1
|
Serdar M, Walther KA, Gallert M, Kempe K, Obst S, Labusek N, Herrmann R, Herz J, Felderhoff-Müser U, Bendix I. Prenatal inflammation exacerbates hyperoxia-induced neonatal brain injury. J Neuroinflammation 2025; 22:57. [PMID: 40022130 PMCID: PMC11871844 DOI: 10.1186/s12974-025-03389-4] [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: 11/25/2024] [Accepted: 02/20/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Premature born infants are at high risk to develop white matter injury (WMI). Hyperoxia and perinatal inflammation are main risk factors for preterm birth and associated brain injury. To date the majority of experimental studies have focused on isolated insults. However, clinically, WMI injury is a multifactorial disorder caused by a variety of triggers. To establish a clinically relevant rodent model of WMI, we combined prenatal inflammation with postnatal hyperoxia to investigate individual, and additive or synergistic effects on inflammatory processes, myelination and grey matter development. METHODS At embryonic day 20, pregnant Wistar rat dams received either a single intraperitoneal injection of 100 µg/ kg lipopolysaccharide (LPS) or sodium chloride. Offspring were either exposed to hyperoxia (80% O2) or normoxia (21% O2) from postnatal day 3 to 5. Animals were sacrificed immediately after hyperoxia or 6 days later, corresponding to term-equivalent age. White and grey matter development and neuroinflammatory responses were investigated at cellular and molecular levels applying immunohistochemistry, western blotting, real time PCR in brain tissues and multiplex protein expression analysis on serum samples. RESULTS Prenatal inflammation combined with postnatal hyperoxia resulted in reduced body weight and length in the offspring, accompanied by increased serum leptin levels at term equivalent age. The altered body parameters, like body weight, were associated with decreased brain volume, thinning of deep cortical layers and hypomyelination. As potential underlying mechanisms, we identified severe myelination deficits and an increased microglia activation associated with elevated inflammatory cytokine expression in brain tissues, while peripheral cytokine levels were reduced. Interestingly, effects on body size were mainly mediated by prenatal LPS, independent of hyperoxia, while oligodendrocyte degeneration was mainly induced by postnatal hyperoxia, independent of prenatal inflammation. However, for the majority of pathological changes, including brain size, myelination deficits, microglia activation and inflammatory cytokine expression, additive or synergistic effects were detected. CONCLUSION Prenatal inflammation combined with postnatal hyperoxia results in aggravated myelination deficits and inflammatory responses compared to single insults, making it an ideal model to improve our understanding of the complex pathophysiology underlying WMI and to evaluate urgently needed therapies.
Collapse
Affiliation(s)
- Meray Serdar
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Kay-Anja Walther
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Markus Gallert
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Karina Kempe
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Stefanie Obst
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Nicole Labusek
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ralf Herrmann
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Josephine Herz
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ursula Felderhoff-Müser
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany.
| | - Ivo Bendix
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
2
|
Basu AP, Low K, Ratnaike T, Rowitch D. Genetic investigations in cerebral palsy. Dev Med Child Neurol 2025; 67:177-185. [PMID: 39208295 PMCID: PMC11695794 DOI: 10.1111/dmcn.16080] [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: 05/08/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
The original description of cerebral palsy (CP) contained case histories suggesting that perinatal environmental stressors resulted in brain injury and neurodevelopmental disability. While there are clear associations between environmental impact on brain development and CP, recent studies indicate an 11% to 40% incidence of monogenic conditions in patients given a diagnosis of CP. A genetic diagnosis supports the delivery of personalized medicine. In this review, we describe how the Wnt pathway exemplifies our understanding of pathophysiology related to a gene variant (CTNNB1) found in some children diagnosed with CP. We cover studies undertaken to establish the baseline prevalence of monogenic conditions in populations attending CP clinics. We list factors indicating increased likelihood of a genomic diagnosis; and we highlight the need for a comprehensive, accurate, genotype-phenotype reference data set to aid variant interpretation in CP cohorts. We also consider the wider societal implications of genomic management of CP including significance of the diagnostic label, benefits and pitfalls of a genetic diagnosis, logistics, and cost.
Collapse
Affiliation(s)
- Anna P. Basu
- Population Health Sciences Institute, Newcastle UniversityNewcastle upon TyneUK
- Paediatric NeurologyGreat North Children's HospitalNewcastle upon TyneUK
| | - Karen Low
- Centre for Academic Child HealthUniversity of BristolBristolUK
- Department of Clinical GeneticsUniversity Hospitals Bristol and Weston NHS TrustBristolUK
| | - Thiloka Ratnaike
- Department of PaediatricsUniversity of CambridgeCambridgeUK
- PaediatricsColchester Hospital, East Suffolk and North Essex NHS Foundation TrustColchesterUK
| | - David Rowitch
- Department of PaediatricsUniversity of CambridgeCambridgeUK
| |
Collapse
|
3
|
Wang X, Zang J, Yang Y, Li K, Ye D, Wang Z, Wang Q, Wu Y, Luan Z. Human neural stem cells transplanted during the sequelae phase alleviate motor deficits in a rat model of cerebral palsy. Cytotherapy 2024; 26:1491-1504. [PMID: 39186025 DOI: 10.1016/j.jcyt.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 08/27/2024]
Abstract
AIMS Cerebral palsy (CP) is the most common physical disability in children, yet lacks an ideal animal model or effective treatment. This study aimed to develop a reliable CP model in neonatal rats and explore the effectiveness and underlying mechanisms of human neural stem cells (hNSCs) transplantation during the sequelae phase of CP. METHODS Vasoconstrictor endothelin-1 (ET-1) was administered intracranially to the motor cortex and striatum of rats on postnatal day 5 to establish a CP model. hNSCs (5 × 105/5 μL) pretreated with hypoxia (5% O2 for 24 h) were transplanted near the infarct 3 weeks after ET-1 injury (the sequelae phase). The distribution and differentiation of hNSCs were observed after transplantation. Changes in neurotrophic factors, neurogenesis, angiogenesis, axonal plasticity, and motor function were analyzed. RESULTS Neurobehavioral tests showed poor muscle strength and postural control in young ET-1 rats. Motor deficits of the left forelimb and gait abnormalities persisted into adulthood. Histopathological findings and MRI indicated the atrophy of the cortex, striatum, and adjacent corpus callosum in ET-1 rats. At 56 days after transplantation, hNSCs were widely distributed in the ipsilateral hemisphere, and differentiated into neurons, oligodendrocytes and astrocytes. Transplantation of hNSCs increased BDNF and VEGF expression, EdU+ cell number in the SVZ area, RECA-1+ vessel density and GAP-43 intensity around the lesion in ET-1 rats. The cylinder test revealed a significant increase in the left forelimb motor function from 28 days after transplantation, and the staircase and CatWalk tests showed improvements in fine motor function and gait parameters. CONCLUSIONS Intracerebral injection of ET-1 modelled key functional and histopathological features of CP. hNSCs transplanted during the sequelae phase of CP resulted in long-term improvement in motor performance, possibly attributed to its capacity to stimulate neurotrophic factors, facilitate neurogenesis, angiogenesis, and promote axonal plasticity.
Collapse
Affiliation(s)
- Xiaohua Wang
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China, 100048; Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China, 226001
| | - Jing Zang
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China, 100048
| | - Yinxiang Yang
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China, 100048
| | - Ke Li
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China, 100048
| | - Dou Ye
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China, 100048
| | - Zhaoyan Wang
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China, 100048
| | - Qian Wang
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China, 100048
| | - Youjia Wu
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China, 226001.
| | - Zuo Luan
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China, 100048.
| |
Collapse
|
4
|
Alart JA, Álvarez A, Catalan A, Herrero de la Parte B, Alonso-Alconada D. Dimethyl Fumarate Strongly Ameliorates Gray and White Matter Brain Injury and Modulates Glial Activation after Severe Hypoxia-Ischemia in Neonatal Rats. Antioxidants (Basel) 2024; 13:1122. [PMID: 39334781 PMCID: PMC11428775 DOI: 10.3390/antiox13091122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Neonatal hypoxia-ischemia is a major cause of infant death and disability. The only clinically accepted treatment is therapeutic hypothermia; however, cooling is less effective in the most severely encephalopathic infants. Here, we wanted to test the neuroprotective effect of the antioxidant dimethyl fumarate after severe hypoxia-ischemia in neonatal rats. We used a modified Rice-Vannucci model to generate severe hypoxic-ischemic brain damage in day 7 postnatal rats, which were randomized into four experimental groups: Sham, Sham + DMF, non-treated HI, and HI + DMF. We analyzed brain tissue loss, global and regional (cortex and hippocampus) neuropathological scores, white matter injury, and microglial and astroglial reactivity. Compared to non-treated HI animals, HI + DMF pups showed a reduced brain area loss (p = 0.0031), an improved neuropathological score (p = 0.0016), reduced white matter injuries by preserving myelin tracts (p < 0.001), and diminished astroglial (p < 0.001) and microglial (p < 0.01) activation. After severe hypoxia-ischemia in neonatal rats, DMF induced a strong neuroprotective response, reducing cerebral infarction, gray and white matter damage, and astroglial and microglial activation. Although further molecular studies are needed and its translation to human babies would need to evaluate the molecule in piglets or lambs, DMF may be a potential treatment against neonatal encephalopathy.
Collapse
Affiliation(s)
- Jon Ander Alart
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Antonia Álvarez
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Ana Catalan
- Psychiatry Department, OSI Bilbao-Basurto, Basurto University Hospital, 48013 Bilbao, Spain
- Neuroscience Department, University of the Basque Country (UPV/EHU), 48013 Leioa, Spain
- Biobizkaia Health Research Institute, 48903 Barakaldo, Spain
- CIBERSAM, Centro Investigación Biomédica en Red de Salud Mental, 28007 Madrid, Spain
- Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Borja Herrero de la Parte
- Department of Surgery and Radiology and Physical Medicine, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Daniel Alonso-Alconada
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| |
Collapse
|
5
|
Xu L, Yuan Z, Zhou J, Zhao Y, Liu W, Lu S, He Z, Qiang B, Shu P, Chen Y, Peng X. Temporal transcriptomic dynamics in developing macaque neocortex. eLife 2024; 12:RP90325. [PMID: 38415809 PMCID: PMC10911584 DOI: 10.7554/elife.90325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024] Open
Abstract
Despite intense research on mice, the transcriptional regulation of neocortical neurogenesis remains limited in humans and non-human primates. Cortical development in rhesus macaque is known to recapitulate multiple facets of cortical development in humans, including the complex composition of neural stem cells and the thicker supragranular layer. To characterize temporal shifts in transcriptomic programming responsible for differentiation from stem cells to neurons, we sampled parietal lobes of rhesus macaque at E40, E50, E70, E80, and E90, spanning the full period of prenatal neurogenesis. Single-cell RNA sequencing produced a transcriptomic atlas of developing parietal lobe in rhesus macaque neocortex. Identification of distinct cell types and neural stem cells emerging in different developmental stages revealed a terminally bifurcating trajectory from stem cells to neurons. Notably, deep-layer neurons appear in the early stages of neurogenesis, while upper-layer neurons appear later. While these different lineages show overlap in their differentiation program, cell fates are determined post-mitotically. Trajectories analysis from ventricular radial glia (vRGs) to outer radial glia (oRGs) revealed dynamic gene expression profiles and identified differential activation of BMP, FGF, and WNT signaling pathways between vRGs and oRGs. These results provide a comprehensive overview of the temporal patterns of gene expression leading to different fates of radial glial progenitors during neocortex layer formation.
Collapse
Affiliation(s)
- Longjiang Xu
- Institute of Medical Biology Chinese Academy of Medical Sciences, Chinese Academy of Medical Science and Peking Union Medical CollegeKunmingChina
| | - Zan Yuan
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- Agricultural Bioinformatics Key Laboratory of Hubei Province, Hubei Engineering Technology Research Center of Agricultural Big Data, College of Informatics, Huazhong Agricultural UniversityWuhanChina
| | - Jiafeng Zhou
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Common Mechanism Research for Major Diseases, Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Yuan Zhao
- Institute of Medical Biology Chinese Academy of Medical Sciences, Chinese Academy of Medical Science and Peking Union Medical CollegeKunmingChina
| | - Wei Liu
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Common Mechanism Research for Major Diseases, Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Shuaiyao Lu
- Institute of Medical Biology Chinese Academy of Medical Sciences, Chinese Academy of Medical Science and Peking Union Medical CollegeKunmingChina
| | - Zhanlong He
- Institute of Medical Biology Chinese Academy of Medical Sciences, Chinese Academy of Medical Science and Peking Union Medical CollegeKunmingChina
| | - Boqin Qiang
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Common Mechanism Research for Major Diseases, Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Pengcheng Shu
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Common Mechanism Research for Major Diseases, Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
- Chinese Institute for Brain ResearchBeijingChina
| | - Yang Chen
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Common Mechanism Research for Major Diseases, Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Xiaozhong Peng
- Institute of Medical Biology Chinese Academy of Medical Sciences, Chinese Academy of Medical Science and Peking Union Medical CollegeKunmingChina
- Department of Molecular Biology and Biochemistry, Institute of Basic Medical Sciences, Medical Primate Research Center, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical CollegeBeijingChina
- State Key Laboratory of Respiratory Health and Multimorbidity, Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
- Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| |
Collapse
|
6
|
Tavasoli S, Tavasoli M, Shojaeefard M, Farahmand F. Analysis of cerebral palsy gait based on movement primitives. Clin Biomech (Bristol, Avon) 2023; 104:105947. [PMID: 37030255 DOI: 10.1016/j.clinbiomech.2023.105947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/03/2023] [Accepted: 03/22/2023] [Indexed: 04/10/2023]
Abstract
BACKGROUND Cerebral palsy is the most prevalent motor disorder among children. Despite extensive studies on motor modularity of gait of children with cerebral palsy, kinematic modularity of their gait has not been addressed which is the main goal of this study. METHODS The kinematics of the gait of 13 typical development children and 188 children with cerebral palsy was captured and analyzed, where the cerebral palsy children were grouped into True, Jump, Apparent, and Crouch. Non-negative matrix factorization method was used to extract the kinematic modulus of each group, which were then clustered to find their characteristic movement primitives. The movement primitives of groups were then matched based on the similarity of their activation profiles. FINDINGS The number of movement primitives was three for the Crouch group, four for the other cerebral palsy groups, and five for the typical development group. Compared to the typical development children, the kinematic modules and activations of the cerebral palsy groups involved higher variability and co-activation, respectively (P < 0.05). Three temporally matched movement primitives were shared by all groups, but with altered structures. INTERPRETATION The gait of children with cerebral palsy involved lower complexity and higher variability due to the reduced and inconsistent kinematic modularity. Three basic movement primitives were sufficient to prodcue the overall gait kinematics, as observed in the Crouch group. Other movement primitives, were responsible for providing smooth transitions between basic movement primitives, as seen in more complex gait patterns.
