1
|
Lear CA, Dhillon SK, Nakao M, Lear BA, Georgieva A, Ugwumadu A, Stone PR, Bennet L, Gunn AJ. The peripheral chemoreflex and fetal defenses against intrapartum hypoxic-ischemic brain injury at term gestation. Semin Fetal Neonatal Med 2024:101543. [PMID: 39455374 DOI: 10.1016/j.siny.2024.101543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2024]
Abstract
Fetal hypoxemia is ubiquitous during labor and, when severe, is associated with perinatal death and long-term neurodevelopmental disability. Adverse outcomes are highly associated with barriers to care, such that developing countries have a disproportionate burden of perinatal injury. The prevalence of hypoxemia and its link to injury can be obscure, simply because the healthy fetus has robust coordinated defense mechanisms, spearheaded by the peripheral chemoreflex, such that hypoxemia only becomes apparent in the minority of cases associated with stillbirth, severe metabolic acidemia or adverse neurodevelopmental outcomes. This represents only the extreme end of the spectrum, when defense mechanisms have failed due to severe/prolonged hypoxemia, or the fetal defenses are compromised by additional risk factors. Understanding the fetal defenses to hypoxemia and when the fetus begins to decompensate is crucial to understanding perinatal health and disease, by linking antenatal health, intrapartum events, the neonatal trajectory and ultimately life-long neurodevelopmental health.
Collapse
Affiliation(s)
- Christopher A Lear
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand; Auckland City Hospital, Auckland, New Zealand.
| | - Simerdeep K Dhillon
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Masahiro Nakao
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand; Department of Obstetrics and Gynecology, Mie University Graduate School of Medicine, Mie, Japan
| | - Benjamin A Lear
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Antoniya Georgieva
- Nuffield Department of Women's and Reproductive Health, The John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Austin Ugwumadu
- Department of Obstetrics and Gynaecology, St George's Hospital, London, United Kingdom
| | - Peter R Stone
- Department of Obstetrics and Gynaecology, The University of Auckland, Auckland, New Zealand
| | - Laura Bennet
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand
| | - Alistair J Gunn
- Fetal Physiology and Neuroscience Group, Department of Physiology, The University of Auckland, Auckland, New Zealand; Starship Children's Hospital, Auckland, New Zealand
| |
Collapse
|
2
|
Vanderhasselt T, Naeyaert M, Buls N, Allemeersch GJ, Raeymaeckers S, Raeymaekers H, Smeets N, Cools F, de Mey J, Dudink J. Synthetic magnetic resonance-based relaxometry and brain volume: cutoff values for predicting neurocognitive outcomes in very preterm infants. Pediatr Radiol 2024; 54:1523-1531. [PMID: 38980354 PMCID: PMC11324712 DOI: 10.1007/s00247-024-05981-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/20/2024] [Accepted: 06/23/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND Early neurorehabilitation can enhance neurocognitive outcomes in very preterm infants (<32 weeks), and conventional magnetic resonance imaging (MRI) is commonly used to assess neonatal brain injury; however, the predictive value for neurodevelopmental delay is limited. Timely predictive quantitative biomarkers are needed to improve early identification and management of infants at risk of neurodevelopmental delay. OBJECTIVE To evaluate the potential of quantitative synthetic MRI measurements at term-equivalent age as predictive biomarkers of neurodevelopmental impairment and establish practical cutoff values to guide clinical decision-making. MATERIALS AND METHODS This retrospective study included 93 very preterm infants who underwent synthetic MRI at term-equivalent age between January 2017 and September 2020. Clinical outcomes were assessed using the Bayley-III scale of infant development (mean age 2.1 years). The predictive value for impaired development was analyzed using receiver operating characteristic curves for synthetic MRI-based volumetry and T1 and T2 relaxation measurements. RESULTS The T1 relaxation time in the posterior limb of the internal capsule was a potent predictor of severe (sensitivity, 92%; specificity, 80%; area under the curve (AUC), 0.91) and mild or severe (AUC, 0.75) developmental impairment. T2 relaxation time in the posterior limb of the internal capsule was a significant predictor of severe impairment (AUC, 0.76), whereas the brain parenchymal volume was a significant predictor of severe (AUC, 0.72) and mild or severe impairment (AUC, 0.71) outperforming the reported qualitative MRI scores (AUC, 0.66). CONCLUSION The proposed cutoff values for T1 relaxation time in the posterior limb of the internal capsule and for total brain volume measurements, derived from synthetic MRI, show promise as predictors of both mild and severe neurodevelopmental impairment in very preterm infants.
Collapse
Affiliation(s)
- Tim Vanderhasselt
- Department of Radiology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium.
| | - Maarten Naeyaert
- Department of Radiology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Nico Buls
- Department of Radiology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Gert-Jan Allemeersch
- Department of Radiology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Steven Raeymaeckers
- Department of Radiology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Hubert Raeymaekers
- Department of Radiology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Nathalie Smeets
- Department of Pediatric Neurology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Filip Cools
- Department of Neonatology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Johan de Mey
- Department of Radiology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Jeroen Dudink
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
3
|
Tran NT, Penny TR, Chan KY, Tang T, Papagianis PC, Sepehrizadeh T, Nekkanti L, Zahra VA, Pham Y, Yawno T, Nitsos I, Kelly SB, Thiel AM, de Veer M, Alahmari DM, Fahey MC, Jenkin G, Miller SL, Galinsky R, Polglase GR, McDonald CA. Early administration of umbilical cord blood cells following brief high tidal volume ventilation in preterm sheep: a cautionary tale. J Neuroinflammation 2024; 21:121. [PMID: 38720368 PMCID: PMC11077893 DOI: 10.1186/s12974-024-03053-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 02/23/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Umbilical cord blood (UCB) cells are a promising treatment for preterm brain injury. Access to allogeneic sources of UCB cells offer the potential for early administration to optimise their therapeutic capacities. As preterm infants often require ventilatory support, which can contribute to preterm brain injury, we investigated the efficacy of early UCB cell administration following ventilation to reduce white matter inflammation and injury. METHODS Preterm fetal sheep (0.85 gestation) were randomly allocated to no ventilation (SHAM; n = 5) or 15 min ex utero high tidal volume ventilation. One hour following ventilation, fetuses were randomly allocated to i.v. administration of saline (VENT; n = 7) or allogeneic term-derived UCB cells (24.5 ± 5.0 million cells/kg; VENT + UCB; n = 7). Twenty-four hours after ventilation, lambs were delivered for magnetic resonance imaging and post-mortem brain tissue collected. Arterial plasma was collected throughout the experiment for cytokine analyses. To further investigate the results from the in vivo study, mononuclear cells (MNCs) isolated from human UCB were subjected to in vitro cytokine-spiked culture medium (TNFα and/or IFNγ; 10 ng/mL; n = 3/group) for 16 h then supernatant and cells collected for protein and mRNA assessments respectively. RESULTS In VENT + UCB lambs, systemic IFNγ levels increased and by 24 h, there was white matter neuroglial activation, vascular damage, reduced oligodendrocytes, and increased average, radial and mean diffusivity compared to VENT and SHAM. No evidence of white matter inflammation or injury was present in VENT lambs, except for mRNA downregulation of OCLN and CLDN1 compared to SHAM. In vitro, MNCs subjected to TNFα and/or IFNγ displayed both pro- and anti-inflammatory characteristics indicated by changes in cytokine (IL-18 & IL-10) and growth factor (BDNF & VEGF) gene and protein expression compared to controls. CONCLUSIONS UCB cells administered early after brief high tidal volume ventilation in preterm fetal sheep causes white matter injury, and the mechanisms underlying these changes are likely dysregulated responses of the UCB cells to the degree of injury/inflammation already present. If immunomodulatory therapies such as UCB cells are to become a therapeutic strategy for preterm brain injury, especially after ventilation, our study suggests that the inflammatory state of the preterm infant should be considered when timing UCB cells administration.
