1
|
Demmings MD, da Silva Chagas L, Traetta ME, Rodrigues RS, Acutain MF, Barykin E, Datusalia AK, German-Castelan L, Mattera VS, Mazengenya P, Skoug C, Umemori H. (Re)building the nervous system: A review of neuron-glia interactions from development to disease. J Neurochem 2025; 169:e16258. [PMID: 39680483 DOI: 10.1111/jnc.16258] [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: 08/13/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 12/18/2024]
Abstract
Neuron-glia interactions are fundamental to the development and function of the nervous system. During development, glia, including astrocytes, microglia, and oligodendrocytes, influence neuronal differentiation and migration, synapse formation and refinement, and myelination. In the mature brain, glia are crucial for maintaining neural homeostasis, modulating synaptic activity, and supporting metabolic functions. Neurons, inherently vulnerable to various stressors, rely on glia for protection and repair. However, glia, in their reactive state, can also promote neuronal damage, which contributes to neurodegenerative and neuropsychiatric diseases. Understanding the dual role of glia-as both protectors and potential aggressors-sheds light on their complex contributions to disease etiology and pathology. By appropriately modulating glial activity, it may be possible to mitigate neurodegeneration and restore neuronal function. In this review, which originated from the International Society for Neurochemistry (ISN) Advanced School in 2019 held in Montreal, Canada, we first describe the critical importance of glia in the development and maintenance of a healthy nervous system as well as their contributions to neuronal damage and neurological disorders. We then discuss potential strategies to modulate glial activity during disease to protect and promote a properly functioning nervous system. We propose that targeting glial cells presents a promising therapeutic avenue for rebuilding the nervous system.
Collapse
Affiliation(s)
- Matthew D Demmings
- Neuroscience Program, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Luana da Silva Chagas
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil
| | - Marianela E Traetta
- Instituto de Biología Celular y Neurociencia (IBCN), Facultad de Medicina, Conicet, Buenos Aires, Argentina
| | - Rui S Rodrigues
- University of Bordeaux, INSERM, Neurocentre Magendie U1215, Bordeaux, France
| | - Maria Florencia Acutain
- Instituto de Biología Celular y Neurociencia (IBCN), Facultad de Medicina, Conicet, Buenos Aires, Argentina
| | - Evgeny Barykin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Ashok Kumar Datusalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER Raebareli), Raebareli, UP, India
| | - Liliana German-Castelan
- Neuroscience Program, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Vanesa S Mattera
- Instituto de Química y Fisicoquímica Biológica (IQUIFIB-FFyB-UBA), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Pedzisai Mazengenya
- Center of Medical and bio-Allied Health Sciences Research, College of Medicine, Ajman University, Ajman, United Arab Emirates
| | - Cecilia Skoug
- Department of Neuroscience, Physiology & Pharmacology, Centre for Cardiovascular and Metabolic Neuroscience, University College London, London, UK
| | - Hisashi Umemori
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
2
|
Al‐Kharboosh R, Bechtle A, Tzeng SY, Zheng J, Mondal SK, Wilson DR, Perez‐Vega C, Green JJ, Quiñones‐Hinojosa A. Therapeutic potential and impact of nanoengineered patient-derived mesenchymal stem cells in a murine resection and recurrence model of human glioblastoma. Bioeng Transl Med 2024; 9:e10675. [PMID: 39545093 PMCID: PMC11558202 DOI: 10.1002/btm2.10675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 11/17/2024] Open
Abstract
Confounding results of engineered mesenchymal stem cells (MSCs) used as cellular vehicles has plagued technologies whereby success or failure of novel approaches may be dismissed or inaccurately ascribed solely to the biotechnology platform rather than suitability of the human donor. Polymeric materials were screened for non-viral engineering of MSCs from multiple human donors to deliver bone morphogenic protein-4 (BMP4), a protein previously investigated in clinical trials for glioblastoma (GBM) to combat a subpopulation of highly invasive and tumorigenic clones. A "smart technology" that target the migratory and stem-like nature of GBM will require: (1) a cellular vehicle (MSC) which can scavenge and target residual cells left behind after surgical debulking and deliver; (2) anti-glioma cargo (BMP4). Multiple MSC donors are safely engineered, though varied in susceptibility to accept BMP4 due to intrinsic characteristics revealed by their molecular signatures. Efficiency is compared via secretion, downstream signaling, differentiation, and anti-proliferative properties across all donors. In a clinically relevant resection and recurrence model of patient-derived human GBM, we demonstrate that nanoengineered MSCs are not "donor agnostic" and efficacy is influenced by the inherent suitability of the MSC to the cargo. Therefore, donor profiles hold greater influence in determining downstream outcomes than the technical capabilities of the engineering technology.
