-presentation, but are not capable of functional antigen presentation on MHC II. We also determined EGC cell surface MHC I and MHC II expression levels by flow cytometry during intestinal inflammation during Dextran Sodium Sulfate-induced colitis or acute Toxoplasma gondii infection. We found that EGCs upregulate MHC I during acute T. gondii infection and induce low-level MHC II expression. These findings suggest that EGCs may be important in the regulation of CD8+ T cell responses via MHC I mediated antigen (cross) presentation but may not be relevant for MHC II-mediated antigen presentation.
Collapse
Affiliation(s)
- Ryan M Brown
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Neuroscience Graduate Program, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Helen H Le
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Isaac W Babcock
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Neuroscience Graduate Program, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Tajie H Harris
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Alban Gaultier
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
7
|
Beiter RM, Raghavan TP, Suchocki O, Ennerfelt HE, Rivet-Noor CR, Merchak AR, Phillips JL, Bathe T, Lukens JR, Prokop S, Dupree JL, Gaultier A. Clusterin induced by OPC phagocytosis blocks IL-9 secretion to inhibit myelination in a model of Alzheimer's disease. Heliyon 2025; 11:e41635. [PMID: 39866464 PMCID: PMC11761289 DOI: 10.1016/j.heliyon.2025.e41635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 01/01/2025] [Accepted: 01/01/2025] [Indexed: 01/28/2025] Open
Abstract
Background Variants in the CLUSTERIN gene have been identified as a risk factor for late-onset Alzheimer's disease and are linked to decreased white matter integrity in healthy adults. However, the specific role for clusterin in myelin maintenance in the context of Alzheimer's disease remains unclear. Methods We employed a combination of immunofluorescence and transmission electron microscopy techniques, primary culture of OPCs, and an animal model of Alzheimer's disease. Results We found that phagocytosis of debris such as amyloid beta, myelin, and apoptotic cells, increases clusterin expression in oligodendrocyte progenitors. We further discovered that exposure to clusterin inhibits differentiation of oligodendrocyte progenitors. Mechanistically, clusterin blunts production of IL-9 and addition of exogenous IL-9 can rescue clusterin-inhibited myelination. Lastly, we demonstrate that clusterin deletion in mice prevents myelin loss in the 5XFAD model. Discussion Our data suggest that clusterin could play a key role in Alzheimer's disease myelin pathology.
Collapse
Affiliation(s)
- Rebecca M. Beiter
- Center for Brain Immunology and Glia, Department of Neuroscience, Charlottesville, VA 22908, USA
- Graduate Program in Neuroscience, Charlottesville, VA 22908, USA
- Department of Neurobiology, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Tula P. Raghavan
- Center for Brain Immunology and Glia, Department of Neuroscience, Charlottesville, VA 22908, USA
- Graduate Program in Neuroscience, Charlottesville, VA 22908, USA
- Medical Scientist Training Program, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Olivia Suchocki
- Center for Brain Immunology and Glia, Department of Neuroscience, Charlottesville, VA 22908, USA
- Graduate Program in Neuroscience, Charlottesville, VA 22908, USA
| | - Hannah E. Ennerfelt
- Center for Brain Immunology and Glia, Department of Neuroscience, Charlottesville, VA 22908, USA
- Graduate Program in Neuroscience, Charlottesville, VA 22908, USA
| | - Courtney R. Rivet-Noor
- Center for Brain Immunology and Glia, Department of Neuroscience, Charlottesville, VA 22908, USA
- Graduate Program in Neuroscience, Charlottesville, VA 22908, USA
| | - Andrea R. Merchak
- Center for Brain Immunology and Glia, Department of Neuroscience, Charlottesville, VA 22908, USA
- Graduate Program in Neuroscience, Charlottesville, VA 22908, USA
| | - Jennifer L. Phillips
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
- Department of Pathology, College of Medicine, University of Florida, Gainesville, 32610, USA
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Tim Bathe
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
- Department of Pathology, College of Medicine, University of Florida, Gainesville, 32610, USA
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - John R. Lukens
- Center for Brain Immunology and Glia, Department of Neuroscience, Charlottesville, VA 22908, USA
- Graduate Program in Neuroscience, Charlottesville, VA 22908, USA
| | - Stefan Prokop
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
- Department of Pathology, College of Medicine, University of Florida, Gainesville, 32610, USA
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Jeffrey L. Dupree
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Alban Gaultier
- Center for Brain Immunology and Glia, Department of Neuroscience, Charlottesville, VA 22908, USA
- Graduate Program in Neuroscience, Charlottesville, VA 22908, USA
| |
Collapse
|
8
|
Kala S, Strutz AG, Katt ME. The Rise of Pluripotent Stem Cell-Derived Glia Models of Neuroinflammation. Neurol Int 2025; 17:6. [PMID: 39852770 PMCID: PMC11767680 DOI: 10.3390/neurolint17010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/02/2025] [Accepted: 01/09/2025] [Indexed: 01/26/2025] Open
Abstract
Neuroinflammation is a blanket term that describes the body's complex inflammatory response in the central nervous system (CNS). It encompasses a phenotype shift to a proinflammatory state, the release of cytokines, the recruitment of peripheral immune cells, and a wide variety of other processes. Neuroinflammation has been implicated in nearly every major CNS disease ranging from Alzheimer's disease to brain cancer. Understanding and modeling neuroinflammation is critical for the identification of novel therapeutic targets in the treatment of CNS diseases. Unfortunately, the translation of findings from non-human models has left much to be desired. This review systematically discusses the role of human pluripotent stem cell (hPSC)-derived glia and supporting cells within the CNS, including astrocytes, microglia, oligodendrocyte precursor cells, pericytes, and endothelial cells, to describe the state of the field and hope for future discoveries. hPSC-derived cells offer an expanded potential to study the pathobiology of neuroinflammation and immunomodulatory cascades that impact disease progression. While much progress has been made in the development of models, there is much left to explore in the application of these models to understand the complex inflammatory response in the CNS.
Collapse
Affiliation(s)
- Srishti Kala
- Cancer Cell Biology Graduate Education Program, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA;
| | - Andrew G. Strutz
- Department of Microbiology, Immunology, and Cell Biology, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA;
| | - Moriah E. Katt
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA
- Department of Neuroscience, School of Medicine, West Virginia University Health Science Center, Morgantown, WV 26506, USA
| |
Collapse
|
9
|
Wang Y, Ghimire S, Mangalam A, Kang Z. RiboTag-based RNA-Seq uncovers oligodendroglial lineage-specific inflammation in autoimmune encephalomyelitis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.24.630168. [PMID: 39764033 PMCID: PMC11703255 DOI: 10.1101/2024.12.24.630168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Oligodendroglial lineage cells (OLCs) are critical for neuronal support functions, including myelination and remyelination. Emerging evidence reveals their active roles in neuroinflammation, particularly in conditions like Multiple Sclerosis (MS). This study explores the inflammatory translatome of OLCs during the early onset of experimental autoimmune encephalomyelitis (EAE), an established MS model. Using RiboTag-based RNA sequencing in genetically modified Olig2-Cre RiboTag mice, we identified 1,556 upregulated and 683 downregulated genes in EAE OLCs. Enrichment analysis indicated heightened immune-related pathways, such as cytokine signaling, interferon responses, and antigen presentation, while downregulated genes were linked to neuronal development and myelination. Notably, OLCs expressed cytokines/chemokines, and their receptor, highlighting their active involvement in neuroinflammatory signaling. Functional studies demonstrated that interferon-gamma (IFN-γ) signaling in OLCs exacerbates EAE pathology by enhancing antigen presentation and chemokine production, whereas interferon-beta (IFN-β) signaling showed minimal impact. These findings provide novel insights into the inflammatory role of OLCs in EAE and suggest therapeutic potential in targeting OLC-mediated neuroinflammation for MS and related disorders.
Collapse
Affiliation(s)
- Yuhang Wang
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Sudeep Ghimire
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Ashutosh Mangalam
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Zizhen Kang
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
10
|
Wang S, Li C, Kang X, Su X, Liu Y, Wang Y, Liu S, Deng X, Huang H, Li T, Lu D, Cai W, Lu Z, Wei L, Lu T. Agomelatine promotes differentiation of oligodendrocyte precursor cells and preserves white matter integrity after cerebral ischemic stroke. J Cereb Blood Flow Metab 2024; 44:1487-1500. [PMID: 38853430 PMCID: PMC11574932 DOI: 10.1177/0271678x241260100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 05/14/2024] [Accepted: 05/18/2024] [Indexed: 06/11/2024]
Abstract
White matter injury contributes to neurological disorders after acute ischemic stroke (AIS). The repair of white matter injury is dependent on the re-myelination by oligodendrocytes. Both melatonin and serotonin antagonist have been proved to protect against post-stroke white matter injury. Agomelatine (AGM) is a multi-functional treatment which is both a melatonin receptor agonist and selective serotonin receptor antagonist. Whether AGM protects against white matter injury after stroke and the underlying mechanisms remain elusive. Here, using the transient middle cerebral artery occlusion (tMCAO) model, we evaluated the therapeutic effects of AGM in stroke mice. Sensorimotor and cognitive functions, white matter integrity, oligodendroglial regeneration and re-myelination in stroke hemisphere after AGM treatment were analyzed. We found that AGM efficiently preserved white matter integrity, reduced brain tissue loss, attenuated long-term sensorimotor and cognitive deficits in tMCAO models. AGM treatment promoted OPC differentiation and enhanced re-myelination both in vitro, ex vivo and in vivo, although OPC proliferation was unaffected. Mechanistically, AGM activated low density lipoprotein receptor related protein 1 (LRP1), peroxisome proliferator-activated receptor γ (PPARγ) signaling thus promoted OPC differentiation and re-myelination after stroke. Inhibition of PPARγ or knock-down of LRP1 in OPCs reversed the beneficial effects of AGM. Altogether, our data indicate that AGM represents a novel therapy against white matter injury after cerebral ischemia.
Collapse
Affiliation(s)
- Shisi Wang
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Chunyi Li
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xinmei Kang
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaotao Su
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuxin Liu
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuge Wang
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Sanxin Liu
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaohui Deng
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Huipeng Huang
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tiemei Li
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Danli Lu
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wei Cai
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, China
| | - Zhengqi Lu
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Lei Wei
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tingting Lu
- Department of Neurology, Mental and Neurological Disease Research Center, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
11
|
Li X, Tong H, Xu S, Zhou G, Yang T, Yin S, Yang S, Li X, Li S. Neuroinflammatory Proteins in Huntington's Disease: Insights into Mechanisms, Diagnosis, and Therapeutic Implications. Int J Mol Sci 2024; 25:11787. [PMID: 39519337 PMCID: PMC11546928 DOI: 10.3390/ijms252111787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Huntington's disease (HD) is a hereditary neurodegenerative disorder caused by a CAG tract expansion in the huntingtin gene (HTT). HD is characterized by involuntary movements, cognitive decline, and behavioral changes. Pathologically, patients with HD show selective striatal neuronal vulnerability at the early disease stage, although the mutant protein is ubiquitously expressed. Activation of the immune system and glial cell-mediated neuroinflammatory responses are early pathological features and have been found in all neurodegenerative diseases (NDDs), including HD. However, the role of inflammation in HD, as well as its therapeutic significance, has been less extensively studied compared to other NDDs. This review highlights the significantly elevated levels of inflammatory proteins and cellular markers observed in various HD animal models and HD patient tissues, emphasizing the critical roles of microglia, astrocytes, and oligodendrocytes in mediating neuroinflammation in HD. Moreover, it expands on recent discoveries related to the peripheral immune system's involvement in HD. Although current immunomodulatory treatments and inflammatory biomarkers for adjunctive diagnosis in HD are limited, targeting inflammation in combination with other therapies, along with comprehensive personalized treatment approaches, shows promising therapeutic potential.