Collapse
Affiliation(s)
- Nhi T Tran
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia.
| | - Tayla R Penny
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Kyra Yy Chan
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Tanya Tang
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Paris C Papagianis
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Tara Sepehrizadeh
- Monash Biomedical Imaging, Monash University, Clayton, VIC, Australia
| | - Lakshmi Nekkanti
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Valerie A Zahra
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Yen Pham
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Tamara Yawno
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
- Department of Paediatrics, Monash University, Clayton, VIC, Australia
| | - Ilias Nitsos
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Sharmony B Kelly
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Alison M Thiel
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Michael de Veer
- Monash Biomedical Imaging, Monash University, Clayton, VIC, Australia
| | - Dhafer M Alahmari
- Monash Biomedical Imaging, Monash University, Clayton, VIC, Australia
- Monash Biomedicine Discovery Institute and Department of Medical Imaging and Radiation Sciences, Monash University, Clayton, VIC, Australia
- Department of Diagnostic Imaging, Kind Saud Medical City, Riyadh, Saudi Arabia
| | - Michael C Fahey
- Department of Paediatrics, Monash University, Clayton, VIC, Australia
| | - Graham Jenkin
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Suzanne L Miller
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Robert Galinsky
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
| | - Graeme R Polglase
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia.
| | - Courtney A McDonald
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia.
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
4
|
Deferm W, Tang T, Moerkerke M, Daniels N, Steyaert J, Alaerts K, Ortibus E, Naulaers G, Boets B. Subtle microstructural alterations in white matter tracts involved in socio-emotional processing after very preterm birth. Neuroimage Clin 2024; 41:103580. [PMID: 38401459 PMCID: PMC10944182 DOI: 10.1016/j.nicl.2024.103580] [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: 12/18/2023] [Revised: 02/10/2024] [Accepted: 02/10/2024] [Indexed: 02/26/2024]
Abstract
Children born very preterm (VPT, < 32 weeks of gestation) have an increased risk of developing socio-emotional difficulties. Possible neural substrates for these socio-emotional difficulties are alterations in the structural connectivity of the social brain due to premature birth. The objective of the current study was to study microstructural white matter integrity in VPT versus full-term (FT) born school-aged children along twelve white matter tracts involved in socio-emotional processing. Diffusion MRI scans were obtained from a sample of 35 VPT and 38 FT 8-to-12-year-old children. Tractography was performed using TractSeg, a state-of-the-art neural network-based approach, which offers investigation of detailed tract profiles of fractional anisotropy (FA). Group differences in FA along the tracts were investigated using both a traditional and complementary functional data analysis approach. Exploratory correlations were performed between the Social Responsiveness Scale (SRS-2), a parent-report questionnaire assessing difficulties in social functioning, and FA along the tract. Both analyses showed significant reductions in FA for the VPT group along the middle portion of the right SLF I and an anterior portion of the left SLF II. These group differences possibly indicate altered white matter maturation due to premature birth and may contribute to altered functional connectivity in the Theory of Mind network which has been documented in earlier work with VPT samples. Apart from reduced social motivation in the VPT group, there were no significant group differences in reported social functioning, as assessed by SRS-2. We found that in the VPT group higher FA values in segments of the left SLF I and right SLF II were associated with better social functioning. Surprisingly, the opposite was found for segments in the right IFO, where higher FA values were associated with worse reported social functioning. Since no significant correlations were found for the FT group, this relationship may be specific for VPT children. The current study overcomes methodological limitations of previous studies by more accurately segmenting white matter tracts using constrained spherical deconvolution based tractography, by applying complementary tractometry analysis approaches to estimate changes in FA more accurately, and by investigating the FA profile along the three components of the SLF.
Collapse
Affiliation(s)
- Ward Deferm
- Center for Developmental Psychiatry, KU Leuven, Belgium.
| | - Tiffany Tang
- Center for Developmental Psychiatry, KU Leuven, Belgium
| | | | - Nicky Daniels
- Neuromotor Rehabilitation Research Group, KU Leuven, Belgium
| | - Jean Steyaert
- Center for Developmental Psychiatry, KU Leuven, Belgium; Child Psychiatry, UZ Leuven, Belgium
| | - Kaat Alaerts
- Neuromotor Rehabilitation Research Group, KU Leuven, Belgium
| | | | - Gunnar Naulaers
- Neonatal Intensive Care Unit - Neonatology, UZ Leuven, Belgium; UZ Leuven & Center for Developmental Disorders, Belgium
| | - Bart Boets
- Center for Developmental Psychiatry, KU Leuven, Belgium
| |
Collapse
|
5
|
Chao AS, Matak P, Pegram K, Powers J, Hutson C, Jo R, Dubois L, Thompson JW, Smith PB, Jain V, Liu C, Younge NE, Rikard B, Reyes EY, Shinohara ML, Gregory SG, Goldberg RN, Benner EJ. 20-αHydroxycholesterol, an oxysterol in human breast milk, reverses mouse neonatal white matter injury through Gli-dependent oligodendrogenesis. Cell Stem Cell 2023; 30:1054-1071.e8. [PMID: 37541211 PMCID: PMC10625465 DOI: 10.1016/j.stem.2023.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 05/21/2023] [Accepted: 07/12/2023] [Indexed: 08/06/2023]
Abstract
White matter injuries (WMIs) are the leading cause of neurologic impairment in infants born premature. There are no treatment options available. The most common forms of WMIs in infants occur prior to the onset of normal myelination, making its pathophysiology distinctive, thus requiring a tailored approach to treatment. Neonates present a unique opportunity to repair WMIs due to a transient abundance of neural stem/progenitor cells (NSPCs) present in the germinal matrix with oligodendrogenic potential. We identified an endogenous oxysterol, 20-αHydroxycholesterol (20HC), in human maternal breast milk that induces oligodendrogenesis through a sonic hedgehog (shh), Gli-dependent mechanism. Following WMI in neonatal mice, injection of 20HC induced subventricular zone-derived oligodendrogenesis and improved myelination in the periventricular white matter, resulting in improved motor outcomes. Targeting the oligodendrogenic potential of postnatal NSPCs in neonates with WMIs may be further developed into a novel approach to mitigate this devastating complication of preterm birth.
Collapse
Affiliation(s)
- Agnes S Chao
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Pavle Matak
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Kelly Pegram
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - James Powers
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Collin Hutson
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Rebecca Jo
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Laura Dubois
- Duke Proteomics and Metabolomics Shared Resource, Center for Genomics and Computational Biology, School of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - J Will Thompson
- Duke Proteomics and Metabolomics Shared Resource, Center for Genomics and Computational Biology, School of Medicine, Duke University Medical Center, Durham, NC 27710, USA; Department of Pharmacology and Cancer Biology, School of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - P Brian Smith
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Vaibhav Jain
- Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA; Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Chunlei Liu
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Noelle E Younge
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Blaire Rikard
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Estefany Y Reyes
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Mari L Shinohara
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Simon G Gregory
- Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA; Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Ronald N Goldberg
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA
| | - Eric J Benner
- Division of Neonatology, Department of Pediatrics, Duke University Medical Center, The Jean and George Brumley, Jr. Neonatal-Perinatal Institute, Durham, NC 27710, USA; Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
6
|
Marchman VA, Ashland MD, Loi EC, Munévar M, Shannon KA, Fernald A, Feldman HM. Associations between early efficiency in language processing and language and cognitive outcomes in children born full term and preterm: similarities and differences. Child Neuropsychol 2023; 29:886-905. [PMID: 36324057 PMCID: PMC10151433 DOI: 10.1080/09297049.2022.2138304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022]
Abstract
Associations between children's early language processing efficiency and later verbal and non-verbal outcomes shed light on the extent to which early information processing skills support later learning across different domains of function. Examining whether the strengths of associations are similar in typically developing and at-risk populations provides an additional lens into the varying routes to learning that children may take across development. In this follow-up study, children born full-term (FT, n = 49) and preterm (PT, n = 45, ≤32 weeks gestational age, birth weight <1800 g) were assessed in the Looking While Listening (LWL) task at 18 months (corrected for degree of prematurity in PT group). This eye-tracking task assesses efficiency of real-time spoken language comprehension as accuracy and speed (RT) of processing. At 4 ½ years, children were assessed on standardized tests of receptive vocabulary, expressive language, and non-verbal IQ. Language processing efficiency was associated with both language outcomes (r2-change: 7.0-19.7%, p < 0.01), after covariates. Birth group did not moderate these effects, suggesting similar mechanisms of learning in these domains for PT and FT children. However, birth group moderated the association between speed and non-verbal IQ (r2-change: 4.5%, p < 0.05), such that an association was found in the PT but not the FT group. This finding suggests that information processing skills reflected in efficiency of real-time language processing may be recruited to support learning in a broader range of verbal and non-verbal domains in the PT compared to the FT group.
