1
|
Serneels PJ, De Schutter JD, De Groef L, Moons L, Bergmans S. Oligodendroglial heterogeneity in health, disease, and recovery: deeper insights into myelin dynamics. Neural Regen Res 2025; 20:3179-3192. [PMID: 39665821 DOI: 10.4103/nrr.nrr-d-24-00694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 10/17/2024] [Indexed: 12/13/2024] Open
Abstract
Decades of research asserted that the oligodendroglial lineage comprises two cell types: oligodendrocyte precursor cells and oligodendrocytes. However, recent studies employing single-cell RNA sequencing techniques have uncovered novel cell states, prompting a revision of the existing terminology. Going forward, the oligodendroglial lineage should be delineated into five distinct cell states: oligodendrocyte precursor cells, committed oligodendrocyte precursor cells, newly formed oligodendrocytes, myelin-forming oligodendrocytes, and mature oligodendrocytes. This new classification system enables a deeper understanding of the oligodendroglia in both physiological and pathological contexts. Adopting this uniform terminology will facilitate comparison and integration of data across studies. This, including the consolidation of findings from various demyelinating models, is essential to better understand the pathogenesis of demyelinating diseases. Additionally, comparing injury models across species with varying regenerative capacities can provide insights that may lead to new therapeutic strategies to overcome remyelination failure. Thus, by standardizing terminology and synthesizing data from diverse studies across different animal models, we can enhance our understanding of myelin pathology in central nervous system disorders such as multiple sclerosis, Alzheimer's disease, and amyotrophic lateral sclerosis, all of which involve oligodendroglial and myelin dysfunction.
Collapse
Affiliation(s)
- Pieter-Jan Serneels
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Neural Circuit Development & Regeneration Research Group, Leuven, Belgium
| | - Julie D De Schutter
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Neural Circuit Development & Regeneration Research Group, Leuven, Belgium
| | - Lies De Groef
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Cellular Communication & Neurodegeneration Research Group, Leuven, Belgium
| | - Lieve Moons
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Neural Circuit Development & Regeneration Research Group, Leuven, Belgium
| | - Steven Bergmans
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Neural Circuit Development & Regeneration Research Group, Leuven, Belgium
| |
Collapse
|
2
|
Margolis ET, Gabard‐Durnam LJ. Prenatal influences on postnatal neuroplasticity: Integrating DOHaD and sensitive/critical period frameworks to understand biological embedding in early development. INFANCY 2025; 30:e12588. [PMID: 38449347 PMCID: PMC11647198 DOI: 10.1111/infa.12588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 01/26/2024] [Accepted: 02/13/2024] [Indexed: 03/08/2024]
Abstract
Early environments can have significant and lasting effects on brain, body, and behavior across the lifecourse. Here, we address current research efforts to understand how experiences impact neurodevelopment with a new perspective integrating two well-known conceptual frameworks - the Developmental Origins of Health and Disease (DOHaD) and sensitive/critical period frameworks. Specifically, we consider how prenatal experiences characterized in the DOHaD model impact two key neurobiological mechanisms of sensitive/critical periods for adapting to and learning from the postnatal environment. We draw from both animal and human research to summarize the current state of knowledge on how particular prenatal substance exposures (psychoactive substances and heavy metals) and nutritional profiles (protein-energy malnutrition and iron deficiency) each differentially impact brain circuits' excitation/GABAergic inhibition balance and myelination. Finally, we highlight new research directions that emerge from this integrated framework, including testing how prenatal environments alter sensitive/critical period timing and learning and identifying potential promotional/buffering prenatal exposures to impact postnatal sensitive/critical periods. We hope this integrative framework considering prenatal influences on postnatal neuroplasticity will stimulate new research to understand how early environments have lasting consequences on our brains, behavior, and health.
Collapse
Affiliation(s)
- Emma T. Margolis
- Department of PsychologyNortheastern UniversityBostonMassachusettsUSA
| | | |
Collapse
|
3
|
Yanai H, Adachi H, Hakoshima M, Katsuyama H. Pathology and Treatments of Alzheimer’s Disease Based on Considering Changes in Brain Energy Metabolism Due to Type 2 Diabetes. Molecules 2024; 29:5936. [PMCID: PMC11677283 DOI: 10.3390/molecules29245936] [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: 10/22/2024] [Revised: 11/22/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder with cognitive dysfunction, memory decline, and behavioral disturbance, and it is pathologically characterized by the accumulation of amyloid plaques and neurofibrillary tangles in the brain. Although various hypotheses have been proposed to explain the pathogenesis of AD, including the amyloid beta hypothesis, oxidative stress hypothesis, and abnormal phosphorylation of tau proteins, the exact pathogenic mechanisms underlying AD remain largely undefined. Furthermore, effective curative treatments are very limited. Epidemiologic studies provide convincing evidence for a significant association between type 2 diabetes and AD. Here, we showed energy metabolism using glucose, lactate, ketone bodies, and lipids as energy substrates in a normal brain, and changes in such energy metabolism due to type 2 diabetes. We also showed the influences of such altered energy metabolism due to type 2 diabetes on the pathology of AD. Furthermore, we comprehensively searched for risk factors related with type 2 diabetes for AD and showed possible therapeutic interventions based on considering risk factors and altered brain energy metabolism due to type 2 diabetes for the development of AD.
Collapse
Affiliation(s)
- Hidekatsu Yanai
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, 1-7-1 Kohnodai, Ichikawa 272-8516, Chiba, Japan; (H.A.); (M.H.); (H.K.)
| | | | | | | |
Collapse
|
4
|
Bernstein HG, Nussbaumer M, Vasilevska V, Dobrowolny H, Nickl-Jockschat T, Guest PC, Steiner J. Glial cell deficits are a key feature of schizophrenia: implications for neuronal circuit maintenance and histological differentiation from classical neurodegeneration. Mol Psychiatry 2024:10.1038/s41380-024-02861-6. [PMID: 39639174 DOI: 10.1038/s41380-024-02861-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 11/19/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
Dysfunctional glial cells play a pre-eminent role in schizophrenia pathophysiology. Post-mortem studies have provided evidence for significantly decreased glial cell numbers in different brain regions of individuals with schizophrenia. Reduced glial cell numbers are most pronounced in oligodendroglia, but reduced astrocyte cell densities have also been reported. This review highlights that oligo- and astroglial deficits are a key histopathological feature in schizophrenia, distinct from typical changes seen in neurodegenerative disorders. Significant deficits of oligodendrocytes in schizophrenia may arise in two ways: (i) demise of mature functionally compromised oligodendrocytes; and (ii) lack of mature oligodendrocytes due to failed maturation of progenitor cells. We also analyse in detail the controversy regarding deficits of astrocytes. Regardless of their origin, glial cell deficits have several pathophysiological consequences. Among these, myelination deficits due to a reduced number of oligodendrocytes may be the most important factor, resulting in the disconnectivity between neurons and different brain regions observed in schizophrenia. When glial cells die, it appears to be through degeneration, a process which is basically reversible. Thus, therapeutic interventions that (i) help rescue glial cells (ii) or improve their maturation might be a viable option. Since antipsychotic treatment alone does not seem to prevent glial cell loss or maturation deficits, there is intense search for new therapeutic options. Current proposals range from the application of antidepressants and other chemical agents as well as physical exercise to engrafting healthy glial cells into brains of schizophrenia patients.
Collapse
Affiliation(s)
- Hans-Gert Bernstein
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Madeleine Nussbaumer
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Veronika Vasilevska
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Department of Radiotherapy, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Henrik Dobrowolny
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Thomas Nickl-Jockschat
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa, IA, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa, IA, USA
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa, IA, USA
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- German Center for Mental Health (DZPG), Partner Site Halle-Jena-Magdeburg, Magdeburg, Germany
| | - Paul C Guest
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Johann Steiner
- Department of Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.
- Laboratory of Translational Psychiatry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany.
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany.
- German Center for Mental Health (DZPG), Partner Site Halle-Jena-Magdeburg, Magdeburg, Germany.
| |
Collapse
|
5
|
Vahidi H, Kowalczyk A, Stubbs K, Musabi M, Roychaudhuri S, Kent M, Bhattacharya S, de Ribaupierre S, Lawrence KS, Mohsenzadeh Y, Duerden EG. Investigating Task-Free Functional Connectivity Patterns in Newborns Using Functional Near-Infrared Spectroscopy. Brain Behav 2024; 14:e70180. [PMID: 39690863 DOI: 10.1002/brb3.70180] [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: 07/02/2024] [Revised: 11/11/2024] [Accepted: 11/15/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Resting-state networks (RSNs), particularly the sensorimotor network, begin to strengthe in the third trimester of pregnancy and mature extensively by term age. The integrity and structure of these networks have been repeatedly linked to neurological health outcomes in neonates, highlighting the importance of understanding the normative variations in RSNs in healthy development. Specifically, robust bilateral functional connectivity in the sensorimotor RSN has been linked to optimal neurodevelopmental outcomes in neonates. AIM In the current study, we aimed to map the developmental trajectory of the sensorimotor RSN in awake neonates using functional near-infrared spectroscopy (fNIRS). MATERIALS & METHODS We acquired fNIRS resting-state data from 41 healthy newborns (17 females, gestational age ranging from 36 + 0 to 42 + 1 weeks) within the first week after birth. We performed both single channel and hemispheric analyses to investigate the relationship between functional connectivity and both gestational and postnatal age. RESULTS We observed robust positive connectivity in numerous channel-pairs across the sensorimotor network, especially in the left hemisphere. Next, we examined the relationship between functional connectivity, gestational age, and postnatal age, while controlling for sex and subject effects. We found both gestational and postnatal age to be significantly associated with changes in functional connectivity in the sensorimotor RSN. In our hemispheric analysis (Ninterhemispheric = 10, Nleft intrahemispheric = 15, and Nright intrahemispheric = 9), we observed a significant positive relationship between interhemispheric connectivity and postnatal age. DISCUSSION AND CONCLUSION In summary, our findings demonstrate the utility of fNIRS for monitoring early developmental changes in functional networks in awake newborns.
Collapse
Affiliation(s)
- Homa Vahidi
- Department of Neuroscience, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Alexandra Kowalczyk
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, London Health Sciences Centre, London, Ontario, Canada
| | - Kevin Stubbs
- Brain and Mind Centre, Western University, London, Ontario, Canada
| | - Melab Musabi
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, London Health Sciences Centre, London, Ontario, Canada
| | - Sriya Roychaudhuri
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, London Health Sciences Centre, London, Ontario, Canada
| | - Michaela Kent
- Department of Neuroscience, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Soume Bhattacharya
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, London Health Sciences Centre, London, Ontario, Canada
| | - Sandrine de Ribaupierre
- Department of Clinical Neurological Sciences, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Keith St Lawrence
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Yalda Mohsenzadeh
- Brain and Mind Centre, Western University, London, Ontario, Canada
- Department of Computer Science, Western University, London, Ontario, Canada
- Vector Institute for Artificial Intelligence, Toronto, Ontario, Canada
| | - Emma G Duerden
- Brain and Mind Centre, Western University, London, Ontario, Canada
- Applied Psychology, Faculty of Education, Western University, London, Ontario, Canada
| |
Collapse
|
6
|
Griflyuk AV, Postnikova TY, Zaitsev AV. Animal Models of Febrile Seizures: Limitations and Recent Advances in the Field. Cells 2024; 13:1895. [PMID: 39594643 PMCID: PMC11592604 DOI: 10.3390/cells13221895] [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: 10/11/2024] [Revised: 11/05/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Febrile seizures (FSs) are defined as seizures occurring in children aged 6 months to 5 years with a background of elevated body temperature. It is one of the most common neurological disorders of childhood, emphasizing the importance of understanding the causes of FSs and their impact on the developing nervous system. However, there are significant limitations to the technologies currently available for studying the etiology and pathophysiology of seizures in humans. It is currently not possible to adequately capture the subtle molecular and structural rearrangements of the nervous system that can occur after seizures in humans. The use of animal models can be invaluable for these purposes. The most commonly used models in modern research are hyperthermic models in rats and mice aged 10-12 days. While these models can reproduce many of the characteristics of FSs, they have certain limitations. This review outlines the key considerations when working with models of FSs, provides an overview of current approaches to producing seizures in different model subjects, and presents a summary of key findings regarding morphological and functional changes in the brain and behavioral alterations that have been identified in studies using animal models of FSs.
