1
|
Zhu H, Hu E, Guo X, Yuan Z, Jiang H, Zhang W, Tang T, Wang Y, Li T. Promoting remyelination in central nervous system diseases: Potentials and prospects of natural products and herbal medicine. Pharmacol Res 2024; 210:107533. [PMID: 39617281 DOI: 10.1016/j.phrs.2024.107533] [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: 10/09/2024] [Revised: 11/22/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024]
Abstract
Myelin damage is frequently associated with central nervous system (CNS) diseases and is a critical factor influencing neurological function and disease prognosis. Nevertheless, the majority of current treatments for the CNS concentrate on gray matter injury and repair strategies, while clinical interventions specifically targeting myelin repair remain unavailable. In recent years, natural products and herbal medicine have achieved considerable progress in the domain of myelin repair, given their remarkable curative effect and low toxic side effects, demonstrating significant therapeutic potential. In this review, we present a rather comprehensive account of the mechanisms underlying myelin formation, injury, and repair, with a particular emphasis on the interactions between oligodendrocytes and other glial cells. Furthermore, we summarize the natural products and herbal medicine currently employed in remyelination along with their mechanisms of action, highlighting the potential and challenges of certain natural compounds to enhance myelin repair. This review aims to facilitate the expedited development of innovative therapeutics derived from natural products and herbal medicine and furnish novel insights into myelin repair in the CNS.
Collapse
Affiliation(s)
- Haonan Zhu
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - En Hu
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China
| | - Xin Guo
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Zhiqiang Yuan
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China
| | - Haoying Jiang
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Wei Zhang
- The College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China
| | - Tao Tang
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China
| | - Yang Wang
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China
| | - Teng Li
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China.
| |
Collapse
|
2
|
Plug BC, Revers IM, Breur M, González GM, Timmerman JA, Meijns NRC, Hamberg D, Wagendorp J, Nutma E, Wolf NI, Luchicchi A, Mansvelder HD, van Til NP, van der Knaap MS, Bugiani M. Human post-mortem organotypic brain slice cultures: a tool to study pathomechanisms and test therapies. Acta Neuropathol Commun 2024; 12:83. [PMID: 38822428 PMCID: PMC11140981 DOI: 10.1186/s40478-024-01784-1] [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] [Accepted: 04/16/2024] [Indexed: 06/03/2024] Open
Abstract
Human brain experimental models recapitulating age- and disease-related characteristics are lacking. There is urgent need for human-specific tools that model the complex molecular and cellular interplay between different cell types to assess underlying disease mechanisms and test therapies. Here we present an adapted ex vivo organotypic slice culture method using human post-mortem brain tissue cultured at an air-liquid interface to also study brain white matter. We assessed whether these human post-mortem brain slices recapitulate the in vivo neuropathology and if they are suitable for pathophysiological, experimental and pre-clinical treatment development purposes, specifically regarding leukodystrophies. Human post-mortem brain tissue and cerebrospinal fluid were obtained from control, psychiatric and leukodystrophy donors. Slices were cultured up to six weeks, in culture medium with or without human cerebrospinal fluid. Human post-mortem organotypic brain slice cultures remained viable for at least six weeks ex vivo and maintained tissue structure and diversity of (neural) cell types. Supplementation with cerebrospinal fluid could improve slice recovery. Patient-derived organotypic slice cultures recapitulated and maintained known in vivo neuropathology. The cultures also showed physiologic multicellular responses to lysolecithin-induced demyelination ex vivo, indicating their suitability to study intrinsic repair mechanisms upon injury. The slice cultures were applicable for various experimental studies, as multi-electrode neuronal recordings. Finally, the cultures showed successful cell-type dependent transduction with gene therapy vectors. These human post-mortem organotypic brain slice cultures represent an adapted ex vivo model suitable for multifaceted studies of brain disease mechanisms, boosting translation from human ex vivo to in vivo. This model also allows for assessing potential treatment options, including gene therapy applications. Human post-mortem brain slice cultures are thus a valuable tool in preclinical research to study the pathomechanisms of a wide variety of brain diseases in living human tissue.
Collapse
Affiliation(s)
- Bonnie C Plug
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Ilma M Revers
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Marjolein Breur
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Gema Muñoz González
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam University Medical Centre, VU University, Amsterdam Neuroscience, De Boelelaan 1108, Amsterdam, 1081 HZ, The Netherlands
| | - Jaap A Timmerman
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam Neuroscience, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Niels R C Meijns
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam University Medical Centre, VU University, Amsterdam Neuroscience, De Boelelaan 1108, Amsterdam, 1081 HZ, The Netherlands
| | - Daniek Hamberg
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Jikke Wagendorp
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Erik Nutma
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
| | - Nicole I Wolf
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
| | - Antonio Luchicchi
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam University Medical Centre, VU University, Amsterdam Neuroscience, De Boelelaan 1108, Amsterdam, 1081 HZ, The Netherlands
| | - Huibert D Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam Neuroscience, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Niek P van Til
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam Neuroscience, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Marjo S van der Knaap
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam Neuroscience, De Boelelaan 1085, Amsterdam, 1081 HV, The Netherlands
| | - Marianna Bugiani
- Department of Paediatrics and Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands.