Collapse
Affiliation(s)
- Virginia A. Marchman
- Department of Psychology, 450 Jane Stanford Way, Stanford University, Stanford, CA 94305, USA
- Division of Developmental-Behavioral Pediatrics, Department of Pediatrics, Stanford University School of Medicine, 3145 Porter Drive, Palo Alto, CA 94304, USA
| | - Melanie D. Ashland
- Department of Psychology, 450 Jane Stanford Way, Stanford University, Stanford, CA 94305, USA
| | - Elizabeth C. Loi
- Division of Developmental-Behavioral Pediatrics, Department of Pediatrics, Stanford University School of Medicine, 3145 Porter Drive, Palo Alto, CA 94304, USA
| | - Mónica Munévar
- Department of Psychology, 450 Jane Stanford Way, Stanford University, Stanford, CA 94305, USA
| | - Katherine A. Shannon
- Department of Psychology, 450 Jane Stanford Way, Stanford University, Stanford, CA 94305, USA
| | - Anne Fernald
- Department of Psychology, 450 Jane Stanford Way, Stanford University, Stanford, CA 94305, USA
| | - Heidi M. Feldman
- Division of Developmental-Behavioral Pediatrics, Department of Pediatrics, Stanford University School of Medicine, 3145 Porter Drive, Palo Alto, CA 94304, USA
| |
Collapse
|
7
|
Yuzkan S, Emecen Sanli M, Balci M, Cennetoglu P, Kafadar I, Kocak B. Use of Thalamus L-Sign to Differentiate Periventricular Leukomalacia From Neurometabolic Disorders. J Child Neurol 2023; 38:446-453. [PMID: 37128731 DOI: 10.1177/08830738231168973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
PURPOSE To assess the diagnostic value of the thalamus L-sign on magnetic resonance imaging (MRI) in distinguishing between periventricular leukomalacia and neurometabolic disorders in pediatric patients. METHODS In this retrospective study, clinical and imaging information was collected from 50 children with periventricular leukomalacia and 52 children with neurometabolic disorders. MRI was used to evaluate the L-sign of the thalamus (ie, injury to the posterolateral thalamus) and the lobar distribution of signal intensity changes. Age, sex, gestational age, and level of Gross Motor Function Classification System (only for periventricular leukomalacia) constituted the clinical parameters. Statistical evaluation of group differences for imaging and clinical variables were conducted using univariable statistical methods. The intra- and inter-observer agreement was evaluated using Cohen's kappa. Univariable or multivariable logistic regression was employed for selection of variables, determining independent predictors, and modeling. RESULTS The thalamus L-sign was observed in 70% (35/50) of patients in the periventricular leukomalacia group, but in none of the patients with neurometabolic disorder (P < .001). The gestational age between groups varied significantly (P < .001). Involvement of frontal, parietal, and occipital lobes differed significantly between groups (P < .001). In the logistic regression, the best model included negative thalamus L-sign and gestational age, yielding an area under the curve, accuracy, sensitivity, specificity, and precision values of 0.995, 96.1%, 96%, 96.2%, and 96%, respectively. Both the lack of thalamus L-sign and gestational age were independent predictors (P < .001). CONCLUSIONS The thalamus L-sign and gestational age may be useful in distinguishing between periventricular leukomalacia and neurometabolic disorders.
Collapse
Affiliation(s)
- Sabahattin Yuzkan
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey
| | - Merve Emecen Sanli
- Department of Pediatric Inherited Metabolic Diseases, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey
| | - Merve Balci
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey
| | - Pakize Cennetoglu
- Department of Pediatric Neurology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey
| | - Ihsan Kafadar
- Department of Pediatric Neurology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey
| | - Burak Kocak
- Department of Radiology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital, Basaksehir, Istanbul, Turkey
| |
Collapse
|
8
|
Torres Y, Celis C, Acurio J, Escudero C. Language Impairment in Children of Mothers with Gestational Diabetes, Preeclampsia, and Preterm Delivery: Current Hypothesis and Potential Underlying Mechanisms : Language Impartment and Pregnancy Complications. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:245-267. [PMID: 37466777 DOI: 10.1007/978-3-031-32554-0_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Many conditions may impair or delay language development, including socioeconomic status, parent's education, or intrauterine environment. Accordingly, increasing evidence has described that pregnancy complications, including gestational diabetes mellitus (GDM), preeclampsia, and preterm delivery, are associated with the offspring's impaired neurodevelopment. Since language is one of the high brain functions, alterations in this function are another sign of neurodevelopment impairment. How these maternal conditions may generate language impairment has yet to be entirely understood. However, since language development requires adequate structural formation and function/connectivity of the brain, these processes must be affected by alterations in maternal conditions. However, the underlying mechanisms of these structural alterations are largely unknown. This manuscript critically analyzes the literature focused on the risk of developing language impairment in children of mothers with GDM, preeclampsia, and preterm delivery. Furthermore, we highlight potential underlying molecular mechanisms associated with these alterations, such as neuroinflammatory and metabolic and cerebrovascular alterations.
Collapse
Affiliation(s)
- Yesenia Torres
- Vascular Physiology Laboratory, Department of Basic Science, Faculty of Sciences, Universidad of Bio Bio, Chillán, Chile
- Brainlab-Cognitive Neuroscience Research Group, Department of Clinical Psychology and Psychobiology, University of Barcelona, Barcelona, Catalonia, Spain
| | - Cristian Celis
- Vascular Physiology Laboratory, Department of Basic Science, Faculty of Sciences, Universidad of Bio Bio, Chillán, Chile
- Centro terapéutico , ABCfonoaudiologia, Santiago, Chile
| | - Jesenia Acurio
- Vascular Physiology Laboratory, Department of Basic Science, Faculty of Sciences, Universidad of Bio Bio, Chillán, Chile
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Science, Faculty of Sciences, Universidad of Bio Bio, Chillán, Chile.
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile.
| |
Collapse
|
9
|
The Role of Melatonin in Pregnancy and the Health Benefits for the Newborn. Biomedicines 2022; 10:biomedicines10123252. [PMID: 36552008 PMCID: PMC9775355 DOI: 10.3390/biomedicines10123252] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/08/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
In the last few years, there have been significant evolutions in the understanding of the hormone melatonin in terms of its physiology, regulatory role, and potential utility in various domains of clinical medicine. Melatonin's properties include, among others, the regulation of mitochondrial function, anti-inflammatory, anti-oxidative and neuro-protective effects, sleep promotion and immune enhancement. As it is also bioavailable and has little or no toxicity, it has been proposed as safe and effective for the treatment of numerous diseases and to preserve human health. In this manuscript, we tried to evaluate the role of melatonin at the beginning of human life, in pregnancy, in the fetus and in newborns through newly published literature studies.
Collapse
|
10
|
Jang YH, Kim H, Lee JY, Ahn JH, Chung AW, Lee HJ. Altered development of structural MRI connectome hubs at near-term age in very and moderately preterm infants. Cereb Cortex 2022; 33:5507-5523. [PMID: 36408630 DOI: 10.1093/cercor/bhac438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 11/22/2022] Open
Abstract
Abstract
Preterm infants may exhibit altered developmental patterns of the brain structural network by endogenous and exogenous stimuli, which are quantifiable through hub and modular network topologies that develop in the third trimester. Although preterm brain networks can compensate for white matter microstructural abnormalities of core connections, less is known about how the network developmental characteristics of preterm infants differ from those of full-term infants. We identified 13 hubs and 4 modules and revealed subtle differences in edgewise connectivity and local network properties between 134 preterm and 76 full-term infants, identifying specific developmental patterns of the brain structural network in preterm infants. The modules of preterm infants showed an imbalanced composition. The edgewise connectivity in preterm infants showed significantly decreased long- and short-range connections and local network properties in the dorsal superior frontal gyrus. In contrast, the fusiform gyrus and several nonhub regions showed significantly increased wiring of short-range connections and local network properties. Our results suggested that decreased local network in the frontal lobe and excessive development in the occipital lobe may contribute to the understanding of brain developmental deviances in preterm infants.
