1
|
Ostrem BEL, Gano D. Novel Approaches to the Treatment of Preterm White Matter Injury through Targeting Remyelination. Clin Perinatol 2025; 52:289-306. [PMID: 40350212 DOI: 10.1016/j.clp.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Preterm white matter injury (WMI) is a common cause of cerebral palsy and cognitive disability after premature birth. Preterm WMI is caused by a differentiation arrest in the oligodendrocyte (OL) lineage, and a failure of myelination. As there are no specific treatments, care is supportive and focused on rehabilitation. However, novel high-throughput screening methods have enabled the identification of "pro-myelinating" compounds that promote OL differentiation and myelination. Many of these compounds stimulate remyelination in animal models and patients with demyelinating disorders. The shared mechanisms of remyelination and developmental myelination suggest that pro-myelinating compounds may have potential utility in preterm WMI.
Collapse
Affiliation(s)
- Bridget E L Ostrem
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, 675 Nelson Rising Lane, Suite 411, San Francisco, CA 94158, USA.
| | - Dawn Gano
- Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, 675 Nelson Rising Lane, Suite 402, San Francisco, CA 94158, USA
| |
Collapse
|
2
|
Zeng J, Zhou H, Wan H, Yang J. Single-cell omics: moving towards a new era in ischemic stroke research. Eur J Pharmacol 2025; 1000:177725. [PMID: 40350018 DOI: 10.1016/j.ejphar.2025.177725] [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: 06/27/2024] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
Ischemic stroke (IS) is a highly complex and heterogeneous disease involving multiple pathophysiological events. A better understanding of the pathophysiology of IS will enhance preventive, diagnostic and therapeutic strategies. Despite significant advances in modern medicine, the molecular mechanisms of IS are still largely unknown. The high-throughput omics approach opens new avenues for identifying IS biomarkers and elucidating disease pathogenesis mechanisms. Single-cell omics enables a more thorough and in-depth analysis of the cellular interactions and properties in IS. This will lead to a better understanding of the onset, treatment and prognosis of IS. In this paper, we first reviewed the disease signatures and mechanisms research of IS. Subsequently, the use of single-cell omics to comprehend the mechanisms of IS was discussed, along with some recent developments in the field. To further delineate the upstream pathogenic alterations and downstream molecular impacts of IS, we also discussed the current use of machine learning approaches to single-cell omics data analysis. Particularly, single-cell omics is being used to inform risk assessment, early patient diagnosis and treatment strategies, and their potential impact on precision medicine. Thus, we summarized the role of single-cell omics in precision medicine. Despite the relative youth of the field, the development of single-cell omics promises to provide a powerful tool for elucidating the pathogenesis of IS.
Collapse
Affiliation(s)
- Jieqiong Zeng
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Ecological and Environmental, Hubei Industrial Polytechnic, Shiyan, 442000, China
| | - Huifen Zhou
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Haitong Wan
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Jiehong Yang
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
3
|
Huang Q, Tang J, Xiang Y, Shang X, Li K, Chen L, Hu J, Li H, Pi Y, Yang H, Zhang H, Tan H, Xiyang Y, Jin H, Li X, Chen M, Mao R, Wang Q. 4-Benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione rescues oligodendrocytes ferroptosis leading to myelin loss and ameliorates neuronal injury facilitating memory in neonatal hypoxic-ischemic brain damage. Exp Neurol 2025; 390:115262. [PMID: 40246011 DOI: 10.1016/j.expneurol.2025.115262] [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: 11/28/2024] [Revised: 04/08/2025] [Accepted: 04/13/2025] [Indexed: 04/19/2025]
Abstract
Neonatal brain hypoxia-ischemia (HI) is proved to cause white matter injury (WMI), which resulted in behavioral disturbance. Myelin formed by oligodendrocytes vulnerable to hypoxia-ischemia (HI), regulating motor and cognitive function, is easily damaged by HI causing myelin loss. 4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione (TDZD-8) has a potential rescue role in neuronal death post HI. Studies reported that neuronal ferroptosis could be induced by HI and linked to behavioral abnormalities. However, the effect of TDZD-8 on WMI and its involvement in memory recovery remains unclear. In this study, our HIBD model showed impaired memory function caused by neuronal injury and myelin loss. TDZD-8 effectively reversed this pathology. Underlying mechanistic exploration implied that TDZD-8 ameliorating myelin loss via ferroptosis pathway was involved in the process of TDZD-8 treating neonatal HIBD. In conclusion, our data demonstrated that combined effect of white matter repairment and neuronal protection achieved the therapeutic role of TDZD-8 in neonatal HIBD, and suggested that white matter repairment also could be a considerable clinical therapy for neonatal HIBD.
Collapse
Affiliation(s)
- Qiyi Huang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Jiahang Tang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - You Xiang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Xinying Shang
- Department of Emergency Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming 650500, China
| | - Kunlin Li
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Lijia Chen
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Junnan Hu
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Han Li
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Yanxiong Pi
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Haiyan Yang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Huijia Zhang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Heng Tan
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Yanbin Xiyang
- Institution of Neuroscience, Kunming Medical University, Kunming 650500, China
| | - Huiyan Jin
- Department of Functional Experiment, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Xia Li
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Manjun Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming 650500, China
| | - Rongrong Mao
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China.
| | - Qian Wang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China.
| |
Collapse
|
4
|
Albertson AJ, Winkler EA, Yang AC, Buckwalter MS, Dingman AL, Fan H, Herson PS, McCullough LD, Perez-Pinzon M, Sansing LH, Sun D, Alkayed NJ. Single-Cell Analysis in Cerebrovascular Research: Primed for Breakthroughs and Clinical Impact. Stroke 2025; 56:1082-1091. [PMID: 39772596 PMCID: PMC11932790 DOI: 10.1161/strokeaha.124.049001] [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] [Indexed: 01/11/2025]
Abstract
Data generated using single-cell RNA-sequencing has the potential to transform understanding of the cerebral circulation and advance clinical care. However, the high volume of data, sometimes generated and presented without proper pathophysiological context, can be difficult to interpret and integrate into current understanding of the cerebral circulation and its disorders. Furthermore, heterogeneity in the representation of brain regions and vascular segments makes it difficult to compare results across studies. There are currently no standards for tissue collection and processing that allow easy comparisons across studies and analytical platforms. There are no standards either for single-cell data analysis and presentation. This topical review introduces single-cell RNA-sequencing to physicians and scientists in the cerebrovascular field, with the goals of highlighting opportunities and challenges of applying this technology in the cerebrovascular field and discussing key concepts and knowledge gaps that can be addressed by single-cell RNA-sequencing.
Collapse
Affiliation(s)
- Asher J. Albertson
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Ethan A. Winkler
- Department of Neurological Surgery, University of California San Francisco, CA
| | - Andrew C. Yang
- Gladstone Institute of Neurological Disease and Department of Neurology, University of California San Francisco, CA
| | - Marion S. Buckwalter
- Department of Neurology and Neurological Sciences, Stanford University, Palo Alto, CA
| | - Andra L. Dingman
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Huihui Fan
- Department of Neurology, University of Texas Health Science Center, Houston, TX
| | - Paco S. Herson
- Department of Neurological Surgery, The Ohio State University College of Medicine, Columbus, OH
| | | | | | - Lauren H. Sansing
- Departments of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Dandan Sun
- Department of Neurology and Pittsburgh Institute of Neurological Degeneration Diseases, University of Pittsburgh, Pittsburgh, PA
| | - Nabil J. Alkayed
- Department of Anesthesiology & Perioperative Medicine and Knight Cardiovascular Institute Portland, OR
| |
Collapse
|
5
|
Malone K, LaCasse E, Beug ST. Cell death in glioblastoma and the central nervous system. Cell Oncol (Dordr) 2025; 48:313-349. [PMID: 39503973 PMCID: PMC11997006 DOI: 10.1007/s13402-024-01007-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2024] [Indexed: 04/15/2025] Open
Abstract
Glioblastoma is the commonest and deadliest primary brain tumor. Glioblastoma is characterized by significant intra- and inter-tumoral heterogeneity, resistance to treatment and dismal prognoses despite decades of research in understanding its biological underpinnings. Encompassed within this heterogeneity and therapy resistance are severely dysregulated programmed cell death pathways. Glioblastomas recapitulate many neurodevelopmental and neural injury responses; in addition, glioblastoma cells are composed of multiple different transformed versions of CNS cell types. To obtain a greater understanding of the features underlying cell death regulation in glioblastoma, it is important to understand the control of cell death within the healthy CNS during homeostatic and neurodegenerative conditions. Herein, we review apoptotic control within neural stem cells, astrocytes, oligodendrocytes and neurons and compare them to glioblastoma apoptotic control. Specific focus is paid to the Inhibitor of Apoptosis proteins, which play key roles in neuroinflammation, CNS cell survival and gliomagenesis. This review will help in understanding glioblastoma as a transformed version of a heterogeneous organ composed of multiple varied cell types performing different functions and possessing different means of apoptotic control. Further, this review will help in developing more glioblastoma-specific treatment approaches and will better inform treatments looking at more direct brain delivery of therapeutic agents.
Collapse
Affiliation(s)
- Kyle Malone
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Eric LaCasse
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Shawn T Beug
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada.
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
6
|
Feng Y, Ma X, Zong X, Jordan JD, Wu CYC, Tesic V, Lee RHC, Zhang Q. Clemastine enhances myelin formation in the striatum and medial prefrontal cortex and improves sociability in a neonatal rat hypoxic-ischemic model. Biomed Pharmacother 2025; 185:117916. [PMID: 40058153 DOI: 10.1016/j.biopha.2025.117916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 01/28/2025] [Accepted: 02/14/2025] [Indexed: 03/23/2025] Open
Abstract
Neonatal hypoxia-ischemia (HI) results in gray and white matter injuries, leading to impairments in social behavior and severe neurological deficits. Clemastine treatment has demonstrated efficacy in alleviating behavioral deficits in various neurological disorders by improving myelin formation. It has been suggested that the medial prefrontal cortex (mPFC) and the striatum play a key role in human social behaviors. To test whether clemastine can mitigate sociability deficits by rescuing the myelin damage in these key brain areas, we administered clemastine orally for two weeks following HI insult in neonatal rats. We demonstrated that clemastine successfully ameliorated HI-induced social deficits during adolescence, attenuated hypomyelination and promoted oligodendrocyte maturation in the striatum and mPFC. We also observed that clementine reduced proliferation and apoptosis of oligodendrocyte progenitor cells (OPCs), decreased myelin debris induced by HI in the striatum, and was accompanied by microglia morphological changes in the striatum. Furthermore, our findings revealed a positive correlation between sociability and myelin formation in the striatum and mPFC. In conclusion, our data indicate that clemastine attenuates HI-induced sociability impairments during adolescence, potentially through its role in promoting myelin formation in the striatum and mPFC.
Collapse
Affiliation(s)
- Yu Feng
- Department of Neurology, Louisiana State University Health, 1501 Kings Highway, Shreveport, LA 71103, USA
| | - Xiaohui Ma
- Department of Neurology, Louisiana State University Health, 1501 Kings Highway, Shreveport, LA 71103, USA
| | - Xuemei Zong
- Department of Neurology, Louisiana State University Health, 1501 Kings Highway, Shreveport, LA 71103, USA
| | - J Dedrick Jordan
- Department of Neurology, Louisiana State University Health, 1501 Kings Highway, Shreveport, LA 71103, USA
| | - Celeste Yin-Chieh Wu
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Vesna Tesic
- Department of Neurology, Louisiana State University Health, 1501 Kings Highway, Shreveport, LA 71103, USA
| | - Reggie Hui-Chao Lee
- Department of Pharmaceutical Sciences, Taneja College of Pharmacy, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA.
| | - Quanguang Zhang
- Department of Neurology, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA 30912, USA.
| |
Collapse
|
7
|
Reyes-Corral M, Gil-González L, González-Díaz Á, Tovar-Luzón J, Ayuso MI, Lao-Pérez M, Montaner J, de la Puerta R, Fernández-Torres R, Ybot-González P. Pretreatment with oleuropein protects the neonatal brain from hypoxia-ischemia by inhibiting apoptosis and neuroinflammation. J Cereb Blood Flow Metab 2025; 45:717-734. [PMID: 39157939 DOI: 10.1177/0271678x241270237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
Hypoxic-ischemic (HI) encephalopathy is a cerebrovascular injury caused by oxygen deprivation to the brain and remains a major cause of neonatal mortality and morbidity worldwide. Therapeutic hypothermia is the current standard of care but it does not provide complete neuroprotection. Our aim was to investigate the neuroprotective effect of oleuropein (Ole) in a neonatal (seven-day-old) mouse model of HI. Ole, a secoiridoid found in olive leaves, has previously shown to reduce damage against cerebral and other ischemia/reperfusion injuries. Here, we administered Ole as a pretreatment prior to HI induction at 20 or 100 mg/kg. A week after HI, Ole significantly reduced the infarct area and the histological damage as well as white matter injury, by preserving myelination, microglial activation and the astroglial reactive response. Twenty-four hours after HI, Ole reduced the overexpression of caspase-3 and the proinflammatory cytokines IL-6 and TNF-α. Moreover, using UPLC-MS/MS we found that maternal supplementation with Ole during pregnancy and/or lactation led to the accumulation of its metabolite hydroxytyrosol in the brains of the offspring. Overall, our results indicate that pretreatment with Ole confers neuroprotection and can prevent HI-induced brain damage by modulating apoptosis and neuroinflammation.
Collapse
Affiliation(s)
- Marta Reyes-Corral
- Institute of Biomedicine of Seville (IBiS), CSIC-US-Junta de Andalucía (SAS), Seville, Spain
| | - Laura Gil-González
- Institute of Biomedicine of Seville (IBiS), CSIC-US-Junta de Andalucía (SAS), Seville, Spain
| | - Ángela González-Díaz
- Institute of Biomedicine of Seville (IBiS), CSIC-US-Junta de Andalucía (SAS), Seville, Spain
| | - Javier Tovar-Luzón
- Institute of Biomedicine of Seville (IBiS), CSIC-US-Junta de Andalucía (SAS), Seville, Spain
| | - María Irene Ayuso
- Institute of Biomedicine of Seville (IBiS), CSIC-US-Junta de Andalucía (SAS), Seville, Spain
- CIBERSAM, ISCIII (Spanish Network for Research in Mental Health), Seville, Spain
| | - Miguel Lao-Pérez
- Institute of Biomedicine of Seville (IBiS), CSIC-US-Junta de Andalucía (SAS), Seville, Spain
| | - Joan Montaner
- Institute of Biomedicine of Seville (IBiS), CSIC-US-Junta de Andalucía (SAS), Seville, Spain
- Department of Neurology, Virgen Macarena University Hospital, Seville, Spain
| | - Rocío de la Puerta
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, Seville, Spain
| | - Rut Fernández-Torres
- Departamento de Química Analítica, Facultad de Química, Universidad de Sevilla, Seville, Spain
| | - Patricia Ybot-González
- Institute of Biomedicine of Seville (IBiS), CSIC-US-Junta de Andalucía (SAS), Seville, Spain
- Spanish National Research Council (CSIC), Spain
| |
Collapse
|
8
|
Barda T, Schmitz-Koep B, Menegaux A, Bartmann P, Wolke D, Sorg C, Hedderich DM. The impact of socio-environmental factors on brain structure over the early life course of preterm-born individuals - A systematic review. Neurosci Biobehav Rev 2025; 170:106061. [PMID: 39952335 DOI: 10.1016/j.neubiorev.2025.106061] [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: 10/25/2024] [Revised: 02/03/2025] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND Approximately 11 % of births worldwide are preterm (<37 weeks). While research traditionally focuses on complications of prematurity and brain development, the role of socio-environmental factors has received less attention. Recent studies indicate these factors significantly influence neurocognitive outcomes and brain development, beyond prematurity alone. This review examines the impact of socio-environmental factors on brain structure and function in preterm-born individuals from birth to early adulthood. METHOD We conducted searches in PubMed, Embase, and Web of Science for studies up to August 28th, 2024, examining socio-environmental effects on brain structure or function in preterm-born individuals using magnetic resonance imaging. From 891 articles screened, 23 met the inclusion criteria. RESULTS Socio-environmental factors, including socioeconomic status, prenatal conditions, hospital environment, and early life experiences, notably affect brain structures in preterm-born individuals. Key impacts were found in limbic and associative cortices (e.g., cingulate gyrus, parieto-temporal cortices), white matter tracts involved in executive functioning (e.g., superior longitudinal fasciculus, cingulum), and overall brain volume. Most studies focused on infancy, with 18 of 23 presenting data from the first year of life. CONCLUSION Socio-environmental factors are associated with changes in grey and white matter in the brain, especially in the limbic system and associative areas. These findings underscore the influence of early environments on preterm-born brain development, but long-term impacts remain unclear due to limited data beyond infancy.