Collapse
Affiliation(s)
- Shahab Tavasoli
- Mechanical Engineering Department, Sharif University of Technology, Tehran, Iran
| | - Marzieh Tavasoli
- Mechanical Engineering Department, Sharif University of Technology, Tehran, Iran
| | - Mahya Shojaeefard
- Mechanical Engineering Department, Sharif University of Technology, Tehran, Iran
| | - Farzam Farahmand
- Mechanical Engineering Department, Sharif University of Technology, Tehran, Iran.
| |
Collapse
|
7
|
Holloway RK, Zhang L, Molina-Gonzalez I, Ton K, Nicoll JAR, Boardman JP, Liang Y, Williams A, Miron VE. Localized microglia dysregulation impairs central nervous system myelination in development. Acta Neuropathol Commun 2023; 11:49. [PMID: 36949514 PMCID: PMC10035254 DOI: 10.1186/s40478-023-01543-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/03/2023] [Indexed: 03/24/2023] Open
Abstract
Myelination of neuronal axons is a critical aspect of central nervous system development and function. However, the fundamental cellular and molecular mechanisms influencing human developmental myelination and its failure are not fully understood. Here, we used digital spatial transcriptomics of a rare bank of human developing white matter to uncover that a localized dysregulated innate immune response is associated with impeded myelination. We identified that poorly myelinating areas have a distinct signature of Type II interferon signalling in microglia/macrophages, relative to adjacent myelinating areas. This is associated with a surprising increase in mature oligodendrocytes, which fail to form myelin processes appropriately. We functionally link these findings by showing that conditioned media from interferon-stimulated microglia is sufficient to dysregulate myelin process formation by oligodendrocytes in culture. We identify the Type II interferon inducer, Osteopontin (SPP1), as being upregulated in poorly myelinating brains, indicating a potential biomarker. Our results reveal the importance of microglia-mature oligodendrocyte interaction and interferon signaling in regulating myelination of the developing human brain.
Collapse
Affiliation(s)
- Rebecca K Holloway
- Keenan Research Centre for Biomedial Science at St. Michael's Hospital, 209 Victoria Street, Toronto, ON, M5B 1T8, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, Scotland, UK
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, Scotland, UK
| | - Liang Zhang
- Nanostring Technologies, Inc., Seattle, WA, USA
| | - Irene Molina-Gonzalez
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, Scotland, UK
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, Scotland, UK
| | - Kathy Ton
- Nanostring Technologies, Inc., Seattle, WA, USA
| | - James A R Nicoll
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Department of Cellular Pathology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - James P Boardman
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, Scotland, UK
| | - Yan Liang
- Nanostring Technologies, Inc., Seattle, WA, USA
| | - Anna Williams
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, UK
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, Scotland, UK
| | - Veronique E Miron
- Keenan Research Centre for Biomedial Science at St. Michael's Hospital, 209 Victoria Street, Toronto, ON, M5B 1T8, Canada.
- Department of Immunology, University of Toronto, Toronto, ON, Canada.
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, UK.
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, Scotland, UK.
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, Scotland, UK.
| |
Collapse
|
8
|
Renz P, Schoeberlein A, Haesler V, Maragkou T, Surbek D, Brosius Lutz A. A Novel Murine Multi-Hit Model of Perinatal Acute Diffuse White Matter Injury Recapitulates Major Features of Human Disease. Biomedicines 2022; 10:biomedicines10112810. [PMID: 36359331 PMCID: PMC9687579 DOI: 10.3390/biomedicines10112810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/17/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022] Open
Abstract
The selection of an appropriate animal model is key to the production of results with optimal relevance to human disease. Particularly in the case of perinatal brain injury, a dearth of affected human neonatal tissue available for research purposes increases the reliance on animal models for insight into disease mechanisms. Improvements in obstetric and neonatal care in the past 20 years have caused the pathologic hallmarks of perinatal white matter injury (WMI) to evolve away from cystic necrotic lesions and toward diffuse regions of reactive gliosis and persistent myelin disruption. Therefore, updated animal models are needed that recapitulate the key features of contemporary disease. Here, we report a murine model of acute diffuse perinatal WMI induced through a two-hit inflammatory–hypoxic injury paradigm. Consistent with diffuse human perinatal white matter injury (dWMI), our model did not show the formation of cystic lesions. Corresponding to cellular outcomes of dWMI, our injury protocol produced reactive microgliosis and astrogliosis, disrupted oligodendrocyte maturation, and disrupted myelination.. Functionally, we observed sensorimotor and cognitive deficits in affected mice. In conclusion, we report a novel murine model of dWMI that induces a pattern of brain injury mirroring multiple key aspects of the contemporary human clinical disease scenario.
Collapse
Affiliation(s)
- Patricia Renz
- Department for BioMedical Research, University of Bern and Switzerland, 3010 Bern, Switzerland
- Department of Obstetrics and Gynecology, Division of Feto-Maternal Medicine University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Andreina Schoeberlein
- Department for BioMedical Research, University of Bern and Switzerland, 3010 Bern, Switzerland
- Department of Obstetrics and Gynecology, Division of Feto-Maternal Medicine University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Valérie Haesler
- Department for BioMedical Research, University of Bern and Switzerland, 3010 Bern, Switzerland
- Department of Obstetrics and Gynecology, Division of Feto-Maternal Medicine University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Theoni Maragkou
- Institute of Pathology, University of Bern, 3010 Bern, Switzerland
| | - Daniel Surbek
- Department for BioMedical Research, University of Bern and Switzerland, 3010 Bern, Switzerland
- Department of Obstetrics and Gynecology, Division of Feto-Maternal Medicine University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Amanda Brosius Lutz
- Department for BioMedical Research, University of Bern and Switzerland, 3010 Bern, Switzerland
- Department of Obstetrics and Gynecology, Division of Feto-Maternal Medicine University Hospital, University of Bern, 3010 Bern, Switzerland
- Correspondence:
| |
Collapse
|
9
|
Araneda R, Ebner-Karestinos D, Dricot L, Herman E, Hatem SM, Friel KM, Gordon AM, Bleyenheuft Y. Impact of early brain lesions on the optic radiations in children with cerebral palsy. Front Neurosci 2022; 16:924938. [PMID: 36278011 PMCID: PMC9583910 DOI: 10.3389/fnins.2022.924938] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Due to their early brain lesion, children with unilateral spastic cerebral palsy (USCP) present important changes in brain gray and white matter, often manifested by perturbed sensorimotor functions. We predicted that type and side of the lesion could influence the microstructure of white matter tracts. Using diffusion tensor imaging in 40 children with USCP, we investigated optic radiation (OR) characteristics: fractional anisotropy (FA), mean diffusivity (MD), axial diffusivity (AD) and radial diffusivity (RD). First, we compared the OR of the lesional and non-lesional hemisphere. Then we evaluated the impact of the brain lesion type (periventricular or cortico-subcortical) and side in the differences observed in the lesional and non-lesional OR. Additionally, we examined the relationship between OR characteristics and performance of a visuospatial attention task. We observed alterations in the OR of children with USCP on the lesional hemisphere compared with the non-lesional hemisphere in the FA, MD and RD. These differences were influenced by the type of lesion and by the side of the lesion. A correlation was also observed between FA, MD and RD and the visuospatial assessment mainly in children with periventricular and right lesions. Our results indicate an important role of the timing and side of the lesion in the resulting features of these children’s OR and probably in the compensation resulting from neuroplastic changes.
Collapse
Affiliation(s)
- Rodrigo Araneda
- Institute of Neuroscience, Université Catholique de Louvain, Ottignies-Louvain-la-Neuve, Belgium
- Exercise and Rehabilitation Science Institute, School of Physical Therapy, Faculty of Rehabilitation Science, Universidad Andrés Bello, Santiago, Chile
| | - Daniela Ebner-Karestinos
- Institute of Neuroscience, Université Catholique de Louvain, Ottignies-Louvain-la-Neuve, Belgium
- Exercise and Rehabilitation Science Institute, School of Physical Therapy, Faculty of Rehabilitation Science, Universidad Andrés Bello, Santiago, Chile
| | - Laurance Dricot
- Institute of Neuroscience, Université Catholique de Louvain, Ottignies-Louvain-la-Neuve, Belgium
| | - Enimie Herman
- Institute of Neuroscience, Université Catholique de Louvain, Ottignies-Louvain-la-Neuve, Belgium
| | - Samar M. Hatem
- Institute of Neuroscience, Université Catholique de Louvain, Ottignies-Louvain-la-Neuve, Belgium
- Physical and Rehabilitation Medicine, Brugmann University Hospital, Brussels, Belgium
- Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Brussels, Belgium
- Faculty of Physical Education and Physiotherapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kathleen M. Friel
- Burke-Cornell Medical Research Institute, White Plains, NY, United States
| | - Andrew M. Gordon
- Department of Biobehavioral Sciences, Teachers College, Columbia University, New York, NY, United States
| | - Yannick Bleyenheuft
- Institute of Neuroscience, Université Catholique de Louvain, Ottignies-Louvain-la-Neuve, Belgium
- *Correspondence: Yannick Bleyenheuft,
| |
Collapse
|
10
|
Therapeutic Interventions in Rat Models of Preterm Hypoxic Ischemic Injury: Effects of Hypothermia, Caffeine, and the Influence of Sex. Life (Basel) 2022; 12:life12101514. [PMID: 36294948 PMCID: PMC9605553 DOI: 10.3390/life12101514] [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: 08/18/2022] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 11/30/2022] Open
Abstract
Infants born prematurely have an increased risk of experiencing brain injury, specifically injury caused by Hypoxia Ischemia (HI). There is no approved treatment for preterm infants, in contrast to term infants that experience Hypoxic Ischemic Encephalopathy (HIE) and can be treated with hypothermia. Given this increased risk and lack of approved treatment, it is imperative to explore and model potential treatments in animal models of preterm injury. Hypothermia is one potential treatment, though cooling to current clinical standards has been found to be detrimental for preterm infants. However, mild hypothermia may prove useful. Caffeine is another treatment that is already used in preterm infants to treat apnea of prematurity, and has shown neuroprotective effects. Both of these treatments show sex differences in behavioral outcomes and neuroprotective effects, which are critical to explore when working to translate from animal to human. The effects and research history of hypothermia, caffeine and how sex affects these treatment outcomes will be explored further in this review article.
Collapse
|
11
|
Roumes H, Goudeneche P, Pellerin L, Bouzier-Sore AK. Resveratrol and Some of Its Derivatives as Promising Prophylactic Treatments for Neonatal Hypoxia-Ischemia. Nutrients 2022; 14:nu14183793. [PMID: 36145168 PMCID: PMC9501144 DOI: 10.3390/nu14183793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022] Open
Abstract
Due to the rate of occurrence of neonatal hypoxia-ischemia, its neuronal sequelae, and the lack of effective therapies, the development of new neuroprotective strategies is required. Polyphenols (including resveratrol) are molecules whose anti-apoptotic, anti-inflammatory, and anti-oxidative properties could be effective against the damage induced by neonatal hypoxia-ischemia. In this review article, very recent data concerning the neuroprotective role of polyphenols and the mechanisms at play are detailed, including a boost in brain energy metabolism. The results obtained with innovative approaches, such as maternal supplementation at nutritional doses, suggest that polyphenols could be a promising prophylactic treatment for neonatal hypoxia-ischemia.
Collapse
Affiliation(s)
- Hélène Roumes
- Centre de Résonance Magnétique des Sysytèmes Biologiques (CRMSB), UMR 5536, University of Bordeaux and CNRS, F-33000 Bordeaux, France
- Correspondence:
| | - Pierre Goudeneche
- Centre de Résonance Magnétique des Sysytèmes Biologiques (CRMSB), UMR 5536, University of Bordeaux and CNRS, F-33000 Bordeaux, France
| | - Luc Pellerin
- Ischémie Reperfusion, Métabolisme et Inflammation Stérile en Transplantation (IRMETIST), Inserm U1313, University of Poitiers and CHU Poitiers, F-86021 Poitiers, France
| | - Anne-Karine Bouzier-Sore
- Centre de Résonance Magnétique des Sysytèmes Biologiques (CRMSB), UMR 5536, University of Bordeaux and CNRS, F-33000 Bordeaux, France
| |
Collapse
|
12
|
Achterberg EJM, van Oldeniel RJ, van Tilborg E, Verharen JPH, Nijboer CH, Vanderschuren LJMJ. Cognitive performance during adulthood in a rat model of neonatal diffuse white matter injury. Psychopharmacology (Berl) 2022; 239:745-764. [PMID: 35064798 PMCID: PMC8891199 DOI: 10.1007/s00213-021-06053-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 12/27/2021] [Indexed: 11/25/2022]
Abstract
RATIONALE Infants born prematurely risk developing diffuse white matter injury (WMI), which is associated with impaired cognitive functioning and an increased risk of autism spectrum disorder. Recently, our rat model of preterm diffuse WMI induced by combined fetal inflammation and postnatal hypoxia showed impaired motor performance, anxiety-like behaviour and autism-like behaviour in juvenile rats, especially males. Immunohistochemistry showed delayed myelination in the sensory cortex and impaired oligodendrocyte differentiation. OBJECTIVE To assess long-term cognitive deficits in this double-hit rat model of diffuse WMI, animals were screened on impulsivity, attention and cognitive flexibility in adulthood using the 5-choice serial reaction time task (5CSRTT) and a probabilistic reversal learning task, tests that require a proper functioning prefrontal cortex. Thereafter, myelination deficits were evaluated by immunofluorescent staining in adulthood. RESULTS Overall, little effect of WMI or sex was found in the cognitive tasks. WMI animals showed subtle differences in performance in the 5CSRTT. Manipulating 5CSRTT parameters resulted in performance patterns previously seen in the literature. Sex differences were found in perseverative responses and omitted trials: female WMI rats seem to be less flexible in the 5CSRTT but not in the reversal learning task. Males collected rewards faster in the probabilistic reversal learning task. These findings are explained by temporally rather than permanently affected myelination and by the absence of extensive injury to prefrontal cortical subregions, confirmed by immunofluorescent staining in both adolescence and adulthood. CONCLUSION This rat model of preterm WMI does not lead to long-term cognitive deficits as observed in prematurely born human infants.
