1
|
Tang Y, Zhang L, Huang P, She Z, Luo S, Peng H, Chen Y, Luo J, Duan W, Xiao Y, Liu L, Liu L. Understanding the Intricacies of Cellular Mechanisms in Remyelination: The Role of Circadian Rhythm. Neurochem Int 2025:105929. [PMID: 39756585 DOI: 10.1016/j.neuint.2025.105929] [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: 09/20/2024] [Revised: 12/27/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
The term "circadian rhythm" refers to the 24-hour oscillations found in various physiological processes in organisms, responsible for maintaining bodily homeostasis. Many neurological diseases mainly involve the process of demyelination, and remyelination is crucial for the treatment of neurological diseases. Current research mainly focuses on the key role of circadian clocks in the pathophysiological mechanisms of multiple sclerosis. Various studies have shown that the circadian rhythm regulates various cellular molecular mechanisms and signaling pathways involved in remyelination. The process of remyelination is primarily mediated by oligodendrocyte precursor cells (OPCs), oligodendrocytes, microglia, and astrocytes. OPCs are activated, proliferate, migrate, and ultimately differentiate into oligodendrocytes after demyelination, involving many key signaling pathway and regulatory factors. Activated microglia secretes important cytokines and chemokines, promoting OPC proliferation and differentiation, and phagocytoses myelin debris that inhibits remyelination. Astrocytes play a crucial role in supporting remyelination by secreting signals that promote remyelination or facilitate the phagocytosis of myelin debris by microglia. Additionally, cell-to-cell communication via gap junctions allows for intimate contact between astrocytes and oligodendrocytes, providing metabolic support for oligodendrocytes. Therefore, gaining a deeper understanding of the mechanisms and molecular pathways of the circadian rhythm at various stages of remyelination can help elucidate the fundamental characteristics of remyelination and provide insights into treating demyelinating disorders.
Collapse
Affiliation(s)
- Yufen Tang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Lu Zhang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Peng Huang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Zhou She
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Senlin Luo
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Hong Peng
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Yuqiong Chen
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Jinwen Luo
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Wangxin Duan
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Yangyang Xiao
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Lingjuan Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China.
| | - Liqun Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China.
| |
Collapse
|
2
|
Russo M, Zahaf A, Kassoussi A, Sharif A, Faure H, Traiffort E, Ruat M. Sonic Hedgehog Is an Early Oligodendrocyte Marker During Remyelination. Cells 2024; 13:1808. [PMID: 39513915 PMCID: PMC11545011 DOI: 10.3390/cells13211808] [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: 09/04/2024] [Revised: 10/22/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Failure of myelin regeneration by oligodendrocytes contributes to progressive decline in many neurological diseases. Here, using in vitro and in vivo rodent models, functional blockade, and mouse brain demyelination, we demonstrate that Sonic hedgehog (Shh) expression in a subset of oligodendrocyte progenitor cells precedes the expression of myelin basic protein (MBP), a major myelin sheath protein. Primary cultures of rodent cortical oligodendrocytes show that Shh mRNA and protein are upregulated during oligodendrocyte maturation before the upregulation of MBP expression. Importantly, almost all MBP-positive cells are Shh positive during differentiation. During remyelination, we identify a rapid induction of Shh mRNA and peptide in oligodendroglial cells present in the demyelinated corpus callosum of mice, including a population of PDGFRα-expressing cells. Shh invalidation by an adeno-associated virus strategy demonstrates that the downregulation of Shh impairs the differentiation of oligodendrocytes in vitro and decreases MBP and myelin proteolipid protein expression in the demyelinated mouse brain at late stages of remyelination. We also report a parallel expression of Shh and MBP in oligodendroglial cells during early post-natal myelination of the mouse brain. Thus, we identify a crucial Shh signal involved in oligodendroglial cell differentiation and remyelination, with potential interest in the design of better-targeted remyelinating therapeutic strategies.
Collapse
Affiliation(s)
- Mariagiovanna Russo
- Paris-Saclay University, CNRS, Neuroscience Paris-Saclay Institute, 91400 Saclay, France (A.K.); (H.F.)
| | - Amina Zahaf
- Paris-Saclay University, INSERM, Diseases and Hormones of the Nervous System-U1195, 94276 Le Kremlin-Bicêtre, France; (A.Z.); (E.T.)
| | - Abdelmoumen Kassoussi
- Paris-Saclay University, CNRS, Neuroscience Paris-Saclay Institute, 91400 Saclay, France (A.K.); (H.F.)