Collapse
Affiliation(s)
- Taylor Barda
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine and Health, Technical University of Munich, Ismaninger Str. 22, Munich 81675, Germany; TUM-NIC Neuroimaging Center, School of Medicine and Health, Technical University of Munich, Ismaninger Str. 22, Munich 81675, Germany; Graduate School of Systemic Neurosciences GSN, Ludwig-Maximilians-Universität München, Biocenter, Großhaderner Strasse 2, Munich 82152, Germany.
| | - Benita Schmitz-Koep
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine and Health, Technical University of Munich, Ismaninger Str. 22, Munich 81675, Germany; TUM-NIC Neuroimaging Center, School of Medicine and Health, Technical University of Munich, Ismaninger Str. 22, Munich 81675, Germany
| | - Aurore Menegaux
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine and Health, Technical University of Munich, Ismaninger Str. 22, Munich 81675, Germany; TUM-NIC Neuroimaging Center, School of Medicine and Health, Technical University of Munich, Ismaninger Str. 22, Munich 81675, Germany
| | - Peter Bartmann
- Department of Neonatology and Pediatric Intensive Care, University Hospital Bonn, Venusberg-Campus 1, Bonn 53127, Germany
| | - Dieter Wolke
- Department of Psychology, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Christian Sorg
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine and Health, Technical University of Munich, Ismaninger Str. 22, Munich 81675, Germany; TUM-NIC Neuroimaging Center, School of Medicine and Health, Technical University of Munich, Ismaninger Str. 22, Munich 81675, Germany; Department of Psychiatry, School of Medicine and Health, Technical University of Munich, Ismaninger Str. 22, Munich 81675, Germany
| | - Dennis M Hedderich
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine and Health, Technical University of Munich, Ismaninger Str. 22, Munich 81675, Germany; TUM-NIC Neuroimaging Center, School of Medicine and Health, Technical University of Munich, Ismaninger Str. 22, Munich 81675, Germany
| |
Collapse
|
9
|
Hollingworth BYA, Pallier PN, Jenkins SI, Chen R. Hypoxic Neuroinflammation in the Pathogenesis of Multiple Sclerosis. Brain Sci 2025; 15:248. [PMID: 40149770 PMCID: PMC11940507 DOI: 10.3390/brainsci15030248] [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: 12/16/2024] [Revised: 02/17/2025] [Accepted: 02/22/2025] [Indexed: 03/29/2025] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease that damages the myelin sheath around the central nervous system axons, leading to neurological dysfunction. Although the initial damage is driven by inflammation, hypoxia has been reported in several brain regions of MS patients, but the significance of this for prognosis and treatment remains unclear. Neuroinflammation can induce hypoxia, and hypoxia can induce and exacerbate neuroinflammation, forming a vicious cycle. Within MS lesions, demyelination is often followed by remyelination, which may restore neurological function. However, demyelinated axons are vulnerable to damage, which leads to the accumulation of the permanent neurological dysfunction typical in MS, with this vulnerability heightened during hypoxia. Clinically approved therapies for MS are immunomodulatory, which can reduce relapse frequency/severity, but there is a lack of pro-regenerative therapies for MS, for example promoting remyelination. All tissues have protective responses to hypoxia, which may be relevant to MS lesions, especially during remyelinating episodes. When oxygen levels are reduced in the brain, constitutively expressed hypoxia-inducible factors (HIF) are stabilised, upregulating hundreds of genes, including neuroprotective factors. Furthermore, astrocytes upregulate heparin-binding epidermal growth factor (EGF)-like growth factor (HB-EGF) in the early stage of MS. HB-EGF promotes protective mechanisms and induces oligodendrocyte and neuron differentiation and survival. This review article outlines the neuroinflammation and hypoxia cycle in MS pathology and identifies potential therapeutic targets to limit neurodegeneration and/or promote regeneration. Both HIF and HB-EGF signalling pathways induce endogenous protection mechanisms in the CNS, promoting neuroprotection and remyelination directly, but also indirectly by modulating the immune response in MS. Promoting such endogenous protective signalling pathways could be an effective therapy for MS patients.
Collapse
Affiliation(s)
| | - Patrick N. Pallier
- Centre for Neuroscience, Surgery and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK;
| | - Stuart I. Jenkins
- Neural Tissue Engineering Keele (NTEK), School of Medicine, Keele University, Staffordshire ST5 5BG, UK;
| | - Ruoli Chen
- School of Allied Health Professions and Pharmacy, Keele University, Staffordshire ST5 5BG, UK;
| |
Collapse
|
10
|
Yang M, Wang K, Liu B, Shen Y, Liu G. Hypoxic-Ischemic Encephalopathy: Pathogenesis and Promising Therapies. Mol Neurobiol 2025; 62:2105-2122. [PMID: 39073530 DOI: 10.1007/s12035-024-04398-9] [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: 01/05/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a brain lesion caused by inadequate blood supply and oxygen deprivation, often occurring in neonates. It has emerged as a grave complication of neonatal asphyxia, leading to chronic neurological damage. Nevertheless, the precise pathophysiological mechanisms underlying HIE are not entirely understood. This paper aims to comprehensively elucidate the contributions of hypoxia-ischemia, reperfusion injury, inflammation, oxidative stress, mitochondrial dysfunction, excitotoxicity, ferroptosis, endoplasmic reticulum stress, and apoptosis to the onset and progression of HIE. Currently, hypothermia therapy stands as the sole standard treatment for neonatal HIE, albeit providing only partial neuroprotection. Drug therapy and stem cell therapy have been explored in the treatment of HIE, exhibiting certain neuroprotective effects. Employing drug therapy or stem cell therapy as adjunctive treatments to hypothermia therapy holds great significance. This article presents a systematic review of the pathogenesis and treatment strategies of HIE, with the goal of enhancing the effect of treatment and improving the quality of life for HIE patients.
Collapse
Affiliation(s)
- Mingming Yang
- Department of Pediatrics, Binhai County People's Hospital, Yancheng, Jiangsu Province, 224500, P. R. China
| | - Kexin Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Boya Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China.
| | - Guangliang Liu
- Department of Pediatrics, Binhai County People's Hospital, Yancheng, Jiangsu Province, 224500, P. R. China.
| |
Collapse
|
11
|
Wang LW, Hsiung CW, Chang CP, Lin MT, Chen SJ. Neuroserpin normalization by mesenchymal stem cell therapy after encephalopathy of prematurity in neonatal rats. Pediatr Res 2025; 97:1199-1208. [PMID: 39085403 PMCID: PMC12055564 DOI: 10.1038/s41390-024-03412-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Hypoxic-ischemia (HI), infection/inflammation and reperfusion injury are pathogenic factors of encephalopathy of prematurity, which involves maturational/neurotrophic disturbances in oligodendrocyte progenitor cells (OPC) and neurons/axons. Mesenchymal stem cells (MSCs) might facilitate neuroserpin production, which is neurotrophic for OPC/neurons. This study investigated MSC effects on developmental disturbances after lipopolysaccharide (LPS)-sensitized HI/reperfusion (LHIR) injury and the relation to neuroserpin expression. METHODS Postnatal day 2 (P2) rat pups received intraperitoneal LPS (5 µg/kg) injection followed by HI (unilateral common-carotid-artery ligation and 6.5% oxygen exposure for 90 min) and post-HI reperfusion (release of ligation). MSCs (5 × 104 cells) were injected into the left lateral ventricle at 24 h post-LHIR. Neurological tests and brain tissue examinations were performed between P5 and P56. RESULTS After LHIR injury, MSC therapy significantly reduced cell death in subplate neurons, attenuated axonal damage, and facilitated synaptophysin synthesis in the cortex. It also alleviated OPC maturation arrest and preserved the complexity of myelinated axons in the white matter, leading to cognitive, motor and behavioral functional improvements. These beneficial effects were linked to restored neuroserpin expression in subplate neurons. CONCLUSIONS MSC therapy ameliorated developmental disturbances after LHIR injury through protection of neuroserpin expression, serving as a promising approach for treating encephalopathy of prematurity. IMPACT Neuroserpin is secreted by subplate neurons and may regulate the development of neurons and oligodendrocyte-axon contact for myelination in the premature brain. LPS-sensitized hypoxic-ischemia/reperfusion (LHIR) injury caused the developmental disturbances of neurons/axons and oligodendrocytes, and lowered neuroserpin levels in a neonatal rat model simulating encephalopathy of prematurity. Mesenchymal stem cell therapy alleviated the developmental disturbances after LHIR injury through protection of neuroserpin expression in subplate neurons, offering a new perspective on potential treatment for encephalopathy of prematurity.
Collapse
Affiliation(s)
- Lan-Wan Wang
- Department of Pediatrics, Chi Mei Medical Center, Tainan, Taiwan, ROC.
- Department of Biotechnology and Food Technology, Southern Taiwan University of Science and Technology, Tainan, Taiwan, ROC.
- School of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan, ROC.
| | - Chien-Wei Hsiung
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan, ROC
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Ching-Ping Chang
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan, ROC
| | - Mao-Tsun Lin
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan, ROC
| | - Shyi-Jou Chen
- Department of Pediatrics, Tri-service General Hospital, School of Medicine, National Defense Medical Center, Taipei, Taiwan, ROC.
| |
Collapse
|
12
|
Kundu S, Ghaemmaghami J, Sanidas G, Wolff N, Vij A, Byrd C, Simonti G, Triantafyllou M, Jablonska B, Dean T, Koutroulis I, Gallo V, Kratimenos P. Cerebellar Purkinje Cell Activity Regulates White Matter Response and Locomotor Function after Neonatal Hypoxia. J Neurosci 2025; 45:e0899242024. [PMID: 39472064 DOI: 10.1523/jneurosci.0899-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 01/03/2025] Open
Abstract
Neonatal hypoxia (Hx) causes white matter (WM) injury, particularly in the cerebellum. We previously demonstrated that Hx-induced reduction of cerebellar Purkinje cell (PC) activity results in locomotor deficits. Yet, the mechanism of Hx-induced cerebellar WM injury and associated locomotor abnormalities remains undetermined. Here, we show that the cerebellar WM injury and linked locomotor deficits are driven by PC activity and are reversed when PC activity is restored. Using optogenetics and multielectrode array recordings, we manipulated PC activity and captured the resulting cellular responses in WM oligodendrocyte precursor cells and GABAergic interneurons. To emulate the effects of Hx, we used light-activated halorhodopsin targeted specifically to the PC layer of normal mice. Suppression of PC firing activity at P13 and P21 phenocopied the locomotor deficits observed in Hx. Moreover, histopathologic analysis of the developing cerebellar WM following PC inhibition (P21) revealed a corresponding reduction in oligodendrocyte maturation and myelination, akin to our findings in Hx mice. Conversely, PC stimulation restored PC activity, promoted oligodendrocyte maturation, and enhanced myelination, resulting in reversed Hx-induced locomotor deficits. Our findings highlight the crucial role of PC activity in cerebellar WM development and locomotor performance following neonatal injury.
Collapse
Affiliation(s)
- Srikanya Kundu
- National Institutes of Health, National Center for Advancing Translational Sciences (NCATS), Bethesda, Maryland 20850
- Children's National Research Institute, Washington, DC 20012
| | - Javid Ghaemmaghami
- Children's National Research Institute, Washington, DC 20012
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109
| | | | - Nora Wolff
- Children's National Research Institute, Washington, DC 20012
| | - Abhya Vij
- Boston Children's Hospital, Boston, Massachusetts 02115
- Harvard Medical School, Boston, Massachusetts 02115
| | - Chad Byrd
- Children's National Research Institute, Washington, DC 20012
| | | | | | - Beata Jablonska
- Children's National Research Institute, Washington, DC 20012
- The George Washington University School of Medicine and Health Sciences, Washington, DC 20052
| | - Terry Dean
- Children's National Research Institute, Washington, DC 20012
- The George Washington University School of Medicine and Health Sciences, Washington, DC 20052
- Children's National Hospital, Washington, DC 20010
| | - Ioannis Koutroulis
- Children's National Research Institute, Washington, DC 20012
- The George Washington University School of Medicine and Health Sciences, Washington, DC 20052
- Children's National Hospital, Washington, DC 20010
| | - Vittorio Gallo
- Children's National Research Institute, Washington, DC 20012
- The George Washington University School of Medicine and Health Sciences, Washington, DC 20052
- Children's National Hospital, Washington, DC 20010
- Seattle Children's Research Institute, Seattle, Washington 98105
| | - Panagiotis Kratimenos
- Children's National Research Institute, Washington, DC 20012
- The George Washington University School of Medicine and Health Sciences, Washington, DC 20052
- Children's National Hospital, Washington, DC 20010
| |
Collapse
|
13
|
Prasad J, Van Steenwinckel J, Gunn AJ, Bennet L, Korzeniewski SJ, Gressens P, Dean JM. Chronic Inflammation Offers Hints About Viable Therapeutic Targets for Preeclampsia and Potentially Related Offspring Sequelae. Int J Mol Sci 2024; 25:12999. [PMID: 39684715 PMCID: PMC11640791 DOI: 10.3390/ijms252312999] [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: 08/29/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
The combination of hypertension with systemic inflammation during pregnancy is a hallmark of preeclampsia, but both processes also convey dynamic information about its antecedents and correlates (e.g., fetal growth restriction) and potentially related offspring sequelae. Causal inferences are further complicated by the increasingly frequent overlap of preeclampsia, fetal growth restriction, and multiple indicators of acute and chronic inflammation, with decreased gestational length and its correlates (e.g., social vulnerability). This complexity prompted our group to summarize information from mechanistic studies, integrated with key clinical evidence, to discuss the possibility that sustained or intermittent systemic inflammation-related phenomena offer hints about viable therapeutic targets, not only for the prevention of preeclampsia, but also the neurobehavioral and other developmental deficits that appear to be overrepresented in surviving offspring. Importantly, we feel that carefully designed hypothesis-driven observational studies are necessary if we are to translate the mechanistic evidence into child health benefits, namely because multiple pregnancy disorders might contribute to heightened risks of neuroinflammation, arrested brain development, or dysconnectivity in survivors who exhibit developmental problems later in life.
Collapse
Affiliation(s)
- Jaya Prasad
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| | | | - Alistair J. Gunn
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| | - Laura Bennet
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| | - Steven J. Korzeniewski
- C.S. Mott Center for Human Growth and Development, Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Pierre Gressens
- Inserm, Neurodiderot, Université de Paris, 75019 Paris, France;
- Centre for the Developing Brain, Division of Imaging Sciences and Department of Biomedical Engineering, King’s College London, King’s Health Partners, St. Thomas’ Hospital, London SE1 7EH, UK
| | - Justin M. Dean
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1142, New Zealand; (J.P.); (A.J.G.); (L.B.); (J.M.D.)
| |
Collapse
|
14
|
Shrivastava V, Tyagi S, Dey D, Singh A, Palanichamy JK, Sinha S, Sharma JB, Seth P, Sen S. Glial cholesterol redistribution in hypoxic injury in vitro influences oligodendrocyte maturation and myelination. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167476. [PMID: 39181517 DOI: 10.1016/j.bbadis.2024.167476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024]
Abstract
Hypoxic insult to the fetal brain causes loss of vulnerable premyelinating oligodendrocytes and arrested oligodendrocyte differentiation. Astrocytes influence oligodendrocyte differentiation and the astrocytic response to hypoxia could affect oligodendrocyte maturation under hypoxia. To identify pathways by which astrocytes influence oligodendroglial maturation in hypoxic injury, human fetal neural stem cell-derived astrocytes were exposed to 0.2 % oxygen for 48 hours. Transcriptomic analysis revealed the upregulation of the cholesterol-biosynthesis pathway in hypoxia-exposed astrocytes. Hypoxia-exposed primary astrocytes and astrocytic cell line (SVG) showed increased expression of hydroxy-methyl-glutaryl-CoA reductase (HMGCR), squalene epoxidase (SQLE), apolipoprotein E (apoE) and ATP-binding cassette transporter 1 (ABCA1) on qPCR and Western blot. Hypoxic SVG also showed increased cholesterol content in cells and culture supernatants and increased cell surface expression of ABCA1. Interestingly hypoxia-exposed premyelinating oligodendrocytes (Mo3.13) showed reduced cholesterol along with decreased expression of HMGCR and SQLE on qPCR and Western blot. Exogenous cholesterol increased the differentiation of Mo3.13 as measured by increased expression of myelin basic protein (MBP) on flow cytometry. Hypoxia exposure resulted in increased cholesterol transport from astrocytes to oligodendrocytes in cocultures with BODIPY-cholesterol labelled SVG and membrane-labelled Mo3.13. As exogenous cholesterol enhanced oligodendrocyte differentiation, our findings indicate that increased cholesterol synthesis by astrocytes and transport to oligodendrocytes could supplement oligodendroglial maturation in conditions of hypoxic brain injury in neonates.