Collapse
Affiliation(s)
- E J Marijke Achterberg
- Department of Population Health Sciences, Unit Animals in Science and Society, Division of Behavioural Neuroscience, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 2, 3584CM, Utrecht, The Netherlands.
| | - Ralf J van Oldeniel
- Department of Population Health Sciences, Unit Animals in Science and Society, Division of Behavioural Neuroscience, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 2, 3584CM, Utrecht, The Netherlands
- Department for Developmental Origins of Disease, University Medical Center, Utrecht Brain Center, Wilhelmina Children's Hospital, Utrecht University, Lundlaan 6, 3584EA, Utrecht, The Netherlands
| | - Erik van Tilborg
- Department for Developmental Origins of Disease, University Medical Center, Utrecht Brain Center, Wilhelmina Children's Hospital, Utrecht University, Lundlaan 6, 3584EA, Utrecht, The Netherlands
| | - Jeroen P H Verharen
- Helen Wills Neuroscience Institute, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
| | - Cora H Nijboer
- Department for Developmental Origins of Disease, University Medical Center, Utrecht Brain Center, Wilhelmina Children's Hospital, Utrecht University, Lundlaan 6, 3584EA, Utrecht, The Netherlands
| | - Louk J M J Vanderschuren
- Department of Population Health Sciences, Unit Animals in Science and Society, Division of Behavioural Neuroscience, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 2, 3584CM, Utrecht, The Netherlands
| |
Collapse
|
13
|
Durán-Carabali LE, Odorcyk FK, Sanches EF, de Mattos MM, Anschau F, Netto CA. Effect of environmental enrichment on behavioral and morphological outcomes following neonatal hypoxia-ischemia in rodent models: A systematic review and meta-analysis. Mol Neurobiol 2022; 59:1970-1991. [PMID: 35040041 DOI: 10.1007/s12035-022-02730-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/02/2022] [Indexed: 02/06/2023]
Abstract
Neonatal hypoxia-ischemia (HI) is a major cause of mortality and morbidity in newborns and, despite recent advances in neonatal intensive care, there is no definitive treatment for this pathology. Once preclinical studies have shown that environmental enrichment (EE) seems to be a promising therapy for children with HI, the present study conducts a systematic review and meta-analysis of articles with EE in HI rodent models focusing on neurodevelopmental reflexes, motor and cognitive function as well as brain damage. The protocol was registered a priori at PROSPERO. The search was conducted in PubMed, Embase and PsycINFO databases, resulting in the inclusion of 22 articles. Interestingly, EE showed a beneficial impact on neurodevelopmental reflexes (SMD= -0.73, CI= [-0.98; -0.47], p< 0.001, I2= 0.0%), motor function (SMD= -0.55, CI= [-0.81; -0.28], p< 0.001, I2= 62.6%), cognitive function (SMD= -0.93, CI= [-1.14; -0.72], p< 0.001, I2= 27.8%) and brain damage (SMD= -0.80, CI= [-1.03; -0.58], p< 0.001, I2= 10.7%). The main factors that potentiate EE positive effects were enhanced study quality, earlier age at injury as well as earlier start and longer duration of EE exposure. Overall, EE was able to counteract the behavioral and histological damage induced by the lesion, being a promising therapeutic strategy for HI.
Collapse
Affiliation(s)
- L E Durán-Carabali
- Graduate Program in Biological Sciences: Physiology, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| | - F K Odorcyk
- Graduate Program in Biological Sciences: Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - E F Sanches
- Division of Child Development and Growth, Department of Pediatrics, Gynecology and Obstetrics, School of Medicine, University of Geneva, Geneva, Switzerland
| | - M M de Mattos
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600, anexo, Porto Alegre, RS, CEP 90035-003, Brazil
| | - F Anschau
- Medicine school, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.,Graduation Program on Evaluation and Production of Technologies for the Brazilian National Health System, Porto Alegre, Brazil
| | - C A Netto
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600, anexo, Porto Alegre, RS, CEP 90035-003, Brazil. .,Department of Physiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
14
|
Zen R, Terashima T, Tsuji S, Katagi M, Ohashi N, Nobuta Y, Higuchi A, Kanai H, Murakami T, Kojima H. Ambient Temperature Is Correlated With the Severity of Neonatal Hypoxic-Ischemic Brain Injury via Microglial Accumulation in Mice. Front Pediatr 2022; 10:883556. [PMID: 35601427 PMCID: PMC9120824 DOI: 10.3389/fped.2022.883556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/14/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The pathophysiology of neonatal hypoxic-ischemic encephalopathy (HIE) has been studied in several rodent models to develop novel treatments. Although it is well known that high ambient temperature results in severe HIE, the effect of subtle changes in ambient temperature during a hypoxic-ischemic (HI) insult has not been studied. Therefore, in order to clarify the difference of pathophysiological change among the HIE models due to the influence of small changes in chamber temperature, three-step gradual change of 0.5°C each were prepared in ambient temperature during hypoxic exposure. METHODS Blood flow in the left common carotid artery (CCA) of neonatal mice was interrupted using bipolar electronic forceps under general and local anesthesia. The mice were subsequently subjected to 10% hypoxic exposure for 50 min at 36.0, 36.5, or 37.0°C. A control group was also included in the study. The size of the striatum and hippocampus and the volume reduction rate of the hemisphere in the section containing them on the ischemic side were evaluated using microtubule associated protein 2 (MAP2) immunostaining. The accumulation of Iba1-positive cells was investigated to assess inflammation. Additionally, rotarod and open-field tests were performed 2 weeks after HI insult to assess its effect on physiological conditions. RESULTS MAP2 staining revealed that the higher the temperature during hypoxia, the more severe the volume reduction rate in the hemisphere, striatum, and hippocampus. The number of Iba1-positive cells in the ipsilateral lesion gradually increased with increasing temperature, and there was a significant difference in motor function in the 36.5 and 37.0°C groups compared with the sham group. In the open-field tests, there was a significant decrease in performance in the 37.0°C groups compared with the 36.0°C and sham groups. CONCLUSIONS Even a small gradual change of 0.5°C produced a significant difference in pathological and behavioral changes and contributed to the accumulation of Iba1-positive cells. The arrangement of ambient temperature is useful for creating a rodent model with the appropriate severity of the targeted neuropsychological symptoms to establish a novel therapy for HIE.
Collapse
Affiliation(s)
- Rika Zen
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Otsu, Japan.,Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Tomoya Terashima
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Shunichiro Tsuji
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Miwako Katagi
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Natsuko Ohashi
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Yuri Nobuta
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Asuka Higuchi
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Hirohiko Kanai
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Takashi Murakami
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, Otsu, Japan
| | - Hideto Kojima
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
15
|
Shariati M, Esfahani RJ, Bidkhori HR, Sabouri E, Mehrzad S, Sadr-Nabavi A. Cell-based treatment of cerebral palsy: still a long way ahead. Curr Stem Cell Res Ther 2021; 17:741-749. [PMID: 34727864 DOI: 10.2174/1574888x16666211102090230] [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: 04/21/2021] [Revised: 07/28/2021] [Accepted: 08/19/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cerebral palsy (CP) is a permanent neurodevelopmental disorder with considerable global disability. Various rehabilitation strategies are currently available. However, none represents a convincing curative result. Cellular therapy recently holds much promise as an alternative strategy to repair neurologic defects. METHOD In this narrative review, a comprehensive search of the MEDLINE and ClinicalTrials.gov was made, using the terms: "cell therapy" and "cerebral palsy", including published and registered clinical studies, respectively. RESULTS The early effects of these studies demonstrated that using cell therapy in CP patients is safe and improves the deficits for a variable duration. Despite such hopeful early bird results, the long-term outcomes are not conclusive. CONCLUSIONS Due to the heterogeneous nature of CP, personal factors seem essential to consider. Cell dosage, routes of administration, and repeated dosing are pivotal to establish optimal personalized treatments. Future clinical trials should consider employing other cell types, specific cell modifications before administration, and cell-free platforms.
Collapse
Affiliation(s)
- Mohammad Shariati
- Stem Cells and Regenerative Medicine Research Department, Academic Center for Education, Culture, and Research (ACECR)-Khorasan Razavi, Mashhad. Iran
| | - Reza Jafarzadeh Esfahani
- Blood Borne Infections Research Center, Academic Center for Education, Culture and Research (ACECR)- Khorasan Razavi, Mashhad. Iran
| | - Hamid Reza Bidkhori
- Stem Cells and Regenerative Medicine Research Department, Academic Center for Education, Culture, and Research (ACECR)-Khorasan Razavi, Mashhad. Iran
| | - Ehsan Sabouri
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad. Iran
| | - Shadi Mehrzad
- Stem Cells and Regenerative Medicine Research Department, Academic Center for Education, Culture, and Research (ACECR)-Khorasan Razavi, Mashhad. Iran
| | - Ariane Sadr-Nabavi
- Department of Neurology, School of Medicine, Mashhad University of Medical Sciences, Mashhad. Iran
| |
Collapse
|
16
|
Dose-Dependent Neuroprotective Effects of Bovine Lactoferrin Following Neonatal Hypoxia-Ischemia in the Immature Rat Brain. Nutrients 2021; 13:nu13113880. [PMID: 34836132 PMCID: PMC8618330 DOI: 10.3390/nu13113880] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 01/07/2023] Open
Abstract
Injuries to the developing brain due to hypoxia–ischemia (HI) are common causes of neurological disabilities in preterm babies. HI, with oxygen deprivation to the brain or reduced cerebral blood perfusion due to birth asphyxia, often leads to severe brain damage and sequelae. Injury mechanisms include glutamate excitotoxicity, oxidative stress, blood–brain barrier dysfunction, and exacerbated inflammation. Nutritional intervention is emerging as a therapeutic alternative to prevent and rescue brain from HI injury. Lactoferrin (Lf) is an iron-binding protein present in saliva, tears, and breast milk, which has been shown to have antioxidant, anti-inflammatory and anti-apoptotic properties when administered to mothers as a dietary supplement during pregnancy and/or lactation in preclinical studies of developmental brain injuries. However, despite Lf’s promising neuroprotective effects, there is no established dose. Here, we tested three different doses of dietary maternal Lf supplementation using the postnatal day 3 HI model and evaluated the acute neurochemical damage profile using 1H Magnetic Resonance Spectroscopy (MRS) and long-term microstructure alterations using advanced diffusion imaging (DTI/NODDI) allied to protein expression and histological analysis. Pregnant Wistar rats were fed either control diet or bovine Lf supplemented chow at 0.1, 1, or 10 g/kg/body weight concentration from the last day of pregnancy (embryonic day 21–E21) to weaning. At postnatal day 3 (P3), pups from both sexes had their right common carotid artery permanently occluded and were exposed to 6% oxygen for 30 min. Sham rats had the incision but neither surgery nor hypoxia episode. At P4, MRS was performed on a 9.4 T scanner to obtain the neurochemical profile in the cortex. At P4 and P25, histological analysis and protein expression were assessed in the cortex and hippocampus. Brain volumes and ex vivo microstructural analysis using DTI/NODDI parameters were performed at P25. Acute metabolic disturbance induced in cortical tissue by HIP3 was reversed with all three doses of Lf. However, data obtained from MRS show that Lf neuroprotective effects were modulated by the dose. Through western blotting analysis, we observed that HI pups supplemented with Lf at 0.1 and 1 g/kg were able to counteract glutamatergic excitotoxicity and prevent metabolic failure. When 10 g/kg was administered, we observed reduced brain volumes, increased astrogliosis, and hypomyelination, pointing to detrimental effects of high Lf dose. In conclusion, Lf supplementation attenuates, in a dose-dependent manner, the acute and long-term cerebral injury caused by HI. Lf reached its optimal effects at a dose of 1 g/kg, which pinpoints the need to better understand effects of Lf, the pathways involved and possible harmful effects. These new data reinforce our knowledge regarding neuroprotection in developmental brain injury using Lf through lactation and provide new insights into lactoferrin’s neuroprotection capacities and limitation for immature brains.
Collapse
|
17
|
Niu J, Yu G, Wang X, Xia W, Wang Y, Hoi KK, Mei F, Xiao L, Chan JR, Fancy SPJ. Oligodendroglial ring finger protein Rnf43 is an essential injury-specific regulator of oligodendrocyte maturation. Neuron 2021; 109:3104-3118.e6. [PMID: 34390652 DOI: 10.1016/j.neuron.2021.07.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 04/06/2021] [Accepted: 07/21/2021] [Indexed: 12/31/2022]
Abstract
Oligodendrocyte (OL) maturation arrest in human white matter injury contributes significantly to the failure of endogenous remyelination in multiple sclerosis (MS) and newborn brain injuries such as hypoxic ischemic encephalopathy (HIE) that cause cerebral palsy. In this study, we identify an oligodendroglial-intrinsic factor that controls OL maturation specifically in the setting of injury. We find a requirement for the ring finger protein Rnf43 not in normal development but in neonatal hypoxic injury and remyelination in the adult mammalian CNS. Rnf43, but not the related Znrf3, is potently activated by Wnt signaling in OL progenitor cells (OPCs) and marks activated OPCs in human MS and HIE. Rnf43 is required in an injury-specific context, and it promotes OPC differentiation through negative regulation of Wnt signal strength in OPCs at the level of Fzd1 receptor presentation on the cell surface. Inhibition of Fzd1 using UM206 promotes remyelination following ex vivo and in vivo demyelinating injury.
Collapse
Affiliation(s)
- Jianqin Niu
- Department of Neurology, University of California at San Francisco, San Francisco, CA 94158, USA; Department of Pediatrics, University of California at San Francisco, San Francisco, CA 94158, USA; Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China; Institute of Brain and Intelligence, Third Military Medical University, Chongqing 400038, China.
| | - Guangdan Yu
- Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China
| | - Xiaorui Wang
- Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China
| | - Wenlong Xia
- Department of Neurology, University of California at San Francisco, San Francisco, CA 94158, USA
| | - Yuxin Wang
- Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China
| | - Kimberly K Hoi
- Department of Neurology, University of California at San Francisco, San Francisco, CA 94158, USA
| | - Feng Mei
- Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China; Institute of Brain and Intelligence, Third Military Medical University, Chongqing 400038, China
| | - Lan Xiao
- Department of Histology and Embryology, Third Military Medical University, Chongqing 400038, China; Institute of Brain and Intelligence, Third Military Medical University, Chongqing 400038, China
| | - Jonah R Chan
- Department of Neurology, University of California at San Francisco, San Francisco, CA 94158, USA; Division of Neuroimmunology and Glial Biology, University of California at San Francisco, San Francisco, CA 94158, USA
| | - Stephen P J Fancy
- Department of Neurology, University of California at San Francisco, San Francisco, CA 94158, USA; Department of Pediatrics, University of California at San Francisco, San Francisco, CA 94158, USA; Division of Neuroimmunology and Glial Biology, University of California at San Francisco, San Francisco, CA 94158, USA; Newborn Brain Research Institute, University of California at San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
18
|
Chang J, Lurie RH, Sharma A, Bashir M, Fung CM, Dettman RW, Dizon MLV. Intrauterine growth restriction followed by oxygen support uniquely interferes with genetic regulators of myelination. eNeuro 2021; 8:ENEURO.0263-20.2021. [PMID: 34099489 PMCID: PMC8266217 DOI: 10.1523/eneuro.0263-20.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 03/23/2021] [Accepted: 04/23/2021] [Indexed: 12/24/2022] Open
Abstract
Intrauterine growth restriction (IUGR) and oxygen exposure in isolation and combination adversely affect the developing brain, putting infants at risk for neurodevelopmental disability including cerebral palsy. Rodent models of IUGR and postnatal hyperoxia have demonstrated oligodendroglial injury with subsequent white matter injury (WMI) and motor dysfunction. Here we investigate transcriptomic dysregulation in IUGR with and without hyperoxia exposure to account for the abnormal brain structure and function previously documented. We performed RNA sequencing and analysis using a mouse model of IUGR and found that IUGR, hyperoxia, and the combination of IUGR with hyperoxia (IUGR/hyperoxia) produced distinct changes in gene expression. IUGR in isolation demonstrated the fewest differentially expressed genes compared to control. In contrast, we detected several gene alterations in IUGR/hyperoxia; genes involved in myelination were strikingly downregulated. We also identified changes to specific regulators including TCF7L2, BDNF, SOX2, and DGCR8, through Ingenuity Pathway Analysis, that may contribute to impaired myelination in IUGR/hyperoxia. Our findings show that IUGR with hyperoxia induces unique transcriptional changes in the developing brain. These indicate mechanisms for increased risk for WMI in IUGR infants exposed to oxygen and suggest potential therapeutic targets to improve motor outcomes.Significance StatementThis study demonstrates that perinatal exposures of IUGR and/or postnatal hyperoxia result in distinct transcriptomic changes in the developing brain. In particular, we found that genes involved in normal developmental myelination, myelin maintenance, and remyelination were most dysregulated when IUGR was combined with hyperoxia. Understanding how multiple risk factors lead to WMI is the first step in developing future therapeutic interventions. Additionally, because oxygen exposure is often unavoidable after birth, an understanding of gene perturbations in this setting will increase our awareness of the need for tight control of oxygen use to minimize future motor disability.