- Paris-Saclay University, INSERM, Diseases and Hormones of the Nervous System-U1195, 94276 Le Kremlin-Bicêtre, France; (A.Z.); (E.T.)
| | - Ariane Sharif
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, FHU 1000 Days for Health, INSERM, Université de Lille, CHU Lille, 59000 Lille, France;
| | - Hélène Faure
- Paris-Saclay University, CNRS, Neuroscience Paris-Saclay Institute, 91400 Saclay, France (A.K.); (H.F.)
| | - Elisabeth Traiffort
- Paris-Saclay University, INSERM, Diseases and Hormones of the Nervous System-U1195, 94276 Le Kremlin-Bicêtre, France; (A.Z.); (E.T.)
| | - Martial Ruat
- Paris-Saclay University, CNRS, Neuroscience Paris-Saclay Institute, 91400 Saclay, France (A.K.); (H.F.)
| |
Collapse
|
3
|
Russo M, Pellegrino G, Faure H, Tirou L, Sharif A, Ruat M. Characterization of Sonic Hedgehog transcripts in the adult mouse brain: co-expression with neuronal and oligodendroglial markers. Brain Struct Funct 2024; 229:705-727. [PMID: 38329543 PMCID: PMC10978748 DOI: 10.1007/s00429-023-02756-2] [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: 08/31/2023] [Accepted: 12/29/2023] [Indexed: 02/09/2024]
Abstract
In the adult mammalian brain, astrocytes are proposed to be the major Sonic Hedgehog (Shh)-responsive cells. However, the sources of the Shh molecule mediating activation of the pathway are still poorly characterized. The present work investigates the distribution and phenotype of cells expressing Shh mRNA in the adult mouse brain. Using single-molecule fluorescent in situ hybridization (smfISH), we report much broader expression of Shh transcripts in almost all brain regions than originally reported. We identify Shh mRNA in HuC/D+ neuronal populations, including GABAergic (glutamic acid decarboxylase 67, Gad67), cholinergic (choline acetyltransferase, ChAT), dopaminergic (tyrosine hydroxylase, TH), nitrergic (neuronal nitric oxide synthase, nNOS), and in a small population of oligodendroglial cells expressing Sox10 and Olig2 mRNA transcription factors. Further analysis of Shh mRNA in cerebral cortical and hypothalamic neurons suggests that Shh is also expressed by glutamatergic neurons. Interestingly, we did not observe substantial Desert Hedgehog and Indian Hedgehog mRNA signals, nor Shh signals in S100β+ astrocytes and Iba1+ microglial cells. Collectively, the present work provides the most robust central map of Shh-expressing cells to date and underscores the importance of nitrergic neurons in regulating Shh availability to brain cells. Thus, our study provides a framework for future experiments aimed at better understanding of the functions of Shh signaling in the brain in normal and pathological states, and the characterization of novel regulatory mechanisms of the signaling pathway.
Collapse
Affiliation(s)
- Mariagiovanna Russo
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, 91400, Saclay, France
| | - Giuliana Pellegrino
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, 91400, Saclay, France
| | - Hélène Faure
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, 91400, Saclay, France
| | - Linda Tirou
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, 91400, Saclay, France
| | - Ariane Sharif
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR-S 1172, FHU 1000 Days for Health, Lille, France
| | - Martial Ruat
- CNRS, Paris-Saclay University, UMR-9197, Neuroscience Paris-Saclay Institute, 91400, Saclay, France.
| |
Collapse
|
4
|
Liu X, Adamo AM, Oteiza PI. Marginal Zinc Deficiency during Gestation and Lactation in Rats Affects Oligodendrogenesis, Motor Performance, and Behavior in the Offspring. J Nutr 2023; 153:2778-2796. [PMID: 37648111 DOI: 10.1016/j.tjnut.2023.08.029] [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: 06/24/2023] [Revised: 08/10/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Oligodendrocytes are responsible for myelin production in the central nervous system (CNS). Hypomyelination may slow saltatory nerve signal conduction and affect motor performance and behavior in adults. Gestational marginal zinc deficiency in rats significantly decreases proliferation of neural stem cells (NSCs) in the offspring brain. OBJECTIVES Given that NSCs are precursors of oligodendrocytes, this study investigated if marginal zinc deficiency during early development in rats affects oligodendrogenesis in the offspring's CNS. METHODS Rat dams were fed an adequate (25 μg zinc/g diet) (C) or a marginal zinc diet (MZD) (10 μg zinc/g diet), from gestation day zero until postnatal day (P) 20, and subsequently all offspring was fed the control diet until P60. Oligodendrogenesis was evaluated in the offspring at P2, P5, P10, P20, and P60, by measuring parameters of oligodendrocyte progenitor cells (OPCs) proliferation, differentiation, maturation, and of myelination. RESULTS The expression of 1) proteins that regulate OPC proliferation (Shh, Sox10, Olig2); 2) OPC markers (NG2, PDGFRα); 3) myelin proteins (MBP, MAG, MOG, PLP) were lower in the brain cortex from MZD than C offspring at various stages in development. The amount of myelin after zinc replenishment continued to be low in the MZD young adult at P60. Accordingly, parameters of motor performance and behavior [grip strength, rotarod, elevated T-maze (ETM), and open-field tests] were impaired in the MZD offspring at P60. CONCLUSIONS Results support the concept that maternal and early postnatal exposure to MZD affects oligodendrogenesis causing long-lasting effects on myelination and on motor performance in the young adult offspring.