Collapse
Affiliation(s)
- Xinhui Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Huichun Tong
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Shuying Xu
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Gongke Zhou
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Tianqi Yang
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Shurui Yin
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Sitong Yang
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Xiaojiang Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| | - Shihua Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (X.L.); (H.T.); (S.X.); (G.Z.); (T.Y.); (S.Y.); (S.Y.); (X.L.)
| |
Collapse
|
12
|
Gao R, Song SJ, Tian MY, Wang LB, Zhang Y, Li X. Myelin debris phagocytosis in demyelinating disease. Glia 2024; 72:1934-1954. [PMID: 39073200 DOI: 10.1002/glia.24602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024]
Abstract
Demyelinating diseases are often caused by a variety of triggers, including immune responses, viral infections, malnutrition, hypoxia, or genetic factors, all of which result in the loss of myelin in the nervous system. The accumulation of myelin debris at the lesion site leads to neuroinflammation and inhibits remyelination; therefore, it is crucial to promptly remove the myelin debris. Initially, Fc and complement receptors on cellular surfaces were the primary clearance receptors responsible for removing myelin debris. However, subsequent studies have unveiled the involvement of additional receptors, including Mac-2, TAM receptors, and the low-density lipoprotein receptor-related protein 1, in facilitating the removal process. In addition to microglia and macrophages, which serve as the primary effector cells in the disease phase, a variety of other cell types such as astrocytes, Schwann cells, and vascular endothelial cells have been demonstrated to engage in the phagocytosis of myelin debris. Furthermore, we have concluded that oligodendrocyte precursor cells, as myelination precursor cells, also exhibit this phagocytic capability. Moreover, our research group has innovatively identified the low-density lipoprotein receptor as a potential phagocytic receptor for myelin debris. In this article, we discuss the functional processes of various phagocytes in demyelinating diseases. We also highlight the alterations in signaling pathways triggered by phagocytosis, and provide a comprehensive overview of the various phagocytic receptors involved. Such insights are invaluable for pinpointing potential therapeutic strategies for the treatment of demyelinating diseases by targeting phagocytosis.
Collapse
Affiliation(s)
- Rui Gao
- The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Sheng-Jiao Song
- The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Meng-Yuan Tian
- The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Li-Bin Wang
- Neurosurgery Department, Huazhong University of Science and Technology Union Shenzhen Hospital/Shenzhen Nanshan Hospital, Shenzhen, Guangdong, China
| | - Yuan Zhang
- The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Xing Li
- The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, China
| |
Collapse
|
13
|
Castelo-Branco G, Kukanja P, Guerreiro-Cacais AO, Rubio Rodríguez-Kirby LA. Disease-associated oligodendroglia: a putative nexus in neurodegeneration. Trends Immunol 2024; 45:750-759. [PMID: 39322475 DOI: 10.1016/j.it.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/27/2024]
Abstract
Neural cells in our central nervous system (CNS) have long been thought to be mere targets of neuroinflammatory events in neurodegenerative diseases such as multiple sclerosis (MS) or Alzheimer's disease. While glial populations such as microglia and astrocytes emerged as active responders and modifiers of pathological environments, oligodendroglia and neurons have been associated with altered homeostasis and eventual cell death. The advent of single-cell and spatial omics technologies has demonstrated transitions of CNS-resident glia, including oligodendroglia, into disease-associated (DA) states. Anchored in recent findings of their roles in MS, we propose that DA glia constitute key nexus of disease progression, with DA oligodendroglia contributing to the modulation of neuroinflammation in certain neurodegenerative diseases, constituting novel putative pharmacological targets for such pathologies.
Collapse
Affiliation(s)
- Gonçalo Castelo-Branco
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Biomedicum, Karolinska Institutet, 17177 Stockholm, Sweden.
| | - Petra Kukanja
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Biomedicum, Karolinska Institutet, 17177 Stockholm, Sweden
| | - André O Guerreiro-Cacais
- Center for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Karolinska University Hospital, 171 76 Solna, Sweden
| | - Leslie A Rubio Rodríguez-Kirby
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Biomedicum, Karolinska Institutet, 17177 Stockholm, Sweden
| |
Collapse
|
14
|
Miller MR, Landis HE, Miller RE, Tizabi Y. Intercellular Adhesion Molecule 1 (ICAM-1): An Inflammatory Regulator with Potential Implications in Ferroptosis and Parkinson's Disease. Cells 2024; 13:1554. [PMID: 39329738 PMCID: PMC11430830 DOI: 10.3390/cells13181554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024] Open
Abstract
Intercellular adhesion molecule 1 (ICAM-1/CD54), a transmembrane glycoprotein, has been considered as one of the most important adhesion molecules during leukocyte recruitment. It is encoded by the ICAM1 gene and plays a central role in inflammation. Its crucial role in many inflammatory diseases such as ulcerative colitis and rheumatoid arthritis are well established. Given that neuroinflammation, underscored by microglial activation, is a key element in neurodegenerative diseases such as Parkinson's disease (PD), we investigated whether ICAM-1 has a role in this progressive neurological condition and, if so, to elucidate the underpinning mechanisms. Specifically, we were interested in the potential interaction between ICAM-1, glial cells, and ferroptosis, an iron-dependent form of cell death that has recently been implicated in PD. We conclude that there exist direct and indirect (via glial cells and T cells) influences of ICAM-1 on ferroptosis and that further elucidation of these interactions can suggest novel intervention for this devastating disease.
Collapse
Affiliation(s)
| | - Harold E. Landis
- Integrative Medicine Fellow, University of Arizona College of Medicine, Tucson, AZ 85724, USA
| | | | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, 520 W Street NW, Washington, DC 20059, USA
| |
Collapse
|
15
|
Zveik O, Rechtman A, Ganz T, Vaknin-Dembinsky A. The interplay of inflammation and remyelination: rethinking MS treatment with a focus on oligodendrocyte progenitor cells. Mol Neurodegener 2024; 19:53. [PMID: 38997755 PMCID: PMC11245841 DOI: 10.1186/s13024-024-00742-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Multiple sclerosis (MS) therapeutic goals have traditionally been dichotomized into two distinct avenues: immune-modulatory-centric interventions and pro-regenerative strategies. Oligodendrocyte progenitor cells (OPCs) were regarded for many years solely in concern to their potential to generate oligodendrocytes and myelin in the central nervous system (CNS). However, accumulating data elucidate the multifaceted roles of OPCs, including their immunomodulatory functions, positioning them as cardinal constituents of the CNS's immune landscape. MAIN BODY In this review, we will discuss how the two therapeutic approaches converge. We present a model by which (1) an inflammation is required for the appropriate pro-myelinating immune function of OPCs in the chronically inflamed CNS, and (2) the immune function of OPCs is crucial for their ability to differentiate and promote remyelination. This model highlights the reciprocal interactions between OPCs' pro-myelinating and immune-modulating functions. Additionally, we review the specific effects of anti- and pro-inflammatory interventions on OPCs, suggesting that immunosuppression adversely affects OPCs' differentiation and immune functions. CONCLUSION We suggest a multi-systemic therapeutic approach, which necessitates not a unidimensional focus but a harmonious balance between OPCs' pro-myelinating and immune-modulatory functions.
Collapse
Affiliation(s)
- Omri Zveik
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel
| | - Ariel Rechtman
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel
| | - Tal Ganz
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel
| | - Adi Vaknin-Dembinsky
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel.
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel.
| |
Collapse
|
16
|
Haroon A, Seerapu H, Fang LP, Weß JH, Bai X. Unlocking the Potential: immune functions of oligodendrocyte precursor cells. Front Immunol 2024; 15:1425706. [PMID: 39044821 PMCID: PMC11263107 DOI: 10.3389/fimmu.2024.1425706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/26/2024] [Indexed: 07/25/2024] Open
Abstract
Oligodendrocyte precursor cells (OPCs) have long been regarded as progenitors of oligodendrocytes, yet recent advances have illuminated their multifaceted nature including their emerging immune functions. This review seeks to shed light on the immune functions exhibited by OPCs, spanning from phagocytosis to immune modulation and direct engagement with immune cells across various pathological scenarios. Comprehensive understanding of the immune functions of OPCs alongside their other roles will pave the way for targeted therapies in neurological disorders.
Collapse
Affiliation(s)
- Amr Haroon
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Harsha Seerapu
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Li-Pao Fang
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, Homburg, Germany
| | - Jakob Heinrich Weß
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Xianshu Bai
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
- Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, Homburg, Germany
| |
Collapse
|
17
|
Beiter RM, Raghavan TP, Suchocki O, Ennerfelt HE, Rivet-Noor CR, Merchak AR, Phillips JL, Bathe T, Lukens JR, Prokop S, Dupree JL, Gaultier A. Oligomeric amyloid beta prevents myelination in a clusterin-dependent manner. RESEARCH SQUARE 2024:rs.3.rs-4415143. [PMID: 38853911 PMCID: PMC11160922 DOI: 10.21203/rs.3.rs-4415143/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Background White matter loss is a well-documented phenomenon in Alzheimer's disease (AD) patients that has been recognized for decades. However, the underlying reasons for the failure of oligodendrocyte progenitor cells (OPCs) to repair myelin deficits in these patients remain elusive. A single nucleotide polymorphism (SNP) in Clusterin has been identified as a risk factor for late-onset Alzheimer's disease and linked to a decrease in white matter integrity in healthy adults, but its specific role in oligodendrocyte function and myelin maintenance in Alzheimer's disease pathology remains unclear. Methods To investigate the impact of Clusterin on OPCs in the context of Alzheimer's disease, we employed a combination of immunofluorescence and transmission electron microscopy techniques, primary culture of OPCs, and an animal model of Alzheimer's disease. Results Our findings demonstrate that Clusterin, a risk factor for late-onset AD, is produced by OPCs and inhibits their differentiation into oligodendrocytes. Specifically, we observed upregulation of Clusterin in OPCs in the 5xFAD mouse model of AD. We also found that the phagocytosis of debris, including amyloid beta (Aβ), myelin, and apoptotic cells leads to the upregulation of Clusterin in OPCs. In vivo experiments confirmed that Aβ oligomers stimulate Clusterin upregulation and that OPCs are capable of phagocytosing Aβ. Furthermore, we discovered that Clusterin significantly inhibits OPC differentiation and hinders the production of myelin proteins. Finally, we demonstrate that Clusterin inhibits OPC differentiation by reducing the production of IL-9 by OPCs. Conclusion Our data suggest that Clusterin may play a key role in the impaired myelin repair observed in AD and could serve as a promising therapeutic target for addressing AD-associated cognitive decline.
Collapse
|
18
|
Ma Y, Jiang T, Zhu X, Xu Y, Wan K, Zhang T, Xie M. Efferocytosis in dendritic cells: an overlooked immunoregulatory process. Front Immunol 2024; 15:1415573. [PMID: 38835772 PMCID: PMC11148234 DOI: 10.3389/fimmu.2024.1415573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 05/09/2024] [Indexed: 06/06/2024] Open
Abstract
Efferocytosis, the process of engulfing and removing apoptotic cells, plays an essential role in preserving tissue health and averting undue inflammation. While macrophages are primarily known for this task, dendritic cells (DCs) also play a significant role. This review delves into the unique contributions of various DC subsets to efferocytosis, highlighting the distinctions in how DCs and macrophages recognize and handle apoptotic cells. It further explores how efferocytosis influences DC maturation, thereby affecting immune tolerance. This underscores the pivotal role of DCs in orchestrating immune responses and sustaining immune equilibrium, providing new insights into their function in immune regulation.