Collapse
Affiliation(s)
- Jill Chang
- Children's Hospital of Chicago, Department of Pediatrics, Division of Neonatology, Chicago, Illinois, USA
| | - Robert H Lurie
- Children's Hospital of Chicago, Department of Pediatrics, Division of Neonatology, Chicago, Illinois, USA
| | - Abhineet Sharma
- Children's Hospital of Chicago, Department of Pediatrics, Division of Neonatology, Chicago, Illinois, USA
| | - Mirrah Bashir
- Children's Hospital of Chicago, Department of Pediatrics, Division of Neonatology, Chicago, Illinois, USA
| | - Camille M Fung
- University of Utah, Department of Pediatrics, Salt Lake City, Utah, USA
| | - Robert W Dettman
- Children's Hospital of Chicago, Department of Pediatrics, Division of Neonatology, Chicago, Illinois, USA
| | - Maria L V Dizon
- Children's Hospital of Chicago, Department of Pediatrics, Division of Neonatology, Chicago, Illinois, USA
| |
Collapse
|
19
|
A Brief Review on Erythropoietin and Mesenchymal Stem Cell Therapies for Paediatric Neurological Disorders. CURRENT STEM CELL REPORTS 2021. [DOI: 10.1007/s40778-021-00189-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
20
|
Berlin C, Lange K, Lekaye HC, Hopland K, Phillips S, Piao J, Tabar V. Long-term clinically relevant rodent model of methotrexate-induced cognitive impairment. Neuro Oncol 2021; 22:1126-1137. [PMID: 32242229 DOI: 10.1093/neuonc/noaa086] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND With the enhanced use of chemotherapy and the advent of increased patient survival rates, there are an increasing number of cancer survivors living with chemotherapy-induced cognitive impairment. A growing number of clinical studies have brought to light the association of agents like methotrexate in generating these neurological sequelae, although mechanisms remain unclear. METHODS Here, we use a clinically relevant regimen of several cycles of methotrexate and leucovorin rescue to develop a model of chemotherapy-induced cognitive impairment, and investigate the in vivo long-term (16 mo) impact of high-dose systemic methotrexate on white matter cellular dynamics as assessed by stereology, animal behavior, and diffusion tensor imaging. RESULTS Our results indicate that at 6 and 16 months post-chemotherapy, methotrexate-treated rats exhibit a significant and permanent decrease in the number of oligodendrocytes and their progenitors in the white matter, in corpus callosum volumes, and myelin basic protein. These findings are associated with mostly delayed deficits in performance on Morris Water Maze and Novel Object Recognition tasks. Diffusion tensor imaging demonstrates significantly decreased fractional anisotropy values in the callosum genu, body, and splenium, as well as previously unassessed areas like the fimbria. Interestingly, these white matter changes are preceded by an earlier, transient decrement in white matter microglia at 3 months, and hippocampal neural progenitors at 3 and 6 months. CONCLUSION These results demonstrate a significant negative impact of methotrexate on the oligodendrocyte compartment and white matter, associated with cognitive impairment. The data also support the use of diffusion tensor imaging in monitoring white matter integrity in this context.
Collapse
Affiliation(s)
- Connor Berlin
- Downstate Medical Center, College of Medicine, State University of New York, Brooklyn, New York
| | - Katharine Lange
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - H Carl Lekaye
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kelsey Hopland
- Neuroscience Graduate Program, Weill Cornell Medicine, New York, New York
| | - Samantha Phillips
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, New York
| | - Jinghua Piao
- Department of Neurosurgery and Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Viviane Tabar
- Department of Neurosurgery and Center for Stem Cell Biology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
21
|
Shen Z, Lin Y, Yang J, Jörg DJ, Peng Y, Zhang X, Xu Y, Hernandez L, Ma J, Simons BD, Shi SH. Distinct progenitor behavior underlying neocortical gliogenesis related to tumorigenesis. Cell Rep 2021; 34:108853. [PMID: 33730566 DOI: 10.1016/j.celrep.2021.108853] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 01/20/2021] [Accepted: 02/19/2021] [Indexed: 12/21/2022] Open
Abstract
Radial glial progenitors (RGPs) give rise to the vast majority of neurons and glia in the neocortex. Although RGP behavior and progressive generation of neocortical neurons have been delineated, the exact process of neocortical gliogenesis remains elusive. Here, we report the precise progenitor behavior and gliogenesis program at single-cell resolution in the mouse neocortex. Fractions of dorsal RGPs transition from neurogenesis to gliogenesis progressively, producing astrocytes, oligodendrocytes, or both in well-defined propensities of ∼60%, 15%, and 25%, respectively, by fate-restricted "intermediate" precursor cells (IPCs). Although the total number of IPCs generated by individual RGPs appears stochastic, the output of individual IPCs exhibit clear patterns in number and subtype and form discrete local subclusters. Clonal loss of tumor suppressor Neurofibromatosis type 1 leads to excessive production of glia selectively, especially oligodendrocyte precursor cells. These results quantitatively delineate the cellular program of neocortical gliogenesis and suggest the cellular and lineage origin of primary brain tumor.
Collapse
Affiliation(s)
- Zhongfu Shen
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center of Biological Structure, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yang Lin
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center of Biological Structure, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jiajun Yang
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center of Biological Structure, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - David J Jörg
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK
| | - Yuwei Peng
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center of Biological Structure, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiuli Zhang
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center of Biological Structure, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yifan Xu
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center of Biological Structure, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Luisirene Hernandez
- Neuroscience Graduate Program, Feil Family Brain & Mind Research Institute, Weill Cornell Medical College, New York, NY 10065, USA
| | - Jian Ma
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center of Biological Structure, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Benjamin D Simons
- The Wellcome Trust/Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK; Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, Wilberforce Road, Cambridge CB3 0WA, UK.
| | - Song-Hai Shi
- IDG/McGovern Institute for Brain Research, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center of Biological Structure, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Neuroscience Graduate Program, Feil Family Brain & Mind Research Institute, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
22
|
Therapeutic potential of stem cells for preterm infant brain damage: Can we move from the heterogeneity of preclinical and clinical studies to established therapeutics? Biochem Pharmacol 2021; 186:114461. [PMID: 33571501 DOI: 10.1016/j.bcp.2021.114461] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/27/2021] [Accepted: 02/02/2021] [Indexed: 12/17/2022]
Abstract
Acquired perinatal brain injuries are a set of conditions that remains a key challenge for neonatologists and that have significant social, emotional and financial implications for our communities. In our perspective article, we will introduce perinatal brain injury focusing specifically on the events leading to brain damage in preterm born infants and outcomes for these infants. Then we will summarize and discuss the preclinical and clinical studies testing the efficacy of stem cells as neuroprotectants in the last ten years in perinatal brain injury. There are no therapies to treat brain damage in preterm born infants and a primary finding from this review is that there is a scarcity of stem cell trials focused on overcoming brain injuries in these infants. Overall, across all forms of perinatal brain injury there is a remarkable heterogeneity in previous and on-going preclinical and clinical studies in terms of the stem cell type, animal models/patient selection, route and time of administration. Despite the quality of many of the studies this variation makes it difficult to reach a valid consensus for future developments. However, it is clear that stem cells (and stem cell derived exosomes) can reduce perinatal brain injury and our field needs to work collectively to refine an effective protocol for each type of injury. The use of standardized stem cell products and testing these products across multiple models of injury will provide a stronger framework for clinical trials development.
Collapse
|
23
|
Xu H, Zhang L, Xuan XY, Zhu M, Tang J, Zhao XK. Intrauterine cytomegalovirus infection: a possible risk for cerebral palsy and related to its clinical features, neuroimaging findings: a retrospective study. BMC Pediatr 2020; 20:555. [PMID: 33292171 PMCID: PMC7722436 DOI: 10.1186/s12887-020-02449-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 12/01/2020] [Indexed: 11/10/2022] Open
Abstract
Background Abundant clinical evidences indicate that the increased risk of cerebral palsy (CP) may be associated with the intrauterine exposure to maternal infection. Cytomegalovirus (CMV) is a common cause of CP. However, little is known about the relationship between the intrauterine exposure of the fetus to CMV infection and CP. This study aims to explore the relationships between intrauterine CMV infection and clinical symptoms, classification, intelligence development and brain neuroimaging findings in children with CP. Methods In this study, 147 children with CP in recent 6 years were retrospectively analyzed (average age: 14.76 ± 3.07months; sex (M/F): 103/44). 148 children had CMV IgG and IgM positive sera identified by TORCH examination were selected as the control group (average age: 15.10 ± 3.21months; sex (M/F): 102/46), which also undergo the examination of CMV-DNA in urine. The age and sex of children in the control group were matched with those in the CP group. CMV-DNA in urine was detected by CMV fluorescence quantitative PCR, and t-test was performed to analyze the number of copies. For the CP group, standardized rehabilitation treatment was performed and the function of gross motor was evaluated by GMFM scale before and after treatment. The Gesell developmental scale (GDS) was used to assess the level of intellectual development. The classification of CP was conducted and the results of magnetic resonance imaging were analyzed. Finally, the correlations between the copy number of CMV-DNA and the clinical characteristics of children with CP were evaluated by the method of Pearson and Spearman correlation analysis. Results The level of CMV infection was negatively correlated with the developmental quotient (DQ) of children with CP. Negative association was found between the level of CMV infection and the level of the gross motor development. The level of CMV infection was positively related with the occurrence probability of spastic quadriplegia. However, no associations were found between the abnormalities of brain tissue and the number of CMV copies. Moreover, CMV infection might add the difficulty of the rehabilitation treatment. Conclusions CMV infection is a risk factor for the occurrence of CP in children. Pregnancy examination should be strengthened. Early detection and control of CMV infection may contribute to the rehabilitation of children with CP and reduce the disability and social burden.
Collapse
Affiliation(s)
- H Xu
- Department of Rehabilitation, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - L Zhang
- Department of Rehabilitation, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - X Y Xuan
- Department of Rehabilitation, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - M Zhu
- Department of Rehabilitation, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - J Tang
- Department of Rehabilitation, Children's Hospital of Nanjing Medical University, Nanjing, China.
| | - X K Zhao
- Department of Rehabilitation, Children's Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
24
|
Goulding DS, Vogel RC, Gensel JC, Morganti JM, Stromberg AJ, Miller BA. Acute brain inflammation, white matter oxidative stress, and myelin deficiency in a model of neonatal intraventricular hemorrhage. J Neurosurg Pediatr 2020; 26:613-623. [PMID: 32858507 PMCID: PMC10193502 DOI: 10.3171/2020.5.peds20124] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/18/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Neonatal intraventricular hemorrhage (IVH) leads to posthemorrhagic hydrocephalus (PHH), brain injury, and long-term disability. Current therapy for IVH is based on treating PHH but does not address the underlying brain injury. In order to develop pharmacological treatment for IVH, there must be a better understanding of the underlying pathology of this disease. This study was designed to determine the time course of the acute inflammation and oxidative stress that may underlie the progressive pathology of IVH. The authors sought to understand the temporal relationships among inflammation, oxidative stress, and white matter pathology in a rat model of IVH. METHODS A rat model of IVH consisting of hemoglobin injection into the lateral ventricle was used. Tissue was analyzed via biochemical and histological methods to map the spatiotemporal distribution of innate immune activation and oxidative stress. White matter was quantified using both immunohistochemistry and Western blot for myelin basic protein (MBP) in the corpus callosum. RESULTS IVH led to acute induction of inflammatory cytokines, followed by oxidative stress. Oxidative stress was concentrated in white matter, adjacent to the lateral ventricles. Animals with IVH initially gained weight at a lower rate than control animals and had larger ventricles and less MBP than control animals. CONCLUSIONS Experimental IVH induces global inflammation throughout the brain and oxidative stress concentrated in the white matter. Both of these phenomena occur early after IVH. This has implications for human neonates with immature white matter that is exquisitely sensitive to inflammation and oxidative stress. Antiinflammatory or antioxidant therapy for IVH may need to be initiated early in order to protect developing white matter.
Collapse
Affiliation(s)
- Danielle S. Goulding
- Departments of Neurosurgery
- Spinal Cord and Brain Injury Research Center, University of Kentucky; and
| | - R. Caleb Vogel
- Departments of Neurosurgery
- Spinal Cord and Brain Injury Research Center, University of Kentucky; and
| | - John C. Gensel
- Physiology
- Spinal Cord and Brain Injury Research Center, University of Kentucky; and
| | - Josh M. Morganti
- Neuroscience, and
- Spinal Cord and Brain Injury Research Center, University of Kentucky; and
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky
| | | | - Brandon A. Miller
- Departments of Neurosurgery
- Neuroscience, and
- Spinal Cord and Brain Injury Research Center, University of Kentucky; and
| |
Collapse
|
25
|
Ross-Munro E, Kwa F, Kreiner J, Khore M, Miller SL, Tolcos M, Fleiss B, Walker DW. Midkine: The Who, What, Where, and When of a Promising Neurotrophic Therapy for Perinatal Brain Injury. Front Neurol 2020; 11:568814. [PMID: 33193008 PMCID: PMC7642484 DOI: 10.3389/fneur.2020.568814] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/18/2020] [Indexed: 12/21/2022] Open
Abstract
Midkine (MK) is a small secreted heparin-binding protein highly expressed during embryonic/fetal development which, through interactions with multiple cell surface receptors promotes growth through effects on cell proliferation, migration, and differentiation. MK is upregulated in the adult central nervous system (CNS) after multiple types of experimental injury and has neuroprotective and neuroregenerative properties. The potential for MK as a therapy for developmental brain injury is largely unknown. This review discusses what is known of MK's expression and actions in the developing brain, areas for future research, and the potential for using MK as a therapeutic agent to ameliorate the effects of brain damage caused by insults such as birth-related hypoxia and inflammation.