Collapse
Affiliation(s)
- Xiuzhen Liu
- Department of Nutrition, University of California, Davis, CA, United States; Department of Environmental Toxicology, University of California, Davis, CA, United States
| | - Ana M Adamo
- Departamento de Quimica Biologica, Facultad de Farmacia y Bioquímica, IQUIFIB, Universidad de Buenos Aires-CONICET, Buenos Aires, Argentina.
| | - Patricia I Oteiza
- Department of Nutrition, University of California, Davis, CA, United States; Department of Environmental Toxicology, University of California, Davis, CA, United States.
| |
Collapse
|
5
|
Higashijima T, Shirozu H, Saitsu H, Sonoda M, Fujita A, Masuda H, Yamamoto T, Matsumoto N, Kameyama S. Incomplete hippocampal inversion in patients with mutations in genes involved in sonic hedgehog signaling. Heliyon 2023; 9:e14712. [PMID: 37012904 PMCID: PMC10066535 DOI: 10.1016/j.heliyon.2023.e14712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 03/01/2023] [Accepted: 03/15/2023] [Indexed: 03/28/2023] Open
Abstract
Sonic hedgehog (Shh) signaling pathways are known to play an important role in the morphological development of the hippocampus in vivo, but their actual roles in humans have not been clarified. Hypothalamic hamartoma (HH) is known to be associated with germline or somatic gene mutations of Shh signaling. We hypothesized that patients with HH and mutations of Shh-related genes also show hippocampal maldevelopment and an abnormal hippocampal infolding angle (HIA). We analyzed 45 patients (age: 1-37 years) with HH who underwent stereotactic radiofrequency thermocoagulation and found Shh-related gene mutations in 20 patients. In addition, 44 pediatric patients without HH (age: 2-25 years) who underwent magnetic resonance imaging (MRI) examinations under the same conditions during the same period were included in this study as a control group. HIA evaluated on MRI was compared between patients with gene mutations and the control group. The median HIA at the cerebral peduncle slice in patients with the gene mutation was 74.36° on the left and 76.11° on the right, and these values were significantly smaller than the corresponding values in the control group (80.46° and 80.56°, respectively, p < 0.01). Thus, mutations of Shh-related genes were correlated to incomplete hippocampal inversion. The HIA, particularly at the cerebral peduncle slice, is a potential indicator of abnormalities of the Shh-signaling pathway.
Collapse
|
6
|
Fang M, Tang T, Qiu M, Xu X. Hedgehog Signaling in CNS Remyelination. Cells 2022; 11:cells11142260. [PMID: 35883703 PMCID: PMC9320235 DOI: 10.3390/cells11142260] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/14/2022] [Accepted: 07/20/2022] [Indexed: 12/14/2022] Open
Abstract
Remyelination is a fundamental repair process in the central nervous system (CNS) that is triggered by demyelinating events. In demyelinating diseases, oligodendrocytes (OLs) are targeted, leading to myelin loss, axonal damage, and severe functional impairment. While spontaneous remyelination often fails in the progression of demyelinating diseases, increased understanding of the mechanisms and identification of targets that regulate myelin regeneration becomes crucial. To date, several signaling pathways have been implicated in the remyelination process, including the Hedgehog (Hh) signaling pathway. This review summarizes the current data concerning the complicated roles of the Hh signaling pathway in the context of remyelination. We will highlight the open issues that have to be clarified prior to bringing molecules targeting the Hh signaling to demyelinating therapy.
Collapse
Affiliation(s)
- Minxi Fang
- Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China;
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tao Tang
- Department of Anatomy, Cell Biology & Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA;
| | - Mengsheng Qiu
- Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China;
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
- School of Basic Medicial Sciences, Hangzhou Normal University, Hangzhou 311121, China
- Correspondence: (M.Q.); (X.X.)
| | - Xiaofeng Xu
- Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China;
- Correspondence: (M.Q.); (X.X.)
| |
Collapse
|
7
|
Wang S, Wang Y, Zou S. A Glance at the Molecules That Regulate Oligodendrocyte Myelination. Curr Issues Mol Biol 2022; 44:2194-2216. [PMID: 35678678 PMCID: PMC9164040 DOI: 10.3390/cimb44050149] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 11/16/2022] Open
Abstract
Oligodendrocyte (OL) myelination is a critical process for the neuronal axon function in the central nervous system. After demyelination occurs because of pathophysiology, remyelination makes repairs similar to myelination. Proliferation and differentiation are the two main stages in OL myelination, and most factors commonly play converse roles in these two stages, except for a few factors and signaling pathways, such as OLIG2 (Oligodendrocyte transcription factor 2). Moreover, some OL maturation gene mutations induce hypomyelination or hypermyelination without an obvious function in proliferation and differentiation. Herein, three types of factors regulating myelination are reviewed in sequence.