Collapse
Affiliation(s)
- Vadanya Shrivastava
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Sagar Tyagi
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Devanjan Dey
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Archna Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | | | - Subrata Sinha
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - J B Sharma
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, New Delhi, India
| | - Pankaj Seth
- Department of Molecular and Cellular Neuroscience, National Brain Research Centre, Manesar, Haryana, India
| | - Sudip Sen
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
15
|
Perdaens O, van Pesch V. Should We Consider Neurodegeneration by Itself or in a Triangulation with Neuroinflammation and Demyelination? The Example of Multiple Sclerosis and Beyond. Int J Mol Sci 2024; 25:12637. [PMID: 39684351 PMCID: PMC11641818 DOI: 10.3390/ijms252312637] [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: 10/31/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Neurodegeneration is preeminent in many neurological diseases, and still a major burden we fail to manage in patient's care. Its pathogenesis is complicated, intricate, and far from being completely understood. Taking multiple sclerosis as an example, we propose that neurodegeneration is neither a cause nor a consequence by itself. Mitochondrial dysfunction, leading to energy deficiency and ion imbalance, plays a key role in neurodegeneration, and is partly caused by the oxidative stress generated by microglia and astrocytes. Nodal and paranodal disruption, with or without myelin alteration, is further involved. Myelin loss exposes the axons directly to the inflammatory and oxidative environment. Moreover, oligodendrocytes provide a singular metabolic and trophic support to axons, but do not emerge unscathed from the pathological events, by primary myelin defects and cell apoptosis or secondary to neuroinflammation or axonal damage. Hereby, trophic failure might be an overlooked contributor to neurodegeneration. Thus, a complex interplay between neuroinflammation, demyelination, and neurodegeneration, wherein each is primarily and secondarily involved, might offer a more comprehensive understanding of the pathogenesis and help establishing novel therapeutic strategies for many neurological diseases and beyond.
Collapse
Affiliation(s)
- Océane Perdaens
- Neurochemistry Group, Institute of NeuroScience, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
| | - Vincent van Pesch
- Neurochemistry Group, Institute of NeuroScience, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
- Department of Neurology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| |
Collapse
|
16
|
Pavy CL, Shaw JC, Dyson RM, Palliser HK, Moloney RA, Sixtus RP, Berry MJ, Hirst JJ. Ganaxolone Therapy After Preterm Birth Restores Cerebellar Oligodendrocyte Maturation and Myelination in Guinea Pigs. Dev Psychobiol 2024; 66:e22554. [PMID: 39378309 DOI: 10.1002/dev.22554] [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: 03/26/2024] [Revised: 09/11/2024] [Accepted: 09/15/2024] [Indexed: 10/10/2024]
Abstract
The postnatal environment is challenging for the preterm neonate with exposure to hypoxic and excitotoxic events, amplified by premature loss of placentally derived neurosteroids. Between preterm birth and term equivalent age (TEA), cerebellar development continues despite these challenges. We hypothesize that neurosteroid replacement therapy during this time will support optimal cerebellar development. Guinea pig sows delivered at term (∼69 days gestation) or were induced to deliver preterm (∼62 days), with preterm pups receiving ganaxolone or vehicle until TEA. Postnatal assessments comprised salivary cortisol (corrected postnatal age [CPA] 0, 7, 38), behavioral analysis (CPA7, 38), and tissue collection (CPA0 and CPA40). Neurodevelopmental markers (MBP, Olig2, and NeuN) were assessed in the cerebellum by immunohistochemistry, whereas RT-PCR was utilized to investigate key inhibitory/excitatory pathways and oligodendrocyte lineage markers. Following preterm birth, there was evidence of a hyperactive phenotype, increased salivary cortisol concentrations, and impaired myelination and oligodendrocyte maturation at the protein level. mRNA expressions of key inhibitory/excitatory pathways and myelin stability were also altered following preterm birth. Importantly, we showed that neurosteroid replacement therapy returns cerebellar development and behavior toward a term-like phenotype. Therefore, ganaxolone may reduce the vulnerability of the cerebellum to postnatal challenges arising from preterm birth.
Collapse
Affiliation(s)
- Carlton L Pavy
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Julia C Shaw
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Rebecca M Dyson
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
- Biomedical Research Unit, University of Otago, Wellington, New Zealand
| | - Hannah K Palliser
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Roisin A Moloney
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Ryan P Sixtus
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
- Biomedical Research Unit, University of Otago, Wellington, New Zealand
| | - Mary J Berry
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
- Biomedical Research Unit, University of Otago, Wellington, New Zealand
| | - Jonathan J Hirst
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
- Mothers and Babies Research Centre, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| |
Collapse
|
17
|
Janowska J, Gargas J, Zajdel K, Wieteska M, Lipinski K, Ziemka‐Nalecz M, Frontczak‐Baniewicz M, Sypecka J. Oligodendrocyte progenitor cells' fate after neonatal asphyxia-Puzzling implications for the development of hypoxic-ischemic encephalopathy. Brain Pathol 2024; 34:e13255. [PMID: 38504469 PMCID: PMC11483519 DOI: 10.1111/bpa.13255] [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: 09/14/2023] [Accepted: 03/01/2024] [Indexed: 03/21/2024] Open
Abstract
Premature birth or complications during labor can cause temporary disruption of cerebral blood flow, often followed by long-term disturbances in brain development called hypoxic-ischemic (HI) encephalopathy. Diffuse damage to the white matter is the most frequently detected pathology in this condition. We hypothesized that oligodendrocyte progenitor cell (OPC) differentiation disturbed by mild neonatal asphyxia may affect the viability, maturation, and physiological functioning of oligodendrocytes. To address this issue, we studied the effect of temporal HI in the in vivo model in P7 rats with magnetic resonance imaging (MRI), microscopy techniques and biochemical analyses. Moreover, we recreated the injury in vitro performing the procedure of oxygen-glucose deprivation on rat neonatal OPCs to determine its effect on cell viability, proliferation, and differentiation. In the in vivo model, MRI evaluation revealed changes in the volume of different brain regions, as well as changes in the directional diffusivity of water in brain tissue that may suggest pathological changes to myelinated neuronal fibers. Hypomyelination was observed in the cortex, striatum, and CA3 region of the hippocampus. Severe changes to myelin ultrastructure were observed, including delamination of myelin sheets. Interestingly, shortly after the injury, an increase in oligodendrocyte proliferation was observed, followed by an overproduction of myelin proteins 4 weeks after HI. Results verified with the in vitro model indicate, that in the first days after damage, OPCs do not show reduced viability, intensively proliferate, and overexpress myelin proteins and oligodendrocyte-specific transcription factors. In conclusion, despite the increase in oligodendrocyte proliferation and myelin protein expression after HI, the production of functional myelin sheaths in brain tissue is impaired. Presented study provides a detailed description of oligodendrocyte pathophysiology developed in an effect of HI injury, resulting in an altered CNS myelination. The described models may serve as useful tools for searching and testing effective of effective myelination-supporting therapies for HI injuries.
Collapse
Affiliation(s)
- Justyna Janowska
- Department of NeuroRepairMossakowski Medical Research Institute PASWarsawPoland
| | - Justyna Gargas
- Department of NeuroRepairMossakowski Medical Research Institute PASWarsawPoland
| | - Karolina Zajdel
- NOMATEN Center of Excellence, National Center for Nuclear ResearchOtwockPoland
- Electron Microscopy Research UnitMossakowski Medical Research Institute PASWarsawPoland
| | - Michal Wieteska
- Small Animal Magnetic Resonance Imaging LaboratoryMossakowski Medical Research Institute PASWarsawPoland
| | - Kamil Lipinski
- Division of Nuclear and Medical ElectronicsWarsaw University of TechnologyWarsawPoland
| | | | | | - Joanna Sypecka
- Department of NeuroRepairMossakowski Medical Research Institute PASWarsawPoland
| |
Collapse
|
18
|
Volstad KB, Pripp AH, Ludviksen JA, Stiris T, Saugstad OD, Mollnes TE, Andresen JH. No Short-Term Effect of Low-Dose Nicotine on Inflammation after Global Hypoxia in Newborn Piglets. Neonatology 2024; 122:171-180. [PMID: 39317175 PMCID: PMC11965827 DOI: 10.1159/000541217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 08/29/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION Perinatal asphyxia initiates cytokine release and complement activation with risk of brain damage. We assessed the effect of nicotine on innate immunity and hypothesized that nicotine infusion in a newborn piglet model of asphyxia would decrease the immune response and be neuroprotective. METHODS Newborn piglets (n = 41) were randomized to one of three groups after hypoxia: two groups receiving nicotine, (1) 18 µg/kg/h (n = 17), (2) 46 µg/kg/h (n = 15), and (3) control group receiving saline (n = 9). C3a, IL-6, TNF, and IL-10 were measured in plasma and IL-6 and IL-8 in microdialysis fluid from cerebral periventricular white matter, using immuno-assays. RESULTS Plasma C3a and IL-6 increased significantly from start to end hypoxia (mean 4.4 ± 0.55 to 5.6 ± 0.71 ng/mL and 1.66 ± 1.04 to 2.68 ± 0.71 pg/mL, respectively), while IL-10 and TNF increased significantly after 4 h (mean 1.4 ± 1.08 to 2.9 ± 1.87 and 3.3 ± 0.67 to 4.0 ± 0.58 pg/mL, respectively) (p < 0.001 for all). IL-6 increased significantly (p < 0.001) in microdialysis samples from end hypoxia to end experiment (mean 0.65 ± 0.88 to 2.78 ± 1.84 ng/mL). No significant differences were observed between the nicotine groups and the control group neither in plasma nor in microdialysis samples. CONCLUSION Hypoxia leads to rapid release of cytokines in plasma and cerebral microdialysis fluid, and complement activation measured on C3a. However, low-dose nicotine administration did not affect the immune response. INTRODUCTION Perinatal asphyxia initiates cytokine release and complement activation with risk of brain damage. We assessed the effect of nicotine on innate immunity and hypothesized that nicotine infusion in a newborn piglet model of asphyxia would decrease the immune response and be neuroprotective. METHODS Newborn piglets (n = 41) were randomized to one of three groups after hypoxia: two groups receiving nicotine, (1) 18 µg/kg/h (n = 17), (2) 46 µg/kg/h (n = 15), and (3) control group receiving saline (n = 9). C3a, IL-6, TNF, and IL-10 were measured in plasma and IL-6 and IL-8 in microdialysis fluid from cerebral periventricular white matter, using immuno-assays. RESULTS Plasma C3a and IL-6 increased significantly from start to end hypoxia (mean 4.4 ± 0.55 to 5.6 ± 0.71 ng/mL and 1.66 ± 1.04 to 2.68 ± 0.71 pg/mL, respectively), while IL-10 and TNF increased significantly after 4 h (mean 1.4 ± 1.08 to 2.9 ± 1.87 and 3.3 ± 0.67 to 4.0 ± 0.58 pg/mL, respectively) (p < 0.001 for all). IL-6 increased significantly (p < 0.001) in microdialysis samples from end hypoxia to end experiment (mean 0.65 ± 0.88 to 2.78 ± 1.84 ng/mL). No significant differences were observed between the nicotine groups and the control group neither in plasma nor in microdialysis samples. CONCLUSION Hypoxia leads to rapid release of cytokines in plasma and cerebral microdialysis fluid, and complement activation measured on C3a. However, low-dose nicotine administration did not affect the immune response.
Collapse
Affiliation(s)
- Karianne B. Volstad
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Neonatal Intensive Care, Oslo University Hospital, Oslo, Norway
| | - Are H. Pripp
- Oslo Center of Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| | | | - Tom Stiris
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Neonatal Intensive Care, Oslo University Hospital, Oslo, Norway
| | - Ola D. Saugstad
- Department of Pediatric Research, University of Oslo, Oslo, Norway
- Ann and Robert H. Lurie Children’s Hospital of Chicago, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tom E. Mollnes
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Research Laboratory, Nordland Hospital, Bodø, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | | |
Collapse
|
19
|
Cai XY, Ma SY, Tang MH, Hu L, Wu KD, Zhang Z, Zhang YQ, Lin Y, Patel N, Yang ZC, Mo XM. Atoh1 mediated disturbance of neuronal maturation by perinatal hypoxia induces cognitive deficits. Commun Biol 2024; 7:1121. [PMID: 39261625 PMCID: PMC11390922 DOI: 10.1038/s42003-024-06846-7] [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: 01/12/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024] Open
Abstract
Neurodevelopmental disorders are currently one of the major complications faced by patients with congenital heart disease (CHD). Chronic hypoxia in the prenatal and postnatal preoperative brain may be associated with neurological damage and impaired long-term cognitive function, but the exact mechanisms are unknown. In this study, we find that delayed neuronal migration and impaired synaptic development are attributed to altered Atoh1 under chronic hypoxia. This is due to the fact that excessive Atoh1 facilitates expression of Kif21b, which causes excess in free-state α-tubulin, leading to disrupted microtubule dynamic stability. Furthermore, the delay in neonatal brain maturation induces cognitive disabilities in adult mice. Then, by down-regulating Atoh1 we alleviate the impairment of cell migration and synaptic development, improving the cognitive behavior of mice to some extent. Taken together, our work unveil that Atoh1 may be one of the targets to ameliorate hypoxia-induced neurodevelopmental disabilities and cognitive impairment in CHD.
Collapse
Affiliation(s)
- Xin-Yu Cai
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Si-Yu Ma
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China.
| | - Ming-Hui Tang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Liang Hu
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Ke-de Wu
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Zhen Zhang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Ya-Qi Zhang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Ye Lin
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Nishant Patel
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Zhao-Cong Yang
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Xu-Ming Mo
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, 210008, China.
- Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
20
|
Odell EP, Jabassini N, Schniedewind B, Pease-Raissi SE, Frymoyer A, Christians U, Green AJ, Chan JR, Ostrem BEL. Minimum effective dose of clemastine in a mouse model of preterm white matter injury. Pediatr Res 2024; 96:933-941. [PMID: 38942888 PMCID: PMC11502491 DOI: 10.1038/s41390-024-03326-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/06/2024] [Accepted: 05/31/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND Preterm white matter injury (PWMI) is the most common cause of brain injury in premature neonates. PWMI involves a differentiation arrest of oligodendrocytes, the myelinating cells of the central nervous system. Clemastine was previously shown to induce oligodendrocyte differentiation and myelination in mouse models of PWMI at a dose of 10 mg/kg/day. The minimum effective dose (MED) of clemastine is unknown. Identification of the MED is essential for maximizing safety and efficacy in neonatal clinical trials. We hypothesized that the MED in neonatal mice is lower than 10 mg/kg/day. METHODS Mouse pups were exposed to normoxia or hypoxia (10% FiO2) from postnatal day 3 (P3) through P10. Vehicle or clemastine at one of four doses (0.5, 2, 7.5 or 10 mg/kg/day) was given to hypoxia-exposed pups. Myelination was assessed at age P14 and 10 weeks to determine the MED. Clemastine pharmacokinetics were evaluated at steady-state on day 8 of treatment. RESULTS Clemastine rescued hypoxia-induced hypomyelination with a MED of 7.5 mg/kg/day. Pharmacokinetic analysis of the MED revealed Cmax 44.0 ng/mL, t1/2 4.6 h, and AUC24 280.1 ng*hr/mL. CONCLUSIONS Based on these results, myelination-promoting exposures should be achievable with oral doses of clemastine in neonates with PWMI. IMPACT Preterm white matter injury (PWMI) is the most common cause of brain injury and cerebral palsy in premature neonates. Clemastine, an FDA-approved antihistamine, was recently identified to strongly promote myelination in a mouse model of PWMI and is a possible treatment. The minimum effective dose in neonatal rodents is unknown and is critical for guiding dose selection and balancing efficacy with toxicity in future clinical trials. We identified the minimum effective dose of clemastine and the associated pharmacokinetics in a murine chronic hypoxia model of PWMI, paving the way for a future clinical trial in human neonates.