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centre, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Meibergdreef 9, 1100 DD, Amsterdam, The Netherlands.
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centre, Meibergdreef 9, Amsterdam, 1100 DD, The Netherlands.
| |
Collapse
|
3
|
Elitt CM, Ross MM, Wang J, Fahrni CJ, Rosenberg PA. Developmental regulation of zinc homeostasis in differentiating oligodendrocytes. Neurosci Lett 2024; 831:137727. [PMID: 38467270 DOI: 10.1016/j.neulet.2024.137727] [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: 07/09/2023] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 03/13/2024]
Abstract
Oligodendrocytes develop through sequential stages and understanding pathways regulating their differentiation remains an important area of investigation. Zinc is required for the function of enzymes, proteins and transcription factors, including those important in myelination and mitosis. Our previous studies using the ratiometric zinc sensor chromis-1 demonstrated a reduction in intracellular free zinc concentrations in mature MBP+ oligodendrocytes compared with earlier stages (Bourassa et al., 2018). We performed a more detailed developmental study to better understand the temporal course of zinc homeostasis across the oligodendrocyte lineage. Using chromis-1, we found a transient increase in free zinc after O4+,O1- pre-oligodendrocytes were switched from proliferation medium into terminal differentiation medium. To gather other evidence for dynamic regulation of free zinc during oligodendrocyte development, qPCR was used to evaluate mRNA expression of major zinc storage proteins metallothioneins (MTs) and metal regulatory transcription factor 1 (MTF1), which controls expression of MTs. MT1, MT2 and MTF1 mRNAs were increased several fold in mature oligodendrocytes compared to oligodendrocytes in proliferation medium. To assess the depth of the zinc buffer, we assayed zinc release from intracellular stores using the oxidizing thiol reagent 2,2'-dithiodipyridine (DTDP). Exposure to DTDP resulted in ∼ 100% increase in free zinc in pre-oligodendrocytes but, paradoxically more modest ∼ 60% increase in mature oligodendrocytes despite increased expression of MTs. These results suggest that zinc homeostasis is regulated during oligodendrocyte development, that oligodendrocytes are a useful model for studying zinc homeostasis in the central nervous system, and that regulation of zinc homeostasis may be important in oligodendrocyte differentiation.
Collapse
Affiliation(s)
- Christopher M Elitt
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, United States; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, United States.
| | - Madeline M Ross
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, United States; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, United States
| | - Jianlin Wang
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, United States; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, United States
| | - Christoph J Fahrni
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, United States; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, United States
| | - Paul A Rosenberg
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, United States; F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, United States
| |
Collapse
|
4
|
Sevin C, Mochel F. Hematopoietic stem cell transplantation in leukodystrophies. HANDBOOK OF CLINICAL NEUROLOGY 2024; 204:355-366. [PMID: 39322389 DOI: 10.1016/b978-0-323-99209-1.00017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
More than 50 leukodystrophies have been described. This group of inherited disorders affects myelin development and/or maintenance and can manifest from birth to adulthood. Neuroinflammation is a hallmark of some leukodystrophies, explaining in part the therapeutic benefit of hematopoietic stem cell transplantation (HSCT). Indeed, in addition to supplying the CNS with myelomonocyte donor cells expressing the deficient protein or enzyme, HSCT allows the restoration of normal microglia function, which may act on neuroinflammation. In this chapter, we explore the rationale, indication, and outcome of HSCT in Cerebral Adrenoleukodystrophy (CALD), Metachromatic Leukodystrophy (MLD), Krabbe Disease (KD), and Adult-onset Leukoencephalopathy with Axonal Spheroids and Pigmented Glia (ALSP), which are among the most frequent leukodystrophies. For these leukodystrophies, HSCT may modify notably the natural history and improve CNS-related deficits, provided that the procedure is performed early into the disease course. In addition, we discuss the recent development of ex vivo gene therapy for CALD and MLD as a promising alternative to allograft.