Collapse
Affiliation(s)
- Rawan Al‐Kharboosh
- Department of NeurosurgeryMayo ClinicJacksonvilleFloridaUSA
- Department of NeuroscienceMayo Clinic Graduate SchoolJacksonvilleFloridaUSA
- AtPoint tx Co.WashingtonDistrict of ColumbiaUSA
| | - Alex Bechtle
- Department of NeurosurgeryMayo ClinicJacksonvilleFloridaUSA
| | - Stephany Y. Tzeng
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Johns Hopkins Translational Immuno Engineering Center, Translational Tissue Engineering Center, and Institute for NanobiotechnologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Jiaying Zheng
- Department of NeuroscienceMayo Clinic Graduate SchoolJacksonvilleFloridaUSA
| | | | - David R. Wilson
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Johns Hopkins Translational Immuno Engineering Center, Translational Tissue Engineering Center, and Institute for NanobiotechnologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | | | - Jordan J. Green
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Johns Hopkins Translational Immuno Engineering Center, Translational Tissue Engineering Center, and Institute for NanobiotechnologyJohns Hopkins UniversityBaltimoreMarylandUSA
- Departments of Neurosurgery, Oncology, Ophthalmology, Materials Science & Engineering, and Chemical & Biomolecular EngineeringJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | | |
Collapse
|
3
|
Chen B, Fan P, Song X, Duan M. The role and possible mechanism of the ferroptosis-related SLC7A11/GSH/GPX4 pathway in myocardial ischemia-reperfusion injury. BMC Cardiovasc Disord 2024; 24:531. [PMID: 39354361 PMCID: PMC11445876 DOI: 10.1186/s12872-024-04220-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 09/23/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Myocardial ischemia-reperfusion injury (MI/RI) is an unavoidable risk event for acute myocardial infarction, with ferroptosis showing close involvement. We investigated the mechanism of MI/RI inducing myocardial injury by inhibiting the ferroptosis-related SLC7A11/glutathione (GSH)/glutathione peroxidase 4 (GPX4) pathway and activating mitophagy. METHODS A rat MI/RI model was established, with myocardial infarction area and injury assessed by TTC and H&E staining. Rat cardiomyocytes H9C2 were cultured in vitro, followed by hypoxia/reoxygenation (H/R) modeling and the ferroptosis inhibitor lipoxstatin-1 (Lip-1) treatment, or 3-Methyladenine or rapamycin treatment and overexpression plasmid (oe-SLC7A11) transfection during modeling. Cell viability and death were evaluated by CCK-8 and LDH assays. Mitochondrial morphology was observed by transmission electron microscopy. Mitochondrial membrane potential was detected by fluorescence dye JC-1. Levels of inflammatory factors, reactive oxygen species (ROS), Fe2+, malondialdehyde, lipid peroxidation, GPX4 enzyme activity, glutathione reductase, GSH and glutathione disulfide, and SLC7A11, GPX4, LC3II/I and p62 proteins were determined by ELISA kit, related indicator detection kits and Western blot. RESULTS The ferroptosis-related SLC7A11/GSH/GPX4 pathway was repressed in MI/RI rat myocardial tissues, inducing myocardial injury. H/R affected GSH synthesis and inhibited GPX4 enzyme activity by down-regulating SLC7A11, thus promoting ferroptosis in cardiomyocytes, which was averted by Lip-1. SLC7A11 overexpression improved H/R-induced cardiomyocyte ferroptosis via the GSH/GPX4 pathway. H/R activated mitophagy in cardiomyocytes. Mitophagy inhibition reversed H/R-induced cellular ferroptosis. Mitophagy activation partially averted SLC7A11 overexpression-improved H/R-induced cardiomyocyte ferroptosis. H/R suppressed the ferroptosis-related SLC7A11/GSH/GPX4 pathway by inducing mitophagy, leading to cardiomyocyte injury. CONCLUSIONS Increased ROS under H/R conditions triggered cardiomyocyte injury by inducing mitophagy to suppress the ferroptosis-related SLC7A11/GSH/GPX4 signaling pathway activation.
Collapse
Affiliation(s)
- Bingxin Chen
- Department of Cardiac Function, The First Affiliated Hospital of Xinjiang Medical University, No. 137 Liyushan South Road, High-tech District, Urumqi, Xinjiang Uygur Autonomous Region, 830054, China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, Xinjiang Uygur Autonomous Region, China
| | - Ping Fan
- Department of Cardiac Function, The First Affiliated Hospital of Xinjiang Medical University, No. 137 Liyushan South Road, High-tech District, Urumqi, Xinjiang Uygur Autonomous Region, 830054, China
| | - Xue Song
- Department of Cardiac Function, The First Affiliated Hospital of Xinjiang Medical University, No. 137 Liyushan South Road, High-tech District, Urumqi, Xinjiang Uygur Autonomous Region, 830054, China
| | - Mingjun Duan
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Animal Experimental Center of Xinjiang Medical University, No. 137 Liyushan South Road, High-tech District, Urumqi, Xinjiang Uygur Autonomous Region, 830000, China.
| |
Collapse
|
4
|
Emery B, Wood TL. Regulators of Oligodendrocyte Differentiation. Cold Spring Harb Perspect Biol 2024; 16:a041358. [PMID: 38503504 PMCID: PMC11146316 DOI: 10.1101/cshperspect.a041358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Myelination has evolved as a mechanism to ensure fast and efficient propagation of nerve impulses along axons. Within the central nervous system (CNS), myelination is carried out by highly specialized glial cells, oligodendrocytes. The formation of myelin is a prolonged aspect of CNS development that occurs well into adulthood in humans, continuing throughout life in response to injury or as a component of neuroplasticity. The timing of myelination is tightly tied to the generation of oligodendrocytes through the differentiation of their committed progenitors, oligodendrocyte precursor cells (OPCs), which reside throughout the developing and adult CNS. In this article, we summarize our current understanding of some of the signals and pathways that regulate the differentiation of OPCs, and thus the myelination of CNS axons.
Collapse
Affiliation(s)
- Ben Emery
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Teresa L Wood
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103, USA
| |
Collapse
|
5
|
Clémot M, D’Alterio C, Kwang AC, Jones DL. mTORC1 is required for differentiation of germline stem cells in the Drosophila melanogaster testis. PLoS One 2024; 19:e0300337. [PMID: 38512882 PMCID: PMC10956854 DOI: 10.1371/journal.pone.0300337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 02/26/2024] [Indexed: 03/23/2024] Open
Abstract
Metabolism participates in the control of stem cell function and subsequent maintenance of tissue homeostasis. How this is achieved in the context of adult stem cell niches in coordination with other local and intrinsic signaling cues is not completely understood. The Target of Rapamycin (TOR) pathway is a master regulator of metabolism and plays essential roles in stem cell maintenance and differentiation. In the Drosophila male germline, mTORC1 is active in germline stem cells (GSCs) and early germ cells. Targeted RNAi-mediated downregulation of mTor in early germ cells causes a block and/or a delay in differentiation, resulting in an accumulation of germ cells with GSC-like features. These early germ cells also contain unusually large and dysfunctional autolysosomes. In addition, downregulation of mTor in adult male GSCs and early germ cells causes non-autonomous activation of mTORC1 in neighboring cyst cells, which correlates with a disruption in the coordination of germline and somatic differentiation. Our study identifies a previously uncharacterized role of the TOR pathway in regulating male germline differentiation.