Collapse
Affiliation(s)
- Emily Ross-Munro
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia
| | - Faith Kwa
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia.,School of Health Sciences, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Jenny Kreiner
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia
| | - Madhavi Khore
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Monash University, Clayton, VIC, Australia
| | - Mary Tolcos
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia
| | - Bobbi Fleiss
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia.,Neurodiderot, Inserm U1141, Universita de Paris, Paris, France
| | - David W Walker
- Neurodevelopment in Health and Disease Research Program, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology (RMIT), Melbourne, VIC, Australia
| |
Collapse
|
26
|
Jung DH, Pak ME, Lee HJ, Ahn SM, Yun YJ, Shin YI, Shin HK, Lee SY, Choi BT. Electroacupuncture on the Scalp over the Motor Cortex Ameliorates Behavioral Deficits Following Neonatal Hypoxia-Ischemia in Rats via the Activation of Neural Stem Cells. Life (Basel) 2020; 10:life10100240. [PMID: 33066563 PMCID: PMC7602251 DOI: 10.3390/life10100240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/04/2020] [Accepted: 10/12/2020] [Indexed: 11/16/2022] Open
Abstract
Electroacupuncture (EA) therapy via alternating current stimulation on the scalp over the motor cortex is used for the treatment of brain disorders. Perinatal hypoxia-ischemia (HI), a brain injury in newborns, leads to long-term neurologic complications. Here, we investigated whether EA could promote functional improvements and neurogenesis in a neonatal HI rat model. A neonatal HI rat model was induced by permanent ligation of the left carotid artery in postnatal day 7 pups. EA for neonatal HI rats was performed at 2 Hz (1, 3, or 5 mA; 20 min) from 4–6 weeks after birth. HI rats undergoing EA had improved motor and memory function, with the greatest improvement after 3 mA EA. The corpus callosum was significantly thicker and showed a significant increase in proliferating astrocytes in the 3 mA EA group. We observed proliferating cells and a greater number of newly developed neurons and astrocytes in the subventricular zone and dentate gyrus of the 3 mA EA group than in those of the HI group. These results suggest that EA promotes functional improvements following neonatal HI assault via the proliferation and differentiation of neural stem cells. This effect was the strongest after 3 mA EA, suggesting that this is the optimal treatment dose.
Collapse
Affiliation(s)
- Da Hee Jung
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Gyeongnam, Korea; (D.H.J.); (M.E.P.); (H.J.L.); (H.K.S.)
- Graduate Training Program of Korean Medicine for Healthy-Aging, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Korea
| | - Malk Eun Pak
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Gyeongnam, Korea; (D.H.J.); (M.E.P.); (H.J.L.); (H.K.S.)
- Graduate Training Program of Korean Medicine for Healthy-Aging, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Korea
| | - Hong Ju Lee
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Gyeongnam, Korea; (D.H.J.); (M.E.P.); (H.J.L.); (H.K.S.)
- Graduate Training Program of Korean Medicine for Healthy-Aging, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Korea
| | - Sung Min Ahn
- Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan 50612, Gyeongnam, Korea;
| | - Young Ju Yun
- Department of Integrative Medicine, School of Korean Medicine, Pusan National University, Yangsan 50612, Gyeongnam, Korea;
| | - Yong-Il Shin
- Department of Rehabilitation Medicine, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University, Yangsan 50612, Gyeongnam, Korea;
| | - Hwa Kyoung Shin
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Gyeongnam, Korea; (D.H.J.); (M.E.P.); (H.J.L.); (H.K.S.)
- Graduate Training Program of Korean Medicine for Healthy-Aging, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Korea
- Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan 50612, Gyeongnam, Korea;
| | - Seo-Yeon Lee
- Department of Pharmacology, Wonkwang University School of Medicine, Iksan 54538, Jeonbuk, Korea
- Correspondence: (S.-Y.L.); (B.T.C.)
| | - Byung Tae Choi
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Gyeongnam, Korea; (D.H.J.); (M.E.P.); (H.J.L.); (H.K.S.)
- Graduate Training Program of Korean Medicine for Healthy-Aging, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam 50612, Korea
- Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan 50612, Gyeongnam, Korea;
- Correspondence: (S.-Y.L.); (B.T.C.)
| |
Collapse
|
27
|
Carloni S, Crinelli R, Palma L, Álvarez FJ, Piomelli D, Duranti A, Balduini W, Alonso-Alconada D. The Synthetic Cannabinoid URB447 Reduces Brain Injury and the Associated White Matter Demyelination after Hypoxia-Ischemia in Neonatal Rats. ACS Chem Neurosci 2020; 11:1291-1299. [PMID: 32271539 PMCID: PMC7997380 DOI: 10.1021/acschemneuro.0c00047] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
![]()
The number of functions controlled
by the endocannabinoid system
in health and disease continues growing over the years. In the brain,
these include the modulation of harmful events such as glutamate excitotoxicity,
oxidative stress, and inflammation, mainly regulated by activation/blockade
of CB1/CB2 cannabinoid receptors. In the present
work, we evaluated the capacity of the CB1 antagonist/CB2 agonist synthetic cannabinoid URB447 on reducing neurodegeneration
after brain injury. By using a model of hypoxia-ischemia (HI) in neonatal
rats, we found that URB447 strongly reduced brain injury when administered
before HI. A comparable effect was observed with the CB1 antagonist SR141716A, whereas the CB1 agonist WIN-55,212-2
reduced the effect of URB447. When administered 3 h after HI, which
is considered a clinically feasible therapeutic window to treat perinatal
brain injury in humans, URB447 reduced neurodegeneration and white
matter damage. Markers of astrogliosis and microglial activation also
appeared reduced. These results confirm the important role played
by the endocannabinoid system in the neurodegenerative process and
strongly encourage further research into the mechanisms of URB447-induced
neuroprotection.
Collapse
Affiliation(s)
- Silvia Carloni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Rita Crinelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Linda Palma
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Francisco J. Álvarez
- Biocruces Bizkaia Health Research Institute, Cruces University Hospital, Barakaldo, Bizkaia, Spain
| | - Daniele Piomelli
- Departments of Anatomy and Neurobiology, Pharmaceutical Sciences, and Biological Chemistry, University of California, Irvine, Irvine, California, United States
| | - Andrea Duranti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Walter Balduini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Daniel Alonso-Alconada
- Department of Cell Biology and Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| |
Collapse
|
28
|
Zhong X, Harris G, Smirnova L, Zufferey V, Sá RDCDSE, Baldino Russo F, Baleeiro Beltrao Braga PC, Chesnut M, Zurich MG, Hogberg HT, Hartung T, Pamies D. Antidepressant Paroxetine Exerts Developmental Neurotoxicity in an iPSC-Derived 3D Human Brain Model. Front Cell Neurosci 2020; 14:25. [PMID: 32153365 PMCID: PMC7047331 DOI: 10.3389/fncel.2020.00025] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 01/28/2020] [Indexed: 02/04/2023] Open
Abstract
Selective serotonin reuptake inhibitors (SSRIs) are frequently used to treat depression during pregnancy. Various concerns have been raised about the possible effects of these drugs on fetal development. Current developmental neurotoxicity (DNT) testing conducted in rodents is expensive, time-consuming, and does not necessarily represent human pathophysiology. A human, in vitro testing battery to cover key events of brain development, could potentially overcome these challenges. In this study, we assess the DNT of paroxetine—a widely used SSRI which has shown contradictory evidence regarding effects on human brain development using a versatile, organotypic human induced pluripotent stem cell (iPSC)-derived brain model (BrainSpheres). At therapeutic blood concentrations, which lie between 20 and 60 ng/ml, Paroxetine led to an 80% decrease in the expression of synaptic markers, a 60% decrease in neurite outgrowth and a 40–75% decrease in the overall oligodendrocyte cell population, compared to controls. These results were consistently shown in two different iPSC lines and indicate that relevant therapeutic concentrations of Paroxetine induce brain cell development abnormalities which could lead to adverse effects.
Collapse
Affiliation(s)
- Xiali Zhong
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States.,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Georgina Harris
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Lena Smirnova
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Valentin Zufferey
- Department of Physiology, Lausanne and Swiss Centre for Applied Human Toxicology (SCAHT), University of Lausanne, Lausanne, Switzerland
| | | | - Fabiele Baldino Russo
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Patricia Cristina Baleeiro Beltrao Braga
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Department of Obstetrics, School of Arts Sciences and Humanities, São Paulo, Brazil
| | - Megan Chesnut
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Marie-Gabrielle Zurich
- Department of Physiology, Lausanne and Swiss Centre for Applied Human Toxicology (SCAHT), University of Lausanne, Lausanne, Switzerland
| | - Helena T Hogberg
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Thomas Hartung
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States.,CAAT-Europe, University of Konstanz, Konstanz, Germany
| | - David Pamies
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States.,Department of Physiology, Lausanne and Swiss Centre for Applied Human Toxicology (SCAHT), University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
29
|
Teissier A, Le Magueresse C, Olusakin J, Andrade da Costa BLS, De Stasi AM, Bacci A, Imamura Kawasawa Y, Vaidya VA, Gaspar P. Early-life stress impairs postnatal oligodendrogenesis and adult emotional behaviour through activity-dependent mechanisms. Mol Psychiatry 2020; 25:1159-1174. [PMID: 31439936 PMCID: PMC7244403 DOI: 10.1038/s41380-019-0493-2] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 06/27/2019] [Accepted: 07/09/2019] [Indexed: 12/15/2022]
Abstract
Exposure to stress during early life (infancy/childhood) has long-term effects on the structure and function of the prefrontal cortex (PFC), and increases the risk for adult depression and anxiety disorders. However, little is known about the molecular and cellular mechanisms of these effects. Here, we focused on changes induced by chronic maternal separation during the first 2 weeks of postnatal life. Unbiased mRNA expression profiling in the medial PFC (mPFC) of maternally separated (MS) pups identified an increased expression of myelin-related genes and a decreased expression of immediate early genes. Oligodendrocyte lineage markers and birthdating experiments indicated a precocious oligodendrocyte differentiation in the mPFC at P15, leading to a depletion of the oligodendrocyte progenitor pool in MS adults. We tested the role of neuronal activity in oligodendrogenesis, using designed receptors exclusively activated by designed drugs (DREADDs) techniques. hM4Di or hM3Dq constructs were transfected into mPFC neurons using fast-acting AAV8 viruses. Reduction of mPFC neuron excitability during the first 2 postnatal weeks caused a premature differentiation of oligodendrocytes similar to the MS pups, while chemogenetic activation normalised it in the MS animals. Bidirectional manipulation of neuron excitability in the mPFC during the P2-P14 period had long lasting effects on adult emotional behaviours and on temporal object recognition: hM4Di mimicked MS effects, while hM3Dq prevented the pro-depressive effects and short-term memory impairment of MS. Thus, our results identify neuronal activity as a critical target of early-life stress and demonstrate its function in controlling both postnatal oligodendrogenesis and adult mPFC-related behaviours.
Collapse
Affiliation(s)
- Anne Teissier
- INSERM, Institut du Fer à Moulin, UMR-S 1270, Sorbonne Université, Paris, France. .,Sorbonne Université, Paris, France. .,Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, Université de Paris, Paris, France.
| | - Corentin Le Magueresse
- 0000 0004 0520 8345grid.462192.aINSERM, Institut du Fer à Moulin, UMR-S 1270, Sorbonne Université, Paris, France ,0000 0001 2308 1657grid.462844.8Sorbonne Université, Paris, France
| | - Jimmy Olusakin
- 0000 0004 0520 8345grid.462192.aINSERM, Institut du Fer à Moulin, UMR-S 1270, Sorbonne Université, Paris, France ,0000 0001 2308 1657grid.462844.8Sorbonne Université, Paris, France
| | - Belmira L. S. Andrade da Costa
- 0000 0001 0670 7996grid.411227.3Physiology and Pharmacology Department, Federal University of Pernambuco, Recife, Brazil
| | - Angela M. De Stasi
- 0000 0001 2308 1657grid.462844.8Sorbonne Université, Paris, France ,0000 0004 0620 5939grid.425274.2Institut du Cerveau et de la Moelle épinière, CNRS UMR 7225—Inserm U1127, Paris, France
| | - Alberto Bacci
- 0000 0001 2308 1657grid.462844.8Sorbonne Université, Paris, France ,0000 0004 0620 5939grid.425274.2Institut du Cerveau et de la Moelle épinière, CNRS UMR 7225—Inserm U1127, Paris, France
| | - Yuka Imamura Kawasawa
- 0000 0001 2097 4281grid.29857.31Departments of Pharmacology and Biochemistry and Molecular Biology, Institute for Personalized Medicine, Penn State University College of Medicine, Hershey, PA USA
| | - Vidita A. Vaidya
- 0000 0004 0502 9283grid.22401.35Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, 400005 India
| | - Patricia Gaspar
- INSERM, Institut du Fer à Moulin, UMR-S 1270, Sorbonne Université, Paris, France. .,Sorbonne Université, Paris, France. .,Institut du Cerveau et de la Moelle épinière, CNRS UMR 7225-Inserm U1127, Paris, France.
| |
Collapse
|
30
|
Abbasi H, Unsworth CP. Electroencephalogram studies of hypoxic ischemia in fetal and neonatal animal models. Neural Regen Res 2020; 15:828-837. [PMID: 31719243 PMCID: PMC6990791 DOI: 10.4103/1673-5374.268892] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Alongside clinical achievements, experiments conducted on animal models (including primate or non-primate) have been effective in the understanding of various pathophysiological aspects of perinatal hypoxic/ischemic encephalopathy (HIE). Due to the reasonably fair degree of flexibility with experiments, most of the research around HIE in the literature has been largely concerned with the neurodevelopmental outcome or how the frequency and duration of HI seizures could relate to the severity of perinatal brain injury, following HI insult. This survey concentrates on how EEG experimental studies using asphyxiated animal models (in rodents, piglets, sheep and non-human primate monkeys) provide a unique opportunity to examine from the exact time of HI event to help gain insights into HIE where human studies become difficult.
Collapse
Affiliation(s)
- Hamid Abbasi
- Department of Engineering Science, the University of Auckland, Auckland, New Zealand
| | - Charles P Unsworth
- Department of Engineering Science, the University of Auckland, Auckland, New Zealand
| |
Collapse
|
31
|
Harbuzariu A, Pitts S, Cespedes JC, Harp KO, Nti A, Shaw AP, Liu M, Stiles JK. Modelling heme-mediated brain injury associated with cerebral malaria in human brain cortical organoids. Sci Rep 2019; 9:19162. [PMID: 31844087 PMCID: PMC6914785 DOI: 10.1038/s41598-019-55631-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 11/26/2019] [Indexed: 01/09/2023] Open
Abstract
Human cerebral malaria (HCM), a severe encephalopathy associated with Plasmodium falciparum infection, has a 20-30% mortality rate and predominantly affects African children. The mechanisms mediating HCM-associated brain injury are difficult to study in human subjects, highlighting the urgent need for non-invasive ex vivo human models. HCM elevates the systemic levels of free heme, which damages the blood-brain barrier and neurons in distinct regions of the brain. We determined the effects of heme on induced pluripotent stem cells (iPSCs) and a three-dimensional cortical organoid system and assessed apoptosis and differentiation. We evaluated biomarkers associated with heme-induced brain injury, including a pro-inflammatory chemokine, CXCL-10, and its receptor, CXCR3, brain-derived neurotrophic factor (BDNF) and a receptor tyrosine-protein kinase, ERBB4, in the organoids. We then tested the neuroprotective effect of neuregulin-1 (NRG-1) against heme treatment in organoids. Neural stem and mature cells differentially expressed CXCL-10, CXCR3, BDNF and ERBB4 in the developing organoids and in response to heme-induced neuronal injury. The organoids underwent apoptosis and structural changes that were attenuated by NRG-1. Thus, cortical organoids can be used to model heme-induced cortical brain injury associated with HCM pathogenesis as well as for testing agents that reduce brain injury and neurological sequelae.