Collapse
Affiliation(s)
| | | | - Aleksey V. Zaitsev
- Sechenov Institute of Evolutionary Physiology and Biochemistry of Russian Academy of Sciences, 44, Toreza Prospekt, Saint Petersburg 194223, Russia; (A.V.G.); (T.Y.P.)
| |
Collapse
|
7
|
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
|
8
|
Russo ML, Sousa AMM, Bhattacharyya A. Consequences of trisomy 21 for brain development in Down syndrome. Nat Rev Neurosci 2024; 25:740-755. [PMID: 39379691 DOI: 10.1038/s41583-024-00866-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2024] [Indexed: 10/10/2024]
Abstract
The appearance of cognitive deficits and altered brain morphology in newborns with Down syndrome (DS) suggests that these features are driven by disruptions at the earliest stages of brain development. Despite its high prevalence and extensively characterized cognitive phenotypes, relatively little is known about the cellular and molecular mechanisms that drive the changes seen in DS. Recent technical advances, such as single-cell omics and the development of induced pluripotent stem cell (iPSC) models of DS, now enable in-depth analyses of the biochemical and molecular drivers of altered brain development in DS. Here, we review the current state of knowledge on brain development in DS, focusing primarily on data from human post-mortem brain tissue. We explore the biological mechanisms that have been proposed to lead to intellectual disability in DS, assess the extent to which data from studies using iPSC models supports these hypotheses, and identify current gaps in the field.
Collapse
Affiliation(s)
- Matthew L Russo
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - André M M Sousa
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neuroscience, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Anita Bhattacharyya
- Waisman Center, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
9
|
Xiao J, Sun C, Chen R, Zhao Z, Wang G, Wu D. Reproducibility of Diffusion MRI-Based Tractography in the Fetal Brain. J Magn Reson Imaging 2024; 60:2055-2062. [PMID: 38284561 DOI: 10.1002/jmri.29253] [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: 08/29/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND Tractography based on diffusion MRI (dMRI) is a useful tool to study white matter of the developing brain. However, its application in fetal brains is limited due to motion artifacts and low resolution of in utero dMRI, leading to reduced reliability, which was scarcely investigated in previous studies. PURPOSE To identify reliably traceable fibers in fetal brains and assess whether reproducibility varies with gestational age (GA) and varies between brain regions. STUDY TYPE Prospective cohort study. SUBJECTS A total of 44 healthy fetuses with GAs between 25 and 37 (31 ± 6). FIELD STRENGTH/SEQUENCE 3-T, diffusion-weighted echo-planar imaging sequence (2-5 repeated dMRI scans within the same session per subject). ASSESSMENT We fitted dMRI with constrained spherical deconvolution model and conducted tractography on eight fibers. We extracted volume, fractional anisotropy, and fiber count for each fiber and assessed the reproducibility of these metrics between repeated scans within each subject. Data were divided into two age-based subgroups (≤30 weeks, N = 28, and >30 weeks, N = 16) for further tests. STATISTICAL TESTS The reproducibility were compared between fibers by analysis of variance and two-sample t tests. Multiple comparisons were corrected by the false discovery rate (5% was accepted). RESULTS The reproducibility of the anterior thalamic radiation, inferior longitudinal fasciculus (ILF), genu of the corpus callosum (GCC), and body of the corpus callosum (BCC) significantly decreased with advancing GA (correlation coefficient = 0.525-0.823), as confirmed by group comparisons between fetuses in early GA (≤30 weeks) and late GA (>30 weeks) groups. Corticospinal tract, inferior fronto-occipital fasciculus, and GCC showed high reproducibility for fiber count (weighted dice average = 0.846 vs. 0.814), while BCC and ILF exhibited the lowest reproducibility in both age groups. DATA CONCLUSION The study indicates that the reliability of fetal brain tractography depends on GA and varies among different fibers. LEVEL OF EVIDENCE 2 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Jiaxin Xiao
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Cong Sun
- Department of Radiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Ruike Chen
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Zhiyong Zhao
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Guangbin Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Dan Wu
- Key Laboratory for Biomedical Engineering of Ministry of Education, Department of Biomedical Engineering, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
10
|
Kliesmete Z, Orchard P, Lee VYK, Geuder J, Krauß SM, Ohnuki M, Jocher J, Vieth B, Enard W, Hellmann I. Evidence for compensatory evolution within pleiotropic regulatory elements. Genome Res 2024; 34:1528-1539. [PMID: 39255977 PMCID: PMC11534155 DOI: 10.1101/gr.279001.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 08/19/2024] [Indexed: 09/12/2024]
Abstract
Pleiotropy, measured as expression breadth across tissues, is one of the best predictors for protein sequence and expression conservation. In this study, we investigated its effect on the evolution of cis-regulatory elements (CREs). To this end, we carefully reanalyzed the Epigenomics Roadmap data for nine fetal tissues, assigning a measure of pleiotropic degree to nearly half a million CREs. To assess the functional conservation of CREs, we generated ATAC-seq and RNA-seq data from humans and macaques. We found that more pleiotropic CREs exhibit greater conservation in accessibility, and the mRNA expression levels of the associated genes are more conserved. This trend of higher conservation for higher degrees of pleiotropy persists when analyzing the transcription factor binding repertoire. In contrast, simple DNA sequence conservation of orthologous sites between species tends to be even lower for pleiotropic CREs than for species-specific CREs. Combining various lines of evidence, we propose that the lack of sequence conservation in functionally conserved pleiotropic CREs is owing to within-element compensatory evolution. In summary, our findings suggest that pleiotropy is also a good predictor for the functional conservation of CREs, even though this is not reflected in the sequence conservation of pleiotropic CREs.
Collapse
Affiliation(s)
- Zane Kliesmete
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, 82152 Munich, Germany
| | - Peter Orchard
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, 82152 Munich, Germany
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109-2218, USA
| | - Victor Yan Kin Lee
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, 82152 Munich, Germany
- Section for Molecular Ecology and Evolution, Globe Institute, University of Copenhagen, 1350 Copenhagen, Denmark
| | - Johanna Geuder
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, 82152 Munich, Germany
| | - Simon M Krauß
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, 82152 Munich, Germany
- Department of Hematology, Cell Therapy, Hemostaseology and Infectious Diseases, University Leipzig Medical Center, 04103 Leipzig, Germany
| | - Mari Ohnuki
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, 82152 Munich, Germany
- Faculty of Medicine, Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto 606-8501, Japan
| | - Jessica Jocher
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, 82152 Munich, Germany
| | - Beate Vieth
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, 82152 Munich, Germany
| | - Wolfgang Enard
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, 82152 Munich, Germany
| | - Ines Hellmann
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians Universität München, 82152 Munich, Germany;
| |
Collapse
|
11
|
Rajan A, Fame RM. Brain development and bioenergetic changes. Neurobiol Dis 2024; 199:106550. [PMID: 38849103 PMCID: PMC11495523 DOI: 10.1016/j.nbd.2024.106550] [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: 04/15/2024] [Revised: 05/29/2024] [Accepted: 06/01/2024] [Indexed: 06/09/2024] Open
Abstract
Bioenergetics describe the biochemical processes responsible for energy supply in organisms. When these changes become dysregulated in brain development, multiple neurodevelopmental diseases can occur, implicating bioenergetics as key regulators of neural development. Historically, the discovery of disease processes affecting individual stages of brain development has revealed critical roles that bioenergetics play in generating the nervous system. Bioenergetic-dependent neurodevelopmental disorders include neural tube closure defects, microcephaly, intellectual disability, autism spectrum disorders, epilepsy, mTORopathies, and oncogenic processes. Developmental timing and cell-type specificity of these changes determine the long-term effects of bioenergetic disease mechanisms on brain form and function. Here, we discuss key metabolic regulators of neural progenitor specification, neuronal differentiation (neurogenesis), and gliogenesis. In general, transitions between glycolysis and oxidative phosphorylation are regulated in early brain development and in oncogenesis, and reactive oxygen species (ROS) and mitochondrial maturity play key roles later in differentiation. We also discuss how bioenergetics interface with the developmental regulation of other key neural elements, including the cerebrospinal fluid brain environment. While questions remain about the interplay between bioenergetics and brain development, this review integrates the current state of known key intersections between these processes in health and disease.
Collapse
Affiliation(s)
- Arjun Rajan
- Developmental Biology Graduate Program, Stanford University, Stanford, CA 94305, USA
| | - Ryann M Fame
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
12
|
Fagiani F, Pedrini E, Taverna S, Brambilla E, Murtaj V, Podini P, Ruffini F, Butti E, Braccia C, Andolfo A, Magliozzi R, Smirnova L, Kuhlmann T, Quattrini A, Calabresi PA, Reich DS, Martino G, Panina-Bordignon P, Absinta M. A glia-enriched stem cell 3D model of the human brain mimics the glial-immune neurodegenerative phenotypes of multiple sclerosis. Cell Rep Med 2024; 5:101680. [PMID: 39121861 PMCID: PMC11384947 DOI: 10.1016/j.xcrm.2024.101680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 01/12/2024] [Accepted: 07/16/2024] [Indexed: 08/12/2024]
Abstract
The role of central nervous system (CNS) glia in sustaining self-autonomous inflammation and driving clinical progression in multiple sclerosis (MS) is gaining scientific interest. We applied a single transcription factor (SOX10)-based protocol to accelerate oligodendrocyte differentiation from human induced pluripotent stem cell (hiPSC)-derived neural precursor cells, generating self-organizing forebrain organoids. These organoids include neurons, astrocytes, oligodendroglia, and hiPSC-derived microglia to achieve immunocompetence. Over 8 weeks, organoids reproducibly generated mature CNS cell types, exhibiting single-cell transcriptional profiles similar to the adult human brain. Exposed to inflamed cerebrospinal fluid (CSF) from patients with MS, organoids properly mimic macroglia-microglia neurodegenerative phenotypes and intercellular communication seen in chronic active MS. Oligodendrocyte vulnerability emerged by day 6 post-MS-CSF exposure, with nearly 50% reduction. Temporally resolved organoid data support and expand on the role of soluble CSF mediators in sustaining downstream events leading to oligodendrocyte death and inflammatory neurodegeneration. Such findings support the implementation of this organoid model for drug screening to halt inflammatory neurodegeneration.
Collapse
Affiliation(s)
- Francesca Fagiani
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Edoardo Pedrini
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Stefano Taverna
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Elena Brambilla
- Division of Neuroscience, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Valentina Murtaj
- Division of Neuroscience, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Paola Podini
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Francesca Ruffini
- Division of Neuroscience, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Erica Butti
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Clarissa Braccia
- ProMeFa, Proteomics and Metabolomics Facility, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Annapaola Andolfo
- ProMeFa, Proteomics and Metabolomics Facility, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Roberta Magliozzi
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37124 Verona, Italy
| | - Lena Smirnova
- Center for Alternatives to Animal Testing, Department of Environmental Health and Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital of Münster, 48149 Münster, Germany
| | - Angelo Quattrini
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Division of Neuroscience, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gianvito Martino
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Division of Neuroscience, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Paola Panina-Bordignon
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Division of Neuroscience, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Martina Absinta
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; Division of Neuroscience, Vita-Salute San Raffaele University, 20132 Milan, Italy; Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
13
|
Mastenbroek LJM, Kooistra SM, Eggen BJL, Prins JR. The role of microglia in early neurodevelopment and the effects of maternal immune activation. Semin Immunopathol 2024; 46:1. [PMID: 38990389 PMCID: PMC11239780 DOI: 10.1007/s00281-024-01017-6] [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: 04/04/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024]
Abstract
Activation of the maternal immune system during gestation has been associated with an increased risk for neurodevelopmental disorders in the offspring, particularly schizophrenia and autism spectrum disorder. Microglia, the tissue-resident macrophages of the central nervous system, are implicated as potential mediators of this increased risk. Early in development, microglia start populating the embryonic central nervous system and in addition to their traditional role as immune responders under homeostatic conditions, microglia are also intricately involved in various early neurodevelopmental processes. The timing of immune activation may interfere with microglia functioning during early neurodevelopment, potentially leading to long-term consequences in postnatal life. In this review we will discuss the involvement of microglia in brain development during the prenatal and early postnatal stages of life, while also examining the effects of maternal immune activation on microglia and neurodevelopmental processes. Additionally, we discuss recent single cell RNA-sequencing studies focusing on microglia during prenatal development, and hypothesize how early life microglial priming, potentially through epigenetic reprogramming, may be related to neurodevelopmental disorders.