Collapse
Affiliation(s)
- Yong Hun Jang
- Hanyang University Graduate School of Biomedical Science and Engineering Department of Translational Medicine, , Seoul 04763 , Republic of Korea
| | - Hyuna Kim
- Hanyang University Graduate School of Biomedical Science and Engineering Department of Translational Medicine, , Seoul 04763 , Republic of Korea
| | - Joo Young Lee
- Hanyang University Graduate School of Biomedical Science and Engineering Department of Translational Medicine, , Seoul 04763 , Republic of Korea
| | - Ja-Hye Ahn
- Hanyang University College of Medicine Department of Pediatrics, Hanyang University Hospital, , Seoul 04763 , Republic of Korea
| | - Ai Wern Chung
- Harvard Medical School Fetal Neonatal-Neuroimaging and Developmental Science Center, Boston Children’s Hospital, , Boston, MA 02115 , USA
- Harvard Medical School Department of Pediatrics, Boston Children’s Hospital, , Boston, MA 02115 , USA
| | - Hyun Ju Lee
- Hanyang University College of Medicine Department of Pediatrics, Hanyang University Hospital, , Seoul 04763 , Republic of Korea
| |
Collapse
|
11
|
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
|
12
|
Gonçalves FG, Freeman C, Khrichenko D, Hwang M. Quantitative Evaluation of Brain Echogenicity in Hypoxic-Ischemic Encephalopathy in Term Neonates Compared with Controls. Ultrasound Int Open 2022; 8:E43-E52. [PMID: 36408372 PMCID: PMC9668510 DOI: 10.1055/a-1958-3985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 08/07/2022] [Indexed: 11/18/2022] Open
Abstract
Purpose Neurosonography evaluation of neonatal hypoxic-ischemic encephalopathy (HIE) is mainly qualitative. We aimed to quantitatively compare the echogenicity of several brain regions in patients with HIE to healthy controls. Materials and Methods 20 term neonates with clinical/MRI evidence of HIE and 20 term healthy neonates were evaluated. Seven brain regions were assessed [frontal, parietal, occipital, and perirolandic white matter (WM), caudate nucleus head, lentiform nucleus, and thalamus]. The echogenicity of the calvarial bones (bone) and the choroid plexus (CP) was used for ratio calculation. Differences in the ratios were determined between neonates with HIE and controls. Results Ratios were significantly higher for HIE neonates in each region (p<0.05). The differences were greatest for the perirolandic WM, with CP and bone ratios being 0.23 and 0.22 greater, respectively, for the HIE compared to the healthy neonates (p<0.001). The perirolandic WM had a high AUC, at 0.980 for both the CP and bone ratios. The intra-observer reliability for all ratios was high, with the caudate to bone ratio being the lowest at 0.832 and the anterior WM to CP ratio being the highest at 0.992. Conclusion When coupled with internal controls, quantitative neurosonography represents a potential tool to identify early neonatal HIE changes. Larger cohort studies could reveal whether a quantitative approach can discern between degrees of severity of HIE. Future neurosonography protocols should be tailored to evaluate the perirolandic region, which requires posterior coronal scanning.
Collapse
Affiliation(s)
| | - Colbey Freeman
- Radiology, University of Pennsylvania Perelman School of Medicine,
Philadelphia, United States
| | - Dmitry Khrichenko
- Radiology, The Children’s Hospital of Philadelphia,
Philadelphia, United States
| | - Misun Hwang
- Radiology, The Children’s Hospital of Philadelphia,
Philadelphia, United States
| |
Collapse
|
13
|
Beeraka NM, Vikram PRH, Greeshma MV, Uthaiah CA, Huria T, Liu J, Kumar P, Nikolenko VN, Bulygin KV, Sinelnikov MY, Sukocheva O, Fan R. Recent Investigations on Neurotransmitters' Role in Acute White Matter Injury of Perinatal Glia and Pharmacotherapies-Glia Dynamics in Stem Cell Therapy. Mol Neurobiol 2022; 59:2009-2026. [PMID: 35041139 DOI: 10.1007/s12035-021-02700-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 12/10/2021] [Indexed: 02/05/2023]
Abstract
Periventricular leukomalacia (PVL) and cerebral palsy are two neurological disease conditions developed from the premyelinated white matter ischemic injury (WMI). The significant pathophysiology of these diseases is accompanied by the cognitive deficits due to the loss of function of glial cells and axons. White matter makes up 50% of the brain volume consisting of myelinated and non-myelinated axons, glia, blood vessels, optic nerves, and corpus callosum. Studies over the years have delineated the susceptibility of white matter towards ischemic injury especially during pregnancy (prenatal, perinatal) or immediately after child birth (postnatal). Impairment in membrane depolarization of neurons and glial cells by ischemia-invoked excitotoxicity is mediated through the overactivation of NMDA receptors or non-NMDA receptors by excessive glutamate influx, calcium, or ROS overload and has been some of the well-studied molecular mechanisms conducive to the injury of white matter. Expression of glutamate receptors (GluR) and transporters (GLT1, EACC1, and GST) has significant influence in glial and axonal-mediated injury of premyelinated white matter during PVL and cerebral palsy. Predominantly, the central premyelinated axons express extensive levels of functional NMDA GluR receptors to confer ischemic injury to premyelinated white matter which in turn invoke defects in neural plasticity. Several underlying molecular mechanisms are yet to be unraveled to delineate the complete pathophysiology of these prenatal neurological diseases for developing the novel therapeutic modalities to mitigate pathophysiology and premature mortality of newborn babies. In this review, we have substantially discussed the above multiple pathophysiological aspects of white matter injury along with glial dynamics, and the pharmacotherapies including recent insights into the application of MSCs as therapeutic modality in treating white matter injury.
Collapse
Affiliation(s)
- Narasimha M Beeraka
- Cancer Center, Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, People's Republic of China
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research (JSS AHER), Mysuru, Karnataka, India
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
| | - P R Hemanth Vikram
- Department of Pharmaceutical Chemistry, JSS Pharmacy College, Mysuru, Karnataka, India
| | - M V Greeshma
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research (JSS AHER), Mysuru, Karnataka, India
| | - Chinnappa A Uthaiah
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education and Research (JSS AHER), Mysuru, Karnataka, India
| | - Tahani Huria
- Faculty of Medicine, Benghazi University, Benghazi, Libya
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, LE1 7RH, UK
| | - Junqi Liu
- Cancer Center, Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, People's Republic of China
| | - Pramod Kumar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER-Guwahati), SilaKatamur (Halugurisuk), Changsari, Kamrup, 781101, Assam, India
| | - Vladimir N Nikolenko
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
- Department of Normal and Topographic Anatomy, Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Kirill V Bulygin
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
| | - Mikhail Y Sinelnikov
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University (Sechenov University), St. Trubetskaya, 8, bld. 2, Moscow, 119991, Russia
- Research Institute of Human Morphology, 3 Tsyurupy Street, Moscow, 117418, Russian Federation
| | - Olga Sukocheva
- Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University, Bedford Park, South Australia, 5042, Australia
| | - Ruitai Fan
- Cancer Center, Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, People's Republic of China.
| |
Collapse
|
14
|
Hwang M, Tierradentro-García LO, Hussaini SH, Cajigas-Loyola SC, Kaplan SL, Otero HJ, Bellah RD. Ultrasound imaging of preterm brain injury: fundamentals and updates. Pediatr Radiol 2022; 52:817-836. [PMID: 34648071 DOI: 10.1007/s00247-021-05191-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/22/2021] [Accepted: 08/12/2021] [Indexed: 11/25/2022]
Abstract
Neurosonography has become an essential tool for diagnosis and serial monitoring of preterm brain injury. Preterm infants are at significantly higher risk of hypoxic-ischemic injury, intraventricular hemorrhage, periventricular leukomalacia and post-hemorrhagic hydrocephalus. Neonatologists have become increasingly dependent on neurosonography to initiate medical and surgical interventions because it can be used at the bedside. While brain MRI is regarded as the gold standard for detecting preterm brain injury, neurosonography offers distinct advantages such as its cost-effectiveness, diagnostic utility and convenience. Neurosonographic signatures associated with poor long-term outcomes shape decisions regarding supportive care, medical or behavioral interventions, and family members' expectations. Within the last decade substantial progress has been made in neurosonography techniques, prompting an updated review of the topic. In addition to the up-to-date summary of neurosonography, this review discusses the potential roles of emerging neurosonography techniques that offer new functional insights into the brain, such as superb microvessel imaging, elastography, three-dimensional ventricular volume assessment, and contrast-enhanced US.