Collapse
Affiliation(s)
- Adriana Harbuzariu
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, 720 Westview Dr, Atlanta, GA, 30310, USA.
| | - Sidney Pitts
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, 720 Westview Dr, Atlanta, GA, 30310, USA
| | - Juan Carlos Cespedes
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, 720 Westview Dr, Atlanta, GA, 30310, USA
| | - Keri Oxendine Harp
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, 720 Westview Dr, Atlanta, GA, 30310, USA
| | - Annette Nti
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, 720 Westview Dr, Atlanta, GA, 30310, USA
| | - Andrew P Shaw
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA, 30332, USA
| | - Mingli Liu
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, 720 Westview Dr, Atlanta, GA, 30310, USA
| | - Jonathan K Stiles
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, 720 Westview Dr, Atlanta, GA, 30310, USA.
| |
Collapse
|
32
|
Baldassarro VA, Marchesini A, Giardino L, Calzà L. Differential effects of glucose deprivation on the survival of fetal versus adult neural stem cells-derived oligodendrocyte precursor cells. Glia 2019; 68:898-917. [PMID: 31755592 DOI: 10.1002/glia.23750] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 12/27/2022]
Abstract
Impaired myelination is a key feature in neonatal hypoxia/ischemia (HI), the most common perinatal/neonatal cause of death and permanent disabilities, which is triggered by the establishment of an inflammatory and hypoxic environment during the most critical period of myelin development. This process is dependent on oligodendrocyte precursor cells (OPCs) and their capability to differentiate into mature oligodendrocytes. In this study, we investigated the vulnerability of fetal and adult OPCs derived from neural stem cells (NSCs) to inflammatory and HI insults. The resulting OPCs/astrocytes cultures were exposed to cytokines to mimic inflammation, or to oxygen-glucose deprivation (OGD) to mimic an HI condition. The differentiation of both fetal and adult OPCs is completely abolished following exposure to inflammatory cytokines, while only fetal-derived OPCs degenerate when exposed to OGD. We then investigated possible mechanisms involved in OGD-mediated toxicity: (a) T3-mediated maturation induction; (b) glutamate excitotoxicity; (c) glucose metabolism. We found that while no substantial differences were observed in T3 intracellular content regulation and glutamate-mediated toxicity, glucose deprivation lead to selective OPC cell death and impaired differentiation in fetal cultures only. These results indicate that the biological response of OPCs to inflammation and demyelination is different in fetal and adult cells, and that the glucose metabolism perturbation in fetal central nervous system (CNS) may significantly contribute to neonatal pathologies. An understanding of the underlying molecular mechanism will contribute greatly to differentiating myelination enhancing and neuroprotective therapies for neonatal and adult CNS white matter lesions.
Collapse
Affiliation(s)
- Vito Antonio Baldassarro
- Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy.,Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.,Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
| | | | - Luciana Giardino
- Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy.,IRET Foundation, Ozzano Emilia, Italy.,Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Laura Calzà
- Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy.,Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.,IRET Foundation, Ozzano Emilia, Italy
| |
Collapse
|
33
|
McNamara NB, Miron VE. Microglia in developing white matter and perinatal brain injury. Neurosci Lett 2019; 714:134539. [PMID: 31614181 DOI: 10.1016/j.neulet.2019.134539] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/04/2019] [Accepted: 10/07/2019] [Indexed: 12/13/2022]
Abstract
Perinatal brain injury (PBI) to the developing white matter results in hypomyelination of axons and can cause long-term motor and cognitive deficits e.g. cerebral palsy. There are currently no approved therapies aimed at repairing the white matter following insult, underscoring the need to investigate the mechanisms underlying the pathogenesis of PBI. Microglia have been strongly implicated, but their function and heterogeneity in this context remain poorly understood, posing a barrier to the development of microglia-targeted therapies for white matter repair following PBI. In this review, we discuss the roles of microglia in normal white matter development and in PBI, and potential drug strategies to influence microglial responses in this setting.
Collapse
Affiliation(s)
- Niamh B McNamara
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Veronique E Miron
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
34
|
Niño DF, Zhou Q, Yamaguchi Y, Martin LY, Wang S, Fulton WB, Jia H, Lu P, Prindle T, Zhang F, Crawford J, Hou Z, Mori S, Chen LL, Guajardo A, Fatemi A, Pletnikov M, Kannan RM, Kannan S, Sodhi CP, Hackam DJ. Cognitive impairments induced by necrotizing enterocolitis can be prevented by inhibiting microglial activation in mouse brain. Sci Transl Med 2019; 10:10/471/eaan0237. [PMID: 30541786 DOI: 10.1126/scitranslmed.aan0237] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/21/2018] [Accepted: 07/23/2018] [Indexed: 12/11/2022]
Abstract
Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease of the premature infant. One of the most important long-term complications observed in children who survive NEC early in life is the development of profound neurological impairments. However, the pathways leading to NEC-associated neurological impairments remain unknown, thus limiting the development of prevention strategies. We have recently shown that NEC development is dependent on the expression of the lipopolysaccharide receptor Toll-like receptor 4 (TLR4) on the intestinal epithelium, whose activation by bacteria in the newborn gut leads to mucosal inflammation. Here, we hypothesized that damage-induced production of TLR4 endogenous ligands in the intestine might lead to activation of microglial cells in the brain and promote cognitive impairments. We identified a gut-brain signaling axis in an NEC mouse model in which activation of intestinal TLR4 signaling led to release of high-mobility group box 1 in the intestine that, in turn, promoted microglial activation in the brain and neurological dysfunction. We further demonstrated that an orally administered dendrimer-based nanotherapeutic approach to targeting activated microglia could prevent NEC-associated neurological dysfunction in neonatal mice. These findings shed light on the molecular pathways leading to the development of NEC-associated brain injury, provide a rationale for early removal of diseased intestine in NEC, and indicate the potential of targeted therapies that protect the developing brain in the treatment of NEC in early childhood.
Collapse
Affiliation(s)
- Diego F Niño
- Division of General Pediatric Surgery, Johns Hopkins University and Bloomberg Children's Center, Johns Hopkins Hospital, Baltimore, MD 21287, USA.,Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Johns Hopkins Hospital, Baltimore, MD 21287, USA
| | - Qinjie Zhou
- Division of General Pediatric Surgery, Johns Hopkins University and Bloomberg Children's Center, Johns Hopkins Hospital, Baltimore, MD 21287, USA.,Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Johns Hopkins Hospital, Baltimore, MD 21287, USA
| | - Yukihiro Yamaguchi
- Division of General Pediatric Surgery, Johns Hopkins University and Bloomberg Children's Center, Johns Hopkins Hospital, Baltimore, MD 21287, USA.,Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Johns Hopkins Hospital, Baltimore, MD 21287, USA
| | - Laura Y Martin
- Division of General Pediatric Surgery, Johns Hopkins University and Bloomberg Children's Center, Johns Hopkins Hospital, Baltimore, MD 21287, USA.,Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Johns Hopkins Hospital, Baltimore, MD 21287, USA
| | - Sanxia Wang
- Division of General Pediatric Surgery, Johns Hopkins University and Bloomberg Children's Center, Johns Hopkins Hospital, Baltimore, MD 21287, USA.,Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Johns Hopkins Hospital, Baltimore, MD 21287, USA
| | - William B Fulton
- Division of General Pediatric Surgery, Johns Hopkins University and Bloomberg Children's Center, Johns Hopkins Hospital, Baltimore, MD 21287, USA.,Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Johns Hopkins Hospital, Baltimore, MD 21287, USA
| | - Hongpeng Jia
- Division of General Pediatric Surgery, Johns Hopkins University and Bloomberg Children's Center, Johns Hopkins Hospital, Baltimore, MD 21287, USA.,Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Johns Hopkins Hospital, Baltimore, MD 21287, USA
| | - Peng Lu
- Division of General Pediatric Surgery, Johns Hopkins University and Bloomberg Children's Center, Johns Hopkins Hospital, Baltimore, MD 21287, USA.,Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Johns Hopkins Hospital, Baltimore, MD 21287, USA
| | - Thomas Prindle
- Division of General Pediatric Surgery, Johns Hopkins University and Bloomberg Children's Center, Johns Hopkins Hospital, Baltimore, MD 21287, USA.,Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Johns Hopkins Hospital, Baltimore, MD 21287, USA
| | - Fan Zhang
- Program of Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Joshua Crawford
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Zhipeng Hou
- Department of Biomedical Engineering and The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Susumu Mori
- Department of Biomedical Engineering and The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Liam L Chen
- Division of Neuropathology, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Andrew Guajardo
- Division of Neuropathology, Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Ali Fatemi
- Departments of Neurology and Pediatrics, Kennedy Krieger Institute and Johns Hopkins University School of Medicine, MD 21205, USA
| | - Mikhail Pletnikov
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.,Department of Molecular and Comparative Pathobiology and Solomon H. Snyder Department of Neuroscience, Johns Hopkins University and Johns Hopkins Children's Center, Johns Hopkins Hospital, Baltimore, MD 21205, USA
| | - Rangaramanujam M Kannan
- Center for Nanomedicine, Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sujatha Kannan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chhinder P Sodhi
- Division of General Pediatric Surgery, Johns Hopkins University and Bloomberg Children's Center, Johns Hopkins Hospital, Baltimore, MD 21287, USA. .,Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Johns Hopkins Hospital, Baltimore, MD 21287, USA
| | - David J Hackam
- Division of General Pediatric Surgery, Johns Hopkins University and Bloomberg Children's Center, Johns Hopkins Hospital, Baltimore, MD 21287, USA. .,Division of General Pediatric Surgery, Johns Hopkins University and Johns Hopkins Children's Center, Johns Hopkins Hospital, Baltimore, MD 21287, USA
| |
Collapse
|
35
|
Abbink TEM, Wisse LE, Jaku E, Thiecke MJ, Voltolini-González D, Fritsen H, Bobeldijk S, Ter Braak TJ, Polder E, Postma NL, Bugiani M, Struijs EA, Verheijen M, Straat N, van der Sluis S, Thomas AAM, Molenaar D, van der Knaap MS. Vanishing white matter: deregulated integrated stress response as therapy target. Ann Clin Transl Neurol 2019; 6:1407-1422. [PMID: 31402619 PMCID: PMC6689685 DOI: 10.1002/acn3.50826] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 05/31/2019] [Indexed: 02/06/2023] Open
Abstract
Objective Vanishing white matter (VWM) is a fatal, stress‐sensitive leukodystrophy that mainly affects children and is currently without treatment. VWM is caused by recessive mutations in eukaryotic initiation factor 2B (eIF2B) that is crucial for initiation of mRNA translation and its regulation during the integrated stress response (ISR). Mutations reduce eIF2B activity. VWM pathomechanisms remain unclear. In contrast with the housekeeping function of eIF2B, astrocytes are selectively affected in VWM. One study objective was to test our hypothesis that in the brain translation of specific mRNAs is altered by eIF2B mutations, impacting primarily astrocytes. The second objective was to investigate whether modulation of eIF2B activity could ameliorate this altered translation and improve the disease. Methods Mice with biallelic missense mutations in eIF2B that recapitulate human VWM were used to screen for mRNAs with altered translation in brain using polysomal profiling. Findings were verified in brain tissue from VWM patients using qPCR and immunohistochemistry. The compound ISRIB (for “ISR inhibitor”) was administered to VWM mice to increase eIF2B activity. Its effect on translation, neuropathology, and clinical signs was assessed. Results In brains of VWM compared to wild‐type mice we observed the most prominent changes in translation concerning ISR mRNAs; their expression levels correlated with disease severity. We substantiated these findings in VWM patients’ brains. ISRIB normalized expression of mRNA markers, ameliorated brain white matter pathology and improved motor skills in VWM mice. Interpretation The present findings show that ISR deregulation is central in VWM pathomechanisms and a viable target for therapy.
Collapse
Affiliation(s)
- Truus E M Abbink
- Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Lisanne E Wisse
- Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Ermelinda Jaku
- Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Michiel J Thiecke
- Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Daniel Voltolini-González
- Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Hein Fritsen
- Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Sander Bobeldijk
- Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Timo J Ter Braak
- Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Emiel Polder
- Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Nienke L Postma
- Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Marianna Bugiani
- Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, The Netherlands.,Department of Pathology, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Eduard A Struijs
- Metabolic Unit, Department of Clinical Chemistry, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Mark Verheijen
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Nina Straat
- Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Sophie van der Sluis
- Complex Trait Genetics, Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Adri A M Thomas
- Developmental Biology, Utrecht University, Utrecht, The Netherlands
| | - Douwe Molenaar
- Systems Bioinformatics, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Marjo S van der Knaap
- Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, The Netherlands.,Functional Genomics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
36
|
Vaes JEG, Vink MA, de Theije CGM, Hoebeek FE, Benders MJNL, Nijboer CHA. The Potential of Stem Cell Therapy to Repair White Matter Injury in Preterm Infants: Lessons Learned From Experimental Models. Front Physiol 2019; 10:540. [PMID: 31143126 PMCID: PMC6521595 DOI: 10.3389/fphys.2019.00540] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/17/2019] [Indexed: 12/12/2022] Open
Abstract
Diffuse white matter injury (dWMI) is a major cause of morbidity in the extremely preterm born infant leading to life-long neurological impairments, including deficits in cognitive, motor, sensory, psychological, and behavioral functioning. At present, no treatment options are clinically available to combat dWMI and therefore exploration of novel strategies is urgently needed. In recent years, the pathophysiology underlying dWMI has slowly started to be unraveled, pointing towards the disturbed maturation of oligodendrocytes (OLs) as a key mechanism. Immature OL precursor cells in the developing brain are believed to be highly sensitive to perinatal inflammation and cerebral oxygen fluctuations, leading to impaired OL differentiation and eventually myelination failure. OL lineage development under normal and pathological circumstances and the process of (re)myelination have been studied extensively over the years, often in the context of other adult and pediatric white matter pathologies such as stroke and multiple sclerosis (MS). Various studies have proposed stem cell-based therapeutic strategies to boost white matter regeneration as a potential strategy against a wide range of neurological diseases. In this review we will discuss experimental studies focusing on mesenchymal stem cell (MSC) therapy to reduce white matter injury (WMI) in multiple adult and neonatal neurological diseases. What lessons have been learned from these previous studies and how can we translate this knowledge to application of MSCs for the injured white matter in the preterm infant? A perspective on the current state of stem cell therapy will be given and we will discuss different important considerations of MSCs including cellular sources, timing of treatment and administration routes. Furthermore, we reflect on optimization strategies that could potentially reinforce stem cell therapy, including preconditioning and genetic engineering of stem cells or using cell-free stem cell products, to optimize cell-based strategy for vulnerable preterm infants in the near future.
Collapse
Affiliation(s)
- Josine E G Vaes
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.,Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Marit A Vink
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Caroline G M de Theije
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Freek E Hoebeek
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Manon J N L Benders
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Cora H A Nijboer
- NIDOD Laboratory, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
37
|
Brandenburg JE, Fogarty MJ, Sieck GC. A Critical Evaluation of Current Concepts in Cerebral Palsy. Physiology (Bethesda) 2019; 34:216-229. [PMID: 30968751 PMCID: PMC7938766 DOI: 10.1152/physiol.00054.2018] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 01/11/2019] [Accepted: 01/23/2019] [Indexed: 11/22/2022] Open
Abstract
Spastic cerebral palsy (CP), despite the name, is not consistently identifiable by specific brain lesions. CP animal models focus on risk factors for development of CP, yet few reproduce the diagnostic symptoms. Animal models of CP must advance beyond risk factors to etiologies, including both the brain and spinal cord.