Collapse
Affiliation(s)
- L J M Mastenbroek
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - S M Kooistra
- Department of BioMedical Sciences, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - B J L Eggen
- Department of BioMedical Sciences, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - J R Prins
- Department of Obstetrics and Gynaecology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
14
|
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
|
15
|
Milos RI, Schmidbauer V, Watzenboeck ML, Stuhr F, Gruber GM, Mitter C, Dovjak GO, Milković-Periša M, Kostovic I, Jovanov-Milošević N, Kasprian G, Prayer D. T1-weighted fast fluid-attenuated inversion-recovery sequence (T1-FFLAIR) enables the visualization and quantification of fetal brain myelination in utero. Eur Radiol 2024; 34:4573-4584. [PMID: 38019312 PMCID: PMC11213743 DOI: 10.1007/s00330-023-10401-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 09/03/2023] [Accepted: 09/16/2023] [Indexed: 11/30/2023]
Abstract
OBJECTIVES To investigate the advantage of T1-weighted fast fluid-attenuated inversion-recovery MRI sequence without (T1-FFLAIR) and with compressed sensing technology (T1-FFLAIR-CS), which shows improved T1-weighted contrast, over standard used T1-weighted fast field echo (T1-FFE) sequence for the assessment of fetal myelination. MATERIALS AND METHODS This retrospective single-center study included 115 consecutive fetal brain MRI examinations (63 axial and 76 coronal, mean gestational age (GA) 28.56 ± 5.23 weeks, range 19-39 weeks). Two raters, blinded to GA, qualitatively assessed a fetal myelin total score (MTS) on each T1-weighted sequence at five brain regions (medulla oblongata, pons, mesencephalon, thalamus, central region). One rater performed region-of-interest quantitative analysis (n = 61) at the same five brain regions. Pearson correlation analysis was applied for correlation of MTS and of the signal intensity ratios (relative to muscle) with GA on each T1-weighted sequence. Fetal MRI-based results were compared with myelination patterns of postmortem fetal human brains (n = 46; GA 18 to 42), processed by histological and immunohistochemical analysis. RESULTS MTS positively correlated with GA on all three sequences (all r between 0.802 and 0.908). The signal intensity ratios measured at the five brain regions correlated best with GA on T1-FFLAIR (r between 0.583 and 0.785). T1-FFLAIR demonstrated significantly better correlations with GA than T1-FFE for both qualitative and quantitative analysis (all p < 0.05). Furthermore, T1-FFLAIR enabled the best visualization of myelinated brain structures when compared to histology. CONCLUSION T1-FFLAIR outperforms the standard T1-FFE sequence in the visualization of fetal brain myelination, as demonstrated by qualitative and quantitative methods. CLINICAL RELEVANCE STATEMENT T1-weighted fast fluid-attenuated inversion-recovery sequence (T1-FFLAIR) provided best visualization and quantification of myelination in utero that, in addition to the relatively short acquisition time, makes feasible its routine application in fetal MRI for the assessment of brain myelination. KEY POINTS • So far, the assessment of fetal myelination in utero was limited due to the insufficient contrast. • T1-weighted fast fluid-attenuated inversion-recovery sequence allows a qualitative and quantitative assessment of fetal brain myelination. • T1-weighted fast fluid-attenuated inversion-recovery sequence outperforms the standard used T1-weighted sequence for visualization and quantification of myelination in utero.
Collapse
Affiliation(s)
- Ruxandra-Iulia Milos
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Victor Schmidbauer
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Martin L Watzenboeck
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Friedrich Stuhr
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Gerlinde Maria Gruber
- Department of Anatomy and Biomechanics, Karl Landsteiner University of Health Sciences, 3500, Krems, Austria
| | - Christian Mitter
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Gregor O Dovjak
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Marija Milković-Periša
- Department of Pathology and Cytology, University Hospital Centre Zagreb, Petrova 13, 10000, Zagreb, Croatia
| | - Ivica Kostovic
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Nataša Jovanov-Milošević
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Biology, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Gregor Kasprian
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Daniela Prayer
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
16
|
Payette K. Stripping away the mysteries of the brain's insulation: might T1-FFLAIR improve our understanding of prenatal myelination? Eur Radiol 2024; 34:4570-4572. [PMID: 38032402 DOI: 10.1007/s00330-023-10450-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/23/2023] [Accepted: 10/28/2023] [Indexed: 12/01/2023]
Affiliation(s)
- Kelly Payette
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, 1St Floor South Wing, London, SE1 7EH, UK.
- Biomedical Engineering Department, School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, 1St Floor South Wing, London, SE1 7EH, UK.
| |
Collapse
|
17
|
Fagiani F, Pedrini E, Taverna S, Brambilla E, Murtaj V, Podini P, Ruffini F, Butti E, Braccia C, Andolfo A, Magliozzi R, Smirnova L, Kuhlmann T, Quattrini A, Calabresi PA, Reich DS, Martino G, Panina-Bordignon P, Absinta M. Glia-enriched stem-cell 3D model of the human brain mimics the glial-immune neurodegenerative phenotypes of multiple sclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.20.597748. [PMID: 39372788 PMCID: PMC11451585 DOI: 10.1101/2024.06.20.597748] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
The role of central nervous system (CNS) glia in sustaining self-autonomous inflammation and driving clinical progression in multiple sclerosis (MS) is gaining scientific interest. We applied a single transcription factor ( SOX10 )-based protocol to accelerate oligodendrocyte differentiation from hiPSC-derived neural precursor cells, generating self-organizing forebrain organoids. These organoids include neurons, astrocytes, oligodendroglia, and hiPSC-derived microglia to achieve immunocompetence. Over 8 weeks, organoids reproducibly generated mature CNS cell types, exhibiting single-cell transcriptional profiles similar to the adult human brain. Exposed to inflamed cerebrospinal fluid (CSF) from MS patients, organoids properly mimic macroglia-microglia neuro-degenerative phenotypes and intercellular communication seen in chronic active MS. Oligodendrocyte vulnerability emerged by day 6 post-MS-CSF exposure, with nearly 50% reduction. Temporally-resolved organoid data support and expand on the role of soluble CSF mediators in sustaining downstream events leading to oligodendrocyte death and inflammatory neurodegeneration. Such findings support implementing this organoid model for drug screening to halt inflammatory neurodegeneration.
Collapse
|
18
|
Weiner S, Wu Y, Kapse K, Vozar T, Cheng JJ, Murnick J, Henderson D, Teramoto H, Limperopoulos C, Andescavage N. Prenatal Maternal Psychological Distress During the COVID-19 Pandemic and Newborn Brain Development. JAMA Netw Open 2024; 7:e2417924. [PMID: 38900424 PMCID: PMC11190810 DOI: 10.1001/jamanetworkopen.2024.17924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/17/2024] [Indexed: 06/21/2024] Open
Abstract
Importance Elevated maternal psychological distress during pregnancy is associated with altered fetal brain development. During the COVID-19 pandemic, prenatal maternal psychological distress more than doubled. Objective To examine the association of the pandemic and rising maternal psychological distress with brain growth in newborns using quantitative 3-dimensional volumetric magnetic resonance imaging (MRI). Design, Setting, and Participants This prospective cross-sectional study recruited mother-infant dyads at Children's National Hospital, Washington, DC, during the COVID-19 pandemic (June 1, 2020, to June 30, 2022) into a longitudinal infant brain development study and compared them with an existing normative healthy cohort (recruited March 1, 2014, to December 31, 2019). Exclusion criteria included multiple gestation pregnancy, known or suspected congenital infection, documented chromosomal abnormalities, or any maternal contraindication to MRI, as well as prenatal COVID-19 exposure. Infants with structural brain abnormalities or a postnatal confirmation of a genetic syndrome were excluded. Exposure Psychological distress during COVID-19 pandemic. Main Outcomes and Measures Prenatal maternal mental health was evaluated using the Spielberger State-Trait Anxiety Inventory and the Perceived Stress Scale. Neonates underwent nonsedated brain MRI. An ordinary least squares linear regression model was used to measure the differences in regional brain volumes of neonates born before vs during the pandemic with and without exposure to elevated prenatal maternal psychological distress after adjustment for neonatal sex and gestational age at MRI and maternal age and educational level. Results A total of 159 mother-infant dyads were included in the analysis: 103 before and 56 during the pandemic (median gestational age of infants, 39.6 [IQR, 38.4-40.4] weeks; median maternal age, 34.5 [IQR, 31.0-37.0] years). Eighty-three infants (52.2%) were female. Among the mothers, 130 (81.8%) had a college degree and 87 (54.7%) had a graduate degree. Forty-four mothers (27.7%) identified as Asian, Hispanic, or multiracial; 27 (17.0%), as Black; and 88 (55.3%), as White. Scores on anxiety and stress measures were significantly increased in the pandemic cohort. Infants of mothers with elevated maternal distress showed median reductions in white matter (-0.36 [95% CI, -0.61 to -0.11] cm3; Q < .001), right hippocampal (-0.35 [95% CI, -0.65 to -0.06] cm3; Q = .04), and left amygdala (-0.49 [95% CI, -0.84 to -0.13] cm3; Q = .03) volumes compared with infants of mothers with low distress levels. After adjusting for the cohort effect of the pandemic, elevated trait anxiety remained significantly associated with decreased left amygdalar volumes (-0.71 [95% CI, -1.12 to -0.29]; Q < .001). Conclusions and Relevance In this cross-sectional study of maternal-infant dyads prior to and during the COVID-19 pandemic, regional neonatal brain volumes were associated with elevated maternal psychological distress.
Collapse
Affiliation(s)
- Susan Weiner
- Developing Brain Institute, Children’s National Hospital, Washington, DC
- The Institute for Biomedical Sciences, George Washington University, Washington, DC
| | - Yao Wu
- Developing Brain Institute, Children’s National Hospital, Washington, DC
| | - Kushal Kapse
- Developing Brain Institute, Children’s National Hospital, Washington, DC
| | - Tracy Vozar
- Department of Diagnostic Imaging and Radiology, Children’s National Hospital, Washington, DC
- Department of Psychology, Children’s National Hospital, Washington, DC
| | | | - Jonathan Murnick
- Department of Diagnostic Imaging and Radiology, Children’s National Hospital, Washington, DC
- Department of Radiology, George Washington University, Washington, DC
- Department of Neonatology, Children’s National Hospital, Washington, DC
| | - Diedtra Henderson
- Developing Brain Institute, Children’s National Hospital, Washington, DC
| | - Hironori Teramoto
- Developing Brain Institute, Children’s National Hospital, Washington, DC
| | - Catherine Limperopoulos
- Developing Brain Institute, Children’s National Hospital, Washington, DC
- Department of Diagnostic Imaging and Radiology, Children’s National Hospital, Washington, DC
- Department of Pediatrics, George Washington University, Washington, DC
- Department of Radiology, George Washington University, Washington, DC
| | - Nickie Andescavage
- Developing Brain Institute, Children’s National Hospital, Washington, DC
- Department of Pediatrics, George Washington University, Washington, DC
- Department of Neonatology, Children’s National Hospital, Washington, DC
| |
Collapse
|
19
|
Mencattini A, Daprati E, Della-Morte D, Guadagni F, Sangiuolo F, Martinelli E. Assembloid learning: opportunities and challenges for personalized approaches to brain functioning in health and disease. Front Artif Intell 2024; 7:1385871. [PMID: 38708094 PMCID: PMC11066156 DOI: 10.3389/frai.2024.1385871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/08/2024] [Indexed: 05/07/2024] Open
Affiliation(s)
- Arianna Mencattini
- Department of Electronic Engineering, University of Rome Tor Vergata, Rome, Italy
- Interdisciplinary Center of Advanced Study of Organ-on-Chip and Lab-on-Chip Applications (IC-LOC), University of Rome Tor Vergata, Rome, Italy
| | - Elena Daprati
- Department of System Medicine and Centro di Biomedicina Spaziale (CBMS), University of Rome Tor Vergata, Rome, Italy
| | - David Della-Morte
- Interdisciplinary Center of Advanced Study of Organ-on-Chip and Lab-on-Chip Applications (IC-LOC), University of Rome Tor Vergata, Rome, Italy
- San Raffaele Rome University, Rome, Italy
| | - Fiorella Guadagni
- San Raffaele Rome University, Rome, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, Rome, Italy
| | - Federica Sangiuolo
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Eugenio Martinelli
- Department of Electronic Engineering, University of Rome Tor Vergata, Rome, Italy
- Interdisciplinary Center of Advanced Study of Organ-on-Chip and Lab-on-Chip Applications (IC-LOC), University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
20
|
Rokach M, Portioli C, Brahmachari S, Estevão BM, Decuzzi P, Barak B. Tackling myelin deficits in neurodevelopmental disorders using drug delivery systems. Adv Drug Deliv Rev 2024; 207:115218. [PMID: 38403255 DOI: 10.1016/j.addr.2024.115218] [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: 11/14/2023] [Revised: 01/27/2024] [Accepted: 02/20/2024] [Indexed: 02/27/2024]
Abstract
Interest in myelin and its roles in almost all brain functions has been greatly increasing in recent years, leading to countless new studies on myelination, as a dominant process in the development of cognitive functions. Here, we explore the unique role myelin plays in the central nervous system and specifically discuss the results of altered myelination in neurodevelopmental disorders. We present parallel developmental trajectories involving myelination that correlate with the onset of cognitive impairment in neurodevelopmental disorders and discuss the key challenges in the treatment of these chronic disorders. Recent developments in drug repurposing and nano/micro particle-based therapies are reviewed as a possible pathway to circumvent some of the main hurdles associated with early intervention, including patient's adherence and compliance, side effects, relapse, and faster route to possible treatment of these disorders. The strategy of drug encapsulation overcomes drug solubility and metabolism, with the possibility of drug targeting to a specific compartment, reducing side effects upon systemic administration.