Collapse
Affiliation(s)
- Misun Hwang
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA.
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Luis O Tierradentro-García
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA
| | - Syed H Hussaini
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephanie C Cajigas-Loyola
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Summer L Kaplan
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hansel J Otero
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Richard D Bellah
- Department of Radiology, Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
15
|
Temporal Characterization of Microglia-Associated Pro- and Anti-Inflammatory Genes in a Neonatal Inflammation-Sensitized Hypoxic-Ischemic Brain Injury Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2479626. [PMID: 35281473 PMCID: PMC8906938 DOI: 10.1155/2022/2479626] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/22/2021] [Accepted: 01/27/2022] [Indexed: 02/06/2023]
Abstract
Hypoxic-ischemic encephalopathy (HIE) mainly affects preterm and term newborns, leading to a high risk of brain damage. Coexisting infection/inflammation and birth asphyxia are key factors associated with intracerebral increase of proinflammatory cytokines linked to HIE. Microglia are key mediators of inflammation during perinatal brain injury, characterized by their phenotypic plasticity, which may facilitate their participation in both the progression and resolution of injury-induced inflammation. The purpose of this study was to investigate the temporal expression of genes associated with pro- and anti-inflammatory cytokines as well as the nucleotide-binding domain, leucine-rich repeat protein (NLRP-3) inflammasome from microglia cells. For this purpose, we used our established neonatal rat model of inflammation-sensitized hypoxic-ischemic (HI) brain injury in seven-day-old rats. We assessed gene expression profiles of 11 cytokines and for NLRP-3 using real-time PCR from sorted CD11b/c microglia of brain samples at different time points (3.5 h after LPS injection and 0, 5, 24, 48, and 72 hours post HI) following different treatments: vehicle, E. coli lipopolysaccharide (LPS), vehicle/HI, and LPS/HI. Our results showed that microglia are early key mediators of the inflammatory response and exacerbate the inflammatory response following HI, polarizing into a predominant proinflammatory M1 phenotype in the early hours post HI. The brains only exposed to HI showed a delay in the expression of proinflammatory cytokines. We also demonstrated that NLRP-3 plays a role in the inflammatory resolution with a high expression after HI insult. The combination of both, a preinfection/inflammation condition and hypoxia-ischemia, resulted in a higher proinflammatory cytokine storm, highlighting the significant contribution of acute inflammation sensitizing prior to a hypoxic insult on the severity of perinatal brain damage.
Collapse
|
16
|
Sun X, Xue F, Wen J, Gao L, Li Y, Jiang Q, Yang L, Cui H. Seizure Characteristics and Background Amplitude-Integrated Electroencephalography Activity in Neonatal Rats Subjected to Hypoxia-Ischemia. Front Pediatr 2022; 10:837909. [PMID: 35463911 PMCID: PMC9021695 DOI: 10.3389/fped.2022.837909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/14/2022] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE Perinatal hypoxic-ischemic encephalopathy (HIE) is a major cause of epilepsy and chronic neurologic morbidity in premature infants. This study aimed to investigate the characteristics of acute seizures and the pattern of background activity on amplitude-integrated electroencephalography (aEEG) in neonatal rats with HIE. METHODS Hypoxia-ischemia (HI) was induced in postnatal day (P) 3 neonatal rats (n = 12) by ligation of the left carotid artery and exposure to airtight hypoxia for 2 h. Data regarding seizure type, frequency, and duration and those related to neurobehavioral development were collected, and the integrated power of background EEG was analyzed to evaluate the effect of HI. RESULTS All neonatal rats in the HI group experienced frequent seizures during hypoxia, and 83.3% of rats (10/12) experienced seizures immediately after hypoxia. Seizure frequency and duration gradually decreased with increasing age. The mortality rate of the HI group was 8.33% (1/12); 120 h after HI induction, only 27.3% (3/11) of pups had low-frequency and short-duration electrographic seizures, respectively. HI rats, which presented seizure activities 96 h after HI insult, exhibited an increase in righting reflex time and a decrease in forelimb grip reflex time. Background EEG was significantly inhibited during HI induction and immediately after hypoxia and gradually recovered 72 h after hypoxia. CONCLUSION Seizures caused by HI brain damage in premature infants can be simulated in the P3 neonatal rat model.
Collapse
Affiliation(s)
- Xiaowei Sun
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Fenqin Xue
- Core Facility Center, Capital Medical University, Beijing, China
| | - Jialin Wen
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Limin Gao
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yang Li
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Qianqian Jiang
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lijun Yang
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hong Cui
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
17
|
Ahn JH, Lee HJ, Lee K, Lim J, Hwang JK, Kim CR, Kim HA, Kim HS, Park HK. Effects of Lipopolysaccharide on Oligodendrocyte Differentiation at Different Developmental Stages: an In Vitro Study. J Korean Med Sci 2021; 36:e332. [PMID: 34931496 PMCID: PMC8688345 DOI: 10.3346/jkms.2021.36.e332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 10/25/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Lipopolysaccharide (LPS) exerts cytotoxic effects on brain cells, especially on those belonging to the oligodendrocyte lineage, in preterm infants. The susceptibility of oligodendrocyte lineage cells to LPS-induced inflammation is dependent on the developmental stage. This study aimed to investigate the effect of LPS on oligodendrocyte lineage cells at different developmental stages in a microglial cell and oligodendrocyte co-culture model. METHODS The primary cultures of oligodendrocytes and microglia cells were prepared from the forebrains of 2-day-old Sprague-Dawley rats. The oligodendrocyte progenitor cells (OPCs) co-cultured with microglial cells were treated with 0 (control), 0.01, 0.1, and 1 µg/mL LPS at the D3 stage to determine the dose of LPS that impairs oligodendrocyte differentiation. The co-culture was treated with 0.01 µg/mL LPS, which was the lowest dose that did not impair oligodendrocyte differentiation, at the developmental stages D1 (early LPS group), D3 (late LPS group), or D1 and D3 (double LPS group). On day 7 of differentiation, oligodendrocytes were subjected to neural glial antigen 2 (NG2) and myelin basic protein (MBP) immunostaining to examine the number of OPCs and mature oligodendrocytes, respectively. RESULTS LPS dose-dependently decreased the proportion of mature oligodendrocytes (MBP+ cells) relative to the total number of cells. The number of MBP+ cells in the early LPS group was significantly lower than that in the late LPS group. Compared with those in the control group, the MBP+ cell numbers were significantly lower and the NG2+ cell numbers were significantly higher in the double LPS group, which exhibited impaired oligodendrocyte lineage cell development, on day 7 of differentiation. CONCLUSION Repetitive LPS stimulation during development significantly inhibited brain cell development by impairing oligodendrocyte differentiation. In contrast, brain cell development was not affected in the late LPS group. These findings suggest that inflammation at the early developmental stage of oligodendrocytes increases the susceptibility of the preterm brain to inflammation-induced injury.