Collapse
Affiliation(s)
- Elizabeth P Odell
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Nora Jabassini
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Björn Schniedewind
- iC42 Clinical Research and Development, Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sarah E Pease-Raissi
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Adam Frymoyer
- Department of Pediatrics, Stanford University, Palo Alto, CA, USA
| | - Uwe Christians
- iC42 Clinical Research and Development, Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ari J Green
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | - Jonah R Chan
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Bridget E L Ostrem
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
21
|
Selvanathan T, Guo T, Ufkes S, Chau V, Branson HM, Synnes AR, Ly LG, Kelly E, Grunau RE, Miller SP. Change in Volumes and Location of Preterm White Matter Injury over a Period of 15 Years. J Pediatr 2024; 272:114090. [PMID: 38754774 DOI: 10.1016/j.jpeds.2024.114090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/12/2024] [Accepted: 05/05/2024] [Indexed: 05/18/2024]
Abstract
OBJECTIVE To evaluate whether white matter injury (WMI) volumes and spatial distribution, which are important predictors of neurodevelopmental outcomes in preterm infants, have changed over a period of 15 years. STUDY DESIGN Five hundred and twenty-eight infants born <32 weeks' gestational age from 2 sequential prospective cohorts (cohort 1: 2006 through 2012; cohort 2: 2014 through 2019) underwent early-life (median 32.7 weeks postmenstrual age) and/or term-equivalent-age MRI (median 40.7 weeks postmenstrual age). WMI were manually segmented for quantification of volumes. There were 152 infants with WMI with 74 infants in cohort 1 and 78 in cohort 2. Multivariable linear regression models examined change in WMI volume across cohorts while adjusting for clinical confounders. Lesion maps assessed change in WMI location across cohorts. RESULTS There was a decrease in WMI volume in cohort 2 compared with cohort 1 (β = -0.6, 95% CI [-0.8, -0.3], P < .001) with a shift from more central to posterior location of WMI. There was a decrease in clinical illness severity of infants across cohorts. CONCLUSIONS We found a decrease in WMI volume and shift to more posterior location in very preterm infants over a period of 15 years. This may potentially reflect more advanced maturation of white matter at the time of injury which may be related to changes in clinical practice over time.
Collapse
Affiliation(s)
- Thiviya Selvanathan
- Pediatrics, BC Children's Hospital Research Institute and University of British Columbia, Vancouver, British Columbia, Canada; Pediatrics, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Ting Guo
- Pediatrics, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada; Neuroscience & Mental Health, SickKids Research Institute, Toronto, Ontario, Canada
| | - Steven Ufkes
- Pediatrics, BC Children's Hospital Research Institute and University of British Columbia, Vancouver, British Columbia, Canada
| | - Vann Chau
- Pediatrics, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada; Neuroscience & Mental Health, SickKids Research Institute, Toronto, Ontario, Canada
| | - Helen M Branson
- Diagnostic Imaging, The Hospital for Sick Children and Medical Imaging, University of Toronto, Toronto, Ontario, Canada
| | - Anne R Synnes
- Pediatrics, BC Children's Hospital Research Institute and University of British Columbia, Vancouver, British Columbia, Canada
| | - Linh G Ly
- Pediatrics, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Edmond Kelly
- Pediatrics, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada; Pediatrics, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Ruth E Grunau
- Pediatrics, BC Children's Hospital Research Institute and University of British Columbia, Vancouver, British Columbia, Canada
| | - Steven P Miller
- Pediatrics, BC Children's Hospital Research Institute and University of British Columbia, Vancouver, British Columbia, Canada; Pediatrics, The Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada; Neuroscience & Mental Health, SickKids Research Institute, Toronto, Ontario, Canada.
| |
Collapse
|
22
|
Thalhammer M, Nimpal M, Schulz J, Meedt V, Menegaux A, Schmitz-Koep B, Daamen M, Boecker H, Zimmer C, Priller J, Wolke D, Bartmann P, Hedderich D, Sorg C. Consistently lower volumes across thalamus nuclei in very premature-born adults. Neuroimage 2024; 297:120732. [PMID: 39004408 DOI: 10.1016/j.neuroimage.2024.120732] [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: 03/27/2024] [Revised: 07/08/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024] Open
Abstract
Lasting thalamus volume reduction after preterm birth is a prominent finding. However, whether thalamic nuclei volumes are affected differentially by preterm birth and whether nuclei aberrations are relevant for cognitive functioning remains unknown. Using T1-weighted MR-images of 83 adults born very preterm (≤ 32 weeks' gestation; VP) and/or with very low body weight (≤ 1,500 g; VLBW) as well as of 92 full-term born (≥ 37 weeks' gestation) controls, we compared thalamic nuclei volumes of six subregions (anterior, lateral, ventral, intralaminar, medial, and pulvinar) across groups at the age of 26 years. To characterize the functional relevance of volume aberrations, cognitive performance was assessed by full-scale intelligence quotient using the Wechsler Adult Intelligence Scale and linked to volume reductions using multiple linear regression analyses. Thalamic volumes were significantly lower across all examined nuclei in VP/VLBW adults compared to controls, suggesting an overall rather than focal impairment. Lower nuclei volumes were linked to higher intensity of neonatal treatment, indicating vulnerability to stress exposure after birth. Furthermore, we found that single results for lateral, medial, and pulvinar nuclei volumes were associated with full-scale intelligence quotient in preterm adults, albeit not surviving correction for multiple hypotheses testing. These findings provide evidence that lower thalamic volume in preterm adults is observable across all subregions rather than focused on single nuclei. Data suggest the same mechanisms of aberrant thalamus development across all nuclei after premature birth.
Collapse
Affiliation(s)
- Melissa Thalhammer
- Department of Diagnostic and Interventional Neuroradiology, Technical University of Munich, School of Medicine and Health, Munich, Germany; Technical University of Munich, School of Medicine and Health, TUM-NIC Neuroimaging Center, Munich, Germany.
| | - Mehul Nimpal
- Faculty of Biology, Graduate School of Systemic Neurosciences, Ludwig Maximilian University of Munich
| | - Julia Schulz
- Department of Diagnostic and Interventional Neuroradiology, Technical University of Munich, School of Medicine and Health, Munich, Germany; Technical University of Munich, School of Medicine and Health, TUM-NIC Neuroimaging Center, Munich, Germany
| | - Veronica Meedt
- Faculty of Biology, Ludwig Maximilian University of Munich
| | - Aurore Menegaux
- Department of Diagnostic and Interventional Neuroradiology, Technical University of Munich, School of Medicine and Health, Munich, Germany; Technical University of Munich, School of Medicine and Health, TUM-NIC Neuroimaging Center, Munich, Germany
| | - Benita Schmitz-Koep
- Department of Diagnostic and Interventional Neuroradiology, Technical University of Munich, School of Medicine and Health, Munich, Germany; Technical University of Munich, School of Medicine and Health, TUM-NIC Neuroimaging Center, Munich, Germany
| | - Marcel Daamen
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Clinical Functional Imaging Group, Bonn, Germany; Department of Neonatology and Pediatric Intensive Care, University Hospital Bonn, Bonn, Germany
| | - Henning Boecker
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Clinical Functional Imaging Group, Bonn, Germany
| | - Claus Zimmer
- Department of Diagnostic and Interventional Neuroradiology, Technical University of Munich, School of Medicine and Health, Munich, Germany; Technical University of Munich, School of Medicine and Health, TUM-NIC Neuroimaging Center, Munich, Germany
| | - Josef Priller
- Department of Psychiatry, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Dieter Wolke
- Department of Psychology, University of Warwick, Coventry, UK; Warwick Medical School, University of Warwick, Coventry, UK
| | - Peter Bartmann
- Department of Neonatology and Pediatric Intensive Care, University Hospital Bonn, Bonn, Germany
| | - Dennis Hedderich
- Department of Diagnostic and Interventional Neuroradiology, Technical University of Munich, School of Medicine and Health, Munich, Germany; Technical University of Munich, School of Medicine and Health, TUM-NIC Neuroimaging Center, Munich, Germany
| | - Christian Sorg
- Department of Diagnostic and Interventional Neuroradiology, Technical University of Munich, School of Medicine and Health, Munich, Germany; Technical University of Munich, School of Medicine and Health, TUM-NIC Neuroimaging Center, Munich, Germany; Department of Psychiatry, Technical University of Munich, School of Medicine and Health, Munich, Germany
| |
Collapse
|
23
|
Isasi E, Wajner M, Duarte JA, Olivera-Bravo S. Cerebral White Matter Alterations Associated With Oligodendrocyte Vulnerability in Organic Acidurias: Insights in Glutaric Aciduria Type I. Neurotox Res 2024; 42:33. [PMID: 38963434 DOI: 10.1007/s12640-024-00710-6] [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: 11/01/2023] [Revised: 04/27/2024] [Accepted: 06/14/2024] [Indexed: 07/05/2024]
Abstract
The white matter is an important constituent of the central nervous system, containing axons, oligodendrocytes, and its progenitor cells, astrocytes, and microglial cells. Oligodendrocytes are central for myelin synthesis, the insulating envelope that protects axons and allows normal neural conduction. Both, oligodendrocytes and myelin, are highly vulnerable to toxic factors in many neurodevelopmental and neurodegenerative disorders associated with disturbances of myelination. Here we review the main alterations in oligodendrocytes and myelin observed in some organic acidurias/acidemias, which correspond to inherited neurometabolic disorders biochemically characterized by accumulation of potentially neurotoxic organic acids and their derivatives. The yet incompletely understood mechanisms underlying the high vulnerability of OLs and/or myelin in glutaric acidemia type I, the most prototypical cerebral organic aciduria, are particularly discussed.
Collapse
Affiliation(s)
- Eugenia Isasi
- Laboratorio de Neurobiología Celular y Molecular, Unidad Académica de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Departamento de Neurobiología y Neuropatología, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
| | - Moacir Wajner
- Department of Biochemistry, Instituto de Ciencias Básicas da Saude, Universidade Federal de Río Grande do Sul, Porto Alegre, Brazil
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Juliana Avila Duarte
- Departamento de Medicina Interna, Serviço de Radiología, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Silvia Olivera-Bravo
- Departamento de Neurobiología y Neuropatología, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay.
| |
Collapse
|
24
|
Ren SY, Xia Y, Yu B, Lei QJ, Hou PF, Guo S, Wu SL, Liu W, Yang SF, Jiang YB, Chen JF, Shen KF, Zhang CQ, Wang F, Yan M, Ren H, Yang N, Zhang J, Zhang K, Lin S, Li T, Yang QW, Xiao L, Hu ZX, Mei F. Growth hormone promotes myelin repair after chronic hypoxia via triggering pericyte-dependent angiogenesis. Neuron 2024; 112:2177-2196.e6. [PMID: 38653248 DOI: 10.1016/j.neuron.2024.03.026] [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: 08/31/2023] [Revised: 01/26/2024] [Accepted: 03/27/2024] [Indexed: 04/25/2024]
Abstract
White matter injury (WMI) causes oligodendrocyte precursor cell (OPC) differentiation arrest and functional deficits, with no effective therapies to date. Here, we report increased expression of growth hormone (GH) in the hypoxic neonatal mouse brain, a model of WMI. GH treatment during or post hypoxic exposure rescues hypoxia-induced hypomyelination and promotes functional recovery in adolescent mice. Single-cell sequencing reveals that Ghr mRNA expression is highly enriched in vascular cells. Cell-lineage labeling and tracing identify the GHR-expressing vascular cells as a subpopulation of pericytes. These cells display tip-cell-like morphology with kinetic polarized filopodia revealed by two-photon live imaging and seemingly direct blood vessel branching and bridging. Gain-of-function and loss-of-function experiments indicate that GHR signaling in pericytes is sufficient to modulate angiogenesis in neonatal brains, which enhances OPC differentiation and myelination indirectly. These findings demonstrate that targeting GHR and/or downstream effectors may represent a promising therapeutic strategy for WMI.
Collapse
Affiliation(s)
- Shu-Yu Ren
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yu Xia
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Bin Yu
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China; Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qi-Jing Lei
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Peng-Fei Hou
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Sheng Guo
- Department of Immunology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shuang-Ling Wu
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wei Liu
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shao-Fan Yang
- Brain Research Center, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yi-Bin Jiang
- Department of Neurobiology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jing-Fei Chen
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Kai-Feng Shen
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chun-Qing Zhang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Fei Wang
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Mi Yan
- Department of Pediatrics, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400000, China
| | - Hong Ren
- Department of Emergence, 5(th) People's Hospital of Chongqing, Chongqing 400062, China
| | - Nian Yang
- Department of Physiology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jun Zhang
- Department of Neurobiology, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Kuan Zhang
- Brain Research Center, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Sen Lin
- Department of Neurology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Tao Li
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lan Xiao
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zhang-Xue Hu
- Department of Pediatrics, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400000, China.
| | - Feng Mei
- Department of Histology and Embryology, Chongqing Key Laboratory of Brain Development and Cognition, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| |
Collapse
|
25
|
Ventura GC, Dyshliuk N, Dmytriyeva O, Nordsten MJB, Haugaard MM, Christiansen LI, Thymann T, Sangild PT, Pankratova S. Enteral plasma supports brain repair in newborn pigs after birth asphyxia. Brain Behav Immun 2024; 119:693-708. [PMID: 38677626 DOI: 10.1016/j.bbi.2024.04.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/03/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024] Open
Abstract
Newborns exposed to birth asphyxia transiently experience deficient blood flow and a lack of oxygen, potentially inducing hypoxic-ischaemic encephalopathy and subsequent neurological damage. Immunomodulatory components in plasma may dampen these responses. Using caesarean-delivered pigs as a model, we hypothesized that dietary plasma supplementation improves brain outcomes in pigs exposed to birth asphyxia. Mild birth asphyxia was induced by temporary occlusion of the umbilical cord prior to caesarean delivery. Motor development was assessed in asphyxiated (ASP) and control (CON) piglets using neonatal arousal, physical activity and gait test parameters before euthanasia on Day 4. The ASP pigs exhibited increased plasma lactate at birth, deficient motor skills and increased glial fibrillary acidic protein levels in CSF and astrogliosis in the putamen. The expression of genes related to oxidative stress, inflammation and synaptic functions was transiently altered in the motor cortex and caudate nucleus. The number of apoptotic cells among CTIP2-positive neurons in the motor cortex and striatal medium spiny neurons was increased, and maturation of preoligodendrocytes in the internal capsule was delayed. Plasma supplementation improved gait performance in the beam test, attenuated neuronal apoptosis and affected gene expression related to neuroinflammation, neurotransmission and antioxidants (motor cortex, caudate). We present a new clinically relevant animal model of moderate birth asphyxia inducing structural and functional brain damage. The components in plasma that support brain repair remain to be identified but may represent a therapeutic potential for infants and animals after birth asphyxia.
Collapse
Affiliation(s)
- Gemma Chavarria Ventura
- Section of Comparative Pediatrics and Nutrition, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nadiya Dyshliuk
- Section of Comparative Pediatrics and Nutrition, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; National University of Life and Environmental Sciences of Ukraine, Kyiv, Ukraine
| | - Oksana Dmytriyeva
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mads Jacob Bagi Nordsten
- Section of Comparative Pediatrics and Nutrition, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maria Mathilde Haugaard
- Section of Comparative Pediatrics and Nutrition, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Line Iadsatian Christiansen
- Section of Comparative Pediatrics and Nutrition, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Thymann
- Section of Comparative Pediatrics and Nutrition, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Per Torp Sangild
- Section of Comparative Pediatrics and Nutrition, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Pediatrics and Adolescent Medicine, Rigshospitalet, Copenhagen, Denmark; Department of Pediatrics, Odense University Hospital, Odense, Denmark
| | - Stanislava Pankratova
- Section of Comparative Pediatrics and Nutrition, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
26
|
Selvanathan T, Mabbott C, Au-Young SH, Seed M, Miller SP, Chau V. Antenatal diagnosis, neonatal brain volumes, and neurodevelopment in transposition of the great arteries. Dev Med Child Neurol 2024; 66:882-891. [PMID: 38204357 DOI: 10.1111/dmcn.15840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 11/21/2023] [Accepted: 11/30/2023] [Indexed: 01/12/2024]
Abstract
AIM To examine whether antenatal diagnosis modifies relationships between neonatal brain volumes and 18-month neurodevelopmental outcomes in children with transposition of the great arteries (TGA). METHOD In a retrospective cohort of 139 children with TGA (77 antenatally diagnosed), we obtained total brain volumes (TBVs) on pre- (n = 102) and postoperative (n = 112) magnetic resonance imaging. Eighteen-month neurodevelopmental outcomes were assessed using the Bayley Scales of Infant and Toddler Development, Third Edition. Generalized estimating equations with interaction terms were used to determine whether antenatal diagnosis modified associations between TBVs and neurodevelopmental outcomes accounting for postmenstrual age at scan, brain injury, and ventricular septal defect. RESULTS Infants with postnatal diagnosis had more preoperative hypotension (35% vs 14%, p = 0.004). The interactions between antenatal diagnosis and TBVs were significantly related to cognitive (p = 0.003) outcomes. Specifically, smaller TBVs were associated with lower cognitive scores in infants diagnosed postnatally; this association was attenuated in those diagnosed antenatally. INTERPRETATION Antenatal diagnosis modifies associations between neonatal brain volume and 18-month cognitive outcome in infants with TGA. These findings suggest that antenatal diagnosis may be neuroprotective, possibly through improved preoperative clinical status. These data highlight the need to improve antenatal diagnosis rates. WHAT THIS PAPER ADDS Antenatal diagnosis of transposition of the great arteries modified relationships between neonatal brain volume and neurodevelopment. Smaller brain volumes related to poorer cognitive scores with postnatal diagnosis only. There was more preoperative hypotension in the postnatal diagnosis group.