Collapse
Affiliation(s)
- Shunqi Wang
- Institute of Life Science & School of Life Sciences, Nanchang University, Nanchang 330031, China; (S.W.); (Y.W.)
- School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
| | - Yingxing Wang
- Institute of Life Science & School of Life Sciences, Nanchang University, Nanchang 330031, China; (S.W.); (Y.W.)
| | - Suqi Zou
- Institute of Life Science & School of Life Sciences, Nanchang University, Nanchang 330031, China; (S.W.); (Y.W.)
- School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
- Correspondence:
| |
Collapse
|
8
|
Garcia-Martin G, Alcover-Sanchez B, Wandosell F, Cubelos B. Pathways Involved in Remyelination after Cerebral Ischemia. Curr Neuropharmacol 2022; 20:751-765. [PMID: 34151767 PMCID: PMC9878953 DOI: 10.2174/1570159x19666210610093658] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 05/05/2021] [Accepted: 05/12/2021] [Indexed: 11/22/2022] Open
Abstract
Brain ischemia, also known as ischemic stroke, occurs when there is a lack of blood supply into the brain. When an ischemic insult appears, both neurons and glial cells can react in several ways that will determine the severity and prognosis. This high heterogeneity of responses has been a major obstacle in developing effective treatments or preventive methods for stroke. Although white matter pathophysiology has not been deeply assessed in stroke, its remodelling can greatly influence the clinical outcome and the disability degree. Oligodendrocytes, the unique cell type implied in CNS myelination, are sensible to ischemic damage. Loss of myelin sheaths can compromise axon survival, so new Oligodendrocyte Precursor Cells are required to restore brain function. Stroke can, therefore, enhance oligodendrogenesis to regenerate those new oligodendrocytes that will ensheath the damaged axons. Given that myelination is a highly complex process that requires coordination of multiple pathways such as Sonic Hedgehog, RTKs or Wnt/β-catenin, we will analyse new research highlighting their importance after brain ischemia. In addition, oligodendrocytes are not isolated cells inside the brain, but rather form part of a dynamic environment of interactions between neurons and glial cells. For this reason, we will put some context into how microglia and astrocytes react against stroke and influence oligodendrogenesis to highlight the relevance of remyelination in the ischemic brain. This will help to guide future studies to develop treatments focused on potentiating the ability of the brain to repair the damage.
Collapse
Affiliation(s)
- Gonzalo Garcia-Martin
- Departamento de Biología Molecular and Centro Biología Molecular “Severo Ochoa”, Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
| | - Berta Alcover-Sanchez
- Departamento de Biología Molecular and Centro Biología Molecular “Severo Ochoa”, Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
| | - Francisco Wandosell
- Departamento de Biología Molecular and Centro Biología Molecular “Severo Ochoa”, Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain
| | - Beatriz Cubelos
- Departamento de Biología Molecular and Centro Biología Molecular “Severo Ochoa”, Universidad Autónoma de Madrid-Consejo Superior de Investigaciones Científicas, 28049 Madrid, Spain,Address correspondence to this author at the Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Nicolás Cabrera 1, Universidad Autónoma de Madrid, 28049 Madrid, Spain; Tel: 34-91-1964561; Fax: 34-91-1964420; E-mail:
| |
Collapse
|
9
|
Barak M, Fedorova V, Pospisilova V, Raska J, Vochyanova S, Sedmik J, Hribkova H, Klimova H, Vanova T, Bohaciakova D. Human iPSC-Derived Neural Models for Studying Alzheimer's Disease: from Neural Stem Cells to Cerebral Organoids. Stem Cell Rev Rep 2022; 18:792-820. [PMID: 35107767 PMCID: PMC8930932 DOI: 10.1007/s12015-021-10254-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2021] [Indexed: 12/05/2022]
Abstract
During the past two decades, induced pluripotent stem cells (iPSCs) have been widely used to study mechanisms of human neural development, disease modeling, and drug discovery in vitro. Especially in the field of Alzheimer’s disease (AD), where this treatment is lacking, tremendous effort has been put into the investigation of molecular mechanisms behind this disease using induced pluripotent stem cell-based models. Numerous of these studies have found either novel regulatory mechanisms that could be exploited to develop relevant drugs for AD treatment or have already tested small molecules on in vitro cultures, directly demonstrating their effect on amelioration of AD-associated pathology. This review thus summarizes currently used differentiation strategies of induced pluripotent stem cells towards neuronal and glial cell types and cerebral organoids and their utilization in modeling AD and potential drug discovery.