Collapse
Affiliation(s)
- Yanyan Ma
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Tangxing Jiang
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xun Zhu
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yizhou Xu
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ke Wan
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Tingxuan Zhang
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Miaorong Xie
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
Elkjaer ML, Hartebrodt A, Oubounyt M, Weber A, Vitved L, Reynolds R, Thomassen M, Rottger R, Baumbach J, Illes Z. Single-Cell Multi-Omics Map of Cell Type-Specific Mechanistic Drivers of Multiple Sclerosis Lesions. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200213. [PMID: 38564686 PMCID: PMC11073880 DOI: 10.1212/nxi.0000000000200213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/19/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND AND OBJECTIVES In progressive multiple sclerosis (MS), compartmentalized inflammation plays a pivotal role in the complex pathology of tissue damage. The interplay between epigenetic regulation, transcriptional modifications, and location-specific alterations within white matter (WM) lesions at the single-cell level remains underexplored. METHODS We examined intracellular and intercellular pathways in the MS brain WM using a novel dataset obtained by integrated single-cell multi-omics techniques from 3 active lesions, 3 chronic active lesions, 3 remyelinating lesions, and 3 control WM of 6 patients with progressive MS and 3 non-neurologic controls. Single-nucleus RNA-seq and ATAC-seq were combined and additionally enriched with newly conducted spatial transcriptomics from 1 chronic active lesion. Functional gene modules were then validated in our previously published bulk tissue transcriptome data obtained from 73 WM lesions of patients with progressive MS and 25 WM of non-neurologic disease controls. RESULTS Our analysis uncovered an MS-specific oligodendrocyte genetic signature influenced by the KLF/SP gene family. This modulation has potential associations with the autocrine iron uptake signaling observed in transcripts of transferrin and its receptor LRP2. In addition, an inflammatory profile emerged within these oligodendrocytes. We observed unique cellular endophenotypes both at the periphery and within the chronic active lesion. These include a distinct metabolic astrocyte phenotype, the importance of FGF signaling among astrocytes and neurons, and a notable enrichment of mitochondrial genes at the lesion edge populated predominantly by astrocytes. Our study also identified B-cell coexpression networks indicating different functional B-cell subsets with differential location and specific tendencies toward certain lesion types. DISCUSSION The use of single-cell multi-omics has offered a detailed perspective into the cellular dynamics and interactions in MS. These nuanced findings might pave the way for deeper insights into lesion pathogenesis in progressive MS.
Collapse
Affiliation(s)
- Maria L Elkjaer
- From the Department of Neurology (M.L.E., A.W., Z.I.), Odense University Hospital; BRIDGE (M.L.E., A.W., M.T., Z.I.), Department of Clinical Research; Department of Molecular Medicine (M.L.E., A.W., L.V., Z.I.), University of Southern Denmark, Odense, Denmark; Biomedical Network Science Lab (A.H.), Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Department of Mathematics and Computer Science (A.H., Richard Rottger, J.B.), University of Southern Denmark, Odense, Denmark; Institute for Computational Systems Biology (M.O., J.B.), University of Hamburg, Germany; Department of Brain Sciences (Richard Reynolds), Imperial College, London, United Kingdom; and Clinical Genome Center (M.T.), Research Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Anne Hartebrodt
- From the Department of Neurology (M.L.E., A.W., Z.I.), Odense University Hospital; BRIDGE (M.L.E., A.W., M.T., Z.I.), Department of Clinical Research; Department of Molecular Medicine (M.L.E., A.W., L.V., Z.I.), University of Southern Denmark, Odense, Denmark; Biomedical Network Science Lab (A.H.), Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Department of Mathematics and Computer Science (A.H., Richard Rottger, J.B.), University of Southern Denmark, Odense, Denmark; Institute for Computational Systems Biology (M.O., J.B.), University of Hamburg, Germany; Department of Brain Sciences (Richard Reynolds), Imperial College, London, United Kingdom; and Clinical Genome Center (M.T.), Research Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Mhaned Oubounyt
- From the Department of Neurology (M.L.E., A.W., Z.I.), Odense University Hospital; BRIDGE (M.L.E., A.W., M.T., Z.I.), Department of Clinical Research; Department of Molecular Medicine (M.L.E., A.W., L.V., Z.I.), University of Southern Denmark, Odense, Denmark; Biomedical Network Science Lab (A.H.), Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Department of Mathematics and Computer Science (A.H., Richard Rottger, J.B.), University of Southern Denmark, Odense, Denmark; Institute for Computational Systems Biology (M.O., J.B.), University of Hamburg, Germany; Department of Brain Sciences (Richard Reynolds), Imperial College, London, United Kingdom; and Clinical Genome Center (M.T.), Research Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Anna Weber
- From the Department of Neurology (M.L.E., A.W., Z.I.), Odense University Hospital; BRIDGE (M.L.E., A.W., M.T., Z.I.), Department of Clinical Research; Department of Molecular Medicine (M.L.E., A.W., L.V., Z.I.), University of Southern Denmark, Odense, Denmark; Biomedical Network Science Lab (A.H.), Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Department of Mathematics and Computer Science (A.H., Richard Rottger, J.B.), University of Southern Denmark, Odense, Denmark; Institute for Computational Systems Biology (M.O., J.B.), University of Hamburg, Germany; Department of Brain Sciences (Richard Reynolds), Imperial College, London, United Kingdom; and Clinical Genome Center (M.T.), Research Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Lars Vitved
- From the Department of Neurology (M.L.E., A.W., Z.I.), Odense University Hospital; BRIDGE (M.L.E., A.W., M.T., Z.I.), Department of Clinical Research; Department of Molecular Medicine (M.L.E., A.W., L.V., Z.I.), University of Southern Denmark, Odense, Denmark; Biomedical Network Science Lab (A.H.), Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Department of Mathematics and Computer Science (A.H., Richard Rottger, J.B.), University of Southern Denmark, Odense, Denmark; Institute for Computational Systems Biology (M.O., J.B.), University of Hamburg, Germany; Department of Brain Sciences (Richard Reynolds), Imperial College, London, United Kingdom; and Clinical Genome Center (M.T.), Research Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Richard Reynolds
- From the Department of Neurology (M.L.E., A.W., Z.I.), Odense University Hospital; BRIDGE (M.L.E., A.W., M.T., Z.I.), Department of Clinical Research; Department of Molecular Medicine (M.L.E., A.W., L.V., Z.I.), University of Southern Denmark, Odense, Denmark; Biomedical Network Science Lab (A.H.), Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Department of Mathematics and Computer Science (A.H., Richard Rottger, J.B.), University of Southern Denmark, Odense, Denmark; Institute for Computational Systems Biology (M.O., J.B.), University of Hamburg, Germany; Department of Brain Sciences (Richard Reynolds), Imperial College, London, United Kingdom; and Clinical Genome Center (M.T.), Research Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Mads Thomassen
- From the Department of Neurology (M.L.E., A.W., Z.I.), Odense University Hospital; BRIDGE (M.L.E., A.W., M.T., Z.I.), Department of Clinical Research; Department of Molecular Medicine (M.L.E., A.W., L.V., Z.I.), University of Southern Denmark, Odense, Denmark; Biomedical Network Science Lab (A.H.), Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Department of Mathematics and Computer Science (A.H., Richard Rottger, J.B.), University of Southern Denmark, Odense, Denmark; Institute for Computational Systems Biology (M.O., J.B.), University of Hamburg, Germany; Department of Brain Sciences (Richard Reynolds), Imperial College, London, United Kingdom; and Clinical Genome Center (M.T.), Research Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Richard Rottger
- From the Department of Neurology (M.L.E., A.W., Z.I.), Odense University Hospital; BRIDGE (M.L.E., A.W., M.T., Z.I.), Department of Clinical Research; Department of Molecular Medicine (M.L.E., A.W., L.V., Z.I.), University of Southern Denmark, Odense, Denmark; Biomedical Network Science Lab (A.H.), Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Department of Mathematics and Computer Science (A.H., Richard Rottger, J.B.), University of Southern Denmark, Odense, Denmark; Institute for Computational Systems Biology (M.O., J.B.), University of Hamburg, Germany; Department of Brain Sciences (Richard Reynolds), Imperial College, London, United Kingdom; and Clinical Genome Center (M.T.), Research Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Jan Baumbach
- From the Department of Neurology (M.L.E., A.W., Z.I.), Odense University Hospital; BRIDGE (M.L.E., A.W., M.T., Z.I.), Department of Clinical Research; Department of Molecular Medicine (M.L.E., A.W., L.V., Z.I.), University of Southern Denmark, Odense, Denmark; Biomedical Network Science Lab (A.H.), Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Department of Mathematics and Computer Science (A.H., Richard Rottger, J.B.), University of Southern Denmark, Odense, Denmark; Institute for Computational Systems Biology (M.O., J.B.), University of Hamburg, Germany; Department of Brain Sciences (Richard Reynolds), Imperial College, London, United Kingdom; and Clinical Genome Center (M.T.), Research Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Zsolt Illes
- From the Department of Neurology (M.L.E., A.W., Z.I.), Odense University Hospital; BRIDGE (M.L.E., A.W., M.T., Z.I.), Department of Clinical Research; Department of Molecular Medicine (M.L.E., A.W., L.V., Z.I.), University of Southern Denmark, Odense, Denmark; Biomedical Network Science Lab (A.H.), Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Germany; Department of Mathematics and Computer Science (A.H., Richard Rottger, J.B.), University of Southern Denmark, Odense, Denmark; Institute for Computational Systems Biology (M.O., J.B.), University of Hamburg, Germany; Department of Brain Sciences (Richard Reynolds), Imperial College, London, United Kingdom; and Clinical Genome Center (M.T.), Research Unit of Human Genetics, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
20
|
Kukanja P, Langseth CM, Rubio Rodríguez-Kirby LA, Agirre E, Zheng C, Raman A, Yokota C, Avenel C, Tiklová K, Guerreiro-Cacais AO, Olsson T, Hilscher MM, Nilsson M, Castelo-Branco G. Cellular architecture of evolving neuroinflammatory lesions and multiple sclerosis pathology. Cell 2024; 187:1990-2009.e19. [PMID: 38513664 DOI: 10.1016/j.cell.2024.02.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 12/13/2023] [Accepted: 02/23/2024] [Indexed: 03/23/2024]
Abstract
Multiple sclerosis (MS) is a neurological disease characterized by multifocal lesions and smoldering pathology. Although single-cell analyses provided insights into cytopathology, evolving cellular processes underlying MS remain poorly understood. We investigated the cellular dynamics of MS by modeling temporal and regional rates of disease progression in mouse experimental autoimmune encephalomyelitis (EAE). By performing single-cell spatial expression profiling using in situ sequencing (ISS), we annotated disease neighborhoods and found centrifugal evolution of active lesions. We demonstrated that disease-associated (DA)-glia arise independently of lesions and are dynamically induced and resolved over the disease course. Single-cell spatial mapping of human archival MS spinal cords confirmed the differential distribution of homeostatic and DA-glia, enabled deconvolution of active and inactive lesions into sub-compartments, and identified new lesion areas. By establishing a spatial resource of mouse and human MS neuropathology at a single-cell resolution, our study unveils the intricate cellular dynamics underlying MS.
Collapse
Affiliation(s)
- Petra Kukanja
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Biomedicum, Karolinska Institutet, 17177 Stockholm, Sweden.
| | - Christoffer M Langseth
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, 17154 Stockholm, Sweden.
| | - Leslie A Rubio Rodríguez-Kirby
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Biomedicum, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Eneritz Agirre
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Biomedicum, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Chao Zheng
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Biomedicum, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Amitha Raman
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, 17154 Stockholm, Sweden
| | - Chika Yokota
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, 17154 Stockholm, Sweden
| | - Christophe Avenel
- Department of Information Technology, Uppsala University, 752 37 Uppsala, Sweden; BioImage Informatics Facility, Science for Life Laboratory, SciLifeLab, 751 05 Uppsala, Sweden
| | - Katarina Tiklová
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, 17154 Stockholm, Sweden
| | - André O Guerreiro-Cacais
- Center for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Karolinska University Hospital, 171 76 Solna, Sweden
| | - Tomas Olsson
- Center for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Karolinska University Hospital, 171 76 Solna, Sweden
| | - Markus M Hilscher
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, 17154 Stockholm, Sweden
| | - Mats Nilsson
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, 17154 Stockholm, Sweden.
| | - Gonçalo Castelo-Branco
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Biomedicum, Karolinska Institutet, 17177 Stockholm, Sweden.
| |
Collapse
|
21
|
Li W, Tiedt S, Lawrence JH, Harrington ME, Musiek ES, Lo EH. Circadian Biology and the Neurovascular Unit. Circ Res 2024; 134:748-769. [PMID: 38484026 DOI: 10.1161/circresaha.124.323514] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 02/14/2024] [Indexed: 03/19/2024]
Abstract
Mammalian physiology and cellular function are subject to significant oscillations over the course of every 24-hour day. It is likely that these daily rhythms will affect function as well as mechanisms of disease in the central nervous system. In this review, we attempt to survey and synthesize emerging studies that investigate how circadian biology may influence the neurovascular unit. We examine how circadian clocks may operate in neural, glial, and vascular compartments, review how circadian mechanisms regulate cell-cell signaling, assess interactions with aging and vascular comorbidities, and finally ask whether and how circadian effects and disruptions in rhythms may influence the risk and progression of pathophysiology in cerebrovascular disease. Overcoming identified challenges and leveraging opportunities for future research might support the development of novel circadian-based treatments for stroke.