Collapse
Affiliation(s)
- May Rokach
- Sagol School of Neuroscience, Tel-Aviv University, Israel
| | - Corinne Portioli
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Sayanti Brahmachari
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Bianca Martins Estevão
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Boaz Barak
- Sagol School of Neuroscience, Tel-Aviv University, Israel; Faculty of Social Sciences, The School of Psychological Sciences, Tel-Aviv University, Israel.
| |
Collapse
|
21
|
Salas-Lucia F, Escamilla S, Bianco AC, Dumitrescu A, Refetoff S. Impaired T3 uptake and action in MCT8-deficient cerebral organoids underlie Allan-Herndon-Dudley syndrome. JCI Insight 2024; 9:e174645. [PMID: 38376950 PMCID: PMC11128209 DOI: 10.1172/jci.insight.174645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 02/15/2024] [Indexed: 02/22/2024] Open
Abstract
Patients with mutations in the thyroid hormone (TH) cell transporter monocarboxylate transporter 8 (MCT8) gene develop severe neuropsychomotor retardation known as Allan-Herndon-Dudley syndrome (AHDS). It is assumed that this is caused by a reduction in TH signaling in the developing brain during both intrauterine and postnatal developmental stages, and treatment remains understandably challenging. Given species differences in brain TH transporters and the limitations of studies in mice, we generated cerebral organoids (COs) using human induced pluripotent stem cells (iPSCs) from MCT8-deficient patients. MCT8-deficient COs exhibited (i) altered early neurodevelopment, resulting in smaller neural rosettes with thinner cortical units, (ii) impaired triiodothyronine (T3) transport in developing neural cells, as assessed through deiodinase-3-mediated T3 catabolism, (iii) reduced expression of genes involved in cerebral cortex development, and (iv) reduced T3 inducibility of TH-regulated genes. In contrast, the TH analogs 3,5-diiodothyropropionic acid and 3,3',5-triiodothyroacetic acid triggered normal responses (induction/repression of T3-responsive genes) in MCT8-deficient COs, constituting proof of concept that lack of T3 transport underlies the pathophysiology of AHDS and demonstrating the clinical potential for TH analogs to be used in treating patients with AHDS. MCT8-deficient COs represent a species-specific relevant preclinical model that can be utilized to screen drugs with potential benefits as personalized therapeutics for patients with AHDS.
Collapse
Affiliation(s)
- Federico Salas-Lucia
- Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Sergio Escamilla
- Instituto de Neurociencias de Alicante, Miguel Hernández-CSIC University, Sant Joan d’Alacant, Alicante, Spain
| | - Antonio C. Bianco
- Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Alexandra Dumitrescu
- Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, Illinois, USA
- Committee on Molecular Metabolism and Nutrition
| | - Samuel Refetoff
- Section of Adult and Pediatric Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Chicago, Chicago, Illinois, USA
- Department of Pediatrics, and Committee on Genetics, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
22
|
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
|
23
|
Zarate-Lopez D, Torres-Chávez AL, Gálvez-Contreras AY, Gonzalez-Perez O. Three Decades of Valproate: A Current Model for Studying Autism Spectrum Disorder. Curr Neuropharmacol 2024; 22:260-289. [PMID: 37873949 PMCID: PMC10788883 DOI: 10.2174/1570159x22666231003121513] [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: 08/04/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 10/25/2023] Open
Abstract
Autism Spectrum Disorder (ASD) is a neurodevelopmental disorder with increased prevalence and incidence in recent decades. Its etiology remains largely unclear, but it seems to involve a strong genetic component and environmental factors that, in turn, induce epigenetic changes during embryonic and postnatal brain development. In recent decades, clinical studies have shown that inutero exposure to valproic acid (VPA), a commonly prescribed antiepileptic drug, is an environmental factor associated with an increased risk of ASD. Subsequently, prenatal VPA exposure in rodents has been established as a reliable translational model to study the pathophysiology of ASD, which has helped demonstrate neurobiological changes in rodents, non-human primates, and brain organoids from human pluripotent stem cells. This evidence supports the notion that prenatal VPA exposure is a valid and current model to replicate an idiopathic ASD-like disorder in experimental animals. This review summarizes and describes the current features reported with this animal model of autism and the main neurobiological findings and correlates that help elucidate the pathophysiology of ASD. Finally, we discuss the general framework of the VPA model in comparison to other environmental and genetic ASD models.
Collapse
Affiliation(s)
- David Zarate-Lopez
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima 28040, México
- Physiological Science Ph.D. Program, School of Medicine, University of Colima, Colima 28040, Mexico
| | - Ana Laura Torres-Chávez
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima 28040, México
- Physiological Science Ph.D. Program, School of Medicine, University of Colima, Colima 28040, Mexico
| | - Alma Yadira Gálvez-Contreras
- Department of Neuroscience, Centro Universitario de Ciencias de la Salud, University of Guadalajara, Guadalajara 44340, México
| | - Oscar Gonzalez-Perez
- Laboratory of Neuroscience, School of Psychology, University of Colima, Colima 28040, México
| |
Collapse
|
24
|
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
|
25
|
LeVine SM. Examining the Role of a Functional Deficiency of Iron in Lysosomal Storage Disorders with Translational Relevance to Alzheimer's Disease. Cells 2023; 12:2641. [PMID: 37998376 PMCID: PMC10670892 DOI: 10.3390/cells12222641] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 11/25/2023] Open
Abstract
The recently presented Azalea Hypothesis for Alzheimer's disease asserts that iron becomes sequestered, leading to a functional iron deficiency that contributes to neurodegeneration. Iron sequestration can occur by iron being bound to protein aggregates, such as amyloid β and tau, iron-rich structures not undergoing recycling (e.g., due to disrupted ferritinophagy and impaired mitophagy), and diminished delivery of iron from the lysosome to the cytosol. Reduced iron availability for biochemical reactions causes cells to respond to acquire additional iron, resulting in an elevation in the total iron level within affected brain regions. As the amount of unavailable iron increases, the level of available iron decreases until eventually it is unable to meet cellular demands, which leads to a functional iron deficiency. Normally, the lysosome plays an integral role in cellular iron homeostasis by facilitating both the delivery of iron to the cytosol (e.g., after endocytosis of the iron-transferrin-transferrin receptor complex) and the cellular recycling of iron. During a lysosomal storage disorder, an enzyme deficiency causes undigested substrates to accumulate, causing a sequelae of pathogenic events that may include cellular iron dyshomeostasis. Thus, a functional deficiency of iron may be a pathogenic mechanism occurring within several lysosomal storage diseases and Alzheimer's disease.
Collapse
Affiliation(s)
- Steven M LeVine
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
26
|
Zhang R, Quan H, Wang Y, Luo F. Neurogenesis in primates versus rodents and the value of non-human primate models. Natl Sci Rev 2023; 10:nwad248. [PMID: 38025664 PMCID: PMC10659238 DOI: 10.1093/nsr/nwad248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/21/2023] [Accepted: 09/10/2023] [Indexed: 12/01/2023] Open
Abstract
Neurogenesis, the process of generating neurons from neural stem cells, occurs during both embryonic and adult stages, with each stage possessing distinct characteristics. Dysfunction in either stage can disrupt normal neural development, impair cognitive functions, and lead to various neurological disorders. Recent technological advancements in single-cell multiomics and gene-editing have facilitated investigations into primate neurogenesis. Here, we provide a comprehensive overview of neurogenesis across rodents, non-human primates, and humans, covering embryonic development to adulthood and focusing on the conservation and diversity among species. While non-human primates, especially monkeys, serve as valuable models with closer neural resemblance to humans, we highlight the potential impacts and limitations of non-human primate models on both physiological and pathological neurogenesis research.
Collapse
Affiliation(s)
- Runrui Zhang
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China
| | - Hongxin Quan
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China
| | - Yinfeng Wang
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China
| | - Fucheng Luo
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming 650500, China
| |
Collapse
|
27
|
Matuleviciute R, Akinluyi ET, Muntslag TAO, Dewing JM, Long KR, Vernon AC, Tremblay ME, Menassa DA. Microglial contribution to the pathology of neurodevelopmental disorders in humans. Acta Neuropathol 2023; 146:663-683. [PMID: 37656188 PMCID: PMC10564830 DOI: 10.1007/s00401-023-02629-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/25/2023] [Accepted: 08/26/2023] [Indexed: 09/02/2023]
Abstract
Microglia are the brain's resident macrophages, which guide various developmental processes crucial for brain maturation, activity, and plasticity. Microglial progenitors enter the telencephalic wall by the 4th postconceptional week and colonise the fetal brain in a manner that spatiotemporally tracks key neurodevelopmental processes in humans. However, much of what we know about how microglia shape neurodevelopment comes from rodent studies. Multiple differences exist between human and rodent microglia warranting further focus on the human condition, particularly as microglia are emerging as critically involved in the pathological signature of various cognitive and neurodevelopmental disorders. In this article, we review the evidence supporting microglial involvement in basic neurodevelopmental processes by focusing on the human species. We next concur on the neuropathological evidence demonstrating whether and how microglia contribute to the aetiology of two neurodevelopmental disorders: autism spectrum conditions and schizophrenia. Next, we highlight how recent technologies have revolutionised our understanding of microglial biology with a focus on how these tools can help us elucidate at unprecedented resolution the links between microglia and neurodevelopmental disorders. We conclude by reviewing which current treatment approaches have shown most promise towards targeting microglia in neurodevelopmental disorders and suggest novel avenues for future consideration.
Collapse
Affiliation(s)
- Rugile Matuleviciute
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Elizabeth T Akinluyi
- Division of Medical Sciences, University of Victoria, Victoria, Canada
- Department of Pharmacology and Therapeutics, Afe Babalola University, Ado Ekiti, Nigeria
| | - Tim A O Muntslag
- Princess Maxima Centre for Paediatric Oncology, Utrecht, The Netherlands
| | | | - Katherine R Long
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Anthony C Vernon
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | | | - David A Menassa
- Department of Neuropathology & The Queen's College, University of Oxford, Oxford, UK.
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
28
|
Chaoul V, Dib EY, Bedran J, Khoury C, Shmoury O, Harb F, Soueid J. Assessing Drug Administration Techniques in Zebrafish Models of Neurological Disease. Int J Mol Sci 2023; 24:14898. [PMID: 37834345 PMCID: PMC10573323 DOI: 10.3390/ijms241914898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 10/15/2023] Open
Abstract
Neurological diseases, including neurodegenerative and neurodevelopmental disorders, affect nearly one in six of the world's population. The burden of the resulting deaths and disability is set to rise during the next few decades as a consequence of an aging population. To address this, zebrafish have become increasingly prominent as a model for studying human neurological diseases and exploring potential therapies. Zebrafish offer numerous benefits, such as genetic homology and brain similarities, complementing traditional mammalian models and serving as a valuable tool for genetic screening and drug discovery. In this comprehensive review, we highlight various drug delivery techniques and systems employed for therapeutic interventions of neurological diseases in zebrafish, and evaluate their suitability. We also discuss the challenges encountered during this process and present potential advancements in innovative techniques.