Collapse
Affiliation(s)
- Martin Barak
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Veronika Fedorova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Veronika Pospisilova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Jan Raska
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Simona Vochyanova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Jiri Sedmik
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
- International Clinical Research Center, St. Anne's Faculty Hospital Brno, Brno, Czech Republic
| | - Hana Hribkova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Hana Klimova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Tereza Vanova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
- International Clinical Research Center, St. Anne's Faculty Hospital Brno, Brno, Czech Republic
| | - Dasa Bohaciakova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic.
- International Clinical Research Center, St. Anne's Faculty Hospital Brno, Brno, Czech Republic.
| |
Collapse
|
10
|
Antonelli F, Casciati A, Belles M, Serra N, Linares-Vidal MV, Marino C, Mancuso M, Pazzaglia S. Long-Term Effects of Ionizing Radiation on the Hippocampus: Linking Effects of the Sonic Hedgehog Pathway Activation with Radiation Response. Int J Mol Sci 2021; 22:ijms222212605. [PMID: 34830484 PMCID: PMC8624704 DOI: 10.3390/ijms222212605] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/04/2021] [Accepted: 11/17/2021] [Indexed: 12/29/2022] Open
Abstract
Radiation therapy represents one of the primary treatment modalities for primary and metastatic brain tumors. Although recent advances in radiation techniques, that allow the delivery of higher radiation doses to the target volume, reduce the toxicity to normal tissues, long-term neurocognitive decline is still a detrimental factor significantly affecting quality of life, particularly in pediatric patients. This imposes the need for the development of prevention strategies. Based on recent evidence, showing that manipulation of the Shh pathway carries therapeutic potential for brain repair and functional recovery after injury, here we evaluate how radiation-induced hippocampal alterations are modulated by the constitutive activation of the Shh signaling pathway in Patched 1 heterozygous mice (Ptch1+/-). Our results show, for the first time, an overall protective effect of constitutive Shh pathway activation on hippocampal radiation injury. This activation, through modulation of the proneural gene network, leads to a long-term reduction of hippocampal deficits in the stem cell and new neuron compartments and to the mitigation of radio-induced astrogliosis, despite some behavioral alterations still being detected in Ptch1+/- mice. A better understanding of the pathogenic mechanisms responsible for the neural decline following irradiation is essential for identifying prevention measures to contain the harmful consequences of irradiation. Our data have important translational implications as they suggest a role for Shh pathway manipulation to provide the therapeutic possibility of improving brain repair and functional recovery after radio-induced injury.
Collapse
Affiliation(s)
- Francesca Antonelli
- Division of Health Protection Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (A.C.); (C.M.); (M.M.)
- Correspondence: (F.A.); (S.P.)
| | - Arianna Casciati
- Division of Health Protection Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (A.C.); (C.M.); (M.M.)
| | - Montserrat Belles
- Physiology Unit, School of Medicine, Rovira I Virgili University (URV), 43007 Reus, Spain; (M.B.); (N.S.); (M.V.L.-V.)
| | - Noemi Serra
- Physiology Unit, School of Medicine, Rovira I Virgili University (URV), 43007 Reus, Spain; (M.B.); (N.S.); (M.V.L.-V.)
| | - Maria Victoria Linares-Vidal
- Physiology Unit, School of Medicine, Rovira I Virgili University (URV), 43007 Reus, Spain; (M.B.); (N.S.); (M.V.L.-V.)
| | - Carmela Marino
- Division of Health Protection Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (A.C.); (C.M.); (M.M.)
| | - Mariateresa Mancuso
- Division of Health Protection Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (A.C.); (C.M.); (M.M.)
| | - Simonetta Pazzaglia
- Division of Health Protection Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development (ENEA), 00123 Rome, Italy; (A.C.); (C.M.); (M.M.)
- Correspondence: (F.A.); (S.P.)