Collapse
Affiliation(s)
- Wenlu Li
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston (W.L., E.H.L.)
- Consortium International pour la Recherche Circadienne sur l'AVC, Munich, Germany (W.L., S.T., J.H.L., M.E.H., E.S.M., E.H.L.)
| | - Steffen Tiedt
- Consortium International pour la Recherche Circadienne sur l'AVC, Munich, Germany (W.L., S.T., J.H.L., M.E.H., E.S.M., E.H.L.)
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany (S.T.)
| | - Jennifer H Lawrence
- Consortium International pour la Recherche Circadienne sur l'AVC, Munich, Germany (W.L., S.T., J.H.L., M.E.H., E.S.M., E.H.L.)
- Department of Neurology, Washington University School of Medicine, St. Louis, MO (J.H.L., E.S.M.)
| | - Mary E Harrington
- Consortium International pour la Recherche Circadienne sur l'AVC, Munich, Germany (W.L., S.T., J.H.L., M.E.H., E.S.M., E.H.L.)
- Neuroscience Program, Smith College, Northampton, MA (M.E.H.)
| | - Erik S Musiek
- Consortium International pour la Recherche Circadienne sur l'AVC, Munich, Germany (W.L., S.T., J.H.L., M.E.H., E.S.M., E.H.L.)
- Department of Neurology, Washington University School of Medicine, St. Louis, MO (J.H.L., E.S.M.)
| | - Eng H Lo
- Neuroprotection Research Laboratories, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston (W.L., E.H.L.)
- Consortium International pour la Recherche Circadienne sur l'AVC, Munich, Germany (W.L., S.T., J.H.L., M.E.H., E.S.M., E.H.L.)
| |
Collapse
|
22
|
Hill RA, Nishiyama A, Hughes EG. Features, Fates, and Functions of Oligodendrocyte Precursor Cells. Cold Spring Harb Perspect Biol 2024; 16:a041425. [PMID: 38052500 PMCID: PMC10910408 DOI: 10.1101/cshperspect.a041425] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Oligodendrocyte precursor cells (OPCs) are a central nervous system resident population of glia with a distinct molecular identity and an ever-increasing list of functions. OPCs generate oligodendrocytes throughout development and across the life span in most regions of the brain and spinal cord. This process involves a complex coordination of molecular checkpoints and biophysical cues from the environment that initiate the differentiation and integration of new oligodendrocytes that synthesize myelin sheaths on axons. Outside of their progenitor role, OPCs have been proposed to play other functions including the modulation of axonal and synaptic development and the participation in bidirectional signaling with neurons and other glia. Here, we review OPC identity and known functions and discuss recent findings implying other roles for these glial cells in brain physiology and pathology.
Collapse
Affiliation(s)
- Robert A Hill
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire 03755, USA
| | - Akiko Nishiyama
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Connecticut 06269, USA
| | - Ethan G Hughes
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| |
Collapse
|
23
|
Shinozaki Y, Namekata K, Guo X, Harada T. Glial cells as a promising therapeutic target of glaucoma: beyond the IOP. FRONTIERS IN OPHTHALMOLOGY 2024; 3:1310226. [PMID: 38983026 PMCID: PMC11182302 DOI: 10.3389/fopht.2023.1310226] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/18/2023] [Indexed: 07/11/2024]
Abstract
Glial cells, a type of non-neuronal cell found in the central nervous system (CNS), play a critical role in maintaining homeostasis and regulating CNS functions. Recent advancements in technology have paved the way for new therapeutic strategies in the fight against glaucoma. While intraocular pressure (IOP) is the most well-known modifiable risk factor, a significant number of glaucoma patients have normal IOP levels. Because glaucoma is a complex, multifactorial disease influenced by various factors that contribute to its onset and progression, it is imperative that we consider factors beyond IOP to effectively prevent or slow down the disease's advancement. In the realm of CNS neurodegenerative diseases, glial cells have emerged as key players due to their pivotal roles in initiating and hastening disease progression. The inhibition of dysregulated glial function holds the potential to protect neurons and restore brain function. Consequently, glial cells represent an enticing therapeutic candidate for glaucoma, even though the majority of glaucoma research has historically concentrated solely on retinal ganglion cells (RGCs). In addition to the neuroprotection of RGCs, the proper regulation of glial cell function can also facilitate structural and functional recovery in the retina. In this review, we offer an overview of recent advancements in understanding the non-cell-autonomous mechanisms underlying the pathogenesis of glaucoma. Furthermore, state-of-the-art technologies have opened up possibilities for regenerating the optic nerve, which was previously believed to be incapable of regeneration. We will also delve into the potential roles of glial cells in the regeneration of the optic nerve and the restoration of visual function.
Collapse
Affiliation(s)
- Youichi Shinozaki
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kazuhiko Namekata
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Xiaoli Guo
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takayuki Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
24
|
Zou P, Wu C, Liu TCY, Duan R, Yang L. Oligodendrocyte progenitor cells in Alzheimer's disease: from physiology to pathology. Transl Neurodegener 2023; 12:52. [PMID: 37964328 PMCID: PMC10644503 DOI: 10.1186/s40035-023-00385-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/01/2023] [Indexed: 11/16/2023] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) play pivotal roles in myelin formation and phagocytosis, communicating with neighboring cells and contributing to the integrity of the blood-brain barrier (BBB). However, under the pathological circumstances of Alzheimer's disease (AD), the brain's microenvironment undergoes detrimental changes that significantly impact OPCs and their functions. Starting with OPC functions, we delve into the transformation of OPCs to myelin-producing oligodendrocytes, the intricate signaling interactions with other cells in the central nervous system (CNS), and the fascinating process of phagocytosis, which influences the function of OPCs and affects CNS homeostasis. Moreover, we discuss the essential role of OPCs in BBB formation and highlight the critical contribution of OPCs in forming CNS-protective barriers. In the context of AD, the deterioration of the local microenvironment in the brain is discussed, mainly focusing on neuroinflammation, oxidative stress, and the accumulation of toxic proteins. The detrimental changes disturb the delicate balance in the brain, impacting the regenerative capacity of OPCs and compromising myelin integrity. Under pathological conditions, OPCs experience significant alterations in migration and proliferation, leading to impaired differentiation and a reduced ability to produce mature oligodendrocytes. Moreover, myelin degeneration and formation become increasingly active in AD, contributing to progressive neurodegeneration. Finally, we summarize the current therapeutic approaches targeting OPCs in AD. Strategies to revitalize OPC senescence, modulate signaling pathways to enhance OPC differentiation, and explore other potential therapeutic avenues are promising in alleviating the impact of AD on OPCs and CNS function. In conclusion, this review highlights the indispensable role of OPCs in CNS function and their involvement in the pathogenesis of AD. The intricate interplay between OPCs and the AD brain microenvironment underscores the complexity of neurodegenerative diseases. Insights from studying OPCs under pathological conditions provide a foundation for innovative therapeutic strategies targeting OPCs and fostering neurodegeneration. Future research will advance our understanding and management of neurodegenerative diseases, ultimately offering hope for effective treatments and improved quality of life for those affected by AD and related disorders.
Collapse
Affiliation(s)
- Peibin Zou
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Chongyun Wu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Timon Cheng-Yi Liu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Rui Duan
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| |
Collapse
|
25
|
Mokhtarzadeh Khanghahi A, Rayatpour A, Baharvand H, Javan M. Neuroglial components of brain lesions may provide new therapeutic strategies for multiple sclerosis. Neurol Sci 2023; 44:3795-3807. [PMID: 37410268 DOI: 10.1007/s10072-023-06915-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 06/14/2023] [Indexed: 07/07/2023]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune and demyelinating disease of the central nervous system (CNS) which leads to focal demyelinated lesions in the brain and spinal cord. Failure of remyelination contributes to chronic disability in young adults. Characterization of events occurring during the demyelination and remyelination processes and those of which subsequently limit remyelination or contribute to demyelination can provide the possibility of new therapies development for MS. Most of the currently available therapies and investigations modulate immune responses and mediators. Since most therapeutic strategies have unsatisfied outcomes, developing new therapies that enhance brain lesion repair is a priority. A close look at cellular and chemical components of MS lesions will pave the way to a better understanding of lesions pathology and will provide possible opportunities for repair strategies and targeted pharmacotherapy. This review summarizes the lesion components and features, particularly the detrimental elements, and discusses the possibility of suggesting new potential targets as therapies for demyelinating diseases like MS.
Collapse
Affiliation(s)
- Akram Mokhtarzadeh Khanghahi
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Atefeh Rayatpour
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Institute for Brain and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Hossein Baharvand
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Javan
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
- Institute for Brain and Cognition, Tarbiat Modares University, Tehran, Iran.
- International Collaboration on Repair Discoveries (ICORD), the University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
26
|
Shumilov K, Xiao S, Ni A, Celorrio M, Friess SH. Recombinant Erythropoietin Induces Oligodendrocyte Progenitor Cell Proliferation After Traumatic Brain Injury and Delayed Hypoxemia. Neurotherapeutics 2023; 20:1859-1874. [PMID: 37768487 PMCID: PMC10684442 DOI: 10.1007/s13311-023-01443-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Traumatic brain injury (TBI) can result in axonal loss and demyelination, leading to persistent damage in the white matter. Demyelinated axons are vulnerable to pathologies related to an abnormal myelin structure that expose neurons to further damage. Oligodendrocyte progenitor cells (OPCs) mediate remyelination after recruitment to the injury site. Often this process is inefficient due to inadequate OPC proliferation. To date, no effective treatments are currently available to stimulate OPC proliferation in TBI. Recombinant human erythropoietin (rhEPO) is a pleiotropic neuroprotective cytokine, and its receptor is present in all stages of oligodendroglial lineage cell differentiation. Therefore, we hypothesized that rhEPO administration would enhance remyelination after TBI through the modulation of OPC response. Utilizing a murine model of controlled cortical impact and a primary OPC culture in vitro model, we characterized the impact of rhEPO on remyelination and proliferation of oligodendrocyte lineage cells. Myelin black gold II staining of the peri-contusional corpus callosum revealed an increase in myelinated area in association with an increase in BrdU-positive oligodendrocytes in injured mice treated with rhEPO. Furthermore, morphological analysis of OPCs showed a decrease in process length in rhEPO-treated animals. RhEPO treatment increased OPC proliferation after in vitro CSPG exposure. Erythropoietin receptor (EPOr) gene knockdown using siRNA prevented rhEPO-induced OPC proliferation, demonstrating that the rhEPO effect on OPC response is EPOr activation dependent. Together, our findings demonstrate that rhEPO administration may promote myelination by increasing oligodendrocyte lineage cell proliferation after TBI.