Collapse
Affiliation(s)
- Joline E Brandenburg
- Department of Physical Medicine and Rehabilitation, Mayo Clinic College of Medicine , Rochester, Minnesota
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine , Rochester, Minnesota
| | - Matthew J Fogarty
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine , Rochester, Minnesota
| | - Gary C Sieck
- Department of Physical Medicine and Rehabilitation, Mayo Clinic College of Medicine , Rochester, Minnesota
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine , Rochester, Minnesota
- Department of Anesthesiology, Mayo Clinic College of Medicine , Rochester, Minnesota
| |
Collapse
|
38
|
Abbasi H, Bennet L, Gunn AJ, Unsworth CP. Latent Phase Detection of Hypoxic-Ischemic Spike Transients in the EEG of Preterm Fetal Sheep Using Reverse Biorthogonal Wavelets & Fuzzy Classifier. Int J Neural Syst 2019; 29:1950013. [PMID: 31184228 DOI: 10.1142/s0129065719500138] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hypoxic-ischemic (HI) studies in preterms lack reliable prognostic biomarkers for diagnostic tests of HI encephalopathy (HIE). Our group's observations from in utero fetal sheep models suggest that potential biomarkers of HIE in the form of developing HI micro-scale epileptiform transients emerge along suppressed EEG/ECoG background during a latent phase of 6-7h post-insult. However, having to observe for the whole of the latent phase disqualifies any chance of clinical intervention. A precise automatic identification of these transients can help for a well-timed diagnosis of the HIE and to stop the spread of the injury before it becomes irreversible. This paper reports fusion of Reverse-Biorthogonal Wavelets with Type-1 Fuzzy classifiers, for the accurate real-time automatic identification and quantification of high-frequency HI spike transients in the latent phase, tested over seven in utero preterm sheep. Considerable high performance of 99.78 ± 0.10% was obtained from the Rbio-Wavelet Type-1 Fuzzy classifier for automatic identification of HI spikes tested over 42h of high-resolution recordings (sampling-freq:1024Hz). Data from post-insult automatic time-localization of high-frequency HI spikes reveals a promising trend in the average rate of the HI spikes, even in the animals with shorter occlusion periods, which highlights considerable higher number of transients within the first 2h post-insult.
Collapse
Affiliation(s)
- Hamid Abbasi
- Department of Engineering Science, The University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- Department of Physiology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Charles P Unsworth
- Department of Engineering Science, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
39
|
Chang JL, Bashir M, Santiago C, Farrow K, Fung C, Brown AS, Dettman RW, Dizon MLV. Intrauterine Growth Restriction and Hyperoxia as a Cause of White Matter Injury. Dev Neurosci 2018; 40:344-357. [PMID: 30428455 DOI: 10.1159/000494273] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 10/03/2018] [Indexed: 01/17/2023] Open
Abstract
Intrauterine growth restriction (IUGR) is estimated to occur in 5% of pregnancies, with placental insufficiency being the most common cause in developed countries. While it is known that white matter injury occurs in premature infants, the extent of IUGR on white matter injury is less defined in term infants. We used a novel murine model that utilizes a thromboxane A2 (TXA2) analog (U46619), a potent vasoconstrictor, to induce maternal hypertension and mimic human placental insufficiency-induced IUGR to study the white matter. We also investigated the role of hyperoxia as an additional risk factor for white matter injury, as IUGR infants are at increased risk of respiratory comorbidities leading to increased oxygen exposure. We found that TXA2 analog-induced IUGR results in white matter injury as demonstrated by altered myelin structure and changes in the oligodendroglial cell/oligodendrocyte population. In addition, our study demonstrates that hyperoxia exposure independently results in white matter perturbation. To our knowledge, this is the first study to report single and combined effects of IUGR with hyperoxia impacting the white matter and motor function. These results draw attention to the need for close monitoring of motor development in IUGR babies following hospital discharge as well as highlighting the importance of limiting, as clinically feasible, the degree of oxygen overexposure to potentially improve motor outcomes in this population of infants.
Collapse
Affiliation(s)
- Jill L Chang
- Division of Neonatology, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA,
| | - Mirrah Bashir
- Division of Neonatology, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | | | - Kathryn Farrow
- Division of Neonatology, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Camille Fung
- Division of Neonatology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Ashley S Brown
- Division of Neonatology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Robert W Dettman
- Stanley Manne Children's Research Institute, Chicago, Illinois, USA
| | - Maria L V Dizon
- Division of Neonatology, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
40
|
Muzzi M, Buonvicino D, Urru M, Tofani L, Chiarugi A. Repurposing of dexpramipexole to treatment of neonatal hypoxic/ischemic encephalopathy. Neurosci Lett 2018; 687:234-240. [PMID: 30287306 DOI: 10.1016/j.neulet.2018.09.064] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 09/27/2018] [Accepted: 09/29/2018] [Indexed: 01/28/2023]
Abstract
Dexpramipexole (DEX) is a drug with a good safety profile in humans, known for its ability to increase mitochondrial ATP production and prompt neuroprotection in adult rodents subjected to cerebral ischemia. In the present study we evaluated the effect of DEX in rat pups subjected to common carotid artery occlusion plus hypoxia (CCAoH, the classic Rice-Vannucci model). Because of the wide range of infarct size distribution in the CCAoH model, a priori subanalysis based on the effect of DEX on mild/moderate or severe brain injuries was conducted. The subanalysis showed that the drug (3 mg/kg bid i.p, after the hypoxic insult) decreased the infarction size in pups with mild/moderate injuries. Next, we developed a distal middle cerebral artery occlusion plus hypoxia (dMCAoH) model, characterized by an intra-experimental infarct size variability lower than that of the CCAoH model. Post-ischemic treatment with DEX (3 mg/kg bid i.p, after the hypoxic insult) reduced brain infarcts in pups exposed to dMCAoH. For the first time, we show that DEX reduces brain injury in different models of neonatal HIE. In light of the favorable safety profile of DEX in humans, the drug might have a realistic translational potential to treatment of perinatal cerebrovascular disorders.
Collapse
Affiliation(s)
- Mirko Muzzi
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy.
| | - Daniela Buonvicino
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Matteo Urru
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Lorenzo Tofani
- Clinical Trials Coordinating Center of Istituto Toscano Tumori, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Alberto Chiarugi
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| |
Collapse
|
41
|
Sun L, Xia L, Wang M, Zhu D, Wang Y, Bi D, Song J, Ma C, Gao C, Zhang X, Sun Y, Wang X, Zhu C, Xing Q. Variants of the OLIG2 Gene are Associated with Cerebral Palsy in Chinese Han Infants with Hypoxic-Ischemic Encephalopathy. Neuromolecular Med 2018; 21:75-84. [PMID: 30178266 DOI: 10.1007/s12017-018-8510-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/31/2018] [Indexed: 12/11/2022]
Abstract
Cerebral palsy (CP) is a leading cause of neurological disability among young children. Congenial and adverse perinatal clinical conditions, such as genetic factors, perinatal infection, and asphyxia, are risk factors for CP. Oligodendrocyte transcription factor (OLIG2) is a protein that is expressed in brain oligodendrocyte cells and is involved in neuron repair after brain injury. In this study, we employed a Chinese Han cohort of 763 CP infants and 738 healthy controls to study the association of OLIG2 gene polymorphisms with CP. We found marginal association of the SNP rs6517135 with CP (p = 0.044) at the genotype level, and the association was greatly strengthened when we focused on the subgroup of CP infants who suffered from hypoxic-ischemic encephalopathy (HIE) after birth, with p = 0.003 (OR = 0.558) at the allele level and p = 0.007 at the genotype level, indicating a risk-associated role of the T allele of the SNP rs6517135 under HIE conditions. The haplotype CTTG for rs6517135-rs1005573-rs6517137-rs9653711 in OLIG2 was also significantly associated with the occurrence of CP in infants with HIE (p = 0.01, OR = 0.521). Our results indicate that in the Han Chinese population, the polymorphisms of OLIG2 were associated with CP, especially in patients who had suffered HIE injury. This finding could be used to develop personalized care for infants with high susceptibility to CP.
Collapse
MESH Headings
- Alleles
- Asian People/genetics
- Asphyxia Neonatorum/complications
- Case-Control Studies
- Cerebral Palsy/etiology
- Cerebral Palsy/genetics
- Child
- Child, Preschool
- Female
- Fetal Growth Retardation/epidemiology
- Genetic Predisposition to Disease
- Genotype
- Haplotypes/genetics
- Humans
- Hypoxia-Ischemia, Brain/complications
- Infant
- Infant, Low Birth Weight
- Infant, Newborn
- Infant, Premature
- Infant, Premature, Diseases/epidemiology
- Infant, Premature, Diseases/genetics
- Male
- Oligodendrocyte Transcription Factor 2/deficiency
- Oligodendrocyte Transcription Factor 2/genetics
- Oligodendrocyte Transcription Factor 2/physiology
- Oligodendroglia/metabolism
- Polymorphism, Single Nucleotide
- Pregnancy
- Pregnancy Complications/epidemiology
- Risk
Collapse
Affiliation(s)
- Liya Sun
- Institute of Biomedical Science and Children's Hospital, Fudan University, Shanghai, 201102, China
- Shanghai Center for Women and Children's Health, Shanghai, 200062, China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Lei Xia
- Henan Key Laboratory of Child Brain Injury, Department of Pediatrics, The 3rd Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Mingtai Wang
- Nursing School, Sias International University, Zhengzhou, 451150, China
| | - Dengna Zhu
- Henan Key Laboratory of Child Brain Injury, Department of Pediatrics, The 3rd Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Child Rehabilitation Center, The 3rd Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yangong Wang
- Institute of Biomedical Science and Children's Hospital, Fudan University, Shanghai, 201102, China
| | - Dan Bi
- Henan Key Laboratory of Child Brain Injury, Department of Pediatrics, The 3rd Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Juan Song
- Henan Key Laboratory of Child Brain Injury, Department of Pediatrics, The 3rd Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Caiyun Ma
- Department of Pediatrics, Children's Hospital of Zhengzhou University and Henan Children's Hospital, Zhengzhou, 450053, China
| | - Chao Gao
- Department of Pediatrics, Children's Hospital of Zhengzhou University and Henan Children's Hospital, Zhengzhou, 450053, China
| | - Xiaoli Zhang
- Henan Key Laboratory of Child Brain Injury, Department of Pediatrics, The 3rd Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yanyan Sun
- Henan Key Laboratory of Child Brain Injury, Department of Pediatrics, The 3rd Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury, Department of Pediatrics, The 3rd Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 40530, Gothenburg, Sweden
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury, Department of Pediatrics, The 3rd Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 40530, Gothenburg, Sweden.
- Henan Key Laboratory of Child Brain Injury, Zhengzhou University, Kangfuqian Street 7, Zhengzhou, 450052, China.
| | - Qinghe Xing
- Institute of Biomedical Science and Children's Hospital, Fudan University, Shanghai, 201102, China.
- Shanghai Center for Women and Children's Health, Shanghai, 200062, China.
| |
Collapse
|
42
|
Bruckner D, Kaser-Eichberger A, Bogner B, Runge C, Schrödl F, Strohmaier C, Silva ME, Zaunmair P, Couillard-Despres S, Aigner L, Rivera FJ, Reitsamer HA, Trost A. Retinal Pericytes: Characterization of Vascular Development-Dependent Induction Time Points in an Inducible NG2 Reporter Mouse Model. Curr Eye Res 2018; 43:1274-1285. [DOI: 10.1080/02713683.2018.1493130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Daniela Bruckner
- University Clinic of Ophthalmology and Optometry, Research Program for Experimental Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, Salzburg, Austria
| | - Alexandra Kaser-Eichberger
- University Clinic of Ophthalmology and Optometry, Research Program for Experimental Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, Salzburg, Austria
| | - Barbara Bogner
- University Clinic of Ophthalmology and Optometry, Research Program for Experimental Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, Salzburg, Austria
| | - Christian Runge
- University Clinic of Ophthalmology and Optometry, Research Program for Experimental Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, Salzburg, Austria
| | - Falk Schrödl
- University Clinic of Ophthalmology and Optometry, Research Program for Experimental Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, Salzburg, Austria
- Department of Anatomy, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Clemens Strohmaier
- University Clinic of Ophthalmology and Optometry, Research Program for Experimental Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, Salzburg, Austria
| | - Maria Elena Silva
- Laboratory of Stem Cells and Neuroregeneration, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Institute of Pharmacy, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Pia Zaunmair
- Institute of Experimental Neuroregeneration, Paracelsus Medical University Salzburg, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Sebastien Couillard-Despres
- Institute of Experimental Neuroregeneration, Paracelsus Medical University Salzburg, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Ludwig Aigner
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
- Institute of Mol. Regenerative Medicine, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Francisco J. Rivera
- Laboratory of Stem Cells and Neuroregeneration, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
- Institute of Mol. Regenerative Medicine, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Herbert A. Reitsamer
- University Clinic of Ophthalmology and Optometry, Research Program for Experimental Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, Salzburg, Austria
- Director of the Research Program for Experimental Ophthalmology and Glaucoma Research, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Andrea Trost
- University Clinic of Ophthalmology and Optometry, Research Program for Experimental Ophthalmology and Glaucoma Research, Paracelsus Medical University/SALK, Salzburg, Austria
| |
Collapse
|
43
|
Sanches EF, Van de Looij Y, Toulotte A, da Silva AR, Romero J, Sizonenko SV. Brain Metabolism Alterations Induced by Pregnancy Swimming Decreases Neurological Impairments Following Neonatal Hypoxia-Ischemia in Very Immature Rats. Front Neurol 2018; 9:480. [PMID: 29988536 PMCID: PMC6026645 DOI: 10.3389/fneur.2018.00480] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 06/01/2018] [Indexed: 01/07/2023] Open
Abstract
Introduction: Prematurity, through brain injury and altered development is a major cause of neurological impairments and can result in motor, cognitive and behavioral deficits later in life. Presently, there are no well-established effective therapies for preterm brain injury and the search for new strategies is needed. Intra-uterine environment plays a decisive role in brain maturation and interventions using the gestational window have been shown to influence long-term health in the offspring. In this study, we investigated whether pregnancy swimming can prevent the neurochemical metabolic alterations and damage that result from postnatal hypoxic-ischemic brain injury (HI) in very immature rats. Methods: Female pregnant Wistar rats were divided into swimming (SW) or sedentary (SE) groups. Following a period of adaptation before mating, swimming was performed during the entire gestation. At postnatal day (PND3), rat pups from SW and SE dams had right common carotid artery occluded, followed by systemic hypoxia. At PND4 (24 h after HI), the early neurochemical profile was measured by 1H-magnetic resonance spectroscopy. Astrogliosis, apoptosis and neurotrophins protein expression were assessed in the cortex and hippocampus. From PND45, behavioral testing was performed. Diffusion tensor imaging and neurite orientation dispersion and density imaging were used to evaluate brain microstructure and the levels of proteins were quantified. Results: Pregnancy swimming was able to prevent early metabolic changes induced by HI preserving the energetic balance, decreasing apoptotic cell death and astrogliosis as well as maintaining the levels of neurotrophins. At adult age, swimming preserved brain microstructure and improved the performance in the behavioral tests. Conclusion: Our study points out that swimming during gestation in rats could prevent prematurity related brain damage in progeny with high translational potential and possibly interesting cost-benefits. HIGHLIGHTS- Prematurity is a major cause of neurodevelopmental impairments; - Swimming during pregnancy reduces brain damage after HI injury; - Pregnancy is an important but underestimated preventive window.