Collapse
Affiliation(s)
- Marie Clémot
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, United States of America
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, United States of America
| | - Cecilia D’Alterio
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, United States of America
| | - Alexa C. Kwang
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, United States of America
| | - D. Leanne Jones
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, United States of America
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, United States of America
- Departments of Anatomy, Division of Geriatrics, University of California, San Francisco, San Francisco, CA, United States of America
- Departments of Medicine, Division of Geriatrics, University of California, San Francisco, San Francisco, CA, United States of America
- Eli and Edythe Broad Center for Regeneration Medicine, University of California, San Francisco, San Francisco, CA, United States of America
| |
Collapse
|
6
|
Zhang H, Yang Y, Zhang J, Huang L, Niu Y, Chen H, Liu Q, Wang R. Oligodendrocytes Play a Critical Role in White Matter Damage of Vascular Dementia. Neuroscience 2024; 538:1-10. [PMID: 37913862 DOI: 10.1016/j.neuroscience.2023.10.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/17/2023] [Accepted: 10/21/2023] [Indexed: 11/03/2023]
Abstract
With the deepening of population aging, the treatment of cognitive impairment and dementia is facing increasing challenges. Vascular dementia (VaD) is a cognitive dysfunction caused by brain blood flow damage and one of the most common causes of dementia after Alzheimer's disease. White matter damage in patients with chronic ischemic dementia often occurs before cognitive impairment, and its pathological changes include leukoaraiosis, myelin destruction and oligodendrocyte death. The pathophysiology of vascular dementia is complex, involving a variety of neuronal and vascular lesions. The current proposed mechanisms include calcium overload, oxidative stress, nitrative stress and inflammatory damage, which can lead to hypoxia-ischemia and demyelination. Oligodendrocytes are the only myelinating cells in the central nervous system and closely associated with VaD. In this review article, we intend to further discuss the role of oligodendrocytes in white matter and myelin injury in VaD and the development of anti-myelin injury target drugs.
Collapse
Affiliation(s)
- Hexin Zhang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Yanrong Yang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Jingjing Zhang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Li Huang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Yang Niu
- Key Laboratory of Modernization of Minority Medicine, Ministry of Education, Ningxia medical University, Yinchuan 750004, Ningxia, China
| | - Hua Chen
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia, China
| | - Qibing Liu
- Department of Pharmacy, The First Affiliated Hospital of Hainan Medical University, Haikou 570100, China
| | - Rui Wang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, School of Pharmacy, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| |
Collapse
|
7
|
Narine M, Azmi MA, Umali M, Volz A, Colognato H. The AMPK activator metformin improves recovery from demyelination by shifting oligodendrocyte bioenergetics and accelerating OPC differentiation. Front Cell Neurosci 2023; 17:1254303. [PMID: 37904733 PMCID: PMC10613472 DOI: 10.3389/fncel.2023.1254303] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/11/2023] [Indexed: 11/01/2023] Open
Abstract
Multiple Sclerosis (MS) is a chronic disease characterized by immune-mediated destruction of myelinating oligodendroglia in the central nervous system. Loss of myelin leads to neurological dysfunction and, if myelin repair fails, neurodegeneration of the denuded axons. Virtually all treatments for MS act by suppressing immune function, but do not alter myelin repair outcomes or long-term disability. Excitingly, the diabetes drug metformin, a potent activator of the cellular "energy sensor" AMPK complex, has recently been reported to enhance recovery from demyelination. In aged mice, metformin can restore responsiveness of oligodendrocyte progenitor cells (OPCs) to pro-differentiation cues, enhancing their ability to differentiate and thus repair myelin. However, metformin's influence on young oligodendroglia remains poorly understood. Here we investigated metformin's effect on the temporal dynamics of differentiation and metabolism in young, healthy oligodendroglia and in oligodendroglia following myelin damage in young adult mice. Our findings reveal that metformin accelerates early stages of myelin repair following cuprizone-induced myelin damage. Metformin treatment of both isolated OPCs and oligodendrocytes altered cellular bioenergetics, but in distinct ways, suppressing oxidative phosphorylation and enhancing glycolysis in OPCs, but enhancing oxidative phosphorylation and glycolysis in both immature and mature oligodendrocytes. In addition, metformin accelerated the differentiation of OPCs to oligodendrocytes in an AMPK-dependent manner that was also dependent on metformin's ability to modulate cell metabolism. In summary, metformin dramatically alters metabolism and accelerates oligodendroglial differentiation both in health and following myelin damage. This finding broadens our knowledge of metformin's potential to promote myelin repair in MS and in other diseases with myelin loss or altered myelination dynamics.