| |
Collapse
|
11
|
Niedzwiedz-Massey VM, Douglas JC, Rafferty T, Kane CJ, Drew PD. Ethanol effects on cerebellar myelination in a postnatal mouse model of fetal alcohol spectrum disorders. Alcohol 2021; 96:43-53. [PMID: 34358666 DOI: 10.1016/j.alcohol.2021.07.003] [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: 03/10/2021] [Revised: 06/12/2021] [Accepted: 07/27/2021] [Indexed: 10/20/2022]
Abstract
Fetal alcohol spectrum disorders (FASD) are alarmingly common, result in significant personal and societal loss, and there are no effective treatments for these disorders. Cerebellar neuropathology is common in FASD and can cause impaired cognitive and motor function. The current study evaluates the effects of ethanol on oligodendrocyte-lineage cells, as well as molecules that modulate oligodendrocyte differentiation and function in the cerebellum in a postnatal mouse model of FASD. Neonatal mice were treated with ethanol from P4-P9 (postnatal day), the cerebellum was isolated at P10, and mRNAs encoding oligodendrocyte-associated molecules were quantitated by qRT-PCR. Our studies demonstrated that ethanol significantly reduced the expression of markers for multiple stages of oligodendrocyte maturation, including oligodendrocyte precursor cells, pre-myelinating oligodendrocytes, and mature myelinating oligodendrocytes. Additionally, we determined that ethanol significantly decreased the expression of molecules that play critical roles in oligodendrocyte differentiation. Interestingly, we also observed that ethanol significantly reduced the expression of myelin-associated inhibitors, which may act as a compensatory mechanism to ethanol toxicity. Furthermore, we demonstrate that ethanol alters the expression of a variety of molecules important in oligodendrocyte function and myelination. Collectively, our studies increase our understanding of specific mechanisms by which ethanol modulates myelination in the developing cerebellum, and potentially identify novel targets for FASD therapy.
Collapse
|
12
|
Lucas TA, Zhu L, Buckwalter MS. Spleen glia are a transcriptionally unique glial subtype interposed between immune cells and sympathetic axons. Glia 2021; 69:1799-1815. [DOI: 10.1002/glia.23993] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 02/24/2021] [Accepted: 03/02/2021] [Indexed: 12/11/2022]
Affiliation(s)
- Tawaun A. Lucas
- Department of Neurology and Neurological Sciences Stanford School of Medicine Stanford California USA
| | - Li Zhu
- Department of Neurology and Neurological Sciences Stanford School of Medicine Stanford California USA
| | - Marion S. Buckwalter
- Department of Neurology and Neurological Sciences Stanford School of Medicine Stanford California USA
- Department of Neurosurgery Stanford School of Medicine Stanford California USA
| |
Collapse
|
13
|
Laouarem Y, Kassoussi A, Zahaf A, Hutteau-Hamel T, Mellouk A, Bobé P, Mattern C, Schumacher M, Traiffort E. Functional cooperation of the hedgehog and androgen signaling pathways during developmental and repairing myelination. Glia 2021; 69:1369-1392. [PMID: 33484204 DOI: 10.1002/glia.23967] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/21/2022]
Abstract
Hedgehog morphogens control fundamental cellular processes during tissue development and regeneration. In the central nervous system (CNS), Hedgehog signaling has been implicated in oligodendrocyte and myelin production, where it functions in a concerted manner with other pathways. Since androgen receptor (AR) plays a key role in establishing the sexual phenotype of myelin during development and is required for spontaneous myelin regeneration in the adult CNS, we hypothesized the existence of a possible coordination between Hedgehog and androgen signals in oligodendrocyte and myelin production. Here, we report complementary activities of both pathways during early postnatal oligodendrogenesis further revealing that persistent Hedgehog signaling activation impedes myelin production. The data also uncover prominent pro-myelinating activity of testosterone and involvement of AR in the control of neural stem cell commitment toward the oligodendroglial lineage. In the context of CNS demyelination, we provide evidence for the functional cooperation of the pathways leading to acceleration of myelin regeneration that might be related to their respective role on microglial and astroglial responses, higher preservation of axonal integrity, lower neuroinflammation, and functional improvement of animals in an immune model of CNS demyelination. Strong decreases of deleterious cytokines in the CNS (GM-CSF, TNF-α, IL-17A) and spleen (IL-2, IFN-γ) stand as unique features of the combined drugs while the potent therapeutic activity of testosterone on peripheral immune cells contributes to increase tolerogenic CD11c+ dendritic cells, reduce the clonal expansion of conventional CD4+ T cells and increase CD4+ Foxp3+ regulatory T cells. Altogether, these data might open promising perspectives for demyelinating diseases.