Collapse
Affiliation(s)
- Kirill Shumilov
- Department of Pediatrics, Washington University in St. Louis School of Medicine, Campus Box 8208, One Children's Place, St. Louis, MO, 63110, USA
| | - Sophia Xiao
- Department of Pediatrics, Washington University in St. Louis School of Medicine, Campus Box 8208, One Children's Place, St. Louis, MO, 63110, USA
| | - Allen Ni
- Department of Pediatrics, Washington University in St. Louis School of Medicine, Campus Box 8208, One Children's Place, St. Louis, MO, 63110, USA
| | - Marta Celorrio
- Department of Pediatrics, Washington University in St. Louis School of Medicine, Campus Box 8208, One Children's Place, St. Louis, MO, 63110, USA
| | - Stuart H Friess
- Department of Pediatrics, Washington University in St. Louis School of Medicine, Campus Box 8208, One Children's Place, St. Louis, MO, 63110, USA.
| |
Collapse
|
27
|
Fang LP, Bai X. Oligodendrocyte precursor cells: the multitaskers in the brain. Pflugers Arch 2023; 475:1035-1044. [PMID: 37401986 PMCID: PMC10409806 DOI: 10.1007/s00424-023-02837-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/19/2023] [Accepted: 06/27/2023] [Indexed: 07/05/2023]
Abstract
In the central nervous system, oligodendrocyte precursor cells (OPCs) are recognized as the progenitors responsible for the generation of oligodendrocytes, which play a critical role in myelination. Extensive research has shed light on the mechanisms underlying OPC proliferation and differentiation into mature myelin-forming oligodendrocytes. However, recent advances in the field have revealed that OPCs have multiple functions beyond their role as progenitors, exerting control over neural circuits and brain function through distinct pathways. This review aims to provide a comprehensive understanding of OPCs by first introducing their well-established features. Subsequently, we delve into the emerging roles of OPCs in modulating brain function in both healthy and diseased states. Unraveling the cellular and molecular mechanisms by which OPCs influence brain function holds great promise for identifying novel therapeutic targets for central nervous system diseases.
Collapse
Affiliation(s)
- Li-Pao Fang
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, 66421 Homburg, Germany
| | - Xianshu Bai
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, 66421 Homburg, Germany
| |
Collapse
|
28
|
Buchanan J, da Costa NM, Cheadle L. Emerging roles of oligodendrocyte precursor cells in neural circuit development and remodeling. Trends Neurosci 2023; 46:628-639. [PMID: 37286422 PMCID: PMC10524797 DOI: 10.1016/j.tins.2023.05.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/26/2023] [Accepted: 05/17/2023] [Indexed: 06/09/2023]
Abstract
Oligodendrocyte precursor cells (OPCs) are non-neuronal brain cells that give rise to oligodendrocytes, glia that myelinate the axons of neurons in the brain. Classically known for their contributions to myelination via oligodendrogenesis, OPCs are increasingly appreciated to play diverse roles in the nervous system, ranging from blood vessel formation to antigen presentation. Here, we review emerging literature suggesting that OPCs may be essential for the establishment and remodeling of neural circuits in the developing and adult brain via mechanisms that are distinct from the production of oligodendrocytes. We discuss the specialized features of OPCs that position these cells to integrate activity-dependent and molecular cues to shape brain wiring. Finally, we place OPCs within the context of a growing field focused on understanding the importance of communication between neurons and glia in the contexts of both health and disease.
Collapse
Affiliation(s)
- JoAnn Buchanan
- Allen Institute for Brain Science, Seattle, WA 98109, USA
| | | | - Lucas Cheadle
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
29
|
He Y, Li Z, Shi X, Ding J, Wang X. Roles of NG2 Glia in Cerebral Small Vessel Disease. Neurosci Bull 2023; 39:519-530. [PMID: 36401147 PMCID: PMC10043141 DOI: 10.1007/s12264-022-00976-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 09/02/2022] [Indexed: 11/19/2022] Open
Abstract
Cerebral small vessel disease (CSVD) is one of the most prevalent pathologic processes affecting 5% of people over 50 years of age and contributing to 45% of dementia cases. Increasing evidence has demonstrated the pathological roles of chronic hypoperfusion, impaired cerebral vascular reactivity, and leakage of the blood-brain barrier in CSVD. However, the pathogenesis of CSVD remains elusive thus far, and no radical treatment has been developed. NG2 glia, also known as oligodendrocyte precursor cells, are the fourth type of glial cell in addition to astrocytes, microglia, and oligodendrocytes in the mammalian central nervous system. Many novel functions for NG2 glia in physiological and pathological states have recently been revealed. In this review, we discuss the role of NG2 glia in CSVD and the underlying mechanisms.
Collapse
Affiliation(s)
- Yixi He
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Zhenghao Li
- Institute of Neuroscience, MOE Key Laboratory of Molecular Neurobiology, NMU, Shanghai, 200433, China
| | - Xiaoyu Shi
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Jing Ding
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| | - Xin Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
30
|
Drake SS, Zaman A, Simas T, Fournier AE. Comparing RNA-sequencing datasets from astrocytes, oligodendrocytes, and microglia in multiple sclerosis identifies novel dysregulated genes relevant to inflammation and myelination. WIREs Mech Dis 2023; 15:e1594. [PMID: 36600404 DOI: 10.1002/wsbm.1594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/25/2022] [Accepted: 12/14/2022] [Indexed: 01/06/2023]
Abstract
Central nervous system (CNS) inflammation is a key factor in multiple sclerosis (MS). Invasion of peripheral immune cells into the CNS resulting from an unknown signal or combination of signals results in activation of resident immune cells and the hallmark feature of the disease: demyelinating lesions. These lesion sites are an amalgam of reactive peripheral and central immune cells, astrocytes, damaged and dying oligodendrocytes, and injured neurons and axons. Sustained inflammation affects cells directly located within the lesion site and further abnormalities are apparent diffusely throughout normal-appearing white matter and grey matter. It is only relatively recently, using animal models, new tissue sampling techniques, and next-generation sequencing, that molecular changes occurring in CNS resident cells have been broadly captured. Advances in cell isolation through Fluorescence Activated Cell Sorting (FACS) and laser-capture microdissection together with the emergence of single-cell sequencing have enabled researchers to investigate changes in gene expression in astrocytes, microglia, and oligodendrocytes derived from animal models of MS as well as from primary patient tissue. The contribution of some dysregulated pathways has been followed up in individual studies; however, corroborating results often go unreported between sequencing studies. To this end, we have consolidated results from numerous RNA-sequencing studies to identify and review novel patterns of differentially regulated genes and pathways occurring within CNS glial cells in MS. This article is categorized under: Neurological Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Sienna S Drake
- McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Aliyah Zaman
- McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Tristan Simas
- McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Alyson E Fournier
- McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada
| |
Collapse
|
31
|
Cashion JM, Young KM, Sutherland BA. How does neurovascular unit dysfunction contribute to multiple sclerosis? Neurobiol Dis 2023; 178:106028. [PMID: 36736923 DOI: 10.1016/j.nbd.2023.106028] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 01/17/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
Multiple sclerosis is an inflammatory demyelinating disease of the central nervous system (CNS) and the most common non-traumatic cause of neurological disability in young adults. Multiple sclerosis clinical care has improved considerably due to the development of disease-modifying therapies that effectively modulate the peripheral immune response and reduce relapse frequency. However, current treatments do not prevent neurodegeneration and disease progression, and efforts to prevent multiple sclerosis will be hampered so long as the cause of this disease remains unknown. Risk factors for multiple sclerosis development or severity include vitamin D deficiency, cigarette smoking and youth obesity, which also impact vascular health. People with multiple sclerosis frequently experience blood-brain barrier breakdown, microbleeds, reduced cerebral blood flow and diminished neurovascular reactivity, and it is possible that these vascular pathologies are tied to multiple sclerosis development. The neurovascular unit is a cellular network that controls neuroinflammation, maintains blood-brain barrier integrity, and tightly regulates cerebral blood flow, matching energy supply to neuronal demand. The neurovascular unit is composed of vessel-associated cells such as endothelial cells, pericytes and astrocytes, however neuronal and other glial cell types also comprise the neurovascular niche. Recent single-cell transcriptomics data, indicate that neurovascular cells, particular cells of the microvasculature, are compromised within multiple sclerosis lesions. Large-scale genetic and small-scale cell biology studies also suggest that neurovascular dysfunction could be a primary pathology contributing to multiple sclerosis development. Herein we revisit multiple sclerosis risk factors and multiple sclerosis pathophysiology and highlight the known and potential roles of neurovascular unit dysfunction in multiple sclerosis development and disease progression. We also evaluate the suitability of the neurovascular unit as a potential target for future disease modifying therapies for multiple sclerosis.
Collapse
Affiliation(s)
- Jake M Cashion
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Brad A Sutherland
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia.
| |
Collapse
|
32
|
de Almeida MMA, Watson AES, Bibi S, Dittmann NL, Goodkey K, Sharafodinzadeh P, Galleguillos D, Nakhaei-Nejad M, Kosaraju J, Steinberg N, Wang BS, Footz T, Giuliani F, Wang J, Sipione S, Edgar JM, Voronova A. Fractalkine enhances oligodendrocyte regeneration and remyelination in a demyelination mouse model. Stem Cell Reports 2023; 18:519-533. [PMID: 36608690 PMCID: PMC9968989 DOI: 10.1016/j.stemcr.2022.12.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 01/07/2023] Open
Abstract
Demyelinating disorders of the central nervous system (CNS) occur when myelin and oligodendrocytes are damaged or lost. Remyelination and regeneration of oligodendrocytes can be achieved from endogenous oligodendrocyte precursor cells (OPCs) that reside in the adult CNS tissue. Using a cuprizone mouse model of demyelination, we show that infusion of fractalkine (CX3CL1) into the demyelinated murine brain increases de novo oligodendrocyte formation and enhances remyelination in the corpus callosum and cortical gray matter. This is achieved by increased OPC proliferation in the cortical gray matter as well as OPC differentiation and attenuation of microglia/macrophage activation both in corpus callosum and cortical gray matter. Finally, we show that activated OPCs and microglia/macrophages express fractalkine receptor CX3CR1 in vivo, and that in OPC-microglia co-cultures fractalkine increases in vitro oligodendrocyte differentiation by modulating both OPC and microglia biology. Our results demonstrate a novel pro-regenerative role of fractalkine in a demyelinating mouse model.
Collapse
Affiliation(s)
- Monique M A de Almeida
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2E1, Canada
| | - Adrianne E S Watson
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; Women and Children's Health Research Institute, University of Alberta, 5-083 Edmonton Clinic Health Academy, 11405 87 Avenue NW, Edmonton, AB T6G 1C9, Canada
| | - Sana Bibi
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Nicole L Dittmann
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2E1, Canada
| | - Kara Goodkey
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; Women and Children's Health Research Institute, University of Alberta, 5-083 Edmonton Clinic Health Academy, 11405 87 Avenue NW, Edmonton, AB T6G 1C9, Canada
| | - Pedram Sharafodinzadeh
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Danny Galleguillos
- Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2E1, Canada; Department of Pharmacology, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2H7, Canada
| | - Maryam Nakhaei-Nejad
- Department of Medicine, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2H7, Canada
| | - Jayasankar Kosaraju
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Noam Steinberg
- Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2E1, Canada; Department of Pharmacology, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2H7, Canada
| | - Beatrix S Wang
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; Women and Children's Health Research Institute, University of Alberta, 5-083 Edmonton Clinic Health Academy, 11405 87 Avenue NW, Edmonton, AB T6G 1C9, Canada
| | - Tim Footz
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Fabrizio Giuliani
- Department of Medicine, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2H7, Canada; Multiple Sclerosis Centre and Department of Cell Biology, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2H7, Canada
| | - Jing Wang
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa Brain and Mind Research Institute, Ottawa, ON K1H 8M5, Canada
| | - Simonetta Sipione
- Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2E1, Canada; Department of Pharmacology, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2H7, Canada
| | - Julia M Edgar
- School of Infection and Immunity, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Anastassia Voronova
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; Women and Children's Health Research Institute, University of Alberta, 5-083 Edmonton Clinic Health Academy, 11405 87 Avenue NW, Edmonton, AB T6G 1C9, Canada; Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2E1, Canada; Department of Cell Biology, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2H7, Canada; Multiple Sclerosis Centre and Department of Cell Biology, Faculty of Medicine & Dentistry, Edmonton, AB T6G 2H7, Canada.
| |
Collapse
|
33
|
Hirose S, Wang S, Jaggi U, Matundan HH, Kato M, Song XY, Molesworth-Kenyon SJ, Lausch RN, Ghiasi H. IL-17A expression by both T cells and non-T cells contribute to HSV-IL-2-induced CNS demyelination. Front Immunol 2023; 14:1102486. [PMID: 36817487 PMCID: PMC9931899 DOI: 10.3389/fimmu.2023.1102486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/20/2023] [Indexed: 02/05/2023] Open
Abstract
Previously we reported that a recombinant HSV-1 expressing murine IL-2 (HSV-IL-2) causes CNS demyelination in different strains of mice and in a T cell-dependent manner. Since TH17 cells have been implicated in CNS pathology, in the present study, we looked into the effects of IL-17A-/- and three of its receptors on HSV-IL-2-induced CNS demyelination. IL-17A-/- mice did not develop CNS demyelination, while IL-17RA-/-, IL-17RC-/-, IL-17RD-/- and IL-17RA-/-RC-/- mice developed CNS demyelination. Adoptive transfer of T cells from wild-type (WT) mice to IL-17A-/- mice or T cells from IL-17A-/- mice to Rag-/- mice induced CNS demyelination in infected mice. Adoptive T cell experiments suggest that both T cells and non-T cells expressing IL-17A contribute to HSV-IL-2-induced CNS demyelination with no difference in the severity of demyelination between the two groups of IL-17A producing cells. IL-6, IL-10, or TGFβ did not contribute to CNS demyelination in infected mice. Transcriptome analysis between IL-17A-/- brain and spinal cord of infected mice with and without T cell transfer from WT mice revealed that "neuron projection extension involved in neuron projection guidance" and "ensheathment of neurons" pathways were associated with CNS demyelination. Collectively, the results indicate the importance of IL-17A in CNS demyelination and the possible involvement of more than three of IL-17 receptors in CNS demyelination.