Collapse
Affiliation(s)
- Ja-Hye Ahn
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Korea
| | - Hyun Ju Lee
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Korea
| | - Kyeongmi Lee
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Korea
| | - Jean Lim
- Kangwon National University College of Veterinary Medicine, Chuncheon, Korea
| | - Jae Kyoon Hwang
- Department of Pediatrics, Hanyang University Guri Hospital, Guri, Korea
| | - Chang-Ryul Kim
- Department of Pediatrics, Hanyang University Guri Hospital, Guri, Korea
| | - Hyun A Kim
- Department of Child Psychotherapy, Hanyang University Graduate School of Medicine, Seoul, Korea
| | - Han-Suk Kim
- Department of Pediatrics, Seoul University College of Medicine, Seoul, Korea
| | - Hyun-Kyung Park
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Korea.
| |
Collapse
|
18
|
Tetorou K, Sisa C, Iqbal A, Dhillon K, Hristova M. Current Therapies for Neonatal Hypoxic-Ischaemic and Infection-Sensitised Hypoxic-Ischaemic Brain Damage. Front Synaptic Neurosci 2021; 13:709301. [PMID: 34504417 PMCID: PMC8421799 DOI: 10.3389/fnsyn.2021.709301] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/19/2021] [Indexed: 12/15/2022] Open
Abstract
Neonatal hypoxic-ischaemic brain damage is a leading cause of child mortality and morbidity, including cerebral palsy, epilepsy, and cognitive disabilities. The majority of neonatal hypoxic-ischaemic cases arise as a result of impaired cerebral perfusion to the foetus attributed to uterine, placental, or umbilical cord compromise prior to or during delivery. Bacterial infection is a factor contributing to the damage and is recorded in more than half of preterm births. Exposure to infection exacerbates neuronal hypoxic-ischaemic damage thus leading to a phenomenon called infection-sensitised hypoxic-ischaemic brain injury. Models of neonatal hypoxia-ischaemia (HI) have been developed in different animals. Both human and animal studies show that the developmental stage and the severity of the HI insult affect the selective regional vulnerability of the brain to damage, as well as the subsequent clinical manifestations. Therapeutic hypothermia (TH) is the only clinically approved treatment for neonatal HI. However, the number of HI infants needed to treat with TH for one to be saved from death or disability at age of 18-22 months, is approximately 6-7, which highlights the need for additional or alternative treatments to replace TH or increase its efficiency. In this review we discuss the mechanisms of HI injury to the immature brain and the new experimental treatments studied for neonatal HI and infection-sensitised neonatal HI.
Collapse
Affiliation(s)
| | | | | | | | - Mariya Hristova
- Perinatal Brain Repair Group, Department of Maternal and Fetal Medicine, UCL Institute for Women’s Health, London, United Kingdom
| |
Collapse
|
19
|
Chesnut M, Hartung T, Hogberg H, Pamies D. Human Oligodendrocytes and Myelin In Vitro to Evaluate Developmental Neurotoxicity. Int J Mol Sci 2021; 22:7929. [PMID: 34360696 PMCID: PMC8347131 DOI: 10.3390/ijms22157929] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/14/2021] [Accepted: 07/21/2021] [Indexed: 01/01/2023] Open
Abstract
Neurodevelopment is uniquely sensitive to toxic insults and there are concerns that environmental chemicals are contributing to widespread subclinical developmental neurotoxicity (DNT). Increased DNT evaluation is needed due to the lack of such information for most chemicals in common use, but in vivo studies recommended in regulatory guidelines are not practical for the large-scale screening of potential DNT chemicals. It is widely acknowledged that developmental neurotoxicity is a consequence of disruptions to basic processes in neurodevelopment and that testing strategies using human cell-based in vitro systems that mimic these processes could aid in prioritizing chemicals with DNT potential. Myelination is a fundamental process in neurodevelopment that should be included in a DNT testing strategy, but there are very few in vitro models of myelination. Thus, there is a need to establish an in vitro myelination assay for DNT. Here, we summarize the routes of myelin toxicity and the known models to study this particular endpoint.
Collapse
Affiliation(s)
- Megan Chesnut
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (M.C.); (T.H.)
| | - Thomas Hartung
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (M.C.); (T.H.)
- Center for Alternatives to Animal Testing (CAAT-Europe), University of Konstanz, 78464 Konstanz, Germany
| | - Helena Hogberg
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (M.C.); (T.H.)
| | - David Pamies
- Center for Alternatives to Animal Testing (CAAT), Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (M.C.); (T.H.)
- Department of Physiology, University of Lausanne, 1005 Lausanne, Switzerland
- Swiss Centre for Applied Human Toxicology (SCAHT), 4055 Basel, Switzerland
| |
Collapse
|
20
|
Ogawa S, Hagiwara M, Misumi S, Tajiri N, Shimizu T, Ishida A, Suzumori N, Sugiura-Ogasawara M, Hida H. Transplanted Oligodendrocyte Progenitor Cells Survive in the Brain of a Rat Neonatal White Matter Injury Model but Less Mature in Comparison with the Normal Brain. Cell Transplant 2021; 29:963689720946092. [PMID: 32757665 PMCID: PMC7563029 DOI: 10.1177/0963689720946092] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Preterm infants have a high risk of neonatal white matter injury (WMI) caused by hypoxia-ischemia. Cell-based therapies are promising strategies for neonatal WMI by providing trophic substances and replacing lost cells. Using a rat model of neonatal WMI in which oligodendrocyte progenitors (OPCs) are predominantly damaged, we investigated whether insulin-like growth factor 2 (IGF2) has trophic effects on OPCs in vitro and whether OPC transplantation has potential as a cell replacement therapy. Enhanced expression of Igf2 mRNA was first confirmed in the brain of P5 model rats by real-time polymerase chain reaction. Immunostaining for IGF2 and its receptor IGF2 R revealed that both proteins were co-expressed in OLIG2-positive and GFAP-positive cells in the corpus callosum (CC), indicating autocrine and paracrine effects of IGF2. To investigate the in vitro effect of IGF2 on OPCs, IGF2 (100 ng/ml) was added to the differentiation medium containing ciliary neurotrophic factor (10 ng/ml) and triiodothyronine (20 ng/ml), and IGF2 promoted the differentiation of OPCs into mature oligodendrocytes. We next transplanted rat-derived OPCs that express green fluorescent protein into the CC of neonatal WMI model rats without immunosuppression and investigated the survival of grafted cells for 8 weeks. Although many OPCs survived for at least 8 weeks, the number of mature oligodendrocytes was unexpectedly small in the CC of the model compared with that in the sham-operated control. These findings suggest that the mechanism in the brain that inhibits differentiation should be solved in cell replacement therapy for neonatal WMI as same as trophic support from IGF2.
Collapse
Affiliation(s)
- Shino Ogawa
- Departments of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Obstetrics and Gynecology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Mutsumi Hagiwara
- Departments of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Sachiyo Misumi
- Departments of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Naoki Tajiri
- Departments of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takeshi Shimizu
- Departments of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akimasa Ishida
- Departments of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Nobuhiro Suzumori
- Obstetrics and Gynecology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Mayumi Sugiura-Ogasawara
- Obstetrics and Gynecology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Hideki Hida
- Departments of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
21
|
Structural network performance for early diagnosis of spastic cerebral palsy in periventricular white matter injury. Brain Imaging Behav 2021; 15:855-864. [PMID: 32306282 DOI: 10.1007/s11682-020-00295-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Periventricular white matter injury (PWMI) is a common cause of spastic cerebral palsy (SCP). Diffusion tensor imaging (DTI) shows high sensitivity but moderate specificity for predicting SCP. The limited specificity may be due to the diverse and extensive brain injuries seen in infants with PWMI. We enrolled 72 infants with corrected age from 6 to 18 months in 3 groups: PWMI with SCP (n = 20), non-CP PWMI (n = 19), and control (n = 33) groups. We compared DTI-based brain network properties among the three groups and evaluated the diagnostic performance of brain network properties for SCP in PWMI infants. Our results show abnormal global parameters (reduced global and local efficiency, and increased shortest path length), and local parameters (reduced node efficiency) in the PWMI with SCP group. On logistic regression, the combined node efficiency of the bilateral precentral gyrus and right middle frontal gyrus had a high sensitivity (90%) and specificity (95%) for differentiating PWMI with SCP from non-CP PWMI, and significantly correlated with the Gross Motor Function Classification System scores. This study confirms that DTI-based brain network has great diagnostic performance for SCP in PWMI infants, and the combined node efficiency improves the diagnostic accuracy.