Collapse
Affiliation(s)
- Thiviya Selvanathan
- Department of Pediatrics, The Hospital for Sick Children and University of Toronto, Toronto, ON, Canada
- Department of Pediatrics, BC Children's Hospital Research Institute and the University of British Columbia, Vancouver, BC, Canada
| | - Connor Mabbott
- Neurosciences and Mental Health, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Stephanie H Au-Young
- Department of Pediatrics, The Hospital for Sick Children and University of Toronto, Toronto, ON, Canada
- Neurosciences and Mental Health, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Mike Seed
- Department of Pediatrics, The Hospital for Sick Children and University of Toronto, Toronto, ON, Canada
- Heart Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - Steven P Miller
- Department of Pediatrics, The Hospital for Sick Children and University of Toronto, Toronto, ON, Canada
- Department of Pediatrics, BC Children's Hospital Research Institute and the University of British Columbia, Vancouver, BC, Canada
- Neurosciences and Mental Health, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Vann Chau
- Department of Pediatrics, The Hospital for Sick Children and University of Toronto, Toronto, ON, Canada
- Neurosciences and Mental Health, The Hospital for Sick Children Research Institute, Toronto, ON, Canada
| |
Collapse
|
27
|
Shui X, Chen J, Fu Z, Zhu H, Tao H, Li Z. Microglia in Ischemic Stroke: Pathogenesis Insights and Therapeutic Challenges. J Inflamm Res 2024; 17:3335-3352. [PMID: 38800598 PMCID: PMC11128258 DOI: 10.2147/jir.s461795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/14/2024] [Indexed: 05/29/2024] Open
Abstract
Ischemic stroke is the most common type of stroke, which is the main cause of death and disability on a global scale. As the primary immune cells in the brain that are crucial for preserving homeostasis of the central nervous system microenvironment, microglia have been found to exhibit dual or even multiple effects at different stages of ischemic stroke. The anti-inflammatory polarization of microglia and release of neurotrophic factors may provide benefits by promoting neurological recovery at the lesion in the early phase after ischemic stroke. However, the pro-inflammatory polarization of microglia and secretion of inflammatory factors in the later phase of injury may exacerbate the ischemic lesion, suggesting the therapeutic potential of modulating the balance of microglial polarization to predispose them to anti-inflammatory transformation in ischemic stroke. Microglia-mediated signaling crosstalk with other cells may also be key to improving functional outcomes following ischemic stroke. Thus, this review provides an overview of microglial functions and responses under physiological and ischemic stroke conditions, including microglial activation, polarization, and interactions with other cells. We focus on approaches that promote anti-inflammatory polarization of microglia, inhibit microglial activation, and enhance beneficial cell-to-cell interactions. These targets may hold promise for the creation of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Xinyao Shui
- Clinical Medical College, Southwest Medical University, Luzhou, People’s Republic of China
| | - Jingsong Chen
- Department of Laboratory Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, People’s Republic of China
- Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| | - Ziyue Fu
- Clinical Medical College, Southwest Medical University, Luzhou, People’s Republic of China
| | - Haoyue Zhu
- Clinical Medical College, Southwest Medical University, Luzhou, People’s Republic of China
| | - Hualin Tao
- Department of Laboratory Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, People’s Republic of China
- Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| | - Zhaoyinqian Li
- Department of Laboratory Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, People’s Republic of China
- Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| |
Collapse
|
28
|
Kim H, Kim BJ, Koh S, Cho HJ, Jin X, Kim BG, Choi JY. A primary culture method for the easy, efficient, and effective acquisition of oligodendrocyte lineage cells from neonatal rodent brains. Heliyon 2024; 10:e29359. [PMID: 38655345 PMCID: PMC11036010 DOI: 10.1016/j.heliyon.2024.e29359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/26/2024] Open
Abstract
Oligodendrocytes (OL) are myelin-forming glial cells in the central nervous system. In vitro primary OL culture models offer the benefit of a more readily controlled environment that facilitates the examination of diverse OL stages and their intricate dynamics. Although conventional methods for primary OL culture exist, their performance in terms of simplicity and efficiency can be improved. Here, we introduce a novel method for primary OL culture, namely the E3 (easy, efficient, and effective) method, which greatly improves the simplicity and efficiency of the primary OL culture procedure using neonatal rodent brains. We also provided the optimal media composition for the augmentation of oligodendrocyte progenitor cell (OPC) proliferation and more robust maturation into myelin-forming OLs. Overall, E3 offers an undemanding method for obtaining primary OLs with high yield and quality. Alongside its value as a practical tool, in vitro characteristics of the OL lineage additionally identified during the development of the E3 method have implications for advancing research on OL physiology and pathophysiology.
Collapse
Affiliation(s)
- Hanki Kim
- Department of Brain Science, Ajou University School of Medicine, Suwon, 16499, South Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, 16499, South Korea
| | - Bum Jun Kim
- Department of Brain Science, Ajou University School of Medicine, Suwon, 16499, South Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, 16499, South Korea
| | - Seungyon Koh
- Department of Brain Science, Ajou University School of Medicine, Suwon, 16499, South Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, 16499, South Korea
- Department of Neurology, Ajou University School of Medicine, Suwon, 16499, South Korea
| | - Hyo Jin Cho
- Department of Brain Science, Ajou University School of Medicine, Suwon, 16499, South Korea
| | - Xuelian Jin
- Department of Brain Science, Ajou University School of Medicine, Suwon, 16499, South Korea
- Geriatrics Department, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, 223800, China
| | - Byung Gon Kim
- Department of Brain Science, Ajou University School of Medicine, Suwon, 16499, South Korea
- Department of Neurology, Ajou University School of Medicine, Suwon, 16499, South Korea
| | - Jun Young Choi
- Department of Brain Science, Ajou University School of Medicine, Suwon, 16499, South Korea
- Department of Neurology, Ajou University School of Medicine, Suwon, 16499, South Korea
| |
Collapse
|
29
|
Tscherrig V, Steinfort M, Haesler V, Surbek D, Schoeberlein A, Joerger-Messerli MS. All but Small: miRNAs from Wharton's Jelly-Mesenchymal Stromal Cell Small Extracellular Vesicles Rescue Premature White Matter Injury after Intranasal Administration. Cells 2024; 13:543. [PMID: 38534387 PMCID: PMC10969443 DOI: 10.3390/cells13060543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/07/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
White matter injury (WMI) is a common neurological issue in premature-born neonates, often causing long-term disabilities. We recently demonstrated a key beneficial role of Wharton's jelly mesenchymal stromal cell-derived small extracellular vesicles (WJ-MSC-sEVs) microRNAs (miRNAs) in WMI-related processes in vitro. Here, we studied the functions of WJ-MSC-sEV miRNAs in vivo using a preclinical rat model of premature WMI. Premature WMI was induced in rat pups through inflammation and hypoxia-ischemia. Small EVs were purified from the culture supernatant of human WJ-MSCs. The capacity of WJ-MSC-sEV-derived miRNAs to decrease microglia activation and promote oligodendrocyte maturation was evaluated by knocking down (k.d) DROSHA in WJ-MSCs, releasing sEVs containing significantly less mature miRNAs. Wharton's jelly MSC-sEVs intranasally administrated 24 h upon injury reached the brain within 1 h, remained detectable for at least 24 h, significantly reduced microglial activation, and promoted oligodendrocyte maturation. The DROSHA k.d in WJ-MSCs lowered the therapeutic capabilities of sEVs in experimental premature WMI. Our results strongly indicate the relevance of miRNAs in the therapeutic abilities of WJ-MSC-sEVs in premature WMI in vivo, opening the path to clinical application.
Collapse
Affiliation(s)
- Vera Tscherrig
- Department of Obstetrics and Feto-maternal Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (M.S.)
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, 3012 Bern, Switzerland
| | - Marel Steinfort
- Department of Obstetrics and Feto-maternal Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (M.S.)
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, 3012 Bern, Switzerland
| | - Valérie Haesler
- Department of Obstetrics and Feto-maternal Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (M.S.)
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Daniel Surbek
- Department of Obstetrics and Feto-maternal Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (M.S.)
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Andreina Schoeberlein
- Department of Obstetrics and Feto-maternal Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (M.S.)
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| | - Marianne Simone Joerger-Messerli
- Department of Obstetrics and Feto-maternal Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (M.S.)
- Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
| |
Collapse
|
30
|
Wang XX, Li GS, Wang KH, Hu XS, Hu Y. Positive effect of microvascular proliferation on functional recovery in experimental cervical spondylotic myelopathy. Front Neurosci 2024; 18:1254600. [PMID: 38510463 PMCID: PMC10951064 DOI: 10.3389/fnins.2024.1254600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 02/09/2024] [Indexed: 03/22/2024] Open
Abstract
Background and purpose Cervical Spondylotic Myelopathy (CSM), the most common cause of spinal cord dysfunction globally, is a degenerative disease that results in non-violent, gradual, and long-lasting compression of the cervical spinal cord. The objective of this study was to investigate whether microvascular proliferation could positively affect neural function recovery in experimental cervical spondylotic myelopathy (CSM). Methods A total of 60 male adult Sprague-Dawley (SD) were randomly divided into four groups: Control (CON), Compression (COM), Angiostasis (AS), and Angiogenesis (A G),with 15 rats in each group. Rats in the AS group received SU5416 to inhibit angiogenesis, while rats in the AG group received Deferoxamine (DFO) to promote angiogenesis. Motor and sensory functions were assessed using the Basso Beattie Bresnahan (BBB) scale and somatosensory evoked potential (SEP) examination. Neuropathological degeneration was evaluated by the number of neurons, Nissl bodies (NB), and the de-myelination of white matter detected by Hematoxylin & Eosin(HE), Toluidine Blue (TB), and Luxol Fast Blue (LFB) staining. Immunohistochemical (IHC) staining was used to observe the Neurovascular Unit (NVU). Results Rats in the CON group exhibited normal locomotor function with full BBB score, normal SEP latency and amplitude. Among the other three groups, the AG group had the highest BBB score and the shortest SEP latency, while the AS group had the lowest BBB score and the most prolonged SEP latency. The SEP amplitude showed an opposite performance to the latency. Compared to the COM and AS groups, the AG group demonstrated significant neuronal restoration in gray matter and axonal remyelination in white matter. DFO promoted microvascular proliferation, especially in gray matter, and improved the survival of neuroglial cells. In contrast, SU-5416 inhibited the viability of neuroglial cells by reducing micro vessels. Conclusion The microvascular status was closely related to NVU remodeling an-d functional recovery. Therefore, proliferation of micro vessels contributed to function -al recovery in experimental CSM, which may be associated with NVU remodeling.
Collapse
Affiliation(s)
- Xu-xiang Wang
- Department of Minimally Invasive Spine Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Guang-sheng Li
- Department of Minimally Invasive Spine Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Kang-heng Wang
- Department of Minimally Invasive Spine Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xiao-song Hu
- Department of Minimally Invasive Spine Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Orthopedics Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yong Hu
- Department of Minimally Invasive Spine Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Orthopedics Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
31
|
Odell E, Jabassini N, Schniedewind B, Pease-Raissi SE, Frymoyer A, Christians U, Green AJ, Chan JR, Ostrem BEL. Minimum Effective Dose of Clemastine in a Mouse Model of Preterm White Matter Injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.08.578953. [PMID: 38464078 PMCID: PMC10925142 DOI: 10.1101/2024.02.08.578953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Background Preterm white matter injury (PWMI) is the most common cause of brain injury in premature neonates. PWMI involves a differentiation arrest of oligodendrocytes, the myelinating cells of the central nervous system. Clemastine was previously shown to induce oligodendrocyte differentiation and myelination in mouse models of PWMI at a dose of 10 mg/kg/day. The minimum effective dose (MED) of clemastine is unknown. Identification if the MED is essential for maximizing safety and efficacy in neonatal clinical trials. We hypothesized that the MED in neonatal mice is lower than 10 mg/kg/day. Methods Mouse pups were exposed to normoxia or hypoxia (10% FiO 2 ) from postnatal day 3 (P3) through P10. Vehicle or clemastine fumarate at one of four doses (0.5, 2, 7.5 or 10 mg/kg/day) was given orally to hypoxia-exposed pups. At P14, myelination was assessed by immunohistochemistry and electron microscopy to determine the MED. Clemastine pharmacokinetics were evaluated at steady-state on day 8 of treatment. Results Clemastine rescued hypoxia-induced hypomyelination with a MED of 7.5 mg/kg/day. Pharmacokinetic analysis of the MED revealed C max 44.0 ng/mL, t 1/2 4.6 hours, and AUC 24 280.1 ng*hr/mL. Conclusion Based on these results, myelination-promoting exposures should be achievable with oral doses of clemastine in neonates with PWMI. Key Points Preterm white matter injury (PWMI) is the most common cause of brain injury and cerebral palsy in premature neonates.Clemastine, an FDA-approved antihistamine, was recently identified to strongly promote myelination in a mouse model of PWMI and is a possible treatment.The minimum effective dose in neonatal rodents is unknown and is critical for guiding dose selection and balancing efficacy with toxicity in future clinical trials.We identified the minimum effective dose of clemastine and the associated pharmacokinetics in a murine chronic hypoxia model of PWMI, paving the way for a future clinical trial in human neonates.
Collapse
|
32
|
Moloney RA, Pavy CL, Kahl RGS, Palliser HK, Hirst JJ, Shaw JC. Dual isolation of primary neurons and oligodendrocytes from guinea pig frontal cortex. Front Cell Neurosci 2024; 17:1298685. [PMID: 38269115 PMCID: PMC10806141 DOI: 10.3389/fncel.2023.1298685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/18/2023] [Indexed: 01/26/2024] Open
Abstract
Primary cell culture is a technique that is widely used in neuroscience research to investigate mechanisms that underlie pathologies at a cellular level. Typically, mouse or rat tissue is used for this process; however, altricial rodent species have markedly different neurodevelopmental trajectories comparatively to humans. The use of guinea pig brain tissue presents a novel aspect to this routinely used cell culture method whilst also allowing for dual isolation of two major cell types from a physiologically relevant animal model for studying perinatal neurodevelopment. Primary neuronal and oligodendrocyte cell cultures were derived from fetal guinea pig's frontal cortex brain tissue collected at a gestational age of 62 days (GA62), which is a key time in the neuronal and oligodendrocyte development. The major advantage of this protocol is the ability to acquire both neuronal and oligodendrocyte cellular cultures from the frontal cortex of one fetal brain. Briefly, neuronal cells were grown in 12-well plates initially in a 24-h serum-rich medium to enhance neuronal survival before switching to a serum-free media formulation. Oligodendrocytes were first grown in cell culture flasks using a serum-rich medium that enabled the growth of oligodendrocyte progenitor cells (OPCs) on an astrocyte bed. Following confluency, the shake method of differential adhesion and separation was utilized via horizontally shaking the OPCs off the astrocyte bed overnight. Therefore, OPCs were plated in 12-well plates and were initially expanded in media supplemented with growth hormones, before switching to maturation media to progress the lineage to a mature phenotype. Reverse transcription-polymerase chain reaction (RT-PCR) was performed on both cell culture types to analyze key population markers, and the results were further validated using immunocytochemistry. Primary neurons displayed the mRNA expression of multiple neuronal markers, including those specific to GABAergic populations. These cells also positively stained for microtubule-associated protein 2 (MAP2; a dendritic marker specific to neurons) and NeuN (a marker of neuronal cell bodies). Primary oligodendrocytes expressed all investigated markers of the oligodendrocyte lineage, with a majority of the cells displaying an immature oligodendrocyte phenotype. This finding was further confirmed with positive oligodendrocyte transcription factor (OLIG2) staining, which serves as a marker for the overall oligodendrocyte population. This study demonstrates a novel method for isolating both neurons and oligodendrocytes from the guinea pig brain tissue. These isolated cells display key markers and gene expression that will allow for functional experiments to occur and may be particularly useful in studying neurodevelopmental conditions with perinatal origins.