Collapse
Affiliation(s)
- Caroline Sevin
- AP-HP, Kremlin-Bicêtre University Hospital, Department of Neuropediatrics, Reference Center for Pediatric Leukodystrophies, Paris, France; INSERM U 1127, CNRS UMR 7225, Sorbonne Université, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau, ICM, Paris, France
| | - Fanny Mochel
- INSERM U 1127, CNRS UMR 7225, Sorbonne Université, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau, ICM, Paris, France; AP-HP, Pitié-Salpêtrière University Hospital, Department of Medical Genetics, Reference Centers for Adult Neurometabolic Diseases and Adult Leukodystrophies, Paris, France.
| |
Collapse
|
5
|
Jaunmuktane Z. Neuropathology of white matter disorders. HANDBOOK OF CLINICAL NEUROLOGY 2024; 204:3-20. [PMID: 39322386 DOI: 10.1016/b978-0-323-99209-1.00011-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
The hallmark neuropathologic feature of all leukodystrophies is depletion or alteration of the white matter of the central nervous system; however increasing genetic discoveries highlight the genetic heterogeneity of white matter disorders. These discoveries have significantly helped to advance the understanding of the complexity of molecular mechanisms involved in the biogenesis and maintenance of healthy white matter. Accordingly, genetic discoveries and functional studies have enabled us to firmly establish that multiple distinct structural defects can lead to white matter pathology. Leukodystrophies can develop not only due to defects in proteins essential for myelin biogenesis and maintenance or oligodendrocyte function, but also due to mutations encoding myriad of proteins involved in the function of neurons, astrocytes, microglial cells as well as blood vessels. To a variable extent, some leukodystrophies also show gray matter, peripheral nervous system, or multisystem involvement. Depending on the genetic defect and its role in the formation or maintenance of the white matter, leukodystrophies can present either in early childhood or adulthood. In this chapter, the classification of leukodystrophies will be discussed from the cellular defect point of view, followed by a description of known neuropathologic alterations for all leukodystrophies.
Collapse
Affiliation(s)
- Zane Jaunmuktane
- Division of Neuropathology, National Hospital for Neurology and Neurosurgery, University College London Hospitals, London, United Kingdom; Queen Square Brain Bank for Neurological Disorders, Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, United Kingdom.
| |
Collapse
|
6
|
Oizumi H, Miyamoto Y, Seiwa C, Yamamoto M, Yoshioka N, Iizuka S, Torii T, Ohbuchi K, Mizoguchi K, Yamauchi J, Asou H. Lethal adulthood myelin breakdown by oligodendrocyte-specific Ddx54 knockout. iScience 2023; 26:107448. [PMID: 37720086 PMCID: PMC10502337 DOI: 10.1016/j.isci.2023.107448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/08/2023] [Accepted: 07/18/2023] [Indexed: 09/19/2023] Open
Abstract
Multiple sclerosis (MS) is a leading disease that causes disability in young adults. We have previously shown that a DEAD-box RNA helicase Ddx54 binds to mRNA and protein isoforms of myelin basic protein (MBP) and that Ddx54 siRNA blocking abrogates oligodendrocyte migration and myelination. Herein, we show that MBP-driven Ddx54 knockout mice (Ddx54 fl/fl;MBP-Cre), after the completion of normal postnatal myelination, gradually develop abnormalities in behavioral profiles and learning ability, inner myelin sheath breakdown, loss of myelinated axons, apoptosis of oligodendrocytes, astrocyte and microglia activation, and they die within 7 months but show minimal peripheral immune cell infiltration. Myelin in Ddx54fl/fl;MBP-Cre is highly vulnerable to the neurotoxicant cuprizone and Ddx54 knockdown greatly impairs myelination in vitro. Ddx54 expression in oligodendrocyte-lineage cells decreased in corpus callosum of MS patients. Our results demonstrate that Ddx54 is indispensable for myelin homeostasis, and they provide a demyelinating disease model based on intrinsic disintegration of adult myelin.
Collapse
Affiliation(s)
- Hiroaki Oizumi
- Tsumura Kampo Laboratories, Tsumura & Co, Ami, Ibaraki 300-1192, Japan
| | - Yuki Miyamoto
- Department of Pharmacology, National Research Institute for Child Health and Development, Setagaya-ku, Tokyo 157-8535, Japan
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Chika Seiwa
- Glovia Myelin Research Institute, Tsurumi-ku, Yokohama, Kanagawa 230-0046, Japan
| | - Masahiro Yamamoto
- Tsumura Kampo Laboratories, Tsumura & Co, Ami, Ibaraki 300-1192, Japan
| | - Nozomu Yoshioka
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Seiichi Iizuka
- Tsumura Kampo Laboratories, Tsumura & Co, Ami, Ibaraki 300-1192, Japan
| | - Tomohiro Torii
- Laboratory of Ion Channel Pathophysiology, Graduate School of Brain Science, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| | - Katsuya Ohbuchi
- Tsumura Kampo Laboratories, Tsumura & Co, Ami, Ibaraki 300-1192, Japan
| | | | - Junji Yamauchi
- Department of Pharmacology, National Research Institute for Child Health and Development, Setagaya-ku, Tokyo 157-8535, Japan
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Hiroaki Asou
- Glovia Myelin Research Institute, Tsurumi-ku, Yokohama, Kanagawa 230-0046, Japan
| |
Collapse
|
7
|