Collapse
|
22
|
Prasad JD, Gunn KC, Davidson JO, Galinsky R, Graham SE, Berry MJ, Bennet L, Gunn AJ, Dean JM. Anti-Inflammatory Therapies for Treatment of Inflammation-Related Preterm Brain Injury. Int J Mol Sci 2021; 22:4008. [PMID: 33924540 PMCID: PMC8069827 DOI: 10.3390/ijms22084008] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/09/2021] [Accepted: 04/10/2021] [Indexed: 12/13/2022] Open
Abstract
Despite the prevalence of preterm brain injury, there are no established neuroprotective strategies to prevent or alleviate mild-to-moderate inflammation-related brain injury. Perinatal infection and inflammation have been shown to trigger acute neuroinflammation, including proinflammatory cytokine release and gliosis, which are associated with acute and chronic disturbances in brain cell survival and maturation. These findings suggest the hypothesis that the inhibition of peripheral immune responses following infection or nonspecific inflammation may be a therapeutic strategy to reduce the associated brain injury and neurobehavioral deficits. This review provides an overview of the neonatal immunity, neuroinflammation, and mechanisms of inflammation-related brain injury in preterm infants and explores the safety and efficacy of anti-inflammatory agents as potentially neurotherapeutics.
Collapse
Affiliation(s)
- Jaya D. Prasad
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland 1010, New Zealand; (J.D.P.); (K.C.G.); (J.O.D.); (L.B.); (A.J.G.)
| | - Katherine C. Gunn
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland 1010, New Zealand; (J.D.P.); (K.C.G.); (J.O.D.); (L.B.); (A.J.G.)
| | - Joanne O. Davidson
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland 1010, New Zealand; (J.D.P.); (K.C.G.); (J.O.D.); (L.B.); (A.J.G.)
| | - Robert Galinsky
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia;
| | - Scott E. Graham
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1010, New Zealand;
| | - Mary J. Berry
- Department of Pediatrics and Health Care, University of Otago, Dunedin 9016, New Zealand;
| | - Laura Bennet
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland 1010, New Zealand; (J.D.P.); (K.C.G.); (J.O.D.); (L.B.); (A.J.G.)
| | - Alistair J. Gunn
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland 1010, New Zealand; (J.D.P.); (K.C.G.); (J.O.D.); (L.B.); (A.J.G.)
| | - Justin M. Dean
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, 85 Park Road, Grafton, Auckland 1010, New Zealand; (J.D.P.); (K.C.G.); (J.O.D.); (L.B.); (A.J.G.)
| |
Collapse
|
23
|
White matter injury in infants with intraventricular haemorrhage: mechanisms and therapies. Nat Rev Neurol 2021; 17:199-214. [PMID: 33504979 PMCID: PMC8880688 DOI: 10.1038/s41582-020-00447-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2020] [Indexed: 01/31/2023]
Abstract
Intraventricular haemorrhage (IVH) continues to be a major complication of prematurity that can result in cerebral palsy and cognitive impairment in survivors. No optimal therapy exists to prevent IVH or to treat its consequences. IVH varies in severity and can present as a bleed confined to the germinal matrix, small-to-large IVH or periventricular haemorrhagic infarction. Moderate-to-severe haemorrhage dilates the ventricle and damages the periventricular white matter. This white matter injury results from a constellation of blood-induced pathological reactions, including oxidative stress, glutamate excitotoxicity, inflammation, perturbed signalling pathways and remodelling of the extracellular matrix. Potential therapies for IVH are currently undergoing investigation in preclinical models and evidence from clinical trials suggests that stem cell treatment and/or endoscopic removal of clots from the cerebral ventricles could transform the outcome of infants with IVH. This Review presents an integrated view of new insights into the mechanisms underlying white matter injury in premature infants with IVH and highlights the importance of early detection of disability and immediate intervention in optimizing the outcomes of IVH survivors.
Collapse
|
24
|
Joerger-Messerli MS, Thomi G, Haesler V, Keller I, Renz P, Surbek DV, Schoeberlein A. Human Wharton's Jelly Mesenchymal Stromal Cell-Derived Small Extracellular Vesicles Drive Oligodendroglial Maturation by Restraining MAPK/ERK and Notch Signaling Pathways. Front Cell Dev Biol 2021; 9:622539. [PMID: 33869172 PMCID: PMC8044995 DOI: 10.3389/fcell.2021.622539] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/28/2021] [Indexed: 12/12/2022] Open
Abstract
Peripartum cerebral hypoxia and ischemia, and intrauterine infection and inflammation, are detrimental for the precursor cells of the myelin-forming oligodendrocytes in the prematurely newborn, potentially leading to white matter injury (WMI) with long-term neurodevelopmental sequelae. Previous data show that hypomyelination observed in WMI is caused by arrested oligodendroglial maturation rather than oligodendrocyte-specific cell death. In a rat model of premature WMI, we have recently shown that small extracellular vesicles (sEV) derived from Wharton's jelly mesenchymal stromal cells (WJ-MSC) protect from myelination deficits. Thus, we hypothesized that sEV derived from WJ-MSC directly promote oligodendroglial maturation in oligodendrocyte precursor cells. To test this assumption, sEV were isolated from culture supernatants of human WJ-MSC by ultracentrifugation and co-cultured with the human immortalized oligodendrocyte precursor cell line MO3.13. As many regulatory functions in WMI have been ascribed to microRNA (miR) and as sEV are carriers of functional miR which can be delivered to target cells, we characterized and quantified the miR content of WJ-MSC-derived sEV by next-generation sequencing. We found that WJ-MSC-derived sEV co-localized with MO3.13 cells within 4 h. After 5 days of co-culture, the expression of myelin basic protein (MBP), a marker for mature oligodendrocytes, was significantly increased, while the oligodendrocyte precursor marker platelet-derived growth factor alpha (PDGFRα) was decreased. Notch and MAPK/ERK pathways known to inhibit oligodendrocyte maturation and differentiation were significantly reduced. The pathway enrichment analysis showed that the miR present in WJ-MSC-derived sEV target genes having key roles in the MAPK pathway. Our data strongly suggest that sEV from WJ-MSC directly drive the maturation of oligodendrocyte precursor cells by repressing Notch and MAPK/ERK signaling.
Collapse
Affiliation(s)
- Marianne S Joerger-Messerli
- Department of Obstetrics and Feto-maternal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Gierin Thomi
- Department of Obstetrics and Feto-maternal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Valérie Haesler
- Department of Obstetrics and Feto-maternal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Irene Keller
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Patricia Renz
- Department of Obstetrics and Feto-maternal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Daniel V Surbek
- Department of Obstetrics and Feto-maternal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Andreina Schoeberlein
- Department of Obstetrics and Feto-maternal Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.,Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| |
Collapse
|
25
|
Yang F, Li Y, Sheng X, Liu Y. Paeoniflorin treatment regulates TLR4/NF-κB signaling, reduces cerebral oxidative stress and improves white matter integrity in neonatal hypoxic brain injury. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2021; 25:97-109. [PMID: 33602880 PMCID: PMC7893490 DOI: 10.4196/kjpp.2021.25.2.97] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/14/2020] [Accepted: 04/14/2020] [Indexed: 02/04/2023]
Abstract
Neonatal hypoxia/ischemia (H/I), injures white matter, results in neuronal loss, disturbs myelin formation, and neural network development. Neuroinflammation and oxidative stress have been reported in neonatal hypoxic brain injuries. We investigated whether Paeoniflorin treatment reduced H/I-induced inflammation and oxidative stress and improved white matter integrity in a neonatal rodent model. Seven-day old Sprague–Dawley pups were exposed to H/I. Paeoniflorin (6.25, 12.5, or 25 mg/kg body weight) was administered every day via oral gavage from postpartum day 3 (P3) to P14, and an hour before induction of H/I. Pups were sacrificed 24 h (P8) and 72 h (P10) following H/I. Paeoniflorin reduced the apoptosis of neurons and attenuated cerebral infarct volume. Elevated expression of cleaved caspase-3 and Bad were regulated. Paeoniflorin decreased oxidative stress by lowering levels of malondialdehyde and reactive oxygen species generation and while, and it enhanced glutathione content. Microglial activation and the TLR4/NF-κB signaling were significantly down-regulated. The degree of inflammatory mediators (interleukin 1β and tumor necrosis factor-α) were reduced. Paeoniflorin markedly prevented white matter injury via improving expression of myelin binding protein and increasing O1-positive olidgodendrocyte and O4-positive oligodendrocyte counts. The present investigation demonstrates the potent protective efficiency of paeoniflorin supplementation against H/I-induced brain injury by effectually preventing neuronal loss, microglial activation, and white matter injury via reducing oxidative stress and inflammatory pathways.
Collapse
Affiliation(s)
- Fan Yang
- Department of Clinical Nutrition, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| | - Ya Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China.,Yunnan Institute of Laboratory Diagnosis, Kunming 650032, Yunnan, China.,Yunnan Key Laboratory of Laboratory Medicine, Kunming 650032, Yunnan, China
| | - Xun Sheng
- School of Stomatology, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Yu Liu
- Department of Pharmacy, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, Yunnan, China
| |
Collapse
|
26
|
Role of extracellular vesicles in neurodegenerative diseases. Prog Neurobiol 2021; 201:102022. [PMID: 33617919 DOI: 10.1016/j.pneurobio.2021.102022] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/27/2020] [Accepted: 02/11/2021] [Indexed: 02/08/2023]
Abstract
Extracellular vesicles (EVs) are heterogeneous cell-derived membranous structures that arise from the endosome system or directly detach from the plasma membrane. In recent years, many advances have been made in the understanding of the clinical definition and pathogenesis of neurodegenerative diseases, but translation into effective treatments is hampered by several factors. Current research indicates that EVs are involved in the pathology of diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD). Besides, EVs are also involved in the process of myelin formation, and can also cross the blood-brain barrier to reach the sites of CNS injury. It is suggested that EVs have great potential as a novel therapy for the treatment of neurodegenerative diseases. Here, we reviewed the advances in understanding the role of EVs in neurodegenerative diseases and addressed the critical function of EVs in the CNS. We have also outlined the physiological mechanisms of EVs in myelin regeneration and highlighted the therapeutic potential of EVs in neurodegenerative diseases.
Collapse
|
27
|
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: 2.3] [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
|
28
|
Yates N, Gunn AJ, Bennet L, Dhillon SK, Davidson JO. Preventing Brain Injury in the Preterm Infant-Current Controversies and Potential Therapies. Int J Mol Sci 2021; 22:1671. [PMID: 33562339 PMCID: PMC7915709 DOI: 10.3390/ijms22041671] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 12/12/2022] Open
Abstract
Preterm birth is associated with a high risk of morbidity and mortality including brain damage and cerebral palsy. The development of brain injury in the preterm infant may be influenced by many factors including perinatal asphyxia, infection/inflammation, chronic hypoxia and exposure to treatments such as mechanical ventilation and corticosteroids. There are currently very limited treatment options available. In clinical trials, magnesium sulfate has been associated with a small, significant reduction in the risk of cerebral palsy and gross motor dysfunction in early childhood but no effect on the combined outcome of death or disability, and longer-term follow up to date has not shown improved neurological outcomes in school-age children. Recombinant erythropoietin has shown neuroprotective potential in preclinical studies but two large randomized trials, in extremely preterm infants, of treatment started within 24 or 48 h of birth showed no effect on the risk of severe neurodevelopmental impairment or death at 2 years of age. Preclinical studies have highlighted a number of promising neuroprotective treatments, such as therapeutic hypothermia, melatonin, human amnion epithelial cells, umbilical cord blood and vitamin D supplementation, which may be useful at reducing brain damage in preterm infants. Moreover, refinements of clinical care of preterm infants have the potential to influence later neurological outcomes, including the administration of antenatal and postnatal corticosteroids and more accurate identification and targeted treatment of seizures.
Collapse
Affiliation(s)
- Nathanael Yates
- The Queensland Brain Institute, University of Queensland, St Lucia, QLD 4072, Australia;
- School of Human Sciences, University of Western Australia, Crawley, WA 6009, Australia
| | - Alistair J. Gunn
- The Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (A.J.G.); (L.B.); (S.K.D.)
| | - Laura Bennet
- The Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (A.J.G.); (L.B.); (S.K.D.)
| | - Simerdeep K. Dhillon
- The Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (A.J.G.); (L.B.); (S.K.D.)
| | - Joanne O. Davidson
- The Department of Physiology, University of Auckland, Auckland 1023, New Zealand; (A.J.G.); (L.B.); (S.K.D.)
| |
Collapse
|
29
|
Katti K, Ayasolla KR, Iurcotta T, Potak D, Codipilly C, Weinberger B. Lipid peroxidation products as predictors of oxidant-mediated disease in preterm infants. J Matern Fetal Neonatal Med 2021; 35:4878-4883. [PMID: 33397176 DOI: 10.1080/14767058.2020.1869934] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Preterm infants are susceptible to "oxygen radical diseases" (ORD). 8-isoprostane (8-IP) is a bioactive eicosanoid generated by reactive oxygen species-catalyzed peroxidation of arachidonic acid. Malondialdehyde (MDA) is generated by the decomposition of oxidant-induced lipid hydroperoxides. We hypothesize that the development of ORD is associated with elevated plasma 8-IP on day 0-1, and increasing urine levels of MDA in the first month. METHODS Preterm (<32 weeks, n = 39) and term (n = 39) infants were recruited at birth. Plasma 8-IP was quantified by ELISA on day 0-1, and urine MDA by colorimetric assay of thiobarbituric acid reactive substances (TBARS) on days 0-1, 7, 14, 21, and 28. ORD was defined as retinopathy of prematurity ≥ stage 1, pneumatosis, or oxygen requirement at 36 weeks corrected gestational age. RESULTS Plasma 8-IP was higher on day 0-1 in preterm infants who developed ORD compared to term infants. Urine TBARS levels increased in preterm infants from day 0-1 to day 28 but were not different in infants with or without ORD. Preterm infants who developed ORD demonstrated a significant rise in urine TBARS levels from day 1 to 14. CONCLUSIONS Elevated plasma 8-IP on day 1 is associated with ORD in preterm infants. If validated as a biomarker for ORD, it may be useful in directing antioxidant therapies to the most susceptible infants. Urine TBARS during the first month are not significantly different in term infants, preterm infants with ORD, and preterm infants without ORD, but rapid rise of TBARS in the first 2 weeks may be associated with ORD.
Collapse
Affiliation(s)
- Karishma Katti
- Division of Neonatal-Perinatal Medicine, Cohen Children's Medical Center, Lilling Family Neonatal Research Lab, Feinstein Institute for Medical Research, Northwell Health, New Hyde Park, NY, USA
| | - Kamesh R Ayasolla
- Division of Neonatal-Perinatal Medicine, Cohen Children's Medical Center, Lilling Family Neonatal Research Lab, Feinstein Institute for Medical Research, Northwell Health, New Hyde Park, NY, USA
| | - Toni Iurcotta
- Division of Neonatal-Perinatal Medicine, Cohen Children's Medical Center, Lilling Family Neonatal Research Lab, Feinstein Institute for Medical Research, Northwell Health, New Hyde Park, NY, USA
| | - Debra Potak
- Division of Neonatal-Perinatal Medicine, Cohen Children's Medical Center, Lilling Family Neonatal Research Lab, Feinstein Institute for Medical Research, Northwell Health, New Hyde Park, NY, USA
| | - Champa Codipilly
- Division of Neonatal-Perinatal Medicine, Cohen Children's Medical Center, Lilling Family Neonatal Research Lab, Feinstein Institute for Medical Research, Northwell Health, New Hyde Park, NY, USA
| | - Barry Weinberger
- Division of Neonatal-Perinatal Medicine, Cohen Children's Medical Center, Lilling Family Neonatal Research Lab, Feinstein Institute for Medical Research, Northwell Health, New Hyde Park, NY