Collapse
Affiliation(s)
- Victoria Chaoul
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (V.C.); (J.B.); (O.S.)
| | - Emanuel-Youssef Dib
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat P.O. Box 100, Lebanon; (E.-Y.D.); (C.K.)
| | - Joe Bedran
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (V.C.); (J.B.); (O.S.)
| | - Chakib Khoury
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat P.O. Box 100, Lebanon; (E.-Y.D.); (C.K.)
| | - Omar Shmoury
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (V.C.); (J.B.); (O.S.)
| | - Frédéric Harb
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat P.O. Box 100, Lebanon; (E.-Y.D.); (C.K.)
| | - Jihane Soueid
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut P.O. Box 11-0236, Lebanon; (V.C.); (J.B.); (O.S.)
| |
Collapse
|
29
|
Björnholm L, Orell O, Kerkelä M, Honka U, Laasonen S, Riekki T, Surcel HM, Suvanto E, Veijola J. Maternal Thyroid Function During Pregnancy and Offspring White Matter Microstructure in Early Adulthood: A Prospective Birth Cohort Study. Thyroid 2023; 33:1245-1254. [PMID: 37498774 PMCID: PMC10611975 DOI: 10.1089/thy.2022.0699] [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] [Indexed: 07/29/2023]
Abstract
Background: The fetus is fully dependent on maternal thyroid hormones until mid-gestation and suboptimal maternal thyroid function has been associated with alterations in the neurodevelopment of the offspring. We used maternal free thyroxine (fT4) and thyrotropin (TSH) levels in early gestation to study the association of maternal thyroid function during early pregnancy and offspring brain white matter (WM) integrity in early adulthood. Methods: Our study population consisted of a total of 292 mother-child pairs. Maternal fT4 and TSH were used as predictors and offspring multimodal imaging measures of fractional anisotropy, mean diffusivity, and magnetization transfer ratio (FA, MD, and MTR) as dependent variables. First, as Global analysis, all analyzed 14 WM tracts were studied simultaneously using linear-mixed effect models. Second, if a global effect was detected, a post hoc Tract-wise analysis was carried out using linear models individually in each WM tract. Study population was stratified by sex. Results: We found a positive association between maternal fT4 and offspring Global FA in males when adjusted for all maternal and offspring covariates (n = 114; β = 0.154; confidence interval = 0.045-0.263; p = 0.006). The finding was observed to be driven by multiple WM tracts, of which three projection fiber tracts and the forceps minor survived correcting for multiple comparisons in Tract-wise analysis. Conclusions: Maternal thyroid function in early pregnancy was observed to be associated with WM microstructure in male offspring in early adulthood. Our results suggest that maternal fT4 levels in early pregnancy may modulate axonal characteristics, with a long-term effect on offspring WM development.
Collapse
Affiliation(s)
- Lassi Björnholm
- Research Unit of Clinical Medicine, Department of Psychiatry, University of Oulu, Oulu, Finland
- Department of Psychiatry, Oulu University Hospital, Oulu, Finland
- Medical Research Centre Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Olavi Orell
- Research Unit of Clinical Medicine, Department of Psychiatry, University of Oulu, Oulu, Finland
| | - Martta Kerkelä
- Research Unit of Clinical Medicine, Department of Psychiatry, University of Oulu, Oulu, Finland
| | - Ulriika Honka
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Sini Laasonen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Tiina Riekki
- Research Unit of Clinical Medicine, Department of Psychiatry, University of Oulu, Oulu, Finland
- Department of Psychiatry, Oulu University Hospital, Oulu, Finland
- Medical Research Centre Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | | | - Eila Suvanto
- Department of Obstetrics and Gynecology, Oulu University Hospital and MRC Oulu University, Oulu, Finland
| | - Juha Veijola
- Research Unit of Clinical Medicine, Department of Psychiatry, University of Oulu, Oulu, Finland
- Department of Psychiatry, Oulu University Hospital, Oulu, Finland
- Medical Research Centre Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| |
Collapse
|
30
|
Abraham M, Peterburs J, Mundorf A. Oligodendrocytes matter: a review of animal studies on early adversity. J Neural Transm (Vienna) 2023; 130:1177-1185. [PMID: 37138023 PMCID: PMC10460720 DOI: 10.1007/s00702-023-02643-4] [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: 12/09/2022] [Accepted: 04/26/2023] [Indexed: 05/05/2023]
Abstract
Exposure to adversities in early life appears to affect the development of white matter, especially oligodendrocytes. Furthermore, altered myelination is present in regions subjected to maturation during the developmental time when early adversities are experienced. In this review, studies applying two well-established animal models of early life adversity, namely maternal separation and maternal immune activation, focusing on oligodendrocyte alterations and resulting implications for psychiatric disorders are discussed. Studies revealed that myelination is reduced as a result of altered oligodendrocyte expression. Furthermore, early adversity is associated with increased cell death, a simpler morphology, and inhibited oligodendrocyte maturation. However, these effects seem to be region- specific as some brain regions show increased expression while others show decreased expression of oligodendroglia-related genes, and they occur especially in regions of ongoing development. Some studies furthermore suggest that early adversity leads to premature differentiation of oligodendrocytes. Importantly, especially early exposure results in stronger oligodendrocyte-related impairments. However, resulting alterations are not restricted to exposure during the early pre- and postnatal days as social isolation after weaning leads to fewer internodes and branches and shorter processes of oligodendrocytes in adulthood. Eventually, the found alterations may lead to dysfunction and long-lasting alterations in structural brain development associated with psychiatric disorders. To date, only few preclinical studies have focused on the effects of early adversity on oligodendrocytes. More studies including several developmental stages are needed to further disentangle the role of oligodendrocytes in the development of psychiatric disorders.
Collapse
Affiliation(s)
- Mate Abraham
- Division of Experimental and Molecular Psychiatry, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Jutta Peterburs
- Institute for Systems Medicine and Department of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Annakarina Mundorf
- Institute for Systems Medicine and Department of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany.
| |
Collapse
|
31
|
Ruffaner-Hanson CD, Fernandez-Oropeza AK, Sun MS, Caldwell KK, Allan AM, Savage DD, Valenzuela CF, Noor S, Milligan ED. Prenatal alcohol exposure alters mRNA expression for stress peptides, glucocorticoid receptor function and immune factors in acutely stressed neonatal brain. Front Neurosci 2023; 17:1203557. [PMID: 37425005 PMCID: PMC10326286 DOI: 10.3389/fnins.2023.1203557] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/24/2023] [Indexed: 07/11/2023] Open
Abstract
Background The amygdala, hippocampus and hypothalamus are critical stress regulatory areas that undergo functional maturation for stress responding initially established during gestational and early postnatal brain development. Fetal alcohol spectrum disorder (FASD), a consequence of prenatal alcohol exposure (PAE), results in cognitive, mood and behavioral disorders. Prenatal alcohol exposure negatively impacts components of the brain stress response system, including stress-associated brain neuropeptides and glucocorticoid receptors in the amygdala, hippocampus and hypothalamus. While PAE generates a unique brain cytokine expression pattern, little is known about the role of Toll-like receptor 4 (TLR4) and related proinflammatory signaling factors, as well as anti-inflammatory cytokines in PAE brain stress-responsive regions. We hypothesized that PAE sensitizes the early brain stress response system resulting in dysregulated neuroendocrine and neuroimmune activation. Methods A single, 4-h exposure of maternal separation stress in male and female postnatal day 10 (PND10) C57Bl/6 offspring was utilized. Offspring were from either prenatal control exposure (saccharin) or a limited access (4 h) drinking-in-the-dark model of PAE. Immediately after stress on PND10, the hippocampus, amygdala and hypothalamus were collected, and mRNA expression was analyzed for stress-associated factors (CRH and AVP), glucocorticoid receptor signaling regulators (GAS5, FKBP51 and FKBP52), astrocyte and microglial activation, and factors associated with TLR4 activation including proinflammatory interleukin-1β (IL-1β), along with additional pro- and anti-inflammatory cytokines. Select protein expression analysis of CRH, FKBP and factors associated with the TLR4 signaling cascade from male and female amygdala was conducted. Results The female amygdala revealed increased mRNA expression in stress-associated factors, glucocorticoid receptor signaling regulators and all of the factors critical in the TLR4 activation cascade, while the hypothalamus revealed blunted mRNA expression of all of these factors in PAE following stress. Conversely, far fewer mRNA changes were observed in males, notably in the hippocampus and hypothalamus, but not the amygdala. Statistically significant increases in CRH protein, and a strong trend in increased IL-1β were observed in male offspring with PAE independent of stressor exposure. Conclusion Prenatal alcohol exposure creates stress-related factors and TLR-4 neuroimmune pathway sensitization observed predominantly in females, that is unmasked in early postnatal life by a stress challenge.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Erin D. Milligan
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico
| |
Collapse
|
32
|
Gradisnik L, Velnar T. Astrocytes in the central nervous system and their functions in health and disease: A review. World J Clin Cases 2023; 11:3385-3394. [PMID: 37383914 PMCID: PMC10294192 DOI: 10.12998/wjcc.v11.i15.3385] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 02/19/2023] [Accepted: 04/14/2023] [Indexed: 05/25/2023] Open
Abstract
Astrocytes are key cells in the central nervous system. They are involved in many important functions under physiological and pathological conditions. As part of neuroglia, they have been recognised as cellular elements in their own right. The name astrocyte was first proposed by Mihaly von Lenhossek in 1895 because of the finely branched processes and star-like appearance of these particular cells. As early as the late 19th and early 20th centuries, Ramon y Cajal and Camillo Golgi had noted that although astrocytes have stellate features, their morphology is extremely diverse. Modern research has confirmed the morphological diversity of astrocytes both in vitro and in vivo and their complex, specific, and important roles in the central nervous system. In this review, the functions of astrocytes and their roles are described.
Collapse
Affiliation(s)
- Lidija Gradisnik
- Institute of Biomedical Sciences, Medical Faculty Maribor, Maribor 2000, Slovenia
| | - Tomaz Velnar
- Department of Neurosurgery, University Medical Centre Ljubljana, Ljubljana 1000, Slovenia
- AMEU ECM Maribor, Maribor 2000, Slovenia
| |
Collapse
|
33
|
Seeker LA, Bestard-Cuche N, Jäkel S, Kazakou NL, Bøstrand SMK, Wagstaff LJ, Cholewa-Waclaw J, Kilpatrick AM, Van Bruggen D, Kabbe M, Baldivia Pohl F, Moslehi Z, Henderson NC, Vallejos CA, La Manno G, Castelo-Branco G, Williams A. Brain matters: unveiling the distinct contributions of region, age, and sex to glia diversity and CNS function. Acta Neuropathol Commun 2023; 11:84. [PMID: 37217978 PMCID: PMC10204264 DOI: 10.1186/s40478-023-01568-z] [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: 04/10/2023] [Accepted: 04/15/2023] [Indexed: 05/24/2023] Open
Abstract
The myelinated white matter tracts of the central nervous system (CNS) are essential for fast transmission of electrical impulses and are often differentially affected in human neurodegenerative diseases across CNS region, age and sex. We hypothesize that this selective vulnerability is underpinned by physiological variation in white matter glia. Using single nucleus RNA sequencing of human post-mortem white matter samples from the brain, cerebellum and spinal cord and subsequent tissue-based validation we found substantial glial heterogeneity with tissue region: we identified region-specific oligodendrocyte precursor cells (OPCs) that retain developmental origin markers into adulthood, distinguishing them from mouse OPCs. Region-specific OPCs give rise to similar oligodendrocyte populations, however spinal cord oligodendrocytes exhibit markers such as SKAP2 which are associated with increased myelin production and we found a spinal cord selective population particularly equipped for producing long and thick myelin sheaths based on the expression of genes/proteins such as HCN2. Spinal cord microglia exhibit a more activated phenotype compared to brain microglia, suggesting that the spinal cord is a more pro-inflammatory environment, a difference that intensifies with age. Astrocyte gene expression correlates strongly with CNS region, however, astrocytes do not show a more activated state with region or age. Across all glia, sex differences are subtle but the consistent increased expression of protein-folding genes in male donors hints at pathways that may contribute to sex differences in disease susceptibility. These findings are essential to consider for understanding selective CNS pathologies and developing tailored therapeutic strategies.
Collapse
Affiliation(s)
- Luise A Seeker
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Edinburgh Bioquarter, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Nadine Bestard-Cuche
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Edinburgh Bioquarter, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Sarah Jäkel
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Edinburgh Bioquarter, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
- Institute for Stroke and Dementia Research, Klinikum Der Universität München, Ludwig-Maximilians-Universität, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Nina-Lydia Kazakou
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Edinburgh Bioquarter, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Sunniva M K Bøstrand
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Edinburgh Bioquarter, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Laura J Wagstaff
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Edinburgh Bioquarter, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Justyna Cholewa-Waclaw
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Edinburgh Bioquarter, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Alastair M Kilpatrick
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Edinburgh Bioquarter, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - David Van Bruggen
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Mukund Kabbe
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Fabio Baldivia Pohl
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Zahra Moslehi
- Laboratory of Neurodevelopmental Systems Biology, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Neil C Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Catalina A Vallejos
- MRC Human Genetics Unit, Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh, EH4 2XU, UK
- The Alan Turing Institute, 96 Euston Road, London, NW1 2DB, UK
| | - Gioele La Manno
- Laboratory of Neurodevelopmental Systems Biology, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | - Goncalo Castelo-Branco
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77, Stockholm, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, Stockholm Node, 171 77, Stockholm, Sweden
| | - Anna Williams
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, Edinburgh Bioquarter, University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK.
| |
Collapse
|
34
|
Yi C, Verkhratsky A, Niu J. Pathological potential of oligodendrocyte precursor cells: terra incognita. Trends Neurosci 2023:S0166-2236(23)00103-0. [PMID: 37183154 DOI: 10.1016/j.tins.2023.04.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/12/2023] [Accepted: 04/13/2023] [Indexed: 05/16/2023]
Abstract
Adult oligodendrocyte precursor cells (aOPCs), transformed from fetal OPCs, are idiosyncratic neuroglia of the central nervous system (CNS) that are distinct in many ways from other glial cells. OPCs have been classically studied in the context of their remyelinating capacity. Recent studies, however, revealed that aOPCs not only contribute to post-lesional remyelination but also play diverse crucial roles in multiple neurological diseases. In this review we briefly present the physiology of aOPCs and summarize current knowledge of the beneficial and detrimental roles of aOPCs in different CNS diseases. We discuss unique features of aOPC death, reactivity, and changes during senescence, as well as aOPC interactions with other glial cells and pathological remodeling during disease. Finally, we outline future perspectives for the study of aOPCs in brain pathologies which may instigate the development of aOPC-targeting therapeutic strategies.