Ellezam B, Kaseka ML, Nguyen DK, Michaud J. SCA34 caused by ELOVL4 L168F mutation is a lysosomal lipid storage disease sharing pathology features with neuronal ceroid lipofuscinosis and peroxisomal disorders. Acta Neuropathol 2023; 146:337-352. [PMID: 37184663 DOI: 10.1007/s00401-023-02582-0] [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: 03/16/2023] [Revised: 04/27/2023] [Accepted: 04/27/2023] [Indexed: 05/16/2023]
Abstract
Spinocerebellar ataxia 34 (SCA34) is a late-onset progressive ataxia caused by a mutation in ELOVL4, a gene involved in the biosynthesis of very long-chain fatty acids (VLCFAs). We performed post-mortem neuropathological examinations on four SCA34 patients with the ELOVL4 L168F mutation and compared the findings to age-matched controls. Specific gross findings of SCA34 were limited to pontocerebellar atrophy. On light microscopy, pontine base showed neuronal loss and storage of an autofluorescent lipopigment positive on oil red O, PAS and Hale's colloidal iron and negative on Alcian blue and Luxol fast blue (LFB). Among the swollen neurons were abundant CD68+ /CD163+ /IBA1- macrophages laden with a material with similar histochemical profile as in neurons except for the lack of autofluorescence and oil red O positivity and the presence of needle-like birefringent inclusions. Normal resting IBA1 + microglia were generally absent from pontine base nuclei but present in normal numbers elsewhere in the pons. In dentate nucleus neurons, atrophy was milder than in the pontine base and the coarser storage material was LFB-positive, closely resembling lipofuscin. On electron microscopy, dentate nucleus neurons showed neuronal storage of tridimensionally organized trilaminar spicules within otherwise normal lipofuscin, while in the more affected pontine base neurons, lipofuscin was almost completely replaced by the storage material. Storage macrophages were tightly packed with stacks of unorganized trilaminar spicules, reminiscent of the storage material seen in peroxisomal disorders and thought to represent VLCFAs incorporated in complex polar lipids. In summary, we provide histochemical and ultrastructural evidence that SCA34 is a lipid storage disease, the first among the currently known SCAs, and that the storage lipid is accumulating within neuronal lipofuscin. Our findings suggest that the storage lipid is similar to the one accumulating in non-neuronal cells in peroxisomal disorders and provide the first ultrastructural description of this type of material within neurons.
Collapse
Affiliation(s)
- Benjamin Ellezam
- Division of Pathology, CHU Sainte-Justine, 3175 Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada.
- Department of Pathology and Cell Biology, Université de Montréal, Montréal, QC, Canada.
| | - Matsanga L Kaseka
- Division of Neurology, CHU Sainte-Justine, Montréal, QC, Canada
- Department of Neuroscience, Université de Montréal, Montréal, QC, Canada
| | - Dang Khoa Nguyen
- Department of Neuroscience, Université de Montréal, Montréal, QC, Canada
- Division of Neurology, Centre hospitalier de l'Université de Montréal, Montréal, QC, Canada
| | - Jean Michaud
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
8
|
Elitt CM, Ross MM, Wang J, Fahrni CJ, Rosenberg PA. Developmental regulation of zinc homeostasis in differentiating oligodendrocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.26.550230. [PMID: 37546881 PMCID: PMC10402100 DOI: 10.1101/2023.07.26.550230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Oligodendrocytes develop through well characterized stages and understanding pathways regulating their differentiation remains an active area of investigation. Zinc is required for the function of many enzymes, proteins and transcription factors, including those important in myelination and mitosis. Our previous studies using the ratiometric zinc sensor chromis-1 demonstrated a reduction in intracellular free zinc concentrations in mature oligodendrocytes compared with earlier stages (Bourassa et al., 2018). We performed a more detailed developmental study to better understand the temporal course of zinc homeostasis across the oligodendrocyte lineage. Using chromis-1, we found a transient increase in free zinc after developing oligodendrocytes were switched into differentiation medium. To gather other evidence for dynamic regulation of free zinc during oligodendrocyte development, qPCR was used to evaluate mRNA expression of the major zinc storage proteins metallothioneins (MTs), and metal regulatory transcription factor 1 (MTF-1) which controls expression of MTs. MT-1, MT-2 and MTF1 mRNAs were all increased several fold in mature oligodendrocytes compared to developing oligodendrocytes. To assess the depth of the zinc buffer, we assayed zinc release from intracellular stores using the oxidizing thiol reagent 2,2'-dithiodipyridine (DTDP). Exposure to DTDP resulted in a ∼100% increase in free zinc in developing oligodendrocytes but, paradoxically more modest ∼60% increase in mature oligodendrocytes despite the increased expression of MTs. These results suggest that zinc homeostasis is regulated during oligodendrocyte development, that oligodendrocytes are a useful model for studying zinc homeostasis in the central nervous system, and that regulation of zinc homeostasis may be important in oligodendrocyte differentiation.