Collapse
Affiliation(s)
- Roisin A. Moloney
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Mothers and Babies Research Centre, Newcastle, NSW, Australia
| | - Carlton L. Pavy
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Mothers and Babies Research Centre, Newcastle, NSW, Australia
| | - Richard G. S. Kahl
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Mothers and Babies Research Centre, Newcastle, NSW, Australia
| | - Hannah K. Palliser
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Mothers and Babies Research Centre, Newcastle, NSW, Australia
| | - Jon J. Hirst
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Mothers and Babies Research Centre, Newcastle, NSW, Australia
| | - Julia C. Shaw
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Mothers and Babies Research Centre, Newcastle, NSW, Australia
| |
Collapse
|
33
|
Adil D, Duerden EG, Eagleson R, de Ribaupierre S. Structural Alterations of the Corpus Callosum in Children With Infantile Hydrocephalus. J Child Neurol 2024; 39:66-76. [PMID: 38387869 PMCID: PMC11083734 DOI: 10.1177/08830738241231343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 02/24/2024]
Abstract
This study investigates structural alterations of the corpus callosum in children diagnosed with infantile hydrocephalus. We aim to assess both macrostructural (volume) and microstructural (diffusion tensor imaging metrics) facets of the corpus callosum, providing insights into the nature and extent of alterations associated with this condition. Eighteen patients with infantile hydrocephalus (mean age = 9 years) and 18 age- and sex-matched typically developing healthy children participated in the study. Structural magnetic resonance imaging and diffusion tensor imaging were used to assess corpus callosum volume and microstructure, respectively. Our findings reveal significant alterations in corpus callosum volume, particularly in the posterior area, as well as distinct microstructural disparities, notably pronounced in these same segments. These results highlight the intricate interplay between macrostructural and microstructural aspects in understanding the impact of infantile hydrocephalus. Examining these structural alterations provides an understanding into the mechanisms underlying the effects of infantile hydrocephalus on corpus callosum integrity, given its pivotal role in interhemispheric communication. This knowledge offers a more nuanced perspective on neurologic disorders and underscores the significance of investigating the corpus callosum's health in such contexts.
Collapse
Affiliation(s)
- Derya Adil
- Western Institute for Neuroscience, Western University, London, Ontario, Canada
| | - Emma G. Duerden
- Western Institute for Neuroscience, Western University, London, Ontario, Canada
- Applied Psychology, Faculty of Education, Western University, London, Ontario, Canada
| | - Roy Eagleson
- Western Institute for Neuroscience, Western University, London, Ontario, Canada
- Electrical and Computer Engineering, Faculty of Engineering, Western University, London, Ontario, Canada
| | - Sandrine de Ribaupierre
- Western Institute for Neuroscience, Western University, London, Ontario, Canada
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
34
|
Sun J, Wang W, Ma Q, Pan X, Zhai H, Wang J, Han Y, Li Y, Wang Y. Necrostatin-1s Suppresses RIPK1-driven Necroptosis and Inflammation in Periventricular Leukomalacia Neonatal Mice. Neurochem Res 2024; 49:129-141. [PMID: 37642893 DOI: 10.1007/s11064-023-04013-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/31/2023]
Abstract
Periventricular leukomalacia (PVL), a predominant form of brain injury in preterm survivors, is characterized by hypomyelination and microgliosis and is also the major cause of long-term neurobehavioral abnormalities in premature infants. Receptor-interacting protein kinase 1 (RIPK1) plays a pivotal role in mediating cell death and inflammatory signaling cascade. However, very little is known about the potential effect of RIPK1 in PVL and the underlying mechanism. Herein, we found that the expression level of RIPK1 was drastically increased in the brain of PVL neonatal mice models, and treatment of PVL neonatal mice with Necrostatin-1s (Nec-1s), an inhibitor of RIPK1, greatly ameliorated cerebral ischemic injury, exhibiting an increase of body weights, reduction of cerebral infarct size, neuronal loss, and occurrence of necrosis-like cells, and significantly improved the long-term abnormal neurobehaviors of PVL. In addition, Nec-1s significantly suppressed hypomyelination and promoted the differentiation of oligodendrocyte precursor cells (OPCs), as demonstrated by the increased expression levels of MBP and Olig2, the decreased expression level of GPR17, a significant increase in the number of CC-1-positive cells, and suppression of myelin ultrastructure impairment. Moreover, the mechanism of neuroprotective effects of Nec-1s against PVL is closely associated with its suppression of the RIPK1-mediated necrosis signaling molecules, RIPK1, PIPK3, and MLKL. More importantly, inhibition of RIPK1 could reduce microglial inflammatory injury by triggering the M1 to M2 microglial phenotype, appreciably decreasing the levels of M1 marker CD86 and increasing the levels of M2 markers Arg1 or CD206 in PVL mice. Taken together, inhibition of RIPK1 markedly ameliorates the brain injury and long-term neurobehavioral abnormalities of PVL mice through the reduction of neural cell necroptosis and reversing neuroinflammation.
Collapse
Affiliation(s)
- Jinping Sun
- School of Basic Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, P.R. China
- Department of Pathology, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, P.R. China
| | - Wei Wang
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, P.R. China
| | - Quanrui Ma
- School of Basic Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, P.R. China
| | - Xiaoli Pan
- Department of Pathology, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, P.R. China
| | - Hualiang Zhai
- Department of Pathology, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, P.R. China
| | - Junyan Wang
- School of Basic Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, P.R. China
| | - Yong Han
- School of Basic Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, P.R. China
| | - Yunhong Li
- School of Basic Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, P.R. China.
| | - Yin Wang
- School of Basic Medicine, Ningxia Medical University, Yinchuan, 750004, Ningxia, P.R. China.
| |
Collapse
|
35
|
Deng X, Hu Z, Zhou S, Wu Y, Fu M, Zhou C, Sun J, Gao X, Huang Y. Perspective from single-cell sequencing: Is inflammation in acute ischemic stroke beneficial or detrimental? CNS Neurosci Ther 2024; 30:e14510. [PMID: 37905592 PMCID: PMC10805403 DOI: 10.1111/cns.14510] [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: 07/05/2023] [Revised: 09/24/2023] [Accepted: 10/08/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND Acute ischemic stroke (AIS) is a common cerebrovascular event associated with high incidence, disability, and poor prognosis. Studies have shown that various cell types, including microglia, astrocytes, oligodendrocytes, neurons, and neutrophils, play complex roles in the early stages of AIS and significantly affect its prognosis. Thus, a comprehensive understanding of the mechanisms of action of these cells will be beneficial for improving stroke prognosis. With the rapid development of single-cell sequencing technology, researchers have explored the pathophysiological mechanisms underlying AIS at the single-cell level. METHOD We systematically summarize the latest research on single-cell sequencing in AIS. RESULT In this review, we summarize the phenotypes and functions of microglia, astrocytes, oligodendrocytes, neurons, neutrophils, monocytes, and lymphocytes, as well as their respective subtypes, at different time points following AIS. In particular, we focused on the crosstalk between microglia and astrocytes, oligodendrocytes, and neurons. Our findings reveal diverse and sometimes opposing roles within the same cell type, with the possibility of interconversion between different subclusters. CONCLUSION This review offers a pioneering exploration of the functions of various glial cells and cell subclusters after AIS, shedding light on their regulatory mechanisms that facilitate the transformation of detrimental cell subclusters towards those that are beneficial for improving the prognosis of AIS. This approach has the potential to advance the discovery of new specific targets and the development of drugs, thus representing a significant breakthrough in addressing the challenges in AIS treatment.
Collapse
Affiliation(s)
- Xinpeng Deng
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Ziliang Hu
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Shengjun Zhou
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
| | - Yiwen Wu
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
| | - Menglin Fu
- School of Economics and ManagementChina University of GeosciencesWuhanChina
| | - Chenhui Zhou
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
| | - Jie Sun
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
| | - Xiang Gao
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
| | - Yi Huang
- Department of NeurosurgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| |
Collapse
|
36
|
Benarroch E. What Are the Roles of Oligodendrocyte Precursor Cells in Normal and Pathologic Conditions? Neurology 2023; 101:958-965. [PMID: 37985182 PMCID: PMC10663025 DOI: 10.1212/wnl.0000000000208000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 11/22/2023] Open
|
37
|
Schmitz‐Koep B, Menegaux A, Zimmermann J, Thalhammer M, Neubauer A, Wendt J, Schinz D, Daamen M, Boecker H, Zimmer C, Priller J, Wolke D, Bartmann P, Sorg C, Hedderich DM. Altered gray-to-white matter tissue contrast in preterm-born adults. CNS Neurosci Ther 2023; 29:3199-3211. [PMID: 37365964 PMCID: PMC10580354 DOI: 10.1111/cns.14320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 06/01/2023] [Accepted: 06/10/2023] [Indexed: 06/28/2023] Open
Abstract
AIMS To investigate cortical organization in brain magnetic resonance imaging (MRI) of preterm-born adults using percent contrast of gray-to-white matter signal intensities (GWPC), which is an in vivo proxy measure for cortical microstructure. METHODS Using structural MRI, we analyzed GWPC at different percentile fractions across the cortex (0%, 10%, 20%, 30%, 40%, 50%, and 60%) in a large and prospectively collected cohort of 86 very preterm-born (<32 weeks of gestation and/or birth weight <1500 g, VP/VLBW) adults and 103 full-term controls at 26 years of age. Cognitive performance was assessed by full-scale intelligence quotient (IQ) using the Wechsler Adult Intelligence Scale. RESULTS GWPC was significantly decreased in VP/VLBW adults in frontal, parietal, and temporal associative cortices, predominantly in the right hemisphere. Differences were pronounced at 20%, 30%, and 40%, hence, in middle cortical layers. GWPC was significantly increased in right paracentral lobule in VP/VLBW adults. GWPC in frontal and temporal cortices was positively correlated with birth weight, and negatively with duration of ventilation (p < 0.05). Furthermore, GWPC in right paracentral lobule was negatively correlated with IQ (p < 0.05). CONCLUSIONS Widespread aberrant gray-to-white matter contrast suggests lastingly altered cortical microstructure after preterm birth, mainly in middle cortical layers, with differential effects on associative and primary cortices.
Collapse
Affiliation(s)
- Benita Schmitz‐Koep
- Department of Diagnostic and Interventional NeuroradiologyTechnical University of Munich; School of MedicineMunichGermany
- Technical University of Munich, School of Medicine, TUM‐NIC Neuroimaging CenterMunichGermany
| | - Aurore Menegaux
- Department of Diagnostic and Interventional NeuroradiologyTechnical University of Munich; School of MedicineMunichGermany
- Technical University of Munich, School of Medicine, TUM‐NIC Neuroimaging CenterMunichGermany
| | - Juliana Zimmermann
- Department of Diagnostic and Interventional NeuroradiologyTechnical University of Munich; School of MedicineMunichGermany
- Technical University of Munich, School of Medicine, TUM‐NIC Neuroimaging CenterMunichGermany
| | - Melissa Thalhammer
- Department of Diagnostic and Interventional NeuroradiologyTechnical University of Munich; School of MedicineMunichGermany
- Technical University of Munich, School of Medicine, TUM‐NIC Neuroimaging CenterMunichGermany
| | - Antonia Neubauer
- Department of Diagnostic and Interventional NeuroradiologyTechnical University of Munich; School of MedicineMunichGermany
- Technical University of Munich, School of Medicine, TUM‐NIC Neuroimaging CenterMunichGermany
| | - Jil Wendt
- Department of Diagnostic and Interventional NeuroradiologyTechnical University of Munich; School of MedicineMunichGermany
- Technical University of Munich, School of Medicine, TUM‐NIC Neuroimaging CenterMunichGermany
| | - David Schinz
- Department of Diagnostic and Interventional NeuroradiologyTechnical University of Munich; School of MedicineMunichGermany
- Technical University of Munich, School of Medicine, TUM‐NIC Neuroimaging CenterMunichGermany
| | - Marcel Daamen
- Department of Diagnostic and Interventional RadiologyUniversity Hospital Bonn, Clinical Functional Imaging GroupBonnGermany
- Department of Neonatology and Pediatric Intensive CareUniversity Hospital BonnBonnGermany
| | - Henning Boecker
- Department of Diagnostic and Interventional RadiologyUniversity Hospital Bonn, Clinical Functional Imaging GroupBonnGermany
| | - Claus Zimmer
- Department of Diagnostic and Interventional NeuroradiologyTechnical University of Munich; School of MedicineMunichGermany
- Technical University of Munich, School of Medicine, TUM‐NIC Neuroimaging CenterMunichGermany
| | - Josef Priller
- Department of PsychiatryTechnical University of Munich, School of MedicineMunichGermany
| | - Dieter Wolke
- Department of PsychologyUniversity of WarwickCoventryUK
- Warwick Medical SchoolUniversity of WarwickCoventryUK
| | - Peter Bartmann
- Department of Neonatology and Pediatric Intensive CareUniversity Hospital BonnBonnGermany
| | - Christian Sorg
- Department of Diagnostic and Interventional NeuroradiologyTechnical University of Munich; School of MedicineMunichGermany
- Technical University of Munich, School of Medicine, TUM‐NIC Neuroimaging CenterMunichGermany
- Department of PsychiatryTechnical University of Munich, School of MedicineMunichGermany
| | - Dennis M. Hedderich
- Department of Diagnostic and Interventional NeuroradiologyTechnical University of Munich; School of MedicineMunichGermany
- Technical University of Munich, School of Medicine, TUM‐NIC Neuroimaging CenterMunichGermany
| |
Collapse
|
38
|
Zhou Q, Ong M, Ye XY, Ting JY, Shah PS, Synnes A, Luu TM, Lee S. Long-Term Neurodevelopmental Impairment among Very Preterm Infants with Sepsis, Meningitis, and Intraventricular Hemorrhage. Neonatology 2023; 121:65-73. [PMID: 37866353 DOI: 10.1159/000534178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 09/11/2023] [Indexed: 10/24/2023]
Abstract
INTRODUCTION Sepsis and intraventricular hemorrhage (IVH) are associated with poorer long-term neurodevelopmental outcomes in very preterm infants (VPIs), but less is known about the long-term effect of meningitis and the combined impact of both meningitis and IVH. Our objective was to examine the long-term neurodevelopmental outcomes of VPIs with late onset sepsis and meningitis, with and without IVH, in Canada. METHODS We conducted a retrospective cohort study of all infants <29 weeks GA who were admitted to 26 tertiary-level neonatal intensive care units in the Canadian Neonatal Network (CNN) and Canadian Neonatal Follow-Up Network (CNFUN) databases, from January 1, 2010, to December 31, 2016. RESULTS Of the 6,322 infants in the cohort, 4,575 had no infection, 1,590 had late onset culture-positive bloodstream infection (CPBSI) only, and 157 had late onset meningitis. There was a significant (p < 0.05) trend of increasing rates of significant neurodevelopmental delay (sNDI) when comparing infants with no infection (sNDI rate 15.0%), late onset CPBSI (sNDI rate 22.9%), and late onset meningitis (sNDI rate 32.0%), even after adjustment for infant characteristics. Similar trends were observed for neurodevelopmental impairment, cerebral palsy, and individual Bayley-III scores <85 for cognitive, language, and motor development. There was an additive effect of IVH in all infant categories, but there was no multiplicative effect between IVH and late onset meningitis. CONCLUSION There was an increasing trend of adverse neurodevelopmental outcomes when infants with no infection, late onset CPBSI and late onset meningitis are compared. IVH had an additive effect.