Collapse
Affiliation(s)
- Chenju Yi
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China; Department of Pathology, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China; Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Shenzhen 518107, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, China.
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine, and Health, University of Manchester, Manchester M13 9PL, UK; Achucarro Centre for Neuroscience, Basque Foundation for Science (IKERBASQUE), Bilbao 48011, Spain; Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102 Vilnius, Lithuania; Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China.
| | - Jianqin Niu
- Department of Histology and Embryology, Chongqing Key Laboratory of Neurobiology, Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
35
|
Galinsky R, Dhillon SK, Kelly SB, Wassink G, Davidson JO, Lear CA, van den Heuij LG, Bennet L, Gunn AJ. Magnesium sulphate reduces tertiary gliosis but does not improve EEG recovery or white or grey matter cell survival after asphyxia in preterm fetal sheep. J Physiol 2023; 601:1999-2016. [PMID: 36999348 PMCID: PMC10952359 DOI: 10.1113/jp284381] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/09/2023] [Indexed: 04/01/2023] Open
Abstract
Maternal magnesium sulphate (MgSO4 ) treatment is widely recommended before preterm birth for neuroprotection. However, this is controversial because there is limited evidence that MgSO4 provides long-term neuroprotection. Preterm fetal sheep (104 days gestation; term is 147 days) were assigned randomly to receive sham occlusion with saline infusion (n = 6) or i.v. infusion with MgSO4 (n = 7) or vehicle (saline, n = 6) from 24 h before hypoxia-ischaemia induced by umbilical cord occlusion until 24 h after occlusion. Sheep were killed after 21 days of recovery, for fetal brain histology. Functionally, MgSO4 did not improve long-term EEG recovery. Histologically, in the premotor cortex and striatum, MgSO4 infusion attenuated post-occlusion astrocytosis (GFAP+ ) and microgliosis but did not affect numbers of amoeboid microglia or improve neuronal survival. In the periventricular and intragyral white matter, MgSO4 was associated with fewer total (Olig-2+ ) oligodendrocytes compared with vehicle + occlusion. Numbers of mature (CC1+ ) oligodendrocytes were reduced to a similar extent in both occlusion groups compared with sham occlusion. In contrast, MgSO4 was associated with an intermediate improvement in myelin density in the intragyral and periventricular white matter tracts. In conclusion, a clinically comparable dose of MgSO4 was associated with moderate improvements in white and grey matter gliosis and myelin density but did not improve EEG maturation or neuronal or oligodendrocyte survival. KEY POINTS: Magnesium sulphate is widely recommended before preterm birth for neuroprotection; however, there is limited evidence that magnesium sulphate provides long-term neuroprotection. In preterm fetal sheep exposed to hypoxia-ischaemia (HI), MgSO4 was associated with attenuated astrocytosis and microgliosis in the premotor cortex and striatum but did not improve neuronal survival after recovery to term-equivalent age, 21 days after HI. Magnesium sulphate was associated with loss of total oligodendrocytes in the periventricular and intragyral white matter tracts, whereas mature, myelinating oligodendrocytes were reduced to a similar extent in both occlusion groups. In the same regions, MgSO4 was associated with an intermediate improvement in myelin density. Functionally, MgSO4 did not improve long-term recovery of EEG power, frequency or sleep stage cycling. A clinically comparable dose of MgSO4 was associated with moderate improvements in white and grey matter gliosis and myelin density but did not improve EEG maturation or neuronal or oligodendrocyte survival.
Collapse
Affiliation(s)
- Robert Galinsky
- Department of PhysiologyUniversity of AucklandAucklandNew Zealand
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of Obstetrics and GynaecologyMonash UniversityVictoriaAustralia
| | | | - Sharmony B. Kelly
- The Ritchie CentreHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of Obstetrics and GynaecologyMonash UniversityVictoriaAustralia
| | - Guido Wassink
- Department of PhysiologyUniversity of AucklandAucklandNew Zealand
| | | | | | | | - Laura Bennet
- Department of PhysiologyUniversity of AucklandAucklandNew Zealand
| | - Alistair J. Gunn
- Department of PhysiologyUniversity of AucklandAucklandNew Zealand
| |
Collapse
|
36
|
Sobierajski E, Lauer G, Czubay K, Grabietz H, Beemelmans C, Beemelmans C, Meyer G, Wahle P. Development of myelin in fetal and postnatal neocortex of the pig, the European wild boar Sus scrofa. Brain Struct Funct 2023; 228:947-966. [PMID: 37000250 PMCID: PMC10147765 DOI: 10.1007/s00429-023-02633-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 03/15/2023] [Indexed: 04/01/2023]
Abstract
Myelination of the neocortex of altricial species is mostly a postnatal event, and the appearance of myelin has been associated with the end of the critical period for ocular dominance plasticity in rodent visual cortex. Due to their precocality, ungulates may tell a different story. Here, we analyzed the development of PDGFRα positive oligodendrocyte precursor cells and expression of myelin proteins in the laminar compartments of fetal and postnatal porcine cortex from E45 onwards. Precursor cell density initially increased and then decreased but remained present at P90. MAG and MBP staining were detectable at E70 in subventricular zone and deep white matter, ascending into gyral white matter at E85, and into the gray matter and marginal zone at E100 (birth in pig at E114). Protein blots confirmed the declining expression of PDGFRα from E65 onwards, and the increase of MBP and MAG expression from E80 onwards. Somatosensory input elicited by spontaneous activity is considered important for the formation of the body representation. Indeed, PDGFRα, MBP and MAG expression started earlier in somatosensory than in visual cortex. Taken together, myelination proceeded in white and gray matter and marginal zone of pig cortex before birth with an areal-specific time course, and an almost mature pattern was present at P5 in visual cortex.
Collapse
Affiliation(s)
- Eric Sobierajski
- Faculty of Biology and Biotechnology, Developmental Neurobiology, Ruhr University Bochum, 44870, Bochum, Germany
| | - German Lauer
- Faculty of Biology and Biotechnology, Developmental Neurobiology, Ruhr University Bochum, 44870, Bochum, Germany
| | - Katrin Czubay
- Faculty of Biology and Biotechnology, Developmental Neurobiology, Ruhr University Bochum, 44870, Bochum, Germany
| | - Hannah Grabietz
- Faculty of Biology and Biotechnology, Developmental Neurobiology, Ruhr University Bochum, 44870, Bochum, Germany
| | - Christa Beemelmans
- Regionalverband Ruhr Grün, Forsthof Üfter Mark, Forsthausweg 306, 46514, Schermbeck, Germany
| | - Christoph Beemelmans
- Regionalverband Ruhr Grün, Forsthof Üfter Mark, Forsthausweg 306, 46514, Schermbeck, Germany
| | - Gundela Meyer
- Department of Basic Medical Science, Faculty of Medicine, University of La Laguna, 38200, Santa Cruz de Tenerife, Tenerife, Spain
| | - Petra Wahle
- Faculty of Biology and Biotechnology, Developmental Neurobiology, Ruhr University Bochum, 44870, Bochum, Germany.
| |
Collapse
|
37
|
Li B, Zhao H, Tu Z, Yang W, Han R, Wang L, Luo X, Pan M, Chen X, Zhang J, Xu H, Guo X, Yan S, Yin P, Zhao Z, Liu J, Luo Y, Li Y, Yang Z, Zhang B, Tan Z, Xu H, Jiang T, Jiang YH, Li S, Zhang YQ, Li XJ. CHD8 mutations increase gliogenesis to enlarge brain size in the nonhuman primate. Cell Discov 2023; 9:27. [PMID: 36878905 PMCID: PMC9988832 DOI: 10.1038/s41421-023-00525-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 01/27/2023] [Indexed: 03/08/2023] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition that affects social interaction and behavior. Mutations in the gene encoding chromodomain helicase DNA-binding protein 8 (CHD8) lead to autism symptoms and macrocephaly by a haploinsufficiency mechanism. However, studies of small animal models showed inconsistent findings about the mechanisms for CHD8 deficiency-mediated autism symptoms and macrocephaly. Using the nonhuman primate as a model system, we found that CRISPR/Cas9-mediated CHD8 mutations in the embryos of cynomolgus monkeys led to increased gliogenesis to cause macrocephaly in cynomolgus monkeys. Disrupting CHD8 in the fetal monkey brain prior to gliogenesis increased the number of glial cells in newborn monkeys. Moreover, knocking down CHD8 via CRISPR/Cas9 in organotypic monkey brain slices from newborn monkeys also enhanced the proliferation of glial cells. Our findings suggest that gliogenesis is critical for brain size in primates and that abnormal gliogenesis may contribute to ASD.
Collapse
Grants
- UL1 TR001863 NCATS NIH HHS
- This work was supported by Department of Science and Technology of Guangdong Province (2021ZT09Y007; 2020B121201006, 2018B030337001, X.J. Li), Guangzhou Key Research Program on Brain Science (202007030008, X.J. Li)the National Science Foundation of China to X.J. Li (81830032, 31872779).
- the Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence Fund (2019018, B. Li), the Postdoctoral Science Foundation of China (2019M653275, B. Li)
- the National Science Foundation of China to H. Zhao (32100783)
- the Fundamental Research Funds for the Central Universities (21619104, L. Wang)
- the Strategic Priority Research Program B of the Chinese Academy of Sciences (XDBS1020100 to Y.Q. Zhang), the National Key Research and Development Program (2019YFA0707100 and 2021ZD0203901 to Y.Q. Zhang),the National Science Foundation of China to Y.Q. Zhang (31830036 and 31921002).
Collapse
Affiliation(s)
- Bang Li
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong, China
| | - Hui Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Zhuchi Tu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong, China
| | - Weili Yang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong, China
| | - Rui Han
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong, China
| | - Lu Wang
- Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University & Institute of Molecular and Functional Imaging, Jinan University, Guangzhou, Guangdong, China
| | - Xiaopeng Luo
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong, China
| | - Mingtian Pan
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong, China
| | - Xiusheng Chen
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong, China
| | - Jiawei Zhang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong, China
| | - Huijuan Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Xiangyu Guo
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong, China
| | - Sen Yan
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong, China
| | - Peng Yin
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong, China
| | - Zhiguang Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Jianrong Liu
- Yuanxi Biotech Inc., Guangzhou, Guangdong, China
| | - Yafeng Luo
- Yuanxi Biotech Inc., Guangzhou, Guangdong, China
| | - Yuefeng Li
- Guangdong Landau Biotechnology Co. Ltd., Guangzhou, Guangdong, China
| | - Zhengyi Yang
- Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Baogui Zhang
- Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Zhiqiang Tan
- Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University & Institute of Molecular and Functional Imaging, Jinan University, Guangzhou, Guangdong, China
| | - Hao Xu
- Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University & Institute of Molecular and Functional Imaging, Jinan University, Guangzhou, Guangdong, China
| | - Tianzi Jiang
- Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Yong-Hui Jiang
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Shihua Li
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong, China
| | - Yong Q Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.
| | - Xiao-Jiang Li
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong, China.
| |
Collapse
|
38
|
Dwivedi S, Choudhary P, Gupta A, Singh S. Therapeutical growth in oligodendroglial fate induction via transdifferentiation of stem cells for neuroregenerative therapy. Biochimie 2023; 211:35-56. [PMID: 36842627 DOI: 10.1016/j.biochi.2023.02.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/20/2022] [Accepted: 02/21/2023] [Indexed: 02/27/2023]
Abstract
The merits of stem cell therapy and research are undisputed due to their widespread usage in the treatment of neurodegenerative diseases and demyelinating disorders. Cell replacement therapy especially revolves around stem cells and their induction into different cell lineages both adult and progenitor - belonging to each germ layer, prior to transplantation or disease modeling studies. The nervous system is abundant in glial cells and among these are oligodendrocytes capable of myelinating new-born neurons and remyelination of axons with lost or damaged myelin sheath. But demyelinating diseases generate tremendous deficit between myelin loss and recovery. To compensate for this loss, analyze the defects in remyelination mechanisms as well as to trigger full recovery in such patients mesenchymal stem cells (MSCs) have been induced to transdifferentiate into oligodendrocytes. But such experiments are riddled with problems like prolonged, tenuous and complicated protocols that stretch longer than the time taken for the spread of demyelination-associated after-effects. This review delves into such protocols and the combinations of different molecules and factors that have been recruited to derive bona fide oligodendrocytes from in vitro differentiation of embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs) and MSCs with special focus on MSC-derived oligodendrocytes.