Collapse
|
9
|
Lotun A, Li D, Xu H, Su Q, Tuncer S, Sanmiguel J, Mooney M, Baer CE, Ulbrich R, Eyles SJ, Strittmatter L, Hayward LJ, Gessler DJ, Gao G. Renewal of oligodendrocyte lineage reverses dysmyelination and CNS neurodegeneration through corrected N-acetylaspartate metabolism. Prog Neurobiol 2023; 226:102460. [PMID: 37149081 PMCID: PMC10330635 DOI: 10.1016/j.pneurobio.2023.102460] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/18/2023] [Accepted: 04/28/2023] [Indexed: 05/08/2023]
Abstract
Myelinating oligodendrocytes are essential for neuronal communication and homeostasis of the central nervous system (CNS). One of the most abundant molecules in the mammalian CNS is N-acetylaspartate (NAA), which is catabolized into L-aspartate and acetate by the enzyme aspartoacylase (ASPA) in oligodendrocytes. The resulting acetate moiety is thought to contribute to myelin lipid synthesis. In addition, affected NAA metabolism has been implicated in several neurological disorders, including leukodystrophies and demyelinating diseases such as multiple sclerosis. Genetic disruption of ASPA function causes Canavan disease, which is hallmarked by increased NAA levels, myelin and neuronal loss, large vacuole formation in the CNS, and early death in childhood. Although NAA's direct role in the CNS is inconclusive, in peripheral adipose tissue, NAA-derived acetate has been found to modify histones, a mechanism known to be involved in epigenetic regulation of cell differentiation. We hypothesize that a lack of cellular differentiation in the brain contributes to the disruption of myelination and neurodegeneration in diseases with altered NAA metabolism, such as Canavan disease. Our study demonstrates that loss of functional Aspa in mice disrupts myelination and shifts the transcriptional expression of neuronal and oligodendrocyte markers towards less differentiated stages in a spatiotemporal manner. Upon re-expression of ASPA, these oligodendrocyte and neuronal lineage markers are either improved or normalized, suggesting that NAA breakdown by Aspa plays an essential role in the maturation of neurons and oligodendrocytes. Also, this effect of ASPA re-expression is blunted in old mice, potentially due to limited ability of neuronal, rather than oligodendrocyte, recovery.
Collapse
Affiliation(s)
- Anoushka Lotun
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Danning Li
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Hongxia Xu
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA; University of Science and Technology of Kunming, People's Republic of China
| | - Qin Su
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | | | - Julio Sanmiguel
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Morgan Mooney
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Christina E Baer
- Sanderson Center for Optical Experimentation, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Russell Ulbrich
- ScientiaLux LLC, Tissue-Gnostics USA-East, Worcester, MA, USA
| | - Stephen J Eyles
- Mass Spectrometry Core, University of Massachusetts, Amherst, MA, USA
| | - Lara Strittmatter
- Electron Microscopy Core, University of Massachusetts Chan Medical School, MA, USA
| | - Lawrence J Hayward
- Department of Neurology, University of Massachusetts Chan Medical School, MA, USA
| | - Dominic J Gessler
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA.
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, USA; Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Microbiology & Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
10
|
Madaan P, Kaushal Y, Srivastava P, Crow YJ, Livingston JH, Ahuja C, Sankhyan N. Delineating the epilepsy phenotype of NRROS-related microgliopathy: A case report and literature review. Seizure 2022; 100:15-20. [PMID: 35716448 DOI: 10.1016/j.seizure.2022.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 06/01/2022] [Accepted: 06/06/2022] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND Negative regulator of reactive oxygen species (NRROS) related microgliopathy, a rare and recently recognized neurodegenerative condition, is caused by pathogenic variants in the NRROS gene, which plays a major role in the regulation of transforming growth factor-beta 1. METHODS We report a child presenting with infantile spasms syndrome (ISS) with subsequent progressive neurodegeneration who was identified to harbour a novel likely pathogenic NRROS variant (c.1359del; p.Ser454Alafs*11). The previously published reports of patients with this disorder were also reviewed systematically. RESULTS Including our index patient, 11 children (6 girls) were identified in total. Early development was normal in seven of these eleven children. All had a history of drug-resistant epilepsy, with 3 having epileptic spasms. The median age at seizure onset and developmental regression was 12 months, and the median age at death was 36 months. Intracranial calcifications were described in eight of eleven children. Neuroimaging revealed progressive cerebral atrophy and white matter loss in all children. The most common reported genetic variation was c.1981delC; (p.Leu661Serfs*97) observed in two families (likely due to a founder effect). CONCLUSIONS Pathogenic variants in NRROS should be suspected in children with neuro-regression and drug-resistant epilepsy including ISS with onset in the first two years of life. Punctate or serpiginous calcifications at the grey-white matter junction and acquired microcephaly are further clues towards the diagnosis.