Collapse
Affiliation(s)
- Qi Zhou
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Melissa Ong
- Department of Acute Medicine, Lewisham and Greenwich Trust, London, UK
| | - Xiang Y Ye
- Department of Biostatistics, Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Joseph Y Ting
- Division of Neonatal-Perinatal Care, Department of Pediatrics, Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Prakesh S Shah
- Department of Pediatrics, Mount Sinai Hospital and University of Toronto, Toronto, Ontario, Canada
| | - Anne Synnes
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Thuy Mai Luu
- Department of Pediatrics, Université de Montréal, Montreal, Québec, Canada
| | - Shoo Lee
- Department of Pediatrics, Mount Sinai Hospital and University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
39
|
Lin Q, Lin L, Li L, Zheng YF, Hu DW, Zhang G. Dynamic changes of oligodendrogenesis in neonatal rats with hypoxic-ischemic white matter injury. Brain Res 2023; 1817:148495. [PMID: 37481153 DOI: 10.1016/j.brainres.2023.148495] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/24/2023]
Abstract
BACKGROUND White matter injury (WMI) is an important type of preterm brain injury, which may result in severe neurological sequelae and lack of effective treatments. It is ascertained that selective vulnerability of oligodendrocytes is closely related to the WMI in preterm infants. But the alteration of the endogenous oligodendrogenesis over long time after hypoxic-ischemic WMI is still not clearly elucidated. METHODS We adopted an animal model of hypoxic-ischemic WMI in 3-day-old neonatal Sprague-Dawley rats. Immunofluorescence staining and western blotting were used to detect dynamic changes of oligodendrogenesis in the white matter region on postoperative day (POD) 1, 3, 7, 14, 28, 56 and 84. RESULTS In the sham group, the oligodendrocyte lineage in the white matter reached a developmental peak from POD 3 to 14. The proliferation and development of oligodendrocyte precursor cells (OPCs) occurred primarily within POD 14. The number of mature oligodendrocytes showed an upward trend and a dynamic change in proliferation over time. While in the WMI group, the oligodendrocyte lineage was upregulated on POD1 and 3 but downregulated on POD 7 and 14. The proliferation of OPCs increased on POD 1 and decreased on POD 3 and 7, with the total number of OPCs significantly reduced from POD 3 to 14. The number of mature oligodendrocytes decreased from POD 3 to 28, and return to the level of the sham group on POD 56 and 84, whereas the MBP expression was still significantly downregulated on POD 56 and 84. CONCLUSIONS Hypoxia-ischemia can have a long-term dynamic effect on the endogenous oligodendrogenesis of neonatal rat brain white matter. The proliferation of OPCs was promoted on POD 1 but inhibited from POD 3 to 14, which may be an early intervention target to improve oligodendrogenesis. The number of mature oligodendrocytes recover to the normal on POD 56 and 84 but the myelination is still blocked, which suggests it is essential to promote the maturation of oligodendrocyte and its function recovery at the same time within POD 28. Such efforts will provide the opportunity to test new interventions in pre-clinical studies for their promising clinical application.
Collapse
Affiliation(s)
- Qing Lin
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou, China; Laboratory of Clinical Applied Anatomy, Department of Human Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Ling Lin
- Public Technology Service Center, Fujian Medical University, Fuzhou, China
| | - Li Li
- Assisted Reproduction Centre, Obstetrics and Gynecology Department, 900TH Hospital of Joint Logistics Support Force, Fuzhou, China
| | - Yu-Fen Zheng
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou, China
| | - Ding-Wang Hu
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou, China; Laboratory of Clinical Applied Anatomy, Department of Human Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| | - Geng Zhang
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou, China; Laboratory of Clinical Applied Anatomy, Department of Human Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
40
|
Schinz D, Schmitz‐Koep B, Zimmermann J, Brandes E, Tahedl M, Menegaux A, Dukart J, Zimmer C, Wolke D, Daamen M, Boecker H, Bartmann P, Sorg C, Hedderich DM. Indirect evidence for altered dopaminergic neurotransmission in very premature-born adults. Hum Brain Mapp 2023; 44:5125-5138. [PMID: 37608591 PMCID: PMC10502650 DOI: 10.1002/hbm.26451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 06/23/2023] [Accepted: 07/28/2023] [Indexed: 08/24/2023] Open
Abstract
While animal models indicate altered brain dopaminergic neurotransmission after premature birth, corresponding evidence in humans is scarce due to missing molecular imaging studies. To overcome this limitation, we studied dopaminergic neurotransmission changes in human prematurity indirectly by evaluating the spatial co-localization of regional alterations in blood oxygenation fluctuations with the distribution of adult dopaminergic neurotransmission. The study cohort comprised 99 very premature-born (<32 weeks of gestation and/or birth weight below 1500 g) and 107 full-term born young adults, being assessed by resting-state functional MRI (rs-fMRI) and IQ testing. Normative molecular imaging dopamine neurotransmission maps were derived from independent healthy control groups. We computed the co-localization of local (rs-fMRI) activity alterations in premature-born adults with respect to term-born individuals to different measures of dopaminergic neurotransmission. We performed selectivity analyses regarding other neuromodulatory systems and MRI measures. In addition, we tested if the strength of the co-localization is related to perinatal measures and IQ. We found selectively altered co-localization of rs-fMRI activity in the premature-born cohort with dopamine-2/3-receptor availability in premature-born adults. Alterations were specific for the dopaminergic system but not for the used MRI measure. The strength of the co-localization was negatively correlated with IQ. In line with animal studies, our findings support the notion of altered dopaminergic neurotransmission in prematurity which is associated with cognitive performance.
Collapse
Affiliation(s)
- David Schinz
- Department of Neuroradiology, School of MedicineTechnical University of MunichMunichGermany
- TUM‐NIC Neuroimaging Center, School of MedicineTechnical University of MunichMunichGermany
| | - Benita Schmitz‐Koep
- Department of Neuroradiology, School of MedicineTechnical University of MunichMunichGermany
- TUM‐NIC Neuroimaging Center, School of MedicineTechnical University of MunichMunichGermany
| | - Juliana Zimmermann
- Department of Neuroradiology, School of MedicineTechnical University of MunichMunichGermany
- TUM‐NIC Neuroimaging Center, School of MedicineTechnical University of MunichMunichGermany
| | - Elin Brandes
- Department of Neuroradiology, School of MedicineTechnical University of MunichMunichGermany
- TUM‐NIC Neuroimaging Center, School of MedicineTechnical University of MunichMunichGermany
| | - Marlene Tahedl
- Department of Neuroradiology, School of MedicineTechnical University of MunichMunichGermany
- TUM‐NIC Neuroimaging Center, School of MedicineTechnical University of MunichMunichGermany
| | - Aurore Menegaux
- Department of Neuroradiology, School of MedicineTechnical University of MunichMunichGermany
- TUM‐NIC Neuroimaging Center, School of MedicineTechnical University of MunichMunichGermany
| | - Juergen Dukart
- Institute of Neuroscience and MedicineBrain & Behaviour (INM‐7), Research Centre JülichJülichGermany
- Institute of Systems Neuroscience, Medical FacultyHeinrich Heine University DüsseldorfDüsseldorfGermany
| | - Claus Zimmer
- Department of Neuroradiology, School of MedicineTechnical University of MunichMunichGermany
- TUM‐NIC Neuroimaging Center, School of MedicineTechnical University of MunichMunichGermany
| | - Dieter Wolke
- Department of PsychologyUniversity of WarwickCoventryUK
- Warwick Medical SchoolUniversity of WarwickCoventryUK
| | - Marcel Daamen
- Clinical Functional Imaging Group, Department of Diagnostic and Interventional RadiologyUniversity Hospital BonnBonnGermany
- Department of NeonatologyUniversity Hospital BonnBonnGermany
| | - Henning Boecker
- Clinical Functional Imaging Group, Department of Diagnostic and Interventional RadiologyUniversity Hospital BonnBonnGermany
| | - Peter Bartmann
- Department of NeonatologyUniversity Hospital BonnBonnGermany
| | - Christian Sorg
- Department of Neuroradiology, School of MedicineTechnical University of MunichMunichGermany
- TUM‐NIC Neuroimaging Center, School of MedicineTechnical University of MunichMunichGermany
- Department of Psychiatry, School of MedicineTechnical University of MunichMunichGermany
| | - Dennis M. Hedderich
- Department of Neuroradiology, School of MedicineTechnical University of MunichMunichGermany
- TUM‐NIC Neuroimaging Center, School of MedicineTechnical University of MunichMunichGermany
| |
Collapse
|
41
|
Viaene AN, Nelson EJ, Santi M. Perinatal hypoxic-ischemic brain injury: What's behind the "ribbon effect"? J Neuropathol Exp Neurol 2023; 82:865-875. [PMID: 37595577 DOI: 10.1093/jnen/nlad067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2023] Open
Abstract
Ribbon effect describes a perceived macroscopic color reversal of the gray and white matter, characterized by a pale cortex and diffusely dusky underlying white matter. This finding is thought to be unique to the perinatal period and indicative of hypoxic-ischemic injury. However, the clinical and microscopic correlates of this macroscopic finding have not been clearly defined. A 21-year retrospective study of autopsies was performed. Ribbon effect was seen in 190 subjects, ages 20 weeks gestation to 9.5 months adjusted age. Clinical associations and radiographic findings were similar in ribbon effect cases and controls. A variety of histologic findings were observed including acute neuronal injury, diffuse white matter gliosis, and white matter necrosis. Only white matter vascular congestion was significantly correlated to the macroscopic severity of ribbon effect; the severity of white matter injury and acute neuronal injury were not significantly correlated to ribbon effect. While hypoxic-ischemic changes were present in nearly all cases of ribbon effect, the location, severity, and chronicity of these changes varied considerably, and similar findings were observed in controls. The presence of ribbon effect therefore does not predict microscopic findings apart from vascular congestion, highlighting the importance of microscopic examination in perinatal brain autopsies.
Collapse
Affiliation(s)
- Angela N Viaene
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine, The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ernest J Nelson
- Department of Pathology and Laboratory Medicine, The Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mariarita Santi
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine, The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
42
|
Fabres RB, Cardoso DS, Aragón BA, Arruda BP, Martins PP, Ikebara JM, Drobyshevsky A, Kihara AH, de Fraga LS, Netto CA, Takada SH. Consequences of oxygen deprivation on myelination and sex-dependent alterations. Mol Cell Neurosci 2023; 126:103864. [PMID: 37268283 DOI: 10.1016/j.mcn.2023.103864] [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: 01/20/2023] [Revised: 05/07/2023] [Accepted: 05/25/2023] [Indexed: 06/04/2023] Open
Abstract
Oxygen deprivation is one of the main causes of morbidity and mortality in newborns, occurring with a higher prevalence in preterm infants, reaching 20 % to 50 % mortality in newborns in the perinatal period. When they survive, 25 % exhibit neuropsychological pathologies, such as learning difficulties, epilepsy, and cerebral palsy. White matter injury is one of the main features found in oxygen deprivation injury, which can lead to long-term functional impairments, including cognitive delay and motor deficits. The myelin sheath accounts for much of the white matter in the brain by surrounding axons and enabling the efficient conduction of action potentials. Mature oligodendrocytes, which synthesize and maintain myelination, also comprise a significant proportion of the brain's white matter. In recent years, oligodendrocytes and the myelination process have become potential therapeutic targets to minimize the effects of oxygen deprivation on the central nervous system. Moreover, evidence indicate that neuroinflammation and apoptotic pathways activated during oxygen deprivation may be influenced by sexual dimorphism. To summarize the most recent research about the impact of sexual dimorphism on the neuroinflammatory state and white matter injury after oxygen deprivation, this review presents an overview of the oligodendrocyte lineage development and myelination, the impact of oxygen deprivation and neuroinflammation on oligodendrocytes in neurodevelopmental disorders, and recent reports about sexual dimorphism regarding the neuroinflammation and white matter injury after neonatal oxygen deprivation.
Collapse
Affiliation(s)
- Rafael Bandeira Fabres
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre 90035-003, Brazil
| | - Débora Sterzeck Cardoso
- Neurogenetics Laboratory, Universidade Federal do ABC, Alameda da Universidade, s/n, São Bernardo do Campo 09606-045, Brazil
| | | | - Bruna Petrucelli Arruda
- Neurogenetics Laboratory, Universidade Federal do ABC, Alameda da Universidade, s/n, São Bernardo do Campo 09606-045, Brazil
| | - Pamela Pinheiro Martins
- Neurogenetics Laboratory, Universidade Federal do ABC, Alameda da Universidade, s/n, São Bernardo do Campo 09606-045, Brazil
| | - Juliane Midori Ikebara
- Neurogenetics Laboratory, Universidade Federal do ABC, Alameda da Universidade, s/n, São Bernardo do Campo 09606-045, Brazil
| | | | - Alexandre Hiroaki Kihara
- Neurogenetics Laboratory, Universidade Federal do ABC, Alameda da Universidade, s/n, São Bernardo do Campo 09606-045, Brazil
| | - Luciano Stürmer de Fraga
- Departamento de Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, Porto Alegre 90050-170, Brazil
| | - Carlos Alexandre Netto
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, Porto Alegre 90035-003, Brazil
| | - Silvia Honda Takada
- Neurogenetics Laboratory, Universidade Federal do ABC, Alameda da Universidade, s/n, São Bernardo do Campo 09606-045, Brazil.
| |
Collapse
|
43
|
Jiang G, Ayaki T, Maki T, Yasuda K, Yoshii D, Kaji S, Takahashi R. Evaluation of BCAS1-positive immature oligodendrocytes after cerebral ischemic stroke and SVD. Neurosci Lett 2023; 812:137405. [PMID: 37479175 DOI: 10.1016/j.neulet.2023.137405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/02/2023] [Accepted: 07/18/2023] [Indexed: 07/23/2023]
Abstract
Ischemic cerebrovascular disease is an important cause of physical disability and dementia. Oligodendrocytes (OLGs), which differentiate from oligodendrocyte precursor cells (OPCs), are crucial for remyelination of the damaged brain and functional recovery. Breast carcinoma amplified sequence 1 (BCAS1) has recently been shown to be highly expressed in newly formed pre-myelinating oligodendrocytes (pre-mOLGs), while its expression level is reduced in mature OLGs. In this study, we analyzed BCAS1 expression by immunohistochemical analysis of human post-mortem brain tissue from six stroke patients (death within 2 months after stroke onset) and eight small vessel disease (SVD) patients. Control post-mortem brain tissue was from eight age-matched patients without any obvious central nervous system (CNS) pathology. The Olig2 expression in the area corresponding to the same section of the BCAS1-stained slice was analyzed to determine the total oligodendrocyte lineage. The percentage of differentiating OPCs in the oligodendrocyte lineage was calculated as the ratio of BCAS1+ to Olig2+ cells (BCAS1+/Olig2+). The stroke and SVD cases showed demyelination with decreased expression of myelin basic protein (MBP, a mature OLG marker). The stroke cases showed significantly increased numbers of early-stage BCAS1+ cells with an immature morphology and Olig2+ cells (pan-oligodendrocyte lineages) in the peri-infarct areas in both the cortex and white matter, but showed no increase in the number of late-stage BCAS1+ cells with a mature morphology. In contrast, the SVD cases showed no significant increase in Olig2+ and BCAS1+ cells. These results indicated that remyelination dysfunction could be attributed to insufficient maturation of OPCs in stroke and impaired recruitment of OPCs in SVD.
Collapse
Affiliation(s)
- Guanhua Jiang
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takashi Ayaki
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Takakuni Maki
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ken Yasuda
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Daisuke Yoshii
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Seiji Kaji
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
44
|
Häusler S, Robertson NJ, Golhen K, van den Anker J, Tucker K, Felder TK. Melatonin as a Therapy for Preterm Brain Injury: What Is the Evidence? Antioxidants (Basel) 2023; 12:1630. [PMID: 37627625 PMCID: PMC10451719 DOI: 10.3390/antiox12081630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 07/28/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Despite significant improvements in survival following preterm birth in recent years, the neurodevelopmental burden of prematurity, with its long-term cognitive and behavioral consequences, remains a significant challenge in neonatology. Neuroprotective treatment options to improve neurodevelopmental outcomes in preterm infants are therefore urgently needed. Alleviating inflammatory and oxidative stress (OS), melatonin might modify important triggers of preterm brain injury, a complex combination of destructive and developmental abnormalities termed encephalopathy of prematurity (EoP). Preliminary data also suggests that melatonin has a direct neurotrophic impact, emphasizing its therapeutic potential with a favorable safety profile in the preterm setting. The current review outlines the most important pathomechanisms underlying preterm brain injury and correlates them with melatonin's neuroprotective potential, while underlining significant pharmacokinetic/pharmacodynamic uncertainties that need to be addressed in future studies.
Collapse
Affiliation(s)
- Silke Häusler
- Division of Neonatology, Department of Pediatrics, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria
| | - Nicola J. Robertson
- EGA Institute for Women’s Health, University College London, London WC1E 6HX, UK; (N.J.R.); (K.T.)
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Klervi Golhen
- Pediatric Pharmacology and Pharmacometrics, University Children’s Hospital Basel (UKBB), University of Basel, 4001 Basel, Switzerland; (K.G.); (J.v.d.A.)
| | - John van den Anker
- Pediatric Pharmacology and Pharmacometrics, University Children’s Hospital Basel (UKBB), University of Basel, 4001 Basel, Switzerland; (K.G.); (J.v.d.A.)