Collapse
Affiliation(s)
- Shrey Dwivedi
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, U.P., India
| | - Princy Choudhary
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, U.P., India
| | - Ayushi Gupta
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, U.P., India
| | - Sangeeta Singh
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, U.P., India.
| |
Collapse
|
39
|
Secondary Degeneration of Oligodendrocyte Precursor Cells Occurs as Early as 24 h after Optic Nerve Injury in Rats. Int J Mol Sci 2023; 24:ijms24043463. [PMID: 36834873 PMCID: PMC9964292 DOI: 10.3390/ijms24043463] [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: 12/22/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
Optic nerve injury causes secondary degeneration, a sequela that spreads damage from the primary injury to adjacent tissue, through mechanisms such as oxidative stress, apoptosis, and blood-brain barrier (BBB) dysfunction. Oligodendrocyte precursor cells (OPCs), a key component of the BBB and oligodendrogenesis, are vulnerable to oxidative deoxyribonucleic acid (DNA) damage by 3 days post-injury. However, it is unclear whether oxidative damage in OPCs occurs earlier at 1 day post-injury, or whether a critical 'window-of-opportunity' exists for therapeutic intervention. Here, a partial optic nerve transection rat model of secondary degeneration was used with immunohistochemistry to assess BBB dysfunction, oxidative stress, and proliferation in OPCs vulnerable to secondary degeneration. At 1 day post-injury, BBB breach and oxidative DNA damage were observed, alongside increased density of DNA-damaged proliferating cells. DNA-damaged cells underwent apoptosis (cleaved caspase3+), and apoptosis was associated with BBB breach. OPCs experienced DNA damage and apoptosis and were the major proliferating cell type with DNA damage. However, the majority of caspase3+ cells were not OPCs. These results provide novel insights into acute secondary degeneration mechanisms in the optic nerve, highlighting the need to consider early oxidative damage to OPCs in therapeutic efforts to limit degeneration following optic nerve injury.
Collapse
|
40
|
Anastasaki C, Gao Y, Gutmann DH. Neurons as stromal drivers of nervous system cancer formation and progression. Dev Cell 2023; 58:81-93. [PMID: 36693322 PMCID: PMC9883043 DOI: 10.1016/j.devcel.2022.12.011] [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: 05/16/2022] [Revised: 06/24/2022] [Accepted: 12/27/2022] [Indexed: 01/24/2023]
Abstract
Similar to their pivotal roles in nervous system development, neurons have emerged as critical regulators of cancer initiation, maintenance, and progression. Focusing on nervous system tumors, we describe the normal relationships between neurons and other cell types relevant to normal nerve function, and discuss how disruptions of these interactions promote tumor evolution, focusing on electrical (gap junctions) and chemical (synaptic) coupling, as well as the establishment of new paracrine relationships. We also review how neuron-tumor communication contributes to some of the complications of cancer, including neuropathy, chemobrain, seizures, and pain. Finally, we consider the implications of cancer neuroscience in establishing risk for tumor penetrance and in the design of future anti-tumoral treatments.
Collapse
Affiliation(s)
- Corina Anastasaki
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yunqing Gao
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
41
|
Romero JC, Berlinicke C, Chow S, Duan Y, Wang Y, Chamling X, Smirnova L. Oligodendrogenesis and myelination tracing in a CRISPR/Cas9-engineered brain microphysiological system. Front Cell Neurosci 2023; 16:1094291. [PMID: 36744062 PMCID: PMC9893511 DOI: 10.3389/fncel.2022.1094291] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/16/2022] [Indexed: 01/20/2023] Open
Abstract
Introduction Oligodendrocytes (OLs) are the myelin-forming cells of the central nervous system (CNS). Although OLs can be differentiated from human-induced pluripotent stem cells (hiPSCs), the in vitro modeling of axon myelination in human cells remains challenging. Brain microphysiological systems (bMPS, e.g. organoids) are complex three-dimensional (3D) cultures that offer an ideal system to study this process as OLs differentiate in a more in vivo-like environment; surrounded by neurons and astrocytes, which support the myelination of axons. Methods Here, we take advantage of CRISPR/Cas9 technology to generate a hiPSC line in which proteolipid protein 1 (PLP1), an OLs marker, is tagged with super-fold GFP (sfGFP). While generating the PLP1-sfGFP reporter, we used reverse transfection and obtained higher Knock-In (KI) efficiency compared to forward transfection (61-72 vs. 46%). Results After validation of the KI and quality control of the PLP1-sfGFP line, selected clones were differentiated into bMPS, and the fidelity, specificity, and function of the tagged PLP protein were verified in this model. We tracked different stages of oligodendrogenesis in the verified lines based on PLP1-sfGFP+ cells' morphology, and the presence of PLP1-sfGFP surrounding axons during bMPS' differentiation. Finally, we challenged the bMPS with cuprizone and quantified changes in both the percentage of PLP1-sfGFP expressing cells and the intensity of GFP expression. Discussion This work demonstrates an efficient method for generating hiPSC KI lines and the description of a new 3D model to study OL differentiation, migration, and maturation both during in vitro neurodevelopment as well as in response to environmental chemicals or disease-associated stressors.
Collapse
Affiliation(s)
- July Carolina Romero
- Bloomberg School of Public Health, Center for Alternatives to Animal Testing, Johns Hopkins University, Baltimore, MD, United States
| | - Cynthia Berlinicke
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sharon Chow
- Bloomberg School of Public Health, Center for Alternatives to Animal Testing, Johns Hopkins University, Baltimore, MD, United States
| | - Yukan Duan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yifei Wang
- Bloomberg School of Public Health, Center for Alternatives to Animal Testing, Johns Hopkins University, Baltimore, MD, United States
| | - Xitiz Chamling
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lena Smirnova
- Bloomberg School of Public Health, Center for Alternatives to Animal Testing, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
42
|
Menassa DA, Kopić J, Junaković A, Kostović I, Krsnik Ž. Microglial Characterization in Transient Human Neurodevelopmental Structures. Dev Neurosci 2023; 45:1-7. [PMID: 36720218 PMCID: PMC10015752 DOI: 10.1159/000528911] [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: 10/28/2022] [Accepted: 12/19/2022] [Indexed: 01/06/2023] Open
Abstract
Human neurodevelopment is characterized by the appearance, development, and disappearance or transformation of various transient structures that underlie the establishment of connectivity within and between future cortical and subcortical areas. Examples of transient structures in the forebrain (among many others) include the subpial granular layer and the subplate zone. We have previously characterized the precise spatiotemporal dynamics of microglia in the human telencephalon. Here, we describe the diversity of microglial morphologies in the subpial granular layer and the subplate zone. Where possible, we couple the predominant morphological phenotype with functional characterizations to infer tentative roles for microglia in a changing neurodevelopmental landscape. We interpret these findings within the context of relevant morphogenetic and neurogenetic events in humans. Due to the unique genetic, molecular, and anatomical features of the human brain and because many human neurological and psychiatric diseases have their origins during development, these structures deserve special attention.
Collapse
Affiliation(s)
- David A. Menassa
- The Queen's College, University of Oxford, Oxford, UK
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Janja Kopić
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Alisa Junaković
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Ivica Kostović
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Željka Krsnik
- Croatian Institute for Brain Research, School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
43
|
Paes-Colli Y, Trindade PMP, Vitorino LC, Piscitelli F, Iannotti FA, Campos RMP, Isaac AR, de Aguiar AFL, Allodi S, de Mello FG, Einicker-Lamas M, de Siqueira-Santos R, Di Marzo V, Tannous BA, Carvalho LA, De Melo Reis RA, Sampaio LS. Activation of cannabinoid type 1 receptor (CB1) modulates oligodendroglial process branching complexity in rat hippocampal cultures stimulated by olfactory ensheathing glia-conditioned medium. Front Cell Neurosci 2023; 17:1134130. [PMID: 37138770 PMCID: PMC10150319 DOI: 10.3389/fncel.2023.1134130] [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: 12/29/2022] [Accepted: 03/16/2023] [Indexed: 05/05/2023] Open
Abstract
The endocannabinoid system (ECS) refers to a complex cell-signaling system highly conserved among species formed by numerous receptors, lipid mediators (endocannabinoids) and synthetic and degradative enzymes. It is widely distributed throughout the body including the CNS, where it participates in synaptic signaling, plasticity and neurodevelopment. Besides, the olfactory ensheathing glia (OEG) present in the olfactory system is also known to play an important role in the promotion of axonal growth and/or myelination. Therefore, both OEG and the ECS promote neurogenesis and oligodendrogenesis in the CNS. Here, we investigated if the ECS is expressed in cultured OEG, by assessing the main markers of the ECS through immunofluorescence, western blotting and qRT-PCR and quantifying the content of endocannabinoids in the conditioned medium of these cells. After that, we investigated whether the production and release of endocannabinoids regulate the differentiation of oligodendrocytes co-cultured with hippocampal neurons, through Sholl analysis in oligodendrocytes expressing O4 and MBP markers. Additionally, we evaluated through western blotting the modulation of downstream pathways such as PI3K/Akt/mTOR and ERK/MAPK, being known to be involved in the proliferation and differentiation of oligodendrocytes and activated by CB1, which is the major endocannabinoid responsive receptor in the brain. Our data show that OEG expresses key genes of the ECS, including the CB1 receptor, FAAH and MAGL. Besides, we were able to identify AEA, 2-AG and AEA related mediators palmitoylethanolamide (PEA) and oleoylethanolamide (OEA), in the conditioned medium of OEG cultures. These cultures were also treated with URB597 10-9 M, a FAAH selective inhibitor, or JZL184 10-9 M, a MAGL selective inhibitor, which led to the increase in the concentrations of OEA and 2-AG in the conditioned medium. Moreover, we found that the addition of OEG conditioned medium (OEGCM) enhanced the complexity of oligodendrocyte process branching in hippocampal mixed cell cultures and that this effect was inhibited by AM251 10-6 M, a CB1 receptor antagonist. However, treatment with the conditioned medium enriched with OEA or 2-AG did not alter the process branching complexity of premyelinating oligodendrocytes, while decreased the branching complexity in mature oligodendrocytes. We also observed no change in the phosphorylation of Akt and ERK 44/42 in any of the conditions used. In conclusion, our data show that the ECS modulates the number and maturation of oligodendrocytes in hippocampal mixed cell cultures.
Collapse
Affiliation(s)
- Yolanda Paes-Colli
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Priscila M. P. Trindade
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Louise C. Vitorino
- Laboratório de Neurobiologia Comparativa e do Desenvolvimento, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabiana Piscitelli
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, CNR, Pozzuoli, Italy
| | - Fabio Arturo Iannotti
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, CNR, Pozzuoli, Italy
| | - Raquel M. P. Campos
- Laboratório de Neurobiologia Comparativa e do Desenvolvimento, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alinny R. Isaac
- Laboratório de Doenças Neurodegenerativas, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Andrey Fabiano Lourenço de Aguiar
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Silvana Allodi
- Laboratório de Neurobiologia Comparativa e do Desenvolvimento, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernando G. de Mello
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo Einicker-Lamas
- Laboratório de Biomembranas, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Raphael de Siqueira-Santos
- Laboratório de Agregação de Proteínas e Amiloidoses, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Istituto di Chimica Biomolecolare, CNR, Pozzuoli, Italy
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis, Laval University, Quebec, QC, Canada
| | - Bakhos A. Tannous
- Experimental Therapeutics and Molecular Imaging Laboratory, Massachusetts General Hospital, Boston, MA, United States
- Neuroscience Program, Harvard Medical School, Boston, MA, United States
| | - Litia A. Carvalho
- Experimental Therapeutics and Molecular Imaging Laboratory, Massachusetts General Hospital, Boston, MA, United States
- Neuroscience Program, Harvard Medical School, Boston, MA, United States
| | - Ricardo A. De Melo Reis
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luzia S. Sampaio
- Laboratório de Neuroquímica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- *Correspondence: Luzia S. Sampaio,
| |
Collapse
|
44
|
Xie Y, Yang Y, Yuan T. Brain Damage in the Preterm Infant: Clinical Aspects and Recent Progress in the Prevention and Treatment. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:27-40. [PMID: 35209835 DOI: 10.2174/1871527321666220223092905] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/16/2022] [Accepted: 01/16/2022] [Indexed: 12/16/2022]
Abstract
Although the prevalence of brain injury and related neurodevelopmental disabilities resulting from preterm birth are major public health concerns, there are no definite neuroprotective strategies to prevent or reduce brain injury. The pattern of brain injury seen in preterm infants has evolved into more subtle lesions that are still essential to diagnose regarding neurodevelopmental outcomes. There is no specific effective method for the treatment of premature infant brain injury, and the focus of clinical treatment is still on prevention. Prevention of this injury requires insight into the pathogenesis, but many gaps exist in our understanding of how neonatal treatment procedures and medications impact cerebral hemodynamics and preterm brain injury. Many studies provide evidence about the prevention of premature infant brain injury, which is related to some drugs (such as erythropoietin, melatonin, mesenchymal stem cells, etc.). However, there are still some controversies about the quality of research and the effectiveness of therapy. This review aims to recapitulate the results of preclinical studies and provide an update on the latest developments around etiological pathways, prevention, and treatment.