Collapse
Affiliation(s)
- Priyanka Madaan
- Pediatric Neurology Unit, Department of Pediatrics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Yashovardhan Kaushal
- Pediatric Neurology Unit, Department of Pediatrics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | | | - Yanick J Crow
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK; Laboratory of Neurogenetics and Neuroinflammation, Institut Imagine, Université de Paris, Paris, France
| | - John H Livingston
- Department of Paediatric Neurology, Leeds Teaching Hospitals, Leeds, UK
| | - Chirag Ahuja
- Department of Radiodiagnosis and Imaging (Section of Neuroimaging and Interventional Radiology), PGIMER, Chandigarh, India
| | - Naveen Sankhyan
- Pediatric Neurology Unit, Department of Pediatrics, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India.
| |
Collapse
|
11
|
Ferrer I. The Primary Microglial Leukodystrophies: A Review. Int J Mol Sci 2022; 23:ijms23116341. [PMID: 35683020 PMCID: PMC9181167 DOI: 10.3390/ijms23116341] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/03/2022] [Indexed: 11/17/2022] Open
Abstract
Primary microglial leukodystrophy or leukoencephalopathy are disorders in which a genetic defect linked to microglia causes cerebral white matter damage. Pigmented orthochromatic leukodystrophy, adult-onset orthochromatic leukodystrophy associated with pigmented macrophages, hereditary diffuse leukoencephalopathy with (axonal) spheroids, and adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP) are different terms apparently used to designate the same disease. However, ALSP linked to dominantly inherited mutations in CSF1R (colony stimulating factor receptor 1) cause CSF-1R-related leukoencephalopathy (CRP). Yet, recessive ALSP with ovarian failure linked to AARS2 (alanyl-transfer (t)RNA synthase 2) mutations (LKENP) is a mitochondrial disease and not a primary microglial leukoencephalopathy. Polycystic membranous lipomembranous osteodysplasia with sclerosing leukoencephalopathy (PLOSL; Nasu–Hakola disease: NHD) is a systemic disease affecting bones, cerebral white matter, selected grey nuclei, and adipose tissue The disease is caused by mutations of one of the two genes TYROBP or TREM2, identified as PLOSL1 and PLOSL2, respectively. TYROBP associates with receptors expressed in NK cells, B and T lymphocytes, dendritic cells, monocytes, macrophages, and microglia. TREM2 encodes the protein TREM2 (triggering receptor expressed on myeloid cells 2), which forms a receptor signalling complex with TYROBP in macrophages and dendritic cells. Rather than pure microglial leukoencephalopathy, NHD can be considered a multisystemic “immunological” disease.
Collapse
Affiliation(s)
- Isidro Ferrer
- Network Centre of Biomedical Research of Neurodegenerative Diseases (CIBERNED), Department of Pathology and Experimental Therapeutics, Bellvitge Biomedical Research Institute (IDIBELL), University of Barcelona, 08907 Barcelona, L'Hospitalet de Llobregat, Spain
| |
Collapse
|
12
|
Komada M, Nishimura Y. Epigenetics and Neuroinflammation Associated With Neurodevelopmental Disorders: A Microglial Perspective. Front Cell Dev Biol 2022; 10:852752. [PMID: 35646933 PMCID: PMC9133693 DOI: 10.3389/fcell.2022.852752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/22/2022] [Indexed: 12/15/2022] Open
Abstract
Neuroinflammation is a cause of neurodevelopmental disorders such as autism spectrum disorders, fetal alcohol syndrome, and cerebral palsy. Converging lines of evidence from basic and clinical sciences suggest that dysregulation of the epigenetic landscape, including DNA methylation and miRNA expression, is associated with neuroinflammation. Genetic and environmental factors can affect the interaction between epigenetics and neuroinflammation, which may cause neurodevelopmental disorders. In this minireview, we focus on neuroinflammation that might be mediated by epigenetic dysregulation in microglia, and compare studies using mammals and zebrafish.
Collapse
Affiliation(s)
- Munekazu Komada
- Mammalian Embryology, Department of Life Science, Faculty of Science and Engineering, Kindai University, Osaka, Japan
| | - Yuhei Nishimura
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu, Japan
- *Correspondence: Yuhei Nishimura,
| |
Collapse
|
13
|
White matter microglia heterogeneity in the CNS. Acta Neuropathol 2022; 143:125-141. [PMID: 34878590 DOI: 10.1007/s00401-021-02389-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/17/2021] [Accepted: 11/28/2021] [Indexed: 02/07/2023]
Abstract
Microglia, the resident myeloid cells in the central nervous system (CNS) play critical roles in shaping the brain during development, responding to invading pathogens, and clearing tissue debris or aberrant protein aggregations during ageing and neurodegeneration. The original concept that like macrophages, microglia are either damaging (pro-inflammatory) or regenerative (anti-inflammatory) has been updated to a kaleidoscope view of microglia phenotypes reflecting their wide-ranging roles in maintaining homeostasis in the CNS and, their contribution to CNS diseases, as well as aiding repair. The use of new technologies including single cell/nucleus RNA sequencing has led to the identification of many novel microglia states, allowing for a better understanding of their complexity and distinguishing regional variations in the CNS. This has also revealed differences between species and diseases, and between microglia and other myeloid cells in the CNS. However, most of the data on microglia heterogeneity have been generated on cells isolated from the cortex or whole brain, whereas white matter changes and differences between white and grey matter have been relatively understudied. Considering the importance of microglia in regulating white matter health, we provide a brief update on the current knowledge of microglia heterogeneity in the white matter, how microglia are important for the development of the CNS, and how microglial ageing affects CNS white matter homeostasis. We discuss how microglia are intricately linked to the classical white matter diseases such as multiple sclerosis and genetic white matter diseases, and their putative roles in neurodegenerative diseases in which white matter is also affected. Understanding the wide variety of microglial functions in the white matter may provide the basis for microglial targeted therapies for CNS diseases.