- Division of Clinical Pharmacology, Children’s National Hospital, Washington, DC 20001, USA
| | - Katie Tucker
- EGA Institute for Women’s Health, University College London, London WC1E 6HX, UK; (N.J.R.); (K.T.)
| | - Thomas K. Felder
- Department of Laboratory Medicine, Paracelsus Medical University, 5020 Salzburg, Austria;
| |
Collapse
|
45
|
Grotheer M, Bloom D, Kruper J, Richie-Halford A, Zika S, Aguilera González VA, Yeatman JD, Grill-Spector K, Rokem A. Human white matter myelinates faster in utero than ex utero. Proc Natl Acad Sci U S A 2023; 120:e2303491120. [PMID: 37549280 PMCID: PMC10438384 DOI: 10.1073/pnas.2303491120] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/27/2023] [Indexed: 08/09/2023] Open
Abstract
The formation of myelin, the fatty sheath that insulates nerve fibers, is critical for healthy brain function. A fundamental open question is what impact being born has on myelin growth. To address this, we evaluated a large (n = 300) cross-sectional sample of newborns from the Developing Human Connectome Project (dHCP). First, we developed software for the automated identification of 20 white matter bundles in individual newborns that is well suited for large samples. Next, we fit linear models that quantify how T1w/T2w (a myelin-sensitive imaging contrast) changes over time at each point along the bundles. We found faster growth of T1w/T2w along the lengths of all bundles before birth than right after birth. Further, in a separate longitudinal sample of preterm infants (N = 34), we found lower T1w/T2w than in full-term peers measured at the same age. By applying the linear models fit on the cross-section sample to the longitudinal sample of preterm infants, we find that their delay in T1w/T2w growth is well explained by the amount of time they spent developing in utero and ex utero. These results suggest that white matter myelinates faster in utero than ex utero. The reduced rate of myelin growth after birth, in turn, explains lower myelin content in individuals born preterm and could account for long-term cognitive, neurological, and developmental consequences of preterm birth. We hypothesize that closely matching the environment of infants born preterm to what they would have experienced in the womb may reduce delays in myelin growth and hence improve developmental outcomes.
Collapse
Affiliation(s)
- Mareike Grotheer
- Department of Psychology, Philipps-Universität Marburg, Marburg35039, Germany
- Center for Mind, Brain and Behavior, Philipps-Universität Marburg and Justus-Liebig-Universität Giessen, Marburg35039, Germany
| | - David Bloom
- Department of Psychology, University of Washington, Seattle, WA98105
- eScience Institute, University of Washington, Seattle, WA98105
| | - John Kruper
- Department of Psychology, University of Washington, Seattle, WA98105
- eScience Institute, University of Washington, Seattle, WA98105
| | - Adam Richie-Halford
- Department of Psychology, University of Washington, Seattle, WA98105
- eScience Institute, University of Washington, Seattle, WA98105
| | - Stephanie Zika
- Department of Psychology, Philipps-Universität Marburg, Marburg35039, Germany
- Center for Mind, Brain and Behavior, Philipps-Universität Marburg and Justus-Liebig-Universität Giessen, Marburg35039, Germany
| | - Vicente A. Aguilera González
- Department of Psychology, Philipps-Universität Marburg, Marburg35039, Germany
- Center for Mind, Brain and Behavior, Philipps-Universität Marburg and Justus-Liebig-Universität Giessen, Marburg35039, Germany
| | - Jason D. Yeatman
- Department of Psychology, Stanford University, Stanford, CA94305
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA94305
- Graduate School of Education, Stanford University, Stanford, CA94305
- Division of Developmental-Behavioral Pediatrics, Stanford University School of Medicine, Stanford, CA94305
| | - Kalanit Grill-Spector
- Department of Psychology, Stanford University, Stanford, CA94305
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA94305
| | - Ariel Rokem
- Department of Psychology, University of Washington, Seattle, WA98105
- eScience Institute, University of Washington, Seattle, WA98105
| |
Collapse
|
46
|
Manukjan N, Majcher D, Leenders P, Caiment F, van Herwijnen M, Smeets HJ, Suidgeest E, van der Weerd L, Vanmierlo T, Jansen JFA, Backes WH, van Oostenbrugge RJ, Staals J, Fulton D, Ahmed Z, Blankesteijn WM, Foulquier S. Hypoxic oligodendrocyte precursor cell-derived VEGFA is associated with blood-brain barrier impairment. Acta Neuropathol Commun 2023; 11:128. [PMID: 37550790 PMCID: PMC10405482 DOI: 10.1186/s40478-023-01627-5] [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: 04/14/2023] [Accepted: 07/23/2023] [Indexed: 08/09/2023] Open
Abstract
Cerebral small vessel disease is characterised by decreased cerebral blood flow and blood-brain barrier impairments which play a key role in the development of white matter lesions. We hypothesised that cerebral hypoperfusion causes local hypoxia, affecting oligodendrocyte precursor cell-endothelial cell signalling leading to blood-brain barrier dysfunction as an early mechanism for the development of white matter lesions. Bilateral carotid artery stenosis was used as a mouse model for cerebral hypoperfusion. Pimonidazole, a hypoxic cell marker, was injected prior to humane sacrifice at day 7. Myelin content, vascular density, blood-brain barrier leakages, and hypoxic cell density were quantified. Primary mouse oligodendrocyte precursor cells were exposed to hypoxia and RNA sequencing was performed. Vegfa gene expression and protein secretion was examined in an oligodendrocyte precursor cell line exposed to hypoxia. Additionally, human blood plasma VEGFA levels were measured and correlated to blood-brain barrier permeability in normal-appearing white matter and white matter lesions of cerebral small vessel disease patients and controls. Cerebral blood flow was reduced in the stenosis mice, with an increase in hypoxic cell number and blood-brain barrier leakages in the cortical areas but no changes in myelin content or vascular density. Vegfa upregulation was identified in hypoxic oligodendrocyte precursor cells, which was mediated via Hif1α and Epas1. In humans, VEGFA plasma levels were increased in patients versus controls. VEGFA plasma levels were associated with increased blood-brain barrier permeability in normal appearing white matter of patients. Cerebral hypoperfusion mediates hypoxia induced VEGFA expression in oligodendrocyte precursor cells through Hif1α/Epas1 signalling. VEGFA could in turn increase BBB permeability. In humans, increased VEGFA plasma levels in cerebral small vessel disease patients were associated with increased blood-brain barrier permeability in the normal appearing white matter. Our results support a role of VEGFA expression in cerebral hypoperfusion as seen in cerebral small vessel disease.
Collapse
Affiliation(s)
- Narek Manukjan
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| | - Daria Majcher
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Peter Leenders
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Florian Caiment
- Department of Toxicogenomics, GROW–School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Marcel van Herwijnen
- Department of Toxicogenomics, GROW–School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Hubert J. Smeets
- Department of Toxicogenomics, GROW–School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Ernst Suidgeest
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, P.O. Box 9500, 2300 RA Leiden, the Netherlands
| | - Louise van der Weerd
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, P.O. Box 9500, 2300 RA Leiden, the Netherlands
- Department of Human Genetics, Leiden University Medical Center, P.O. Box 9500, 2300 RA Leiden, The Netherlands
| | - Tim Vanmierlo
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, European Graduate School of Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Jacobus F. A. Jansen
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Walter H. Backes
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Robert J. van Oostenbrugge
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neurology, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Julie Staals
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neurology, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Daniel Fulton
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| | - Zubair Ahmed
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
- Centre for Trauma Sciences Research, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| | - W. Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Sébastien Foulquier
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neurology, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| |
Collapse
|
47
|
Qiu X, Zhou R, Su X, Ying J, Qu Y, Mu D. Pleiotrophin ameliorates white matter injury of neonatal rats by activating the mTOR/YY1/Id4 signaling pathway. FASEB J 2023; 37:e23082. [PMID: 37462506 DOI: 10.1096/fj.202201766rrr] [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: 10/28/2022] [Revised: 06/09/2023] [Accepted: 06/26/2023] [Indexed: 07/21/2023]
Abstract
Brain white matter injury (WMI) is a serious disease of the central nervous system. Pleiotrophin (PTN) promotes the differentiation and myelination of oligodendrocytes (OLs) in vitro. However, the role of PTN in WMI remains unknown. Therefore, this study aimed to investigate the neuroprotective role and potential mechanisms of PTN function in neonatal rats with WMI. The PTN and mammalian target of rapamycin (mTOR) inhibitor everolimus was used to treat a WMI model in postnatal day 3 Sprague-Dawley rats, in which the right common carotid arteries of these rats were isolated, ligated, and exposed to a hypoxic environment (6% O2 + 94% N2 ) for 2 h. OL differentiation and myelination, as well as the spatial learning and memory abilities of the rats were evaluated to examine the effects of PTN. Two proteins of the mTOR signaling pathway, YingYang1 (YY1) and inhibitor of DNA binding 4 (Id4), were detected and were used to explore the potential mechanisms of PTN in rat WMI experiment and oxygen glucose deprivation (OGD) model. We found that the differentiation and myelination of OLs were impaired after WMI. PTN administration rescued this injury by activating mTOR/YY1 and inhibiting Id4. Everolimus administration inhibited mTOR/YY1 and activated Id4, which blocked the neuroprotective role of PTN in WMI. PTN plays a neuroprotective role in neonatal rats with WMI, which could be involved in the mTOR/YY1/Id4 signaling pathway.
Collapse
Affiliation(s)
- Xia Qiu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Ruixi Zhou
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Xiaojuan Su
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Junjie Ying
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu, China
| |
Collapse
|
48
|
Inoue Y, Shue F, Bu G, Kanekiyo T. Pathophysiology and probable etiology of cerebral small vessel disease in vascular dementia and Alzheimer's disease. Mol Neurodegener 2023; 18:46. [PMID: 37434208 PMCID: PMC10334598 DOI: 10.1186/s13024-023-00640-5] [Citation(s) in RCA: 87] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 06/28/2023] [Indexed: 07/13/2023] Open
Abstract
Vascular cognitive impairment and dementia (VCID) is commonly caused by vascular injuries in cerebral large and small vessels and is a key driver of age-related cognitive decline. Severe VCID includes post-stroke dementia, subcortical ischemic vascular dementia, multi-infarct dementia, and mixed dementia. While VCID is acknowledged as the second most common form of dementia after Alzheimer's disease (AD) accounting for 20% of dementia cases, VCID and AD frequently coexist. In VCID, cerebral small vessel disease (cSVD) often affects arterioles, capillaries, and venules, where arteriolosclerosis and cerebral amyloid angiopathy (CAA) are major pathologies. White matter hyperintensities, recent small subcortical infarcts, lacunes of presumed vascular origin, enlarged perivascular space, microbleeds, and brain atrophy are neuroimaging hallmarks of cSVD. The current primary approach to cSVD treatment is to control vascular risk factors such as hypertension, dyslipidemia, diabetes, and smoking. However, causal therapeutic strategies have not been established partly due to the heterogeneous pathogenesis of cSVD. In this review, we summarize the pathophysiology of cSVD and discuss the probable etiological pathways by focusing on hypoperfusion/hypoxia, blood-brain barriers (BBB) dysregulation, brain fluid drainage disturbances, and vascular inflammation to define potential diagnostic and therapeutic targets for cSVD.
Collapse
Affiliation(s)
- Yasuteru Inoue
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Francis Shue
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| | - Guojun Bu
- SciNeuro Pharmaceuticals, Rockville, MD 20850 USA
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224 USA
| |
Collapse
|
49
|
Christiansen LI, Ventura GC, Holmqvist B, Aasmul-Olsen K, Lindholm SEH, Lycas MD, Mori Y, Secher JBM, Burrin DG, Thymann T, Sangild PT, Pankratova S. Insulin-like growth factor 1 supplementation supports motor coordination and affects myelination in preterm pigs. Front Neurosci 2023; 17:1205819. [PMID: 37404461 PMCID: PMC10315495 DOI: 10.3389/fnins.2023.1205819] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 05/23/2023] [Indexed: 07/06/2023] Open
Abstract
Introduction Preterm infants have increased risk of impaired neurodevelopment to which reduced systemic levels of insulin-like growth factor 1 (IGF-1) in the weeks after birth may play a role. Hence, we hypothesized that postnatal IGF-1 supplementation would improve brain development in preterm pigs, used as a model for preterm infants. Methods Preterm pigs delivered by cesarean section received recombinant human IGF-1/IGF binding protein-3 complex (rhIGF-1/rhIGFBP-3, 2.25 mg/kg/day) or vehicle from birth to postnatal day 19. Motor function and cognition were assessed by monitoring of in-cage and open field activities, balance beam test, gait parameters, novel object recognition and operant conditioning tests. Collected brains were subject to magnetic resonance imaging (MRI), immunohistochemistry, gene expression analyses and protein synthesis measurements. Results The IGF-1 treatment increased cerebellar protein synthesis rates (both in vivo and ex vivo). Performance in the balance beam test was improved by IGF-1 but not in other neurofunctional tests. The treatment decreased total and relative caudate nucleus weights, without any effects to total brain weight or grey/white matter volumes. Supplementation with IGF-1 reduced myelination in caudate nucleus, cerebellum, and white matter regions and decreased hilar synapse formation, without effects to oligodendrocyte maturation or neuron differentiation. Gene expression analyses indicated enhanced maturation of the GABAergic system in the caudate nucleus (decreased NKCC1:KCC2 ratio) with limited effects in cerebellum or hippocampus. Conclusion Supplemental IGF-1 during the first three weeks after preterm birth may support motor function by enhancing GABAergic maturation in the caudate nucleus, despite reduced myelination. Supplemental IGF-1 may support postnatal brain development in preterm infants, but more studies are required to identify optimal treatment regimens for subgroups of very or extremely preterm infants.
Collapse
Affiliation(s)
- Line I. Christiansen
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animals Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Gemma C. Ventura
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animals Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | | | - Karoline Aasmul-Olsen
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animals Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Sandy E. H. Lindholm
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animals Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Matthew D. Lycas
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yuki Mori
- Center for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jan Bojsen-Møller Secher
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Douglas G. Burrin
- United States Department of Agriculture, Agricultural Research Service Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Thomas Thymann
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animals Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Per T. Sangild
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animals Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
- Department of Neonatology, Rigshospitalet, Copenhagen, Denmark
- Department of Pediatrics, Odense University Hospital, Odense, Denmark
- Faculty of Theology, University of Copenhagen, Copenhagen, Denmark
| | - Stanislava Pankratova
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animals Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
50
|
Frazier AP, Mitchell DN, Given KS, Hunn G, Burch AM, Childs CR, Moreno-Garcia M, Corigilano MR, Quillinan N, Macklin WB, Herson PS, Dingman AL. Chronic changes in oligodendrocyte sub-populations after middle cerebral artery occlusion in neonatal mice. Glia 2023; 71:1429-1450. [PMID: 36794545 DOI: 10.1002/glia.24349] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 02/17/2023]
Abstract
Neonatal stroke is common and causes life-long motor and cognitive sequelae. Because neonates with stroke are not diagnosed until days-months after the injury, chronic targets for repair are needed. We evaluated oligodendrocyte maturity and myelination and assessed oligodendrocyte gene expression changes using single cell RNA sequencing (scRNA seq) at chronic timepoints in a mouse model of neonatal arterial ischemic stroke. Mice underwent 60 min of transient right middle cerebral artery occlusion (MCAO) on postnatal day 10 (p10) and received 5-ethynyl-2'-deoxyuridine (EdU) on post-MCAO days 3-7 to label dividing cells. Animals were sacrificed 14 and 28-30 days post-MCAO for immunohistochemistry and electron microscopy. Oligodendrocytes were isolated from striatum 14 days post-MCAO for scRNA seq and differential gene expression analysis. The density of Olig2+ EdU+ cells was significantly increased in ipsilateral striatum 14 days post-MCAO and the majority of oligodendrocytes were immature. Density of Olig2+ EdU+ cells declined significantly between 14 and 28 days post-MCAO without a concurrent increase in mature Olig2+ EdU+ cells. By 28 days post-MCAO there were significantly fewer myelinated axons in ipsilateral striatum. scRNA seq identified a cluster of "disease associated oligodendrocytes (DOLs)" specific to the ischemic striatum, with increased expression of MHC class I genes. Gene ontology analysis suggested decreased enrichment of pathways involved in myelin production in the reactive cluster. Oligodendrocytes proliferate 3-7 days post-MCAO and persist at 14 days, but fail to mature by 28 days. MCAO induces a subset of oligodendrocytes with reactive phenotype, which may be a therapeutic target to promote white matter repair.
Collapse
Affiliation(s)
- Alexandra P Frazier
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Danae N Mitchell
- Department of Pediatrics, Division of Child Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Katherine S Given
- Department of Developmental and Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Genevieve Hunn
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Amelia M Burch
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Christine R Childs
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Myriam Moreno-Garcia
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Michael R Corigilano
- Department of Graduate Medical Education, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Nidia Quillinan
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Wendy B Macklin
- Department of Developmental and Cell Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Paco S Herson
- Department of Neurosurgery, The Ohio State University, Columbus, Ohio, USA
| | - Andra L Dingman
- Department of Pediatrics, Division of Child Neurology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|