Collapse
Affiliation(s)
- Mohanlall Narine
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, United States
- Program in Neurosciences, Stony Brook University, Stony Brook, NY, United States
| | - Maryam A. Azmi
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, United States
| | - Martin Umali
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, United States
| | - Ashley Volz
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, United States
| | - Holly Colognato
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
8
|
Milbocker KA, Williams LT, Caban-Rivera DA, Smith IF, Kurtz S, McGarry MDJ, Wattrisse B, Van Houten EEW, Johnson CL, Klintsova AY. Monitoring lasting changes to brain tissue integrity through mechanical properties following adolescent exercise intervention in a rat model of Fetal Alcohol Spectrum Disorders. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.26.559571. [PMID: 37808633 PMCID: PMC10557734 DOI: 10.1101/2023.09.26.559571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Background Fetal Alcohol Spectrum Disorders (FASD) encompass a group of highly prevalent conditions resulting from prenatal alcohol exposure. Alcohol exposure during the third trimester of pregnancy overlapping with the brain growth spurt is detrimental to white matter growth and myelination, particularly in the corpus callosum, ultimately affecting tissue integrity in adolescence. Traditional neuroimaging techniques have been essential for assessing neurodevelopment in affected youth; however, these methods are limited in their capacity to track subtle microstructural alterations to white matter, thus restricting their effectiveness in monitoring therapeutic intervention. In this preliminary study we use a highly sensitive and clinically translatable Magnetic Resonance Elastography (MRE) protocol for assessing brain tissue microstructure through its mechanical properties following an exercise intervention in a rat model of FASD. Methods Rat pups were divided into two groups: alcohol-exposed (AE) pups which received alcohol in milk substitute (5.25 g/kg/day) via intragastric intubation on postnatal days (PD) four through nine during the rat brain growth spurt (Dobbing and Sands, 1979), or sham-intubated (SI) controls. In adolescence, on PD 30, half AE and SI rats were randomly assigned to either a modified home cage with free access to a running wheel or to a new home cage for 12 days (Gursky and Klintsova, 2017). Previous studies conducted in the lab have shown that 12 days of voluntary exercise intervention in adolescence immediately ameliorated callosal myelination in AE rats (Milbocker et al., 2022, 2023). MRE was used to measure longitudinal changes to mechanical properties of the whole brain and the corpus callosum at intervention termination and one-month post-intervention. Histological quantification of precursor and myelinating oligoglia in corpus callosum was performed one-month post-intervention. Results Prior to intervention, AE rats had lower forebrain stiffness in adolescence compared to SI controls ( p = 0.02). Exercise intervention immediately mitigated this effect in AE rats, resulting in higher forebrain stiffness post-intervention in adolescence. Similarly, we discovered that forebrain damping ratio was lowest in AE rats in adolescence ( p < 0.01), irrespective of intervention exposure. One-month post-intervention in adulthood, AE and SI rats exhibited comparable forebrain stiffness and damping ratio (p > 0.05). Taken together, these MRE data suggest that adolescent exercise intervention supports neurodevelopmental "catch-up" in AE rats. Analysis of the stiffness and damping ratio of the body of corpus callosum revealed that these measures increased with age. Finally, histological quantification of myelinating oligodendrocytes one-month post-intervention revealed a negative rebound effect of exercise cessation on the total estimate of these cells in the body of corpus callosum, irrespective of treatment group which was not convergent with noninvasive MRE measures. Conclusions This is the first application of MRE to measure changes in brain mechanical properties in a rodent model of FASD. MRE successfully captured alcohol-related changes to forebrain stiffness and damping ratio in adolescence. These preliminary findings expand upon results from previous studies which used traditional diffusion neuroimaging to identify structural changes to the adolescent brain in rodent models of FASD (Milbocker et al., 2022; Newville et al., 2017). Additionally, in vivo MRE identified an exercise-related alteration to forebrain stiffness that occurred in adolescence, immediately post-intervention.
Collapse
|
9
|
Dixit A, Savage HS, Greer JM. An appraisal of emerging therapeutic targets for multiple sclerosis derived from current preclinical models. Expert Opin Ther Targets 2023; 27:553-574. [PMID: 37438986 DOI: 10.1080/14728222.2023.2236301] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 07/09/2023] [Indexed: 07/14/2023]
Abstract
INTRODUCTION Multiple sclerosis (MS) is a chronic inflammatory, demyelinating, and neurodegenerative condition affecting the central nervous system (CNS). Although therapeutic approaches have become available over the last 20 years that markedly slow the progression of disease, there is no cure for MS. Furthermore, the capacity to repair existing CNS damage caused by MS remains very limited. AREAS COVERED Several animal models are widely used in MS research to identify potential druggable targets for new treatment of MS. In this review, we look at targets identified since 2019 in studies using these models, and their potential for effecting a cure for MS. EXPERT OPINION Refinement of therapeutic strategies targeting key molecules involved in the activation of immune cells, cytokine, and chemokine signaling, and the polarization of the immune response have dominated recent publications. While some progress has been made in identifying effective targets to combat chronic demyelination and neurodegeneration, much more work is required. Progress is largely limited by the gaps in knowledge of how the immune system and the nervous system interact in MS and its animal models, and whether the numerous targets present in both systems respond in the same way in each system to the same therapeutic manipulation.
Collapse
Affiliation(s)
- Aakanksha Dixit
- The University of Queensland, UQ Centre for Clinical Research, Royal Brisbane & Women's Hospita, Brisbane, QLD, Australia
| | - Hannah S Savage
- The University of Queensland, UQ Centre for Clinical Research, Royal Brisbane & Women's Hospita, Brisbane, QLD, Australia
| | - Judith M Greer
- The University of Queensland, UQ Centre for Clinical Research, Royal Brisbane & Women's Hospita, Brisbane, QLD, Australia
| |
Collapse
|
10
|
Devanand M, V N S, Madhu K. Signaling mechanisms involved in the regulation of remyelination in multiple sclerosis: a mini review. J Mol Med (Berl) 2023:10.1007/s00109-023-02312-9. [PMID: 37084092 DOI: 10.1007/s00109-023-02312-9] [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: 05/14/2022] [Revised: 02/22/2023] [Accepted: 03/28/2023] [Indexed: 04/22/2023]
Abstract
Multiple sclerosis is an autoimmune neurodegenerative disease of the CNS that causes progressive disabilities, owing to CNS axon degeneration as a late result of demyelination. In the search for the prevention of axonal loss, mitigating inflammatory attacks in the CNS and myelin restoration are two possible approaches. As a result, therapies that target diverse signaling pathways involved in neuroprotection and remyelination have the potential to overcome the challenges in the development of multiple sclerosis treatments. LINGO1 (Leucine rich repeat and Immunoglobulin domain containing, Nogo receptor- interaction protein), AKT/PIP3/mTOR, Notch, Wnt, RXR (Retinoid X receptor gamma), and Nrf2 (nuclear factor erythroid 2-related factor 2) signaling pathways are highlighted in this section. This article reviews the present knowledge regarding numerous signaling pathways and their functions in regulating remyelination in multiple sclerosis pathogenesis. These pathways are potential biomarkers and therapeutic targets in MS.
Collapse
Affiliation(s)
- Midhuna Devanand
- Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - Saiprabha V N
- Department of Pharmaceutical Chemistry and Analysis, Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India.
| | - Krishnadas Madhu
- Department of Pharmacology, Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India.
| |
Collapse
|
11
|
Yang Z, Yu Z, Xiao B. Coordinated Regulation of Myelination by Growth Factor and Amino-acid Signaling Pathways. Neurosci Bull 2023; 39:453-465. [PMID: 36352321 PMCID: PMC10043148 DOI: 10.1007/s12264-022-00967-x] [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: 03/11/2022] [Accepted: 06/27/2022] [Indexed: 11/11/2022] Open
Abstract
Myelin-forming oligodendrocytes in the central nervous system (CNS) and Schwann cells in the peripheral nervous system (PNS) are essential for structural and functional homeostasis of nervous tissue. Albeit with certain similarities, the regulation of CNS and PNS myelination is executed differently. Recent advances highlight the coordinated regulation of oligodendrocyte myelination by amino-acid sensing and growth factor signaling pathways. In this review, we discuss novel insights into the understanding of differential regulation of oligodendrocyte and Schwann cell biology in CNS and PNS myelination, with particular focus on the roles of growth factor-stimulated RHEB-mTORC1 and GATOR2-mediated amino-acid sensing/signaling pathways. We also discuss recent progress on the metabolic regulation of oligodendrocytes and Schwann cells and the impact of their dysfunction on neuronal function and disease.