Collapse
|
14
|
Lanciotti A, Brignone MS, Macioce P, Visentin S, Ambrosini E. Human iPSC-Derived Astrocytes: A Powerful Tool to Study Primary Astrocyte Dysfunction in the Pathogenesis of Rare Leukodystrophies. Int J Mol Sci 2021; 23:ijms23010274. [PMID: 35008700 PMCID: PMC8745131 DOI: 10.3390/ijms23010274] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 12/11/2022] Open
Abstract
Astrocytes are very versatile cells, endowed with multitasking capacities to ensure brain homeostasis maintenance from brain development to adult life. It has become increasingly evident that astrocytes play a central role in many central nervous system pathologies, not only as regulators of defensive responses against brain insults but also as primary culprits of the disease onset and progression. This is particularly evident in some rare leukodystrophies (LDs) where white matter/myelin deterioration is due to primary astrocyte dysfunctions. Understanding the molecular defects causing these LDs may help clarify astrocyte contribution to myelin formation/maintenance and favor the identification of possible therapeutic targets for LDs and other CNS demyelinating diseases. To date, the pathogenic mechanisms of these LDs are poorly known due to the rarity of the pathological tissue and the failure of the animal models to fully recapitulate the human diseases. Thus, the development of human induced pluripotent stem cells (hiPSC) from patient fibroblasts and their differentiation into astrocytes is a promising approach to overcome these issues. In this review, we discuss the primary role of astrocytes in LD pathogenesis, the experimental models currently available and the advantages, future evolutions, perspectives, and limitations of hiPSC to study pathologies implying astrocyte dysfunctions.
Collapse
Affiliation(s)
- Angela Lanciotti
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy; (A.L.); (M.S.B.); (P.M.)
| | - Maria Stefania Brignone
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy; (A.L.); (M.S.B.); (P.M.)
| | - Pompeo Macioce
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy; (A.L.); (M.S.B.); (P.M.)
| | - Sergio Visentin
- National Center for Research and Preclinical and Clinical Evaluation of Drugs, Istituto Superiore di Sanità, 00169 Rome, Italy;
| | - Elena Ambrosini
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy; (A.L.); (M.S.B.); (P.M.)
- Correspondence: ; Tel.: +39-064-990-2037
| |
Collapse
|
15
|
Bhattacharya A, Choi WWY, Muffat J, Li Y. Modeling Developmental Brain Diseases Using Human Pluripotent Stem Cells-Derived Brain Organoids - Progress and Perspective. J Mol Biol 2021; 434:167386. [PMID: 34883115 DOI: 10.1016/j.jmb.2021.167386] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 02/07/2023]
Abstract
Developmental brain diseases encompass a group of conditions resulting from genetic or environmental perturbations during early development. Despite the increased research attention in recent years following recognition of the prevalence of these diseases, there is still a significant lack of knowledge of their etiology and treatment options. The genetic and clinical heterogeneity of these diseases, in addition to the limitations of experimental animal models, contribute to this difficulty. In this regard, the advent of brain organoid technology has provided a new means to study the cause and progression of developmental brain diseases in vitro. Derived from human pluripotent stem cells, brain organoids have been shown to recapitulate key developmental milestones of the early human brain. Combined with technological advancements in genome editing, tissue engineering, electrophysiology, and multi-omics analysis, brain organoids have expanded the frontiers of human neurobiology, providing valuable insight into the cellular and molecular mechanisms of normal and pathological brain development. This review will summarize the current progress of applying brain organoids to model human developmental brain diseases and discuss the challenges that need to be overcome to further advance their utility.