Collapse
Affiliation(s)
- Yixuan Xie
- Department of Neonatology, Children\'s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang, P.R. China
| | - Yue Yang
- Department of Neonatology, Children\'s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang, P.R. China
| | - Tianming Yuan
- Department of Neonatology, Children\'s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang, P.R. China
| |
Collapse
|
45
|
Hou J, Chen Y, Grajales-Reyes G, Colonna M. TREM2 dependent and independent functions of microglia in Alzheimer's disease. Mol Neurodegener 2022; 17:84. [PMID: 36564824 PMCID: PMC9783481 DOI: 10.1186/s13024-022-00588-y] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 12/02/2022] [Indexed: 12/25/2022] Open
Abstract
Microglia are central players in brain innate immunity and have been the subject of extensive research in Alzheimer's disease (AD). In this review, we aim to summarize the genetic and functional discoveries that have advanced our understanding of microglia reactivity to AD pathology. Given the heightened AD risk posed by rare variants of the microglial triggering receptor expressed on myeloid cells 2 (TREM2), we will focus on the studies addressing the impact of this receptor on microglia responses to amyloid plaques, tauopathy and demyelination pathologies in mouse and human. Finally, we will discuss the implications of recent discoveries on microglia and TREM2 biology on potential therapeutic strategies for AD.
Collapse
Affiliation(s)
- Jinchao Hou
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Yun Chen
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA ,grid.4367.60000 0001 2355 7002Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Gary Grajales-Reyes
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Marco Colonna
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110 USA
| |
Collapse
|
46
|
Identifying Genes that Affect Differentiation of Human Neural Stem Cells and Myelination of Mature Oligodendrocytes. Cell Mol Neurobiol 2022:10.1007/s10571-022-01313-5. [DOI: 10.1007/s10571-022-01313-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022]
Abstract
AbstractHuman neural stem cells (NSCs) are self-renewing, multipotent cells of the central nervous system (CNS). They are characterized by their ability to differentiate into a range of cells, including oligodendrocytes (OLs), neurons, and astrocytes, depending on exogenous stimuli. An efficient and easy directional differentiation method was developed for obtaining large quantities of high-quality of human OL progenitor cells (OPCs) and OLs from NSCs. RNA sequencing, immunofluorescence staining, flow cytometry, western blot, label-free proteomic sequencing, and qPCR were performed in OL lines differentiated from NSC lines. The changes in the positive rate of typical proteins were analyzed expressed by NSCs, neurons, astrocytes, OPCs, and OLs. We assessed Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways of differentially expressed (DE) messenger RNAs (mRNAs) related to the differentiation of NSCs and the maturation of OLs. The percentage of NSCs differentiated into neurons, astrocytes, and OLs was 82.13%, 80.19%, and 90.15%, respectively. We found that nestin, PAX6, Musashi, and vimentin were highly expressed in NSCs; PDGFR-α, A2B5, NG2, OLIG2, SOX10, and NKX2-2 were highly expressed in OPCs; and CNP, GALC, PLP1, and MBP were highly expressed in OLs. RNA sequencing, western blot and qPCR revealed that ERBB4 and SORL1 gradually increased during NSC–OL differentiation. In conclusion, NSCs can differentiate into neurons, astrocytes, and OLs efficiently. PDGFR-α, APC, ID4, PLLP, and other markers were related to NSC differentiation and OL maturation. Moreover, we refined a screening method for ERBB4 and SORL1, which may underlie NSC differentiation and OL maturation.
Graphical Abstract
Potential unreported genes and proteins may regulate differentiation of human neural stem cells into oligodendrocyte lineage. Neural stem cells (NSCs) can differentiate into neurons, astrocytes, and oligodendrocyte (OLs) efficiently. By analyzing the DE mRNAs and proteins of NSCs and OLs lineage, we could identify reported markers and unreported markers of ERBB4 and SORL1 that may underlie regulate NSC differentiation and OL maturation.
Collapse
|
47
|
Farley SJ, Grishok A, Zeldich E. Shaking up the silence: consequences of HMGN1 antagonizing PRC2 in the Down syndrome brain. Epigenetics Chromatin 2022; 15:39. [PMID: 36463299 PMCID: PMC9719135 DOI: 10.1186/s13072-022-00471-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/11/2022] [Indexed: 12/04/2022] Open
Abstract
Intellectual disability is a well-known hallmark of Down Syndrome (DS) that results from the triplication of the critical region of human chromosome 21 (HSA21). Major studies were conducted in recent years to gain an understanding about the contribution of individual triplicated genes to DS-related brain pathology. Global transcriptomic alterations and widespread changes in the establishment of neural lineages, as well as their differentiation and functional maturity, suggest genome-wide chromatin organization alterations in trisomy. High Mobility Group Nucleosome Binding Domain 1 (HMGN1), expressed from HSA21, is a chromatin remodeling protein that facilitates chromatin decompaction and is associated with acetylated lysine 27 on histone H3 (H3K27ac), a mark correlated with active transcription. Recent studies causatively linked overexpression of HMGN1 in trisomy and the development of DS-associated B cell acute lymphoblastic leukemia (B-ALL). HMGN1 has been shown to antagonize the activity of the Polycomb Repressive Complex 2 (PRC2) and prevent the deposition of histone H3 lysine 27 trimethylation mark (H3K27me3), which is associated with transcriptional repression and gene silencing. However, the possible ramifications of the increased levels of HMGN1 through the derepression of PRC2 target genes on brain cell pathology have not gained attention. In this review, we discuss the functional significance of HMGN1 in brain development and summarize accumulating reports about the essential role of PRC2 in the development of the neural system. Mechanistic understanding of how overexpression of HMGN1 may contribute to aberrant brain cell phenotypes in DS, such as altered proliferation of neural progenitors, abnormal cortical architecture, diminished myelination, neurodegeneration, and Alzheimer's disease-related pathology in trisomy 21, will facilitate the development of DS therapeutic approaches targeting chromatin.
Collapse
Affiliation(s)
- Sean J. Farley
- grid.189504.10000 0004 1936 7558Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
| | - Alla Grishok
- grid.189504.10000 0004 1936 7558Department of Biochemistry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA ,grid.189504.10000 0004 1936 7558Boston University Genome Science Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA USA
| | - Ella Zeldich
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
48
|
Abstract
ABSTRACT
Deconstructing and then reconstructing developmental processes ex vivo is crucial to understanding how organs assemble and how physiology can be disrupted in disease. Human 3D stem cell-derived systems, such as organoids, have facilitated this pursuit; however, they often do not capture inter-tissue or inter-lineage cellular interactions that give rise to emergent tissue properties during development. Assembloids are self-organizing 3D cellular systems that result from the integration of multiple organoids or the combination of organoids with missing cell types or primary tissue explants. Here, we outline the concept and types of assembloids and present their applications for studying the nervous system and other tissues. We describe tools that are used to probe and manipulate assembloids and delineate current challenges and the potential for this new approach to interrogate development and disease.
Collapse
Affiliation(s)
- Sabina Kanton
- Stanford University 1 Department of Psychiatry and Behavioral Sciences , , Stanford, CA 94305, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X 2 , Stanford, CA 94305, USA
| | - Sergiu P. Paşca
- Stanford University 1 Department of Psychiatry and Behavioral Sciences , , Stanford, CA 94305, USA
- Stanford Brain Organogenesis Program, Wu Tsai Neuroscience Institute & Bio-X 2 , Stanford, CA 94305, USA
| |
Collapse
|
49
|
Habibey R, Rojo Arias JE, Striebel J, Busskamp V. Microfluidics for Neuronal Cell and Circuit Engineering. Chem Rev 2022; 122:14842-14880. [PMID: 36070858 PMCID: PMC9523714 DOI: 10.1021/acs.chemrev.2c00212] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Indexed: 02/07/2023]
Abstract
The widespread adoption of microfluidic devices among the neuroscience and neurobiology communities has enabled addressing a broad range of questions at the molecular, cellular, circuit, and system levels. Here, we review biomedical engineering approaches that harness the power of microfluidics for bottom-up generation of neuronal cell types and for the assembly and analysis of neural circuits. Microfluidics-based approaches are instrumental to generate the knowledge necessary for the derivation of diverse neuronal cell types from human pluripotent stem cells, as they enable the isolation and subsequent examination of individual neurons of interest. Moreover, microfluidic devices allow to engineer neural circuits with specific orientations and directionality by providing control over neuronal cell polarity and permitting the isolation of axons in individual microchannels. Similarly, the use of microfluidic chips enables the construction not only of 2D but also of 3D brain, retinal, and peripheral nervous system model circuits. Such brain-on-a-chip and organoid-on-a-chip technologies are promising platforms for studying these organs as they closely recapitulate some aspects of in vivo biological processes. Microfluidic 3D neuronal models, together with 2D in vitro systems, are widely used in many applications ranging from drug development and toxicology studies to neurological disease modeling and personalized medicine. Altogether, microfluidics provide researchers with powerful systems that complement and partially replace animal models.
Collapse
Affiliation(s)
- Rouhollah Habibey
- Department
of Ophthalmology, Universitäts-Augenklinik
Bonn, University of Bonn, Ernst-Abbe-Straße 2, D-53127 Bonn, Germany
| | - Jesús Eduardo Rojo Arias
- Wellcome—MRC
Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge
Biomedical Campus, University of Cambridge, Cambridge CB2 0AW, United Kingdom
| | - Johannes Striebel
- Department
of Ophthalmology, Universitäts-Augenklinik
Bonn, University of Bonn, Ernst-Abbe-Straße 2, D-53127 Bonn, Germany
| | - Volker Busskamp
- Department
of Ophthalmology, Universitäts-Augenklinik
Bonn, University of Bonn, Ernst-Abbe-Straße 2, D-53127 Bonn, Germany
| |
Collapse
|
50
|
Mohammadi A, Higazy R, Gauda EB. PGC-1α activity and mitochondrial dysfunction in preterm infants. Front Physiol 2022; 13:997619. [PMID: 36225305 PMCID: PMC9548560 DOI: 10.3389/fphys.2022.997619] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/09/2022] [Indexed: 11/26/2022] Open
Abstract
Extremely low gestational age neonates (ELGANs) are born in a relatively hyperoxic environment with weak antioxidant defenses, placing them at high risk for mitochondrial dysfunction affecting multiple organ systems including the nervous, respiratory, ocular, and gastrointestinal systems. The brain and lungs are highly affected by mitochondrial dysfunction and dysregulation in the neonate, causing white matter injury (WMI) and bronchopulmonary dysplasia (BPD), respectively. Adequate mitochondrial function is important in providing sufficient energy for organ development as it relates to alveolarization and axonal myelination and decreasing oxidative stress via reactive oxygen species (ROS) and reactive nitrogen species (RNS) detoxification. Peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) is a master regulator of mitochondrial biogenesis and function. Since mitochondrial dysfunction is at the root of WMI and BPD pathobiology, exploring therapies that can regulate PGC-1α activity may be beneficial. This review article describes several promising therapeutic agents that can mitigate mitochondrial dysfunction through direct and indirect activation and upregulation of the PGC-1α pathway. Metformin, resveratrol, omega 3 fatty acids, montelukast, L-citrulline, and adiponectin are promising candidates that require further pre-clinical and clinical studies to understand their efficacy in decreasing the burden of disease from WMI and BPD in preterm infants.
Collapse
Affiliation(s)
- Atefeh Mohammadi
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Randa Higazy
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, Toronto, ON, Canada
| | - Estelle B. Gauda
- The Hospital for Sick Children, Division of Neonatology, Department of Pediatrics and Translational Medicine Program, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- *Correspondence: Estelle B. Gauda,
| |
Collapse
|