Collapse
Affiliation(s)
- Zhiwen Yang
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518000, China
| | - Zongyan Yu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518000, China.
| | - Bo Xiao
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518000, China.
| |
Collapse
|
12
|
Khandker L, Wood TL. Live-cell metabolic analysis of oligodendroglia isolated from postnatal mouse brain and spinal cord. STAR Protoc 2022; 3:101655. [PMID: 36092821 PMCID: PMC9449657 DOI: 10.1016/j.xpro.2022.101655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
This protocol describes isolation and live-cell metabolic analysis of O4+ oligodendroglia from brain and spinal cord of postnatal mice. We have optimized existing protocols for O4+ isolation from neonatal brain and expanded the protocol to include isolation of highly viable oligodendroglia from spinal cords of postnatal mice up to 18 days of age. Isolated oligodendroglia can be used in multiple downstream analyses, and here we describe an optimized real-time metabolic assay using Agilent Seahorse Analyzer to measure mitochondrial respiration. For complete details on the use and execution of this protocol, please refer to Khandker et al. (2022).
Collapse
Affiliation(s)
- Luipa Khandker
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA.
| | - Teresa L Wood
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA.
| |
Collapse
|
13
|
Therapeutic opportunities for targeting cellular senescence in progressive multiple sclerosis. Curr Opin Pharmacol 2022; 63:102184. [DOI: 10.1016/j.coph.2022.102184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/13/2022] [Accepted: 01/15/2022] [Indexed: 11/22/2022]
|
14
|
Khandker L, Jeffries MA, Chang YJ, Mather ML, Evangelou AV, Bourne JN, Tafreshi AK, Ornelas IM, Bozdagi-Gunal O, Macklin WB, Wood TL. Cholesterol biosynthesis defines oligodendrocyte precursor heterogeneity between brain and spinal cord. Cell Rep 2022; 38:110423. [PMID: 35235799 PMCID: PMC8988216 DOI: 10.1016/j.celrep.2022.110423] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 01/04/2022] [Accepted: 02/02/2022] [Indexed: 12/28/2022] Open
Abstract
Brain and spinal cord oligodendroglia have distinct functional characteristics, and cell-autonomous loss of individual genes can result in different regional phenotypes. However, a molecular basis for these distinctions is unknown. Using single-cell analysis of oligodendroglia during developmental myelination, we demonstrate that brain and spinal cord precursors are transcriptionally distinct, defined predominantly by cholesterol biosynthesis. We further identify the mechanistic target of rapamycin (mTOR) as a major regulator promoting cholesterol biosynthesis in oligodendroglia. Oligodendroglia-specific loss of mTOR decreases cholesterol biosynthesis in both the brain and the spinal cord, but mTOR loss in spinal cord oligodendroglia has a greater impact on cholesterol biosynthesis, consistent with more pronounced deficits in developmental myelination. In the brain, mTOR loss results in a later adult myelin deficit, including oligodendrocyte death, spontaneous demyelination, and impaired axonal function, demonstrating that mTOR is required for myelin maintenance in the adult brain. Using single-cell RNA sequencing, Khandker et al. reveal that oligodendroglia in the brain and spinal cord are distinct. These differences arise from mechanisms regulating cholesterol acquisition, necessary for maintenance of the lipid-rich myelin sheath, and involve mTOR in the regulation of cholesterol biosynthesis in oligodendroglia.
Collapse
Affiliation(s)
- Luipa Khandker
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Marisa A Jeffries
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Yun-Juan Chang
- Office of Advance Research Computing, Rutgers University, Piscataway, NJ 08854, USA
| | - Marie L Mather
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Angelina V Evangelou
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Jennifer N Bourne
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Azadeh K Tafreshi
- Department of Psychiatry, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Isis M Ornelas
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Ozlem Bozdagi-Gunal
- Department of Psychiatry, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Wendy B Macklin
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Teresa L Wood
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA.
| |
Collapse
|
15
|
Benardais K, Ornelas IM, Fauveau M, Brown TL, Finseth LT, Panic R, Deboux C, Macklin WB, Wood TL, Nait Oumesmar B. p70S6 kinase regulates oligodendrocyte differentiation and is active in remyelinating lesions. Brain Commun 2022; 4:fcac025. [PMID: 35224490 PMCID: PMC8864467 DOI: 10.1093/braincomms/fcac025] [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: 08/13/2020] [Revised: 09/10/2021] [Accepted: 02/10/2022] [Indexed: 11/15/2022] Open
Abstract
The p70 ribosomal S6 kinases (p70 ribosomal S6 kinase 1 and p70 ribosomal S6 kinase 2) are downstream targets of the mechanistic target of rapamycin signalling pathway. p70 ribosomal S6 kinase 1 specifically has demonstrated functions in regulating cell size in Drosophila and in insulin-sensitive cell populations in mammals. Prior studies demonstrated that the mechanistic target of the rapamycin pathway promotes oligodendrocyte differentiation and developmental myelination; however, how the immediate downstream targets of mechanistic target of rapamycin regulate these processes has not been elucidated. Here, we tested the hypothesis that p70 ribosomal S6 kinase 1 regulates oligodendrocyte differentiation during developmental myelination and remyelination processes in the CNS. We demonstrate that p70 ribosomal S6 kinase activity peaks in oligodendrocyte lineage cells at the time when they transition to myelinating oligodendrocytes during developmental myelination in the mouse spinal cord. We further show p70 ribosomal S6 kinase activity in differentiating oligodendrocytes in acute demyelinating lesions induced by lysophosphatidylcholine injection or by experimental autoimmune encephalomyelitis in mice. In demyelinated lesions, the expression of the p70 ribosomal S6 kinase target, phosphorylated S6 ribosomal protein, was transient and highest in maturing oligodendrocytes. Interestingly, we also identified p70 ribosomal S6 kinase activity in oligodendrocyte lineage cells in active multiple sclerosis lesions. Consistent with its predicted function in promoting oligodendrocyte differentiation, we demonstrate that specifically inhibiting p70 ribosomal S6 kinase 1 in cultured oligodendrocyte precursor cells significantly impairs cell lineage progression and expression of myelin basic protein. Finally, we used zebrafish to show in vivo that inhibiting p70 ribosomal S6 kinase 1 function in oligodendroglial cells reduces their differentiation and the number of myelin internodes produced. These data reveal an essential function of p70 ribosomal S6 kinase 1 in promoting oligodendrocyte differentiation during development and remyelination across multiple species.