Collapse
Affiliation(s)
- Afrin Bhattacharya
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; The University of Toronto, Department of Molecular Genetics, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | - Wendy W Y Choi
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; The University of Toronto, Department of Molecular Genetics, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Julien Muffat
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; The University of Toronto, Department of Molecular Genetics, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Program in Neurosciences and Mental Health, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Yun Li
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, 686 Bay Street, Toronto, ON M5G 0A4, Canada; The University of Toronto, Department of Molecular Genetics, 1 King's College Circle, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
16
|
Berdowski WM, Sanderson LE, van Ham TJ. The multicellular interplay of microglia in health and disease: lessons from leukodystrophy. Dis Model Mech 2021; 14:dmm048925. [PMID: 34282843 PMCID: PMC8319551 DOI: 10.1242/dmm.048925] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Microglia are highly dynamic cells crucial for developing and maintaining lifelong brain function and health through their many interactions with essentially all cellular components of the central nervous system. The frequent connection of microglia to leukodystrophies, genetic disorders of the white matter, has highlighted their involvement in the maintenance of white matter integrity. However, the mechanisms that underlie their putative roles in these processes remain largely uncharacterized. Microglia have also been gaining attention as possible therapeutic targets for many neurological conditions, increasing the demand to understand their broad spectrum of functions and the impact of their dysregulation. In this Review, we compare the pathological features of two groups of genetic leukodystrophies: those in which microglial dysfunction holds a central role, termed 'microgliopathies', and those in which lysosomal or peroxisomal defects are considered to be the primary driver. The latter are suspected to have notable microglia involvement, as some affected individuals benefit from microglia-replenishing therapy. Based on overlapping pathology, we discuss multiple ways through which aberrant microglia could lead to white matter defects and brain dysfunction. We propose that the study of leukodystrophies, and their extensively multicellular pathology, will benefit from complementing analyses of human patient material with the examination of cellular dynamics in vivo using animal models, such as zebrafish. Together, this will yield important insight into the cell biological mechanisms of microglial impact in the central nervous system, particularly in the development and maintenance of myelin, that will facilitate the development of new, and refinement of existing, therapeutic options for a range of brain diseases.
Collapse
Affiliation(s)
| | | | - Tjakko J. van Ham
- Department of Clinical Genetics, Erasmus MC University Medical Center, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| |
Collapse
|
17
|
von Jonquieres G, Rae CD, Housley GD. Emerging Concepts in Vector Development for Glial Gene Therapy: Implications for Leukodystrophies. Front Cell Neurosci 2021; 15:661857. [PMID: 34239416 PMCID: PMC8258421 DOI: 10.3389/fncel.2021.661857] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
Central Nervous System (CNS) homeostasis and function rely on intercellular synchronization of metabolic pathways. Developmental and neurochemical imbalances arising from mutations are frequently associated with devastating and often intractable neurological dysfunction. In the absence of pharmacological treatment options, but with knowledge of the genetic cause underlying the pathophysiology, gene therapy holds promise for disease control. Consideration of leukodystrophies provide a case in point; we review cell type – specific expression pattern of the disease – causing genes and reflect on genetic and cellular treatment approaches including ex vivo hematopoietic stem cell gene therapies and in vivo approaches using adeno-associated virus (AAV) vectors. We link recent advances in vectorology to glial targeting directed towards gene therapies for specific leukodystrophies and related developmental or neurometabolic disorders affecting the CNS white matter and frame strategies for therapy development in future.
Collapse
Affiliation(s)
- Georg von Jonquieres
- Translational Neuroscience Facility, Department of Physiology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Caroline D Rae
- Neuroscience Research Australia, Randwick, NSW, Australia
| | - Gary D Housley
- Translational Neuroscience Facility, Department of Physiology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
18
|
Chowen JA, Garcia-Segura LM. Role of glial cells in the generation of sex differences in neurodegenerative diseases and brain aging. Mech Ageing Dev 2021; 196:111473. [PMID: 33766745 DOI: 10.1016/j.mad.2021.111473] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/14/2021] [Accepted: 03/16/2021] [Indexed: 12/11/2022]
Abstract
Diseases and aging-associated alterations of the nervous system often show sex-specific characteristics. Glial cells play a major role in the endogenous homeostatic response of neural tissue, and sex differences in the glial transcriptome and function have been described. Therefore, the possible role of these cells in the generation of sex differences in pathological alterations of the nervous system is reviewed here. Studies have shown that glia react to pathological insults with sex-specific neuroprotective and regenerative effects. At least three factors determine this sex-specific response of glia: sex chromosome genes, gonadal hormones and neuroactive steroid hormone metabolites. The sex chromosome complement determines differences in the transcriptional responses in glia after brain injury, while gonadal hormones and their metabolites activate sex-specific neuroprotective mechanisms in these cells. Since the sex-specific neuroprotective and regenerative activity of glial cells causes sex differences in the pathological alterations of the nervous system, glia may represent a relevant target for sex-specific therapeutic interventions.
Collapse
Affiliation(s)
- Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutriciόn (CIBEROBN), Instituto de Salud Carlos III, and IMDEA Food Institute, CEIUAM+CSIC, Madrid, Spain.
| | - Luis M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC) and Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|