Collapse
Affiliation(s)
- Satoshi Hirose
- Center for Neurobiology & Vaccine Development, Ophthalmology Research, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Shaohui Wang
- Center for Neurobiology & Vaccine Development, Ophthalmology Research, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Ujjaldeep Jaggi
- Center for Neurobiology & Vaccine Development, Ophthalmology Research, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Harry H. Matundan
- Center for Neurobiology & Vaccine Development, Ophthalmology Research, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Mihoko Kato
- Department of Biology, Pomona College, Claremont, CA, United States
| | - Xue-Ying Song
- Applied Genomics, Computation, and Translational Core, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | | | - Robert N. Lausch
- Department of Microbiology and Immunology, University of South Alabama, College of Medicine, Mobile, Al, United States
| | - Homayon Ghiasi
- Center for Neurobiology & Vaccine Development, Ophthalmology Research, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
34
|
Wang H, Liang L, Yang C, Xiao L, Wang H, Wang G, Zhu Z. The protective role of hippocampal LRP1 knockdown involves synaptic plasticity through the promoting microtubule dynamics and activation of Akt/GSK-3β pathway in depressive rats. J Affect Disord 2023; 322:63-75. [PMID: 36372121 DOI: 10.1016/j.jad.2022.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 09/14/2022] [Accepted: 11/06/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND The mechanism by which synaptic plasticity mediates the occurrence of depression is unknown. Low-density lipoprotein receptor-related protein 1 (LRP1) affects axon growth and neurogenesis in the brain, but its role in depressive-like behaviors is poorly understood. METHODS Adeno-associated virus-mediated small interfering RNA was injected into the bilateral hippocampus 14 days before chronic unpredicted mild stress (CUMS). Behavior performance was assessed for depressive-like behaviors. Western blot was conducted to detect levels of LRP1, neurogenesis-related proteins, synaptic markers, microtubule system molecules and Akt/GSK-3β signaling-related proteins. Immunohistochemical staining was performed for LRP1 protein, immunofluorescence staining was conducted to determine the Sox2 protein, Nissl's staining and transmission electron microscope staining were used to observe hippocampal morphological features. RESULTS The expression of hippocampal LRP1 was positively correlated with depressive-like behaviors. Treatment with iAAV-LRP1 exerted protective effects on depressive-like behaviors. LRP1 Knockdown relieved the inhibition of synaptic plasticity induced by CUMS. Expression of Sox2, GluR2 and SYP was significantly increased in iAAV-LRP1 CUMS rats. LRP1 knockdown reduced the p-tau (Ser262 and Thr404) and Acet-tubule levels in depressed rats. Finally, we found that LRP1 knockdown activated the PI3K/Akt pathway and inhibited GSK-3β signal transduction. LIMITATIONS More neurogenesis markers would be considered, and stereotactic injection into hippocampal DG region could be performed to investigate the effects of LRP1. CONCLUSIONS These findings indicated that hippocampal LRP1 deficiency in stressed rats plays an important protective role in depressive-like behavior by increasing synaptic plasticity mediated by microtubule dynamic and activating Akt/GSK-3β signaling pathway. Therefore, LRP1 may represent a potential therapeutic target for depression.
Collapse
Affiliation(s)
- Hui Wang
- Department of Clinical Psychology, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China
| | - Liang Liang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China
| | - Can Yang
- Department of Clinical Psychology, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China
| | - Ling Xiao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China
| | - Huiling Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China.
| | - Zhixian Zhu
- Department of Clinical Psychology, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China.
| |
Collapse
|
35
|
Dittmann NL, Torabi P, Watson AES, Yuzwa SA, Voronova A. Culture Protocol and Transcriptomic Analysis of Murine SVZ NPCs and OPCs. Stem Cell Rev Rep 2023; 19:983-1000. [PMID: 36617597 DOI: 10.1007/s12015-022-10492-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2022] [Indexed: 01/10/2023]
Abstract
The mammalian adult brain contains two neural stem and precursor (NPC) niches: the subventricular zone [SVZ] lining the lateral ventricles and the subgranular zone [SGZ] in the hippocampus. From these, SVZ NPCs represent the largest NPC pool. While SGZ NPCs typically only produce neurons and astrocytes, SVZ NPCs produce neurons, astrocytes and oligodendrocytes throughout life. Of particular importance is the generation and replacement of oligodendrocytes, the only myelinating cells of the central nervous system (CNS). SVZ NPCs contribute to myelination by regenerating the parenchymal oligodendrocyte precursor cell (OPC) pool and by differentiating into oligodendrocytes in the developing and demyelinated brain. The neurosphere assay has been widely adopted by the scientific community to facilitate the study of NPCs in vitro. Here, we present a streamlined protocol for culturing postnatal and adult SVZ NPCs and OPCs from primary neurosphere cells. We characterize the purity and differentiation potential as well as provide RNA-sequencing profiles of postnatal SVZ NPCs, postnatal SVZ OPCs and adult SVZ NPCs. We show that primary neurospheres cells generated from postnatal and adult SVZ differentiate into neurons, astrocytes and oligodendrocytes concurrently and at comparable levels. SVZ OPCs are generated by subjecting primary neurosphere cells to OPC growth factors fibroblast growth factor (FGF) and platelet-derived growth factor-AA (PDGF-AA). We further show SVZ OPCs can differentiate into oligodendrocytes in the absence and presence of thyroid hormone T3. Transcriptomic analysis confirmed the identities of each cell population and revealed novel immune and signalling pathways expressed in an age and cell type specific manner.
Collapse
Affiliation(s)
- Nicole L Dittmann
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada.,Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Pouria Torabi
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Adrianne E S Watson
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Scott A Yuzwa
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Anastassia Voronova
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada. .,Neuroscience and Mental Health Institute, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2E1, Canada. .,Women and Children's Health Research Institute5-083 Edmonton Clinic Health Academy, University of Alberta, 11405 87 Avenue NW, Edmonton, Alberta, T6G 1C9, Canada. .,Department of Cell Biology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada. .,Multiple Sclerosis Centre, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada.
| |
Collapse
|
36
|
Cabeza-Fernández S, White JA, McMurran CE, Gómez-Sánchez JA, de la Fuente AG. Immune-stem cell crosstalk in the central nervous system: how oligodendrocyte progenitor cells interact with immune cells. Immunol Cell Biol 2023; 101:25-35. [PMID: 36427276 DOI: 10.1111/imcb.12610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/23/2022] [Accepted: 11/24/2022] [Indexed: 11/26/2022]
Abstract
The interaction between immune and stem cells has proven essential for homeostasis and regeneration in a wide range of tissues. However, because the central nervous system was long considered an immune-privileged organ, its immune-stem cell axis was not deeply investigated until recently. Research has shown that oligodendrocyte progenitor cells (OPCs), a highly abundant population of adult brain stem cells, establish bidirectional interactions with the immune system. Here, we provide an overview of the interactions that OPCs have with tissue-resident and recruited immune cells, paying particular attention to the role they play in myelin regeneration and neuroinflammation. We highlight the described role of OPCs as key active players in neuroinflammation, overriding the previous concept that OPCs are mere recipients of immune signals. Understanding the mechanisms behind this bidirectional interaction holds great potential for the development of novel therapeutic approaches limiting neuroinflammation and promoting myelin repair. A better understanding of the central nervous system's immune-stem cell axis will also be key for tackling two important features shared across neurodegenerative diseases, neuroinflammation and myelin loss.
Collapse
Affiliation(s)
- Sonia Cabeza-Fernández
- Instituto Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain.,Instituto de Neurosciencias CSIC-UMH, San Juan de Alicante, Spain
| | - Jessica A White
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Christopher E McMurran
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.,Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - José A Gómez-Sánchez
- Instituto Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain.,Instituto de Neurosciencias CSIC-UMH, San Juan de Alicante, Spain
| | - Alerie G de la Fuente
- Instituto Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain.,Instituto de Neurosciencias CSIC-UMH, San Juan de Alicante, Spain.,Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, UK
| |
Collapse
|
37
|
de Almeida MMA, Goodkey K, Voronova A. Regulation of microglia function by neural stem cells. Front Cell Neurosci 2023; 17:1130205. [PMID: 36937181 PMCID: PMC10014810 DOI: 10.3389/fncel.2023.1130205] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/13/2023] [Indexed: 03/05/2023] Open
Abstract
Neural stem and precursor cells (NPCs) build and regenerate the central nervous system (CNS) by maintaining their pool (self-renewal) and differentiating into neurons, astrocytes, and oligodendrocytes (multipotency) throughout life. This has inspired research into pro-regenerative therapies that utilize transplantation of exogenous NPCs or recruitment of endogenous adult NPCs for CNS regeneration and repair. Recent advances in single-cell RNA sequencing and other "omics" have revealed that NPCs express not just traditional progenitor-related genes, but also genes involved in immune function. Here, we review how NPCs exert immunomodulatory function by regulating the biology of microglia, immune cells that are present in NPC niches and throughout the CNS. We discuss the role of transplanted and endogenous NPCs in regulating microglia fates, such as survival, proliferation, migration, phagocytosis and activation, in the developing, injured and degenerating CNS. We also provide a literature review on NPC-specific mediators that are responsible for modulating microglia biology. Our review highlights the immunomodulatory properties of NPCs and the significance of these findings in the context of designing pro-regenerative therapies for degenerating and diseased CNS.
Collapse
Affiliation(s)
- Monique M. A. de Almeida
- Department of Medical Genetics, Faculty of Medicine & Dentistry, Edmonton, AB, Canada
- Faculty of Medicine & Dentistry, Neuroscience and Mental Health Institute, Edmonton, AB, Canada
| | - Kara Goodkey
- Department of Medical Genetics, Faculty of Medicine & Dentistry, Edmonton, AB, Canada
- Women and Children’s Health Research Institute, 5-083 Edmonton Clinic Health Academy, University of Alberta, Edmonton, AB, Canada
| | - Anastassia Voronova
- Department of Medical Genetics, Faculty of Medicine & Dentistry, Edmonton, AB, Canada
- Faculty of Medicine & Dentistry, Neuroscience and Mental Health Institute, Edmonton, AB, Canada
- Women and Children’s Health Research Institute, 5-083 Edmonton Clinic Health Academy, University of Alberta, Edmonton, AB, Canada
- Department of Cell Biology, Faculty of Medicine & Dentistry, Edmonton, AB, Canada
- Multiple Sclerosis Centre and Department of Cell Biology, Faculty of Medicine & Dentistry, Edmonton, AB, Canada
| |
Collapse
|
38
|
Abstract
The molecular mechanisms that maintain circadian rhythms in mammalian as well as non-mammalian systems are well documented in neuronal populations but comparatively understudied in glia. Glia are highly dynamic in form and function, and the circadian clock provides broad dynamic ranges for the maintenance of this homeostasis, thus glia are key to understanding the role of circadian biology in brain function. Here, we highlight the implications of the molecular circadian clock on the homeodynamic nature of glia, underscoring the current gap in understanding the role of the circadian system in oligodendroglia lineage cells and subsequent myelination. Through this perspective, we will focus on the intersection of circadian and glial biology and how it interfaces with global circadian rhythm maintenance associated with normative and aberrant brain function.