Collapse
Affiliation(s)
- Karelle Benardais
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Isis M. Ornelas
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA 07101
| | - Melissa Fauveau
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Tanya L. Brown
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA 80045
| | - Lisbet T. Finseth
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA 80045
| | - Radmila Panic
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Cyrille Deboux
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Wendy B. Macklin
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA 80045
| | - Teresa L. Wood
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA 07101
| | - Brahim Nait Oumesmar
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA 07101
| |
Collapse
|
16
|
Frondelli MJ, Mather ML, Levison SW. Oligodendrocyte progenitor proliferation is disinhibited following traumatic brain injury in leukemia inhibitory factor heterozygous mice. J Neurosci Res 2021; 100:578-597. [PMID: 34811802 DOI: 10.1002/jnr.24984] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 09/28/2021] [Accepted: 10/01/2021] [Indexed: 01/25/2023]
Abstract
Traumatic brain injury (TBI) is a significant problem that affects over 800,000 children each year. As cell proliferation is disturbed by injury and required for normal brain development, we investigated how a pediatric closed head injury (CHI) would affect the progenitors of the subventricular zone (SVZ). Additionally, we evaluated the contribution of leukemia inhibitory factor (LIF) using germline LIF heterozygous mice (LIF Het), as LIF is an injury-induced cytokine, known to influence neurogenesis and gliogenesis. CHIs were performed on P20 LIF Het and wild-type (WT) mice. Ki-67 immunostaining and stereology revealed that cell proliferation increased ~250% in injured LIF Het mice compared to the 30% increase observed in injured WT mice at 48-hr post-CHI. OLIG2+ cell proliferation increased in the SVZ and white matter of LIF Het injured mice at 48-hr recovery. Using an 8-color flow cytometry panel, the proliferation of three distinct multipotential progenitors and early oligodendrocyte progenitor cell proliferation was significantly increased in LIF Het injured mice compared to WT injured mice. Supporting its cytostatic function, LIF decreased neurosphere progenitor and oligodendrocyte progenitor cell proliferation compared to controls. In highly enriched mouse oligodendrocyte progenitor cell cultures, LIF increased phospho-protein kinase B after 20 min and increased phospho-S6 ribosomal protein at 20 and 40 min of exposure, which are downstream targets of the mammalian target of rapamycin pathway. Altogether, our data provide new insights into the regulatory role of LIF in suppressing neural progenitor cell proliferation and, in particular, oligodendrocyte progenitor cell proliferation after a mild TBI.
Collapse
Affiliation(s)
- Michelle J Frondelli
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Marie L Mather
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Steven W Levison
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| |
Collapse
|
17
|
Jeffries MA, McLane LE, Khandker L, Mather ML, Evangelou AV, Kantak D, Bourne JN, Macklin WB, Wood TL. mTOR Signaling Regulates Metabolic Function in Oligodendrocyte Precursor Cells and Promotes Efficient Brain Remyelination in the Cuprizone Model. J Neurosci 2021; 41:8321-8337. [PMID: 34417330 PMCID: PMC8496195 DOI: 10.1523/jneurosci.1377-20.2021] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/21/2021] [Accepted: 08/10/2021] [Indexed: 02/02/2023] Open
Abstract
In demyelinating diseases, such as multiple sclerosis, primary loss of myelin and subsequent neuronal degeneration throughout the CNS impair patient functionality. While the importance of mechanistic target of rapamycin (mTOR) signaling during developmental myelination is known, no studies have yet directly examined the function of mTOR signaling specifically in the oligodendrocyte (OL) lineage during remyelination. Here, we conditionally deleted Mtor from adult oligodendrocyte precursor cells (OPCs) using Ng2-CreERT in male adult mice to test its function in new OLs responsible for remyelination. During early remyelination after cuprizone-induced demyelination, mice lacking mTOR in adult OPCs had unchanged OL numbers but thinner myelin. Myelin thickness recovered by late-stage repair, suggesting a delay in myelin production when Mtor is deleted from adult OPCs. Surprisingly, loss of mTOR in OPCs had no effect on efficiency of remyelination after lysophosphatidylcholine lesions in either the spinal cord or corpus callosum, suggesting that mTOR signaling functions specifically in a pathway dysregulated by cuprizone to promote remyelination efficiency. We further determined that cuprizone and inhibition of mTOR cooperatively compromise metabolic function in primary rat OLs undergoing differentiation. Together, our results support the conclusion that mTOR signaling in OPCs is required to overcome the metabolic dysfunction in the cuprizone-demyelinated adult brain.SIGNIFICANCE STATEMENT Impaired remyelination by oligodendrocytes contributes to the progressive pathology in multiple sclerosis, so it is critical to identify mechanisms of improving remyelination. The goal of this study was to examine mechanistic target of rapamycin (mTOR) signaling in remyelination. Here, we provide evidence that mTOR signaling promotes efficient remyelination of the brain after cuprizone-mediated demyelination but has no effect on remyelination after lysophosphatidylcholine demyelination in the spinal cord or brain. We also present novel data revealing that mTOR inhibition and cuprizone treatment additively affect the metabolic profile of differentiating oligodendrocytes, supporting a mechanism for the observed remyelination delay. These data suggest that altered metabolic function may underlie failure of remyelination in multiple sclerosis lesions and that mTOR signaling may be of therapeutic potential for promoting remyelination.