Collapse
Affiliation(s)
- Eduardo F Sanches
- Division of Child Development and Growth, Department of Pediatrics, University of Geneva, Geneva, Switzerland
| | - Yohan Van de Looij
- Division of Child Development and Growth, Department of Pediatrics, University of Geneva, Geneva, Switzerland.,Laboratory for Functional and Metabolic Imaging, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Audrey Toulotte
- Division of Child Development and Growth, Department of Pediatrics, University of Geneva, Geneva, Switzerland
| | - Analina R da Silva
- Laboratory for Functional and Metabolic Imaging, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Jacqueline Romero
- Laboratory for Functional and Metabolic Imaging, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Stephane V Sizonenko
- Division of Child Development and Growth, Department of Pediatrics, University of Geneva, Geneva, Switzerland
| |
Collapse
|
44
|
Lu F, Zhu J, Guo S, Wong BJ, Chehab FF, Ferriero DM, Jiang X. Upregulation of cholesterol 24-hydroxylase following hypoxia-ischemia in neonatal mouse brain. Pediatr Res 2018; 83:1218-1227. [PMID: 29718007 PMCID: PMC6019156 DOI: 10.1038/pr.2018.49] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 03/02/2018] [Indexed: 01/07/2023]
Abstract
BackgroundMaintenance of cholesterol homeostasis is crucial for brain development. Brain cholesterol relies on de novo synthesis and is cleared primarily by conversion to 24S-hydroxycholesterol (24S-HC) with brain-specific cholesterol 24-hydroxylase (CYP46A1). We aimed to investigate the impact of hypoxia-ischemia (HI) on brain cholesterol metabolism in the neonatal mice.MethodsPostnatal day 9 C57BL/6 pups were subjected to HI using the Vannucci model. CYP46A1 expression was assessed with western blotting and its cellular localization was determined using immunofluorescence staining. The amount of brain cholesterol, 24S-HC in the cortex and in the serum, was measured with enzyme-linked immunosorbent assay (ELISA).ResultsThere was a transient cholesterol loss at 6 h after HI. CYP46A1 was significantly upregulated at 6 and 24 h following HI with a concomitant increase of 24S-HC in the ipsilateral cortex and in the serum. The serum levels of 24S-HC correlated with those in the brain, as well as with necrotic and apoptotic cell death evaluated by the expression of spectrin breakdown products and cleaved caspase-3 at 6 and 24 h after HI.ConclusionEnhanced cholesterol turnover by activation of CYP46A1 represents disrupted brain cholesterol homeostasis early after neonatal HI. 24S-HC might be a novel blood biomarker for severity of hypoxic-ischemic encephalopathy with potential clinical application.
Collapse
Affiliation(s)
- Fuxin Lu
- Department of Pediatrics, University of California San Francisco, San Francisco, CA
| | - Jun Zhu
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
| | - Selena Guo
- Dougherty Valley High School, San Ramon, CA
| | | | - Farid F. Chehab
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA
| | - Donna M. Ferriero
- Department of Pediatrics, University of California San Francisco, San Francisco, CA,Department of Neurology, University of California San Francisco, San Francisco, CA
| | - Xiangning Jiang
- Department of Pediatrics, University of California San Francisco, San Francisco, CA,Corresponding author: Xiangning Jiang, Department of Pediatrics, University of California, San Francisco 675 Nelson Rising Lane Room 494, San Francisco, CA 94158 Phone: 415-502-7278 Fax: 415-502-7325
| |
Collapse
|
45
|
Odorcyk FK, Kolling J, Sanches EF, Wyse ATS, Netto CA. Experimental neonatal hypoxia ischemia causes long lasting changes of oxidative stress parameters in the hippocampus and the spleen. J Perinat Med 2018; 46:433-439. [PMID: 28841577 DOI: 10.1515/jpm-2017-0070] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 07/25/2017] [Indexed: 11/15/2022]
Abstract
Neonatal hypoxia ischemia (HI) is the main cause of mortality and morbidity in newborns. The mechanisms involved in its progression start immediately and persist for several days. Oxidative stress and inflammation are determinant factors of the severity of the final lesion. The spleen plays a major part in the inflammatory response to HI. This study assessed the temporal progression of HI-induced alterations in oxidative stress parameters in the hippocampus, the most affected brain structure, and in the spleen. HI was induced in Wistar rat pups in post-natal day 7. Production of reactive oxygen species (ROS), and the activity of the anti oxidant enzyme superoxide dismutase and catalase were assessed 24 h, 96 h and 38 days post-HI. Interestingly, both structures showed a similar pattern, with few alterations in the production of ROS species up to 96 h often combined with an increased activity of the anti oxidant enzymes. However, 38 days after the injury, ROS were at the highest in both structures, coupled with a decrease in the activity of the enzymes. Altogether, present results suggest that HI causes long lasting alterations in the hippocampus as well as in the spleen, suggesting a possible target for delayed treatments for HI.
Collapse
Affiliation(s)
- Felipe Kawa Odorcyk
- Post-graduation Program of Neurosciences, Departamento de Bioquímica, Instituto das Ciências da Saúde (ICBS), Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600 anexo 90035-003, Porto Alegre, RS, Brazil, Tel./Fax: 0055-051 33085568
| | - Janaína Kolling
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Eduardo Farias Sanches
- Post-graduation Program of Neurosciences, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Angela T S Wyse
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carlos Alexandre Netto
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
46
|
Dettman RW, Birch D, Fernando A, Kessler JA, Dizon ML. Targeted Knockdown of Bone Morphogenetic Protein Signaling within Neural Progenitors Protects the Brain and Improves Motor Function following Postnatal Hypoxia-Ischemia. Dev Neurosci 2018; 40:23-38. [PMID: 29324456 PMCID: PMC5861030 DOI: 10.1159/000485379] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 11/15/2017] [Indexed: 11/19/2022] Open
Abstract
Hypoxic-ischemic injury (HI) to the neonatal human brain results in myelin loss that, in some children, can manifest as cerebral palsy. Previously, we had found that neuronal overexpression of the bone morphogenic protein (BMP) inhibitor noggin during development increased oligodendroglia and improved motor function in an experimental model of HI utilizing unilateral common carotid artery ligation followed by hypoxia. As BMPs are known to negatively regulate oligodendroglial fate specification of neural stem cells and alter differentiation of committed oligodendroglia, BMP signaling is likely an important mechanism leading to myelin loss. Here, we showed that BMP signaling is upregulated within oligodendroglia of the neonatal brain. We tested the hypothesis that inhibition of BMP signaling specifically within neural progenitor cells (NPCs) is sufficient to protect oligodendroglia. We conditionally deleted the BMP receptor 2 subtype (BMPR2) in NG2-expressing cells after HI. We found that BMPR2 deletion globally protects the brain as assessed by MRI and protects motor function as assessed by digital gait analysis, and that conditional deletion of BMPR2 maintains oligodendrocyte marker expression by immunofluorescence and Western blot and prevents loss of oligodendroglia. Finally, BMPR2 deletion after HI results in an increase in noncompacted myelin. Thus, our data indicate that inhibition of BMP signaling specifically in NPCs may be a tractable strategy to protect the newborn brain from HI.
Collapse
Affiliation(s)
- Robert W. Dettman
- Developmental Biology Program, Stanley Manne Children’s Research Institute, Chicago, IL, USA
| | - Derin Birch
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Augusta Fernando
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - John A. Kessler
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Maria L.V. Dizon
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
47
|
van Tilborg E, de Theije CGM, van Hal M, Wagenaar N, de Vries LS, Benders MJ, Rowitch DH, Nijboer CH. Origin and dynamics of oligodendrocytes in the developing brain: Implications for perinatal white matter injury. Glia 2017; 66:221-238. [PMID: 29134703 PMCID: PMC5765410 DOI: 10.1002/glia.23256] [Citation(s) in RCA: 183] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/17/2017] [Accepted: 10/20/2017] [Indexed: 12/11/2022]
Abstract
Infants born prematurely are at high risk to develop white matter injury (WMI), due to exposure to hypoxic and/or inflammatory insults. Such perinatal insults negatively impact the maturation of oligodendrocytes (OLs), thereby causing deficits in myelination. To elucidate the precise pathophysiology underlying perinatal WMI, it is essential to fully understand the cellular mechanisms contributing to healthy/normal white matter development. OLs are responsible for myelination of axons. During brain development, OLs are generally derived from neuroepithelial zones, where neural stem cells committed to the OL lineage differentiate into OL precursor cells (OPCs). OPCs, in turn, develop into premyelinating OLs and finally mature into myelinating OLs. Recent studies revealed that OPCs develop in multiple waves and form potentially heterogeneous populations. Furthermore, it has been shown that myelination is a dynamic and plastic process with an excess of OPCs being generated and then abolished if not integrated into neural circuits. Myelination patterns between rodents and humans show high spatial and temporal similarity. Therefore, experimental studies on OL biology may provide novel insights into the pathophysiology of WMI in the preterm infant and offers new perspectives on potential treatments for these patients.
Collapse
Affiliation(s)
- Erik van Tilborg
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Caroline G M de Theije
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Maurik van Hal
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Nienke Wagenaar
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Linda S de Vries
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Manon J Benders
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - David H Rowitch
- Department of Pediatrics, Eli and Edythe Broad Center for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, California.,Department of Paediatrics, Wellcome Trust-MRC Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Cora H Nijboer
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
48
|
Multipotency and therapeutic potential of NG2 cells. Biochem Pharmacol 2017; 141:42-55. [DOI: 10.1016/j.bcp.2017.05.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 05/12/2017] [Indexed: 12/20/2022]
|
49
|
van Tilborg E, Achterberg EJM, van Kammen CM, van der Toorn A, Groenendaal F, Dijkhuizen RM, Heijnen CJ, Vanderschuren LJMJ, Benders MNJL, Nijboer CHA. Combined fetal inflammation and postnatal hypoxia causes myelin deficits and autism-like behavior in a rat model of diffuse white matter injury. Glia 2017; 66:78-93. [PMID: 28925578 PMCID: PMC5724703 DOI: 10.1002/glia.23216] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/16/2017] [Accepted: 08/21/2017] [Indexed: 12/12/2022]
Abstract
Diffuse white matter injury (WMI) is a serious problem in extremely preterm infants, and is associated with adverse neurodevelopmental outcome, including cognitive impairments and an increased risk of autism-spectrum disorders. Important risk factors include fetal or perinatal inflammatory insults and fluctuating cerebral oxygenation. However, the exact mechanisms underlying diffuse WMI are not fully understood and no treatment options are currently available. The use of clinically relevant animal models is crucial to advance knowledge on the pathophysiology of diffuse WMI, allowing the definition of novel therapeutic targets. In the present study, we developed a multiple-hit animal model of diffuse WMI by combining fetal inflammation and postnatal hypoxia in rats. We characterized the effects on white matter development and functional outcome by immunohistochemistry, MRI and behavioral paradigms. Combined fetal inflammation and postnatal hypoxia resulted in delayed cortical myelination, microglia activation and astrogliosis at P18, together with long-term changes in oligodendrocyte maturation as observed in 10 week old animals. Furthermore, rats with WMI showed impaired motor performance, increased anxiety and signs of autism-like behavior, i.e. reduced social play behavior and increased repetitive grooming. In conclusion, the combination of fetal inflammation and postnatal hypoxia in rats induces a pattern of brain injury and functional impairments that closely resembles the clinical situation of diffuse WMI. This animal model provides the opportunity to elucidate pathophysiological mechanisms underlying WMI, and can be used to develop novel treatment options for diffuse WMI in preterm infants.
Collapse
Affiliation(s)
- Erik van Tilborg
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, 3584EA, The Netherlands
| | - E J Marijke Achterberg
- Department of Animals in Science and Society, Division of Behavioural Neuroscience, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CM, The Netherlands
| | - Caren M van Kammen
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, 3584EA, The Netherlands
| | - Annette van der Toorn
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht, 3584 CJ, The Netherlands
| | - Floris Groenendaal
- Department of Neonatology, University Medical Center Utrecht, Utrecht, 3584EA, The Netherlands
| | - Rick M Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht, 3584 CJ, The Netherlands
| | - Cobi J Heijnen
- Laboratory of Neuroimmunology, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, Texas, 77030
| | - Louk J M J Vanderschuren
- Department of Animals in Science and Society, Division of Behavioural Neuroscience, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584CM, The Netherlands
| | - Manon N J L Benders
- Department of Neonatology, University Medical Center Utrecht, Utrecht, 3584EA, The Netherlands
| | - Cora H A Nijboer
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, 3584EA, The Netherlands
| |
Collapse
|
50
|
Sanches EF, Durán-Carabali LE, Tosta A, Nicola F, Schmitz F, Rodrigues A, Siebert C, Wyse A, Netto C. Pregnancy swimming causes short- and long-term neuroprotection against hypoxia-ischemia in very immature rats. Pediatr Res 2017; 82:544-553. [PMID: 28426648 DOI: 10.1038/pr.2017.110] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 03/15/2017] [Indexed: 11/09/2022]
Abstract
BackgroundHypoxia-ischemia (HI) is a major cause of neurological damage in preterm newborn. Swimming during pregnancy alters the offspring's brain development. We tested the effects of swimming during pregnancy in the very immature rat brain.MethodsFemale Wistar rats (n=12) were assigned to the sedentary (SE, n=6) or the swimming (SW, n=6) group. From gestational day 0 (GD0) to GD21 the rats in the SW group were made to swim for 20 min/day. HI on postnatal day (PND) 3 rats caused sensorimotor and cognitive impairments. Animals were distributed into SE sham (SESH), sedentary HIP3 (SEHI), swimming sham (SWSH), and swimming HIP3 (SWHI) groups. At PND4 and PND5, Na+/K+-ATPase activity and brain-derived neurotrophic factor (BDNF) levels were assessed. During lactation and adulthood, neurological reflexes, sensorimotor, anxiety-related, and cognitive evaluations were made, followed by histological assessment at PND60.ResultsAt early stages, swimming caused an increase in hippocampal BDNF levels and in the maintenance of Na+/K+-ATPase function in the SWHI group. The SWHI group showed smaller lesions and the preservation of white matter tracts. SEHI animals showed a delay in reflex maturation, which was reverted in the SWHI group. HIP3 induced spatial memory deficits and hypomyelination in SEHI rats, which was reverted in the SWHI group.ConclusionSwimming during pregnancy neuroprotected the brains against HI in very immature neonatal rats.
Collapse
Affiliation(s)
- Eduardo Farias Sanches
- Post-Graduation Program of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Luz Elena Durán-Carabali
- Post-Graduation Program of Phisiology, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Andrea Tosta
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fabrício Nicola
- Post-Graduation Program of Neurosciences, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Felipe Schmitz
- Post-Graduation Program of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - André Rodrigues
- Post-Graduation Program of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Cassiana Siebert
- Post-Graduation Program of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Angela Wyse
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Carlos Netto
- Department of Biochemistry, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|