Collapse
Affiliation(s)
- Yousra Laouarem
- U1195 Inserm, University Paris-Saclay, Kremlin-Bicêtre, France
| | | | - Amina Zahaf
- U1195 Inserm, University Paris-Saclay, Kremlin-Bicêtre, France
| | | | - Amine Mellouk
- UMR996 Inserm, University Paris-Saclay, Clamart, France
| | - Pierre Bobé
- UMR996 Inserm, University Paris-Saclay, Clamart, France
| | - Claudia Mattern
- M et P Pharma AG, Emmetten, Switzerland.,Oceanographic Center, Nova Southeastern University, Fort Lauderdal, Florida, USA
| | | | | |
Collapse
|
14
|
Malik P, Muthusamy K, Mankad K, Shroff M, Sudhakar S. Solving the hypomyelination conundrum - Imaging perspectives. Eur J Paediatr Neurol 2020; 27:9-24. [PMID: 32418752 DOI: 10.1016/j.ejpn.2020.04.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 03/25/2020] [Accepted: 04/14/2020] [Indexed: 11/26/2022]
Abstract
Hypomyelinating Leukodystrophies (HLDs) are a genetically heterogeneous, clinically overlapping group of disorders with the unifying MR imaging appearance of myelin deficit in the brain. In fact, it is the MRI phenotype that typically raises the diagnostic suspicion in this single largest group of undiagnosed leukodystrophies and guides gene testing for confirmation. This article reviews the neurobiology of myelination, focussing on the complex interplay of molecular genetic pathways and presents a practical clinico-radiological diagnostic algorithm based on the neuroimaging patterns of the common hypomyelinating disorders. The authors also address the current controversies about the definition and use of the term 'hypomyelination'.
Collapse
|
15
|
Tirou L, Russo M, Faure H, Pellegrino G, Sharif A, Ruat M. C9C5 positive mature oligodendrocytes are a source of Sonic Hedgehog in the mouse brain. PLoS One 2020; 15:e0229362. [PMID: 32078657 PMCID: PMC7032736 DOI: 10.1371/journal.pone.0229362] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 02/04/2020] [Indexed: 12/12/2022] Open
Abstract
In the mature rodent brain, Sonic Hedgehog (Shh) signaling regulates stem and progenitor cell maintenance, neuronal and glial circuitry and brain repair. However, the sources and distribution of Shh mediating these effects are still poorly characterized. Here, we report in the adult mouse brain, a broad expression pattern of Shh recognized by the specific monoclonal C9C5 antibody in a subset (11–12%) of CC1+ mature oligodendrocytes that do not express carbonic anhydrase II. These cells express also Olig2 and Sox10, two oligodendrocyte lineage-specific markers, but not PDGFRα, a marker of oligodendrocyte progenitors. In agreement with oligodendroglial cells being a source of Shh in the adult mouse brain, we identify Shh transcripts by single molecule fluorescent in situ hybridization in a subset of cells expressing Olig2 and Sox10 mRNAs. These findings also reveal that Shh expression is more extensive than originally reported. The Shh-C9C5-associated signal labels the oligodendroglial cell body and decorates by intense puncta the processes. C9C5+ cells are distributed in a grid-like manner. They constitute small units that could deliver locally Shh to its receptor Patched expressed in GFAP+ and S100β+ astrocytes, and in HuC/D+ neurons as shown in PtcLacZ/+ reporter mice. Postnatally, C9C5 immunoreactivity overlaps the myelination peak that occurs between P10 and P20 and is down regulated during ageing. Thus, our data suggest that C9C5+CC1+ oligodendroglial cells are a source of Shh in the mouse postnatal brain.
Collapse
Affiliation(s)
- Linda Tirou
- UMR-9197, Neuroscience Paris-Saclay Institute, CNRS, Paris Saclay University, Gif-sur-Yvette, France
| | - Mariagiovanna Russo
- UMR-9197, Neuroscience Paris-Saclay Institute, CNRS, Paris Saclay University, Gif-sur-Yvette, France
| | - Helene Faure
- UMR-9197, Neuroscience Paris-Saclay Institute, CNRS, Paris Saclay University, Gif-sur-Yvette, France
| | - Giuliana Pellegrino
- UMR-9197, Neuroscience Paris-Saclay Institute, CNRS, Paris Saclay University, Gif-sur-Yvette, France
| | | | - Martial Ruat
- UMR-9197, Neuroscience Paris-Saclay Institute, CNRS, Paris Saclay University, Gif-sur-Yvette, France
- * E-mail:
| |
Collapse
|
16
|
Aberrant Oligodendrogenesis in Down Syndrome: Shift in Gliogenesis? Cells 2019; 8:cells8121591. [PMID: 31817891 PMCID: PMC6953000 DOI: 10.3390/cells8121591] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/28/2019] [Accepted: 12/04/2019] [Indexed: 12/25/2022] Open
Abstract
Down syndrome (DS), or trisomy 21, is the most prevalent chromosomal anomaly accounting for cognitive impairment and intellectual disability (ID). Neuropathological changes of DS brains are characterized by a reduction in the number of neurons and oligodendrocytes, accompanied by hypomyelination and astrogliosis. Recent studies mainly focused on neuronal development in DS, but underestimated the role of glial cells as pathogenic players. Aberrant or impaired differentiation within the oligodendroglial lineage and altered white matter functionality are thought to contribute to central nervous system (CNS) malformations. Given that white matter, comprised of oligodendrocytes and their myelin sheaths, is vital for higher brain function, gathering knowledge about pathways and modulators challenging oligodendrogenesis and cell lineages within DS is essential. This review article discusses to what degree DS-related effects on oligodendroglial cells have been described and presents collected evidence regarding induced cell-fate switches, thereby resulting in an enhanced generation of astrocytes. Moreover, alterations in white matter formation observed in mouse and human post-mortem brains are described. Finally, the rationale for a better understanding of pathways and modulators responsible for the glial cell imbalance as a possible source for future therapeutic interventions is given based on current experience on pro-oligodendroglial treatment approaches developed for demyelinating diseases, such as multiple sclerosis.