Collapse
Affiliation(s)
- Daniela Rojo
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Anna Badner
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Erin M. Gibson
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA 94305, USA,Corresponding Author: Erin M. Gibson, PhD, 3165 Porter Drive, #2178, Palo Alto, CA 94304, (650)725-6659,
| |
Collapse
|
39
|
Abstract
Neurons in the developing brain undergo extensive structural refinement as nascent circuits adopt their mature form. This physical transformation of neurons is facilitated by the engulfment and degradation of axonal branches and synapses by surrounding glial cells, including microglia and astrocytes. However, the small size of phagocytic organelles and the complex, highly ramified morphology of glia have made it difficult to define the contribution of these and other glial cell types to this crucial process. Here, we used large-scale, serial section transmission electron microscopy (TEM) with computational volume segmentation to reconstruct the complete 3D morphologies of distinct glial types in the mouse visual cortex, providing unprecedented resolution of their morphology and composition. Unexpectedly, we discovered that the fine processes of oligodendrocyte precursor cells (OPCs), a population of abundant, highly dynamic glial progenitors, frequently surrounded small branches of axons. Numerous phagosomes and phagolysosomes (PLs) containing fragments of axons and vesicular structures were present inside their processes, suggesting that OPCs engage in axon pruning. Single-nucleus RNA sequencing from the developing mouse cortex revealed that OPCs express key phagocytic genes at this stage, as well as neuronal transcripts, consistent with active axon engulfment. Although microglia are thought to be responsible for the majority of synaptic pruning and structural refinement, PLs were ten times more abundant in OPCs than in microglia at this stage, and these structures were markedly less abundant in newly generated oligodendrocytes, suggesting that OPCs contribute substantially to the refinement of neuronal circuits during cortical development.
Collapse
|
40
|
Plastini MJ, Desu HL, Ascona MC, Lang AL, Saporta MA, Brambilla R. Transcriptional abnormalities in induced pluripotent stem cell-derived oligodendrocytes of individuals with primary progressive multiple sclerosis. Front Cell Neurosci 2022; 16:972144. [PMID: 36246526 PMCID: PMC9554611 DOI: 10.3389/fncel.2022.972144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is the most common neurological disorder in young adults and is classically defined as a chronic inflammatory demyelinating disease of the central nervous system (CNS). Although MS affects millions of people worldwide, its underlying cause remains unknown making discovery of effective treatments challenging. Whether intrinsic or extrinsic factors contribute to MS initiation and progression is still unclear. This is especially true for primary progressive MS (PPMS), the rarest form of the disease, in which progressive and irreversible loss of neurological function is often observed in the absence of an overt immune-inflammatory response. To test the hypothesis that intrinsic dysfunction in oligodendrocytes (OLs), the primary targets of damage in MS, may contribute to PPMS etiopathology, we differentiated human induced pluripotent stem cell (hiPSC) lines derived from PPMS and healthy individuals into mature OLs to compare their transcriptional profile. PPMS derived OLs displayed hundreds of differentially expressed genes compared to control OLs, many associated with cell adhesion, apoptosis and inflammation, including the inflammasome component Nlrp2, which was highly upregulated. NLRP2 immunoreactivity in OLs was confirmed in post-mortem PPMS brain tissues, with higher expression than in control tissues. Altogether, our findings suggest that mature OLs in PPMS affected individuals carry intrinsic abnormalities that could contribute, at least in part, to the pathophysiology of this form of the disease.
Collapse
Affiliation(s)
- Melanie J. Plastini
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
- The Neuroscience Graduate Program, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Haritha L. Desu
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
- The Neuroscience Graduate Program, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Maureen C. Ascona
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
- The Neuroscience Graduate Program, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Anna L. Lang
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Mario A. Saporta
- The Neuroscience Graduate Program, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Roberta Brambilla
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
- The Neuroscience Graduate Program, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- BRIDGE-Brain Research-Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- *Correspondence: Roberta Brambilla,
| |
Collapse
|
41
|
Gao Y, Xie D, Wang Y, Niu L, Jiang H. Short-Chain Fatty Acids Reduce Oligodendrocyte Precursor Cells Loss by Inhibiting the Activation of Astrocytes via the SGK1/IL-6 Signalling Pathway. Neurochem Res 2022; 47:3476-3489. [PMID: 36098889 PMCID: PMC9546972 DOI: 10.1007/s11064-022-03710-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 12/09/2022]
Abstract
Short-chain fatty acids (SCFAs) are known to be actively involved in neurological diseases, but their roles in hypoxic-ischaemic brain injury (HIBI) are unclear. In this study, a rat model of HIBI was established, and this study measured the changes in IL-6 and NOD-like receptor thermal protein domain associated protein 3 (NLRP3), in addition to proliferation and apoptosis indicators of oligodendrocyte precursor cells (OPCs). The mechanism of action of SCFA on astrocytes was also investigated. Astrocytes were subjected to hypoxia in vitro, and OPCs were treated with IL-6. The results showed that SCFAs significantly alleviated HIBI-induced activation of astrocytes and loss of OPCs. SCFA pretreatment (1) downregulated the expression of NLRP3, IL-6, CCL2, and IP-10; (2) had no effect on the proliferation of OPCs; (3) ameliorated the abnormal expression of Bax and Bcl-2; and (4) regulated IL-6 expression via the SGK1-related pathway in astrocytes. Our findings revealed that SCFAs alleviated the loss of OPCs by regulating astrocyte activation through the SGK1/IL-6 signalling pathway.
Collapse
Affiliation(s)
- Yanmin Gao
- Department of General Practice, Shanghai East Hospital, School of Medicine, Tongji University, No.150, Jimo Road, Pudong New District, Shanghai, 200120, China.,Department of General Practice, Kongjiang Community Health Service Center, No. 100, Yanji West Road, Yangpu District, Shanghai, 200093, China
| | - Di Xie
- Emergency Department, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Yang Wang
- Department of General Practice, Shanghai East Hospital, School of Medicine, Tongji University, No.150, Jimo Road, Pudong New District, Shanghai, 200120, China.,Emergency Department, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Lei Niu
- Department of General Practice, Shanghai East Hospital, School of Medicine, Tongji University, No.150, Jimo Road, Pudong New District, Shanghai, 200120, China.,Emergency Department, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Hua Jiang
- Department of General Practice, Shanghai East Hospital, School of Medicine, Tongji University, No.150, Jimo Road, Pudong New District, Shanghai, 200120, China. .,Emergency Department, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No.1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China.
| |
Collapse
|
42
|
Beiter RM, Rivet-Noor C, Merchak AR, Bai R, Johanson DM, Slogar E, Sol-Church K, Overall CC, Gaultier A. Evidence for oligodendrocyte progenitor cell heterogeneity in the adult mouse brain. Sci Rep 2022; 12:12921. [PMID: 35902669 PMCID: PMC9334628 DOI: 10.1038/s41598-022-17081-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/20/2022] [Indexed: 12/25/2022] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) account for approximately 5% of the adult brain and have been historically studied for their role in myelination. In the adult brain, OPCs maintain their proliferative capacity and ability to differentiate into oligodendrocytes throughout adulthood, even though relatively few mature oligodendrocytes are produced post-developmental myelination. Recent work has begun to demonstrate that OPCs likely perform multiple functions in both homeostasis and disease and can significantly impact behavioral phenotypes such as food intake and depressive symptoms. However, the exact mechanisms through which OPCs might influence brain function remain unclear. The first step in further exploration of OPC function is to profile the transcriptional repertoire and assess the heterogeneity of adult OPCs. In this work, we demonstrate that adult OPCs are transcriptionally diverse and separate into two distinct populations in the homeostatic brain. These two groups show distinct transcriptional signatures and enrichment of biological processes unique to individual OPC populations. We have validated these OPC populations using multiple methods, including multiplex RNA in situ hybridization and RNA flow cytometry. This study provides an important resource that profiles the transcriptome of adult OPCs and will provide a toolbox for further investigation into novel OPC functions.
Collapse
Affiliation(s)
- Rebecca M Beiter
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.,Graduate Program in Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Courtney Rivet-Noor
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.,Graduate Program in Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Andrea R Merchak
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.,Graduate Program in Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Robin Bai
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - David M Johanson
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Erica Slogar
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Katia Sol-Church
- Genome Analysis and Technology Core, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Christopher C Overall
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Alban Gaultier
- Department of Neuroscience, Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA. .,Graduate Program in Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.
| |
Collapse
|
43
|
Rivet-Noor CR, Merchak AR, Li S, Beiter RM, Lee S, Thomas JA, Fernández-Castañeda A, Shin JB, Gaultier A. Stress-induced despair behavior develops independently of the Ahr-RORγt axis in CD4 + cells. Sci Rep 2022; 12:8594. [PMID: 35597802 PMCID: PMC9124178 DOI: 10.1038/s41598-022-12464-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 05/04/2022] [Indexed: 02/07/2023] Open
Abstract
Current treatments for major depressive disorder are limited to neuropharmacological approaches and are ineffective for large numbers of patients. Recently, alternative means have been explored to understand the etiology of depression. Specifically, changes in the microbiome and immune system have been observed in both clinical settings and in mouse models. As such, microbial supplements and probiotics have become a target for potential therapeutics. A current hypothesis for the mechanism of action of these supplements is via the aryl hydrocarbon receptor's (Ahr) modulation of the T helper 17 cell (Th17) and T regulatory cell axis. As inflammatory RORγt + CD4 + Th17 T cells and their primary cytokine IL-17 have been implicated in the development of stress-induced depression, the connection between stress, the Ahr, Th17s and depression remains critical to understanding mood disorders. Here, we utilize genetic knockouts to examine the role of the microbial sensor Ahr in the development of stressinduced despair behavior. We observe an Ahr-independent increase in gut-associated Th17s in stressed mice, indicating that the Ahr is not responsible for this communication. Further, we utilized a CD4-specific RAR Related Orphan Receptor C (Rorc) knockout line to disrupt the production of Th17s. Mice lacking Rorc-produced IL-17 did not show any differences in behavior before or after stress when compared to controls. Finally, we utilize an unsupervised machine learning system to examine minute differences in behavior that could not be observed by traditional behavioral assays. Our data demonstrate that neither CD4 specific Ahr nor Rorc are necessary for the development of stress-induced anxiety- or depressive-like behaviors. These data suggest that research approaches should focus on other sources or sites of IL-17 production in stress-induced depression.
Collapse
Affiliation(s)
- Courtney R Rivet-Noor
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
- Graduate Program in Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Andrea R Merchak
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
- Graduate Program in Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Sihan Li
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
- Graduate Program in Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Rebecca M Beiter
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
- Graduate Program in Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Sangwoo Lee
- Undergraduate Department of Computer Science, University of Virginia School of Engineering and Applied Science, Charlottesville, VA, 22904, USA
| | - Jalon Aaron Thomas
- Undergraduate Department of Computer Science, University of Virginia School of Engineering and Applied Science, Charlottesville, VA, 22904, USA
| | - Anthony Fernández-Castañeda
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
- Graduate Program in Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Jung-Bum Shin
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
- Graduate Program in Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Alban Gaultier
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.