Collapse
Affiliation(s)
- Marisa A Jeffries
- Department of Pharmacology, Physiology, and Neuroscience and Center for Cell Signaling, Rutgers New Jersey Medical School, Newark, New Jersey 07103
| | - Lauren E McLane
- Department of Pharmacology, Physiology, and Neuroscience and Center for Cell Signaling, Rutgers New Jersey Medical School, Newark, New Jersey 07103
| | - Luipa Khandker
- Department of Pharmacology, Physiology, and Neuroscience and Center for Cell Signaling, Rutgers New Jersey Medical School, Newark, New Jersey 07103
| | - Marie L Mather
- Department of Pharmacology, Physiology, and Neuroscience and Center for Cell Signaling, Rutgers New Jersey Medical School, Newark, New Jersey 07103
| | - Angelina V Evangelou
- Department of Pharmacology, Physiology, and Neuroscience and Center for Cell Signaling, Rutgers New Jersey Medical School, Newark, New Jersey 07103
| | - Divyangi Kantak
- Department of Pharmacology, Physiology, and Neuroscience and Center for Cell Signaling, Rutgers New Jersey Medical School, Newark, New Jersey 07103
| | - Jennifer N Bourne
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Wendy B Macklin
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Teresa L Wood
- Department of Pharmacology, Physiology, and Neuroscience and Center for Cell Signaling, Rutgers New Jersey Medical School, Newark, New Jersey 07103
| |
Collapse
|
18
|
Hughes EG, Stockton ME. Premyelinating Oligodendrocytes: Mechanisms Underlying Cell Survival and Integration. Front Cell Dev Biol 2021; 9:714169. [PMID: 34368163 PMCID: PMC8335399 DOI: 10.3389/fcell.2021.714169] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/30/2021] [Indexed: 12/31/2022] Open
Abstract
In the central nervous system, oligodendrocytes produce myelin sheaths that enwrap neuronal axons to provide trophic support and increase conduction velocity. New oligodendrocytes are produced throughout life through a process referred to as oligodendrogenesis. Oligodendrogenesis consists of three canonical stages: the oligodendrocyte precursor cell (OPC), the premyelinating oligodendrocyte (preOL), and the mature oligodendrocyte (OL). However, the generation of oligodendrocytes is inherently an inefficient process. Following precursor differentiation, a majority of premyelinating oligodendrocytes are lost, likely due to apoptosis. If premyelinating oligodendrocytes progress through this survival checkpoint, they generate new myelinating oligodendrocytes in a process we have termed integration. In this review, we will explore the intrinsic and extrinsic signaling pathways that influence preOL survival and integration by examining the intrinsic apoptotic pathways, metabolic demands, and the interactions between neurons, astrocytes, microglia, and premyelinating oligodendrocytes. Additionally, we will discuss similarities between the maturation of newly generated neurons and premyelinating oligodendrocytes. Finally, we will consider how increasing survival and integration of preOLs has the potential to increase remyelination in multiple sclerosis. Deepening our understanding of premyelinating oligodendrocyte biology may open the door for new treatments for demyelinating disease and will help paint a clearer picture of how new oligodendrocytes are produced throughout life to facilitate brain function.
Collapse
Affiliation(s)
- Ethan G Hughes
- Department of Cell and Developmental Biology, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Michael E Stockton
- Department of Cell and Developmental Biology, School of Medicine, University of Colorado, Aurora, CO, United States
| |
Collapse
|
19
|
Vaes JEG, Brandt MJV, Wanders N, Benders MJNL, de Theije CGM, Gressens P, Nijboer CH. The impact of trophic and immunomodulatory factors on oligodendrocyte maturation: Potential treatments for encephalopathy of prematurity. Glia 2020; 69:1311-1340. [PMID: 33595855 PMCID: PMC8246971 DOI: 10.1002/glia.23939] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022]
Abstract
Encephalopathy of prematurity (EoP) is a major cause of morbidity in preterm neonates, causing neurodevelopmental adversities that can lead to lifelong impairments. Preterm birth-related insults, such as cerebral oxygen fluctuations and perinatal inflammation, are believed to negatively impact brain development, leading to a range of brain abnormalities. Diffuse white matter injury is a major hallmark of EoP and characterized by widespread hypomyelination, the result of disturbances in oligodendrocyte lineage development. At present, there are no treatment options available, despite the enormous burden of EoP on patients, their families, and society. Over the years, research in the field of neonatal brain injury and other white matter pathologies has led to the identification of several promising trophic factors and cytokines that contribute to the survival and maturation of oligodendrocytes, and/or dampening neuroinflammation. In this review, we discuss the current literature on selected factors and their therapeutic potential to combat EoP, covering a wide range of in vitro, preclinical and clinical studies. Furthermore, we offer a future perspective on the translatability of these factors into clinical practice.
Collapse
Affiliation(s)
- Josine E G Vaes
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands.,Department of Neonatology, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Myrna J V Brandt
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Nikki Wanders
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Manon J N L Benders
- Department of Neonatology, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | - Caroline G M de Theije
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| | | | - Cora H Nijboer
- Department for Developmental Origins of Disease, University Medical Center Utrecht Brain Center and Wilhelmina Children's Hospital, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
20
|
Adams KL, Dahl KD, Gallo V, Macklin WB. Intrinsic and extrinsic regulators of oligodendrocyte progenitor proliferation and differentiation. Semin Cell Dev Biol 2020; 116:16-24. [PMID: 34110985 DOI: 10.1016/j.semcdb.2020.10.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/30/2020] [Accepted: 10/01/2020] [Indexed: 12/14/2022]
Abstract
Oligodendrocytes are highly specialized glial cells, responsible for producing myelin in the central nervous system (CNS). The multi-stage process of oligodendrocyte development is tightly regulated to ensure proper lineage progression of oligodendrocyte progenitor cells (OPCs) to mature myelin producing oligodendrocytes. This developmental process involves complex interactions between several intrinsic signaling pathways that are modulated by an array of extrinsic factors. Understanding these regulatory processes is of crucial importance, as it may help to identify specific molecular targets both to enhance plasticity in the normal CNS and to promote endogenous recovery following injury or disease. This review describes two major regulators that play important functional roles in distinct phases of oligodendrocyte development: OPC proliferation and differentiation. Specifically, we highlight the roles of the extracellular astrocyte/radial glia-derived protein Endothelin-1 in OPC proliferation and the intracellular Akt/mTOR pathway in OPC differentiation. Lastly, we reflect on how recent advances in neuroscience and scientific technology will enable greater understanding into how intrinsic and extrinsic regulators interact to generate oligodendrocyte diversity.