Collapse
|
17
|
Xu X, Yu Q, Fang M, Yi M, Yang A, Xie B, Yang J, Zhang Z, Dai Z, Qiu M. Stage-specific regulation of oligodendrocyte development by Hedgehog signaling in the spinal cord. Glia 2019; 68:422-434. [PMID: 31605511 DOI: 10.1002/glia.23729] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 08/16/2019] [Accepted: 09/13/2019] [Indexed: 01/31/2023]
Abstract
Elucidation of signaling pathways that control oligodendrocyte (OL) development is a prerequisite for developing novel strategies for myelin repair in neurological diseases. Despite the extensive work outlining the importance of Hedgehog (Hh) signaling in the commitment and generation of OL progenitor cells (OPCs), there are conflicting reports on the role of Hh signaling in regulating OL differentiation and maturation. In the present study, we systematically investigated OPC specification and differentiation in genetically modified mouse models of Smoothened (Smo), an essential component of the Hh signaling pathway in vertebrates. Through conditional gain-of-function strategy, we demonstrated that hyperactivation of Smo in neural progenitors induced transient ectopic OPC generation and precocious OL differentiation accompanied by the co-induction of Olig2 and Nkx2.2. After the commitment of OL lineage, Smo activity is not required for OL differentiation, and sustained expression of Smo in OPCs stimulated cell proliferation but inhibited terminal differentiation. These findings have uncovered the stage-specific regulation of OL development by Smo-mediated Hh signaling, providing novel insights into the molecular regulation of OL differentiation and myelin repair.
Collapse
Affiliation(s)
- Xiaofeng Xu
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Qian Yu
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Minxi Fang
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Min Yi
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Aifen Yang
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Binghua Xie
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Junlin Yang
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Zunyi Zhang
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Zhongmin Dai
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Mengsheng Qiu
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China.,Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, Kentucky
| |
Collapse
|
18
|
Scheuer T, Klein LS, Bührer C, Endesfelder S, Schmitz T. Transient Improvement of Cerebellar Oligodendroglial Development in a Neonatal Hyperoxia Model by PDGFA Treatment. Dev Neurobiol 2019; 79:222-235. [PMID: 30674088 DOI: 10.1002/dneu.22667] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/10/2019] [Accepted: 01/12/2019] [Indexed: 12/13/2022]
Abstract
In preterm infants, the changes from fetal life to ex-utero conditions often coincide with reduced growth and white matter damage of the cerebellum. The premature increase in arterial oxygen tension caused by preterm birth may dysregulate cerebellar development. In a hyperoxia rat model of white matter damage to mimic a steep increase in oxygen levels by 24 h exposure to 80% O2 from postnatal day 6 (P6) to day 7, we analyzed growth factor (GF) synthesis of cerebellar astrocytes. Determination of GF production was performed in astrocytes after Magnetic-activated cell sorting (MACS) isolation from cerebelli after hyperoxia exposure ex vivo, and also in astroglial cultures. Oligodendrocyte progenitor cell (OPC) function was analyzed in cerebellar OPCs isolated by MACS after hyperoxia. Administration of PDGFA from P6 to P11, during hyperoxia and during 4 days recovery, was finally tested for protection of oligodendroglia and myelination. As a result, expression of the GFs Pdgfa, Fgf2, and Bdnf was diminished in cerebellar astrocytes in vitro and in vivo. Gene expression of Olig1, Olig2, Sox9, Sox10, and Cnp was reduced in OPCs in vivo. Nasal PDGFA application improved oligodendroglial proliferation after hyperoxia at P7. However, this treatment effect vanished until P9. Impaired MBP expression after hyperoxia was attenuated by PDGFA treatment until P11, but not beyond when PDGFA supply was stopped. In this study on neonatal cerebellar injury, it is documented for the first time that improvement of oligodendroglial proliferation and of myelination can be achieved by PDGFA treatment. However, the treatment benefit is not maintained long term.
Collapse
Affiliation(s)
- Till Scheuer
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| | - Luisa Sophie Klein
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| | - Christoph Bührer
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| | | | - Thomas Schmitz
- Department for Neonatology, Charité University Medical Center, Berlin, Germany
| |
Collapse
|