- Graduate Program in Neuroscience, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.
| |
Collapse
|
44
|
Meijer M, Agirre E, Kabbe M, van Tuijn CA, Heskol A, Zheng C, Mendanha Falcão A, Bartosovic M, Kirby L, Calini D, Johnson MR, Corces MR, Montine TJ, Chen X, Chang HY, Malhotra D, Castelo-Branco G. Epigenomic priming of immune genes implicates oligodendroglia in multiple sclerosis susceptibility. Neuron 2022; 110:1193-1210.e13. [PMID: 35093191 PMCID: PMC9810341 DOI: 10.1016/j.neuron.2021.12.034] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 11/05/2021] [Accepted: 12/27/2021] [Indexed: 01/05/2023]
Abstract
Multiple sclerosis (MS) is characterized by a targeted attack on oligodendroglia (OLG) and myelin by immune cells, which are thought to be the main drivers of MS susceptibility. We found that immune genes exhibit a primed chromatin state in single mouse and human OLG in a non-disease context, compatible with transitions to immune-competent states in MS. We identified BACH1 and STAT1 as transcription factors involved in immune gene regulation in oligodendrocyte precursor cells (OPCs). A subset of immune genes presents bivalency of H3K4me3/H3K27me3 in OPCs, with Polycomb inhibition leading to their increased activation upon interferon gamma (IFN-γ) treatment. Some MS susceptibility single-nucleotide polymorphisms (SNPs) overlap with these regulatory regions in mouse and human OLG. Treatment of mouse OPCs with IFN-γ leads to chromatin architecture remodeling at these loci and altered expression of interacting genes. Thus, the susceptibility for MS may involve OLG, which therefore constitutes novel targets for immunological-based therapies for MS.
Collapse
Affiliation(s)
- Mandy Meijer
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Eneritz Agirre
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Mukund Kabbe
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Cassandra A van Tuijn
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Abeer Heskol
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden; Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
| | - Chao Zheng
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Ana Mendanha Falcão
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's Associate Laboratory, PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Marek Bartosovic
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Leslie Kirby
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Daniela Calini
- Roche Pharma Research and Early Development, 4070 Basel, Switzerland
| | - Michael R Johnson
- Faculty of Medicine, Department of Brain Sciences, Imperial College of London, SW7 2AZ London, UK
| | - M Ryan Corces
- Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA; Center for Personal Dynamic Regulomes and Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA; Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas J Montine
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Xingqi Chen
- Center for Personal Dynamic Regulomes and Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA; Department of Immunology, Genetics, and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes and Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305-5101, USA
| | - Dheeraj Malhotra
- Roche Pharma Research and Early Development, 4070 Basel, Switzerland
| | - Gonçalo Castelo-Branco
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden; Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, 171 77 Stockholm, Sweden.
| |
Collapse
|
45
|
Sun N, Kellis M. Immune genes outside immune cells for multiple sclerosis. Neuron 2022; 110:1090-1092. [PMID: 35390286 DOI: 10.1016/j.neuron.2022.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In this issue of Neuron, Meijer and Agirre et al. (2022) demonstrate that immune genes exhibit a primed chromatin state in healthy oligodendroglia and are transcriptionally activated in MS through a series of epigenetic activations including histone modification deposition, transcription factor binding, and chromatin reconfiguration.
Collapse
Affiliation(s)
- Na Sun
- MIT Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA, USA
| | - Manolis Kellis
- MIT Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA, USA.
| |
Collapse
|
46
|
Mazur RA, Yokosawa R, VandeVord PJ, Lampe KJ. The Need for Tissue Engineered Models to Facilitate the Study of Oligodendrocyte Progenitor Cells in Traumatic Brain Injury and Repair. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2022. [DOI: 10.1016/j.cobme.2022.100378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
47
|
Li H, Chen Y, Niu J, Yi C. New insights into the immunologic role of oligodendrocyte lineage cells in demyelination diseases. J Biomed Res 2022; 36:343-352. [PMID: 35578762 PMCID: PMC9548433 DOI: 10.7555/jbr.36.20220016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Oligodendrocyte lineage cells (OL-lineage cells) are a cell population that are crucial for mammalian central nervous system (CNS) myelination. OL-lineage cells go through developmental stages, initially differentiating into oligodendrocyte precursor cells (OPCs), before becoming immature oligodendrocytes, then mature oligodendrocytes (OLs). While the main function of cell lineage is in myelin formation, and increasing number of studies have turned to explore the immunological characteristics of these cells. Initially, these studies focused on discovering how OPCs and OLs are affected by the immune system, and then, how these immunological changes influence the myelination process. However, recent studies have uncovered another feature of OL-lineage cells in our immune systems. It would appear that OL-lineage cells also express immunological factors such as cytokines and chemokines in response to immune activation, and the expression of these factors changes under various pathologic conditions. Evidence suggests that OL-lineage cells actually modulate immune functions. Indeed, OL-lineage cells appear to play both "victim" and "agent" in the CNS which raises a number of questions. Here, we summarize immunologic changes in OL-lineage cells and their effects, as well as consider OL-lineage cell changes which influence immune cells under pathological conditions. We also describe some of the underlying mechanisms of these changes and their effects. Finally, we describe several studies which use OL-lineage cells as immunotherapeutic targets for demyelination diseases.
Collapse
Affiliation(s)
- Hui Li
- Research Centre, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
| | - Yang Chen
- Research Centre, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Jianqin Niu
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China
- Jianqin Niu, Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Gaotanyan Main street, Chongqing 400038, China. Tel: +86-13668016001, E-mail:
| | - Chenju Yi
- Research Centre, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
- Chenju Yi, Research Centre, the Seventh Affiliated Hospital of Sun Yat-sen University, 628 Zhenyuan Road, Guangming (New) District, Shenzhen 518107, China. Tel: +86-13419189905, E-mail:
| |
Collapse
|
48
|
Marangon D, Caporale N, Boccazzi M, Abbracchio MP, Testa G, Lecca D. Novel in vitro Experimental Approaches to Study Myelination and Remyelination in the Central Nervous System. Front Cell Neurosci 2021; 15:748849. [PMID: 34720882 PMCID: PMC8551863 DOI: 10.3389/fncel.2021.748849] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/22/2021] [Indexed: 12/15/2022] Open
Abstract
Myelin is the lipidic insulating structure enwrapping axons and allowing fast saltatory nerve conduction. In the central nervous system, myelin sheath is the result of the complex packaging of multilamellar extensions of oligodendrocyte (OL) membranes. Before reaching myelinating capabilities, OLs undergo a very precise program of differentiation and maturation that starts from OL precursor cells (OPCs). In the last 20 years, the biology of OPCs and their behavior under pathological conditions have been studied through several experimental models. When co-cultured with neurons, OPCs undergo terminal maturation and produce myelin tracts around axons, allowing to investigate myelination in response to exogenous stimuli in a very simple in vitro system. On the other hand, in vivo models more closely reproducing some of the features of human pathophysiology enabled to assess the consequences of demyelination and the molecular mechanisms of remyelination, and they are often used to validate the effect of pharmacological agents. However, they are very complex, and not suitable for large scale drug discovery screening. Recent advances in cell reprogramming, biophysics and bioengineering have allowed impressive improvements in the methodological approaches to study brain physiology and myelination. Rat and mouse OPCs can be replaced by human OPCs obtained by induced pluripotent stem cells (iPSCs) derived from healthy or diseased individuals, thus offering unprecedented possibilities for personalized disease modeling and treatment. OPCs and neural cells can be also artificially assembled, using 3D-printed culture chambers and biomaterial scaffolds, which allow modeling cell-to-cell interactions in a highly controlled manner. Interestingly, scaffold stiffness can be adopted to reproduce the mechanosensory properties assumed by tissues in physiological or pathological conditions. Moreover, the recent development of iPSC-derived 3D brain cultures, called organoids, has made it possible to study key aspects of embryonic brain development, such as neuronal differentiation, maturation and network formation in temporal dynamics that are inaccessible to traditional in vitro cultures. Despite the huge potential of organoids, their application to myelination studies is still in its infancy. In this review, we shall summarize the novel most relevant experimental approaches and their implications for the identification of remyelinating agents for human diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Davide Marangon
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Nicolò Caporale
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Human Technopole, Milan, Italy
| | - Marta Boccazzi
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Maria P. Abbracchio
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Giuseppe Testa
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Human Technopole, Milan, Italy
| | - Davide Lecca
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
49
|
Pavic G, Petzsch P, Jansen R, Raba K, Rychlik N, Simiantonakis I, Küry P, Göttle P, Köhrer K, Hartung HP, Meuth SG, Jander S, Gliem M. Microglia contributes to remyelination in cerebral but not spinal cord ischemia. Glia 2021; 69:2739-2751. [PMID: 34390590 DOI: 10.1002/glia.24068] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/02/2021] [Accepted: 07/07/2021] [Indexed: 11/07/2022]
Abstract
Inflammation after injury of the central nervous system (CNS) is increasingly viewed as a therapeutic target. However, comparative studies in different CNS compartments are sparse. To date only few studies based on immunohistochemical data and all referring to mechanical injury have directly compared inflammation in different CNS compartments. These studies revealed that inflammation is more pronounced in spinal cord than in brain. Therefore, it is unclear whether concepts and treatments established in the cerebral cortex can be transferred to spinal cord lesions and vice versa or whether immunological treatments must be adapted to different CNS compartments. By use of transcriptomic and flow cytometry analysis of equally sized photothrombotically induced lesions in the cerebral cortex and the spinal cord, we could document an overall comparable inflammatory reaction and repair activity in brain and spinal cord between day 1 and day 7 after ischemia. However, remyelination was increased after cerebral versus spinal cord ischemia which is in line with increased remyelination in gray matter in previous analyses and was accompanied by microglia dominated inflammation opposed to monocytes/macrophages dominated inflammation after spinal cord ischemia. Interestingly remyelination could be reduced by microglia and not hematogenous macrophage depletion. Our results show that despite different cellular composition of the postischemic infiltrate the inflammatory response in cerebral cortex and spinal cord are comparable between day 1 and day 7. A striking difference was higher remyelination capacity in the cerebral cortex, which seems to be supported by microglia dominance.
Collapse
Affiliation(s)
- Goran Pavic
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Patrick Petzsch
- Biological and Medical Research Center (BMFZ), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Robin Jansen
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Katharina Raba
- Institute for Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Nicole Rychlik
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | | | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Peter Göttle
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Karl Köhrer
- Biological and Medical Research Center (BMFZ), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Sebastian Jander
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Michael Gliem
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
50
|
Psenicka MW, Smith BC, Tinkey RA, Williams JL. Connecting Neuroinflammation and Neurodegeneration in Multiple Sclerosis: Are Oligodendrocyte Precursor Cells a Nexus of Disease? Front Cell Neurosci 2021; 15:654284. [PMID: 34234647 PMCID: PMC8255483 DOI: 10.3389/fncel.2021.654284] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 05/20/2021] [Indexed: 12/14/2022] Open
Abstract
The pathology in neurodegenerative diseases is often accompanied by inflammation. It is well-known that many cells within the central nervous system (CNS) also contribute to ongoing neuroinflammation, which can promote neurodegeneration. Multiple sclerosis (MS) is both an inflammatory and neurodegenerative disease in which there is a complex interplay between resident CNS cells to mediate myelin and axonal damage, and this communication network can vary depending on the subtype and chronicity of disease. Oligodendrocytes, the myelinating cell of the CNS, and their precursors, oligodendrocyte precursor cells (OPCs), are often thought of as the targets of autoimmune pathology during MS and in several animal models of MS; however, there is emerging evidence that OPCs actively contribute to inflammation that directly and indirectly contributes to neurodegeneration. Here we discuss several contributors to MS disease progression starting with lesion pathology and murine models amenable to studying particular aspects of disease. We then review how OPCs themselves can play an active role in promoting neuroinflammation and neurodegeneration, and how other resident CNS cells including microglia, astrocytes, and neurons can impact OPC function. Further, we outline the very complex and pleiotropic role(s) of several inflammatory cytokines and other secreted factors classically described as solely deleterious during MS and its animal models, but in fact, have many neuroprotective functions and promote a return to homeostasis, in part via modulation of OPC function. Finally, since MS affects patients from the onset of disease throughout their lifespan, we discuss the impact of aging on OPC function and CNS recovery. It is becoming clear that OPCs are not simply a bystander during MS progression and uncovering the active roles they play during different stages of disease will help uncover potential new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Morgan W. Psenicka
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Brandon C. Smith
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH, United States
| | - Rachel A. Tinkey
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Jessica L. Williams
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Brain Health Research Institute, Kent State University, Kent, OH, United States
| |
Collapse
|