Collapse
Affiliation(s)
- Katrina L Adams
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010, USA
| | - Kristin D Dahl
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Vittorio Gallo
- Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC 20010, USA
| | - Wendy B Macklin
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| |
Collapse
|
21
|
Ren Y, Yin ZH, Dai JX, Yang Z, Ye BB, Ma YS, Zhang TE, Shi YY. Evidence-Based Complementary and Alternative Medicine Exploring Active Components and Mechanism of Jinhua Qinggan Granules in Treatment of COVID-19 Based on Virus-Host Interaction. Nat Prod Commun 2020. [DOI: 10.1177/1934578x20947213] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
This study aimed at exploring the active components and mechanisms of Jinhua Qinggan granules (JQG) in the prevention and treatment of coronavirus disease 2019 (COVID-19) using network pharmacology and molecular docking technology. These efforts were accomplished by employing the holistic approach of traditional Chinese medicine (TCM) and considering the virus-host interaction consisting of viral characteristics, the entry pathway into the host, and the resulting immune response. The chemical constituents and molecular targets of the 12 herbs from JQG were obtained using the TCM Systems Pharmacology database and analysis platform. UniProt was used to search for genes corresponding to JQG protein targets and Cytoscape 3.7.2 to construct the component-target (gene) network. Database for Annotation, Visualization and Integrated Discovery was used to perform enrichment analysis of gene ontology functions and the Kyoto Encyclopedia of Genes and Genomes pathways to predict the mechanism of action. The components ranked high in the network, and the major active components of the principal medicines, based on published literature, were docked with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) 3CL hydrolase, SARS-CoV-2 spike glycoprotein (S protein), angiotensin conversion enzyme II (ACE2), and suppressor of cytokine signaling 1 (SOCS1). Visualization analysis demonstrated that the core active components of JQG had a strong affinity for SARS-CoV-2 3CL hydrolase, SARS-CoV-2 S protein, ACE2, and SOCS1. These data imply that the potential active components of JQG may act on multiple signaling pathways by binding to targets such as SARS-CoV-2 3CL hydrolase, S protein, ACE2, and SOCS1, thereby inhibiting virus replication and targeting cell binding, reducing host inflammation, and activating antiviral immunity to a certain extent.
Collapse
Affiliation(s)
- Yan Ren
- College of Pharmacy,Institute of Ethnomedicine, Southwest Minzu University, Chengdu, China
| | - Zheng-hao Yin
- College of Pharmacy,Institute of Ethnomedicine, Southwest Minzu University, Chengdu, China
| | | | - Zhuo Yang
- College of Pharmacy,Institute of Ethnomedicine, Southwest Minzu University, Chengdu, China
| | - Bin-bin Ye
- College of Pharmacy,Institute of Ethnomedicine, Southwest Minzu University, Chengdu, China
| | - Yi-sha Ma
- College of Pharmacy,Institute of Ethnomedicine, Southwest Minzu University, Chengdu, China
| | - Tian-e Zhang
- Key Laboratory of Sichuan University Biology Laboratories Research on Zang-Xiang of TCM, Chinese Medical Center of Chengdu University of TCM, Chengdu, China
| | - Yuan-yuan Shi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
22
|
Mechanistic Target of Rapamycin Regulates the Oligodendrocyte Cytoskeleton during Myelination. J Neurosci 2020; 40:2993-3007. [PMID: 32139584 DOI: 10.1523/jneurosci.1434-18.2020] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 02/23/2020] [Accepted: 02/26/2020] [Indexed: 12/22/2022] Open
Abstract
During differentiation, oligodendrocyte precursor cells (OPCs) extend a network of processes that make contact with axons and initiate myelination. Recent studies revealed that actin polymerization is required for initiation of myelination whereas actin depolymerization promotes myelin wrapping. Here, we used primary OPCs in culture isolated from neonatal rat cortices of both sexes and young male and female mice with oligodendrocyte-specific deletion of mechanistic target of rapamycin (mTOR) to demonstrate that mTOR regulates expression of specific cytoskeletal targets and actin reorganization in oligodendrocytes during developmental myelination. Loss or inhibition of mTOR reduced expression of profilin2 and ARPC3, actin polymerizing factors, and elevated levels of active cofilin, which mediates actin depolymerization. The deficits in actin polymerization were revealed in reduced phalloidin and deficits in oligodendrocyte cellular branching complexity at the peak of morphologic differentiation and a delay in initiation of myelination. We further show a critical role for mTOR in expression and localization of myelin basic protein (Mbp) mRNA and MBP protein to the cellular processes where it is necessary at the myelin membrane for axon wrapping. Mbp mRNA transport deficits were confirmed by single molecule RNA FISH. Moreover, expression of the kinesin family member 1B, an Mbp mRNA transport protein, was reduced in CC1+ cells in the mTOR cKO and in mTOR inhibited oligodendrocytes undergoing differentiation in vitro These data support the conclusion that mTOR regulates both initiation of myelination and axon wrapping by targeting cytoskeletal reorganization and MBP localization to oligodendrocyte processes.SIGNIFICANCE STATEMENT Myelination is essential for normal CNS development and adult axon preservation and function. The mechanistic target of rapamycin (mTOR) signaling pathway has been implicated in promoting CNS myelination; however, there is a gap in our understanding of the mechanisms by which mTOR promotes developmental myelination through regulating specific downstream targets. Here, we present evidence that mTOR promotes the initiation of myelination through regulating specific cytoskeletal targets and cellular process expansion by oligodendrocyte precursor cells as well as expression and cellular localization of myelin basic protein.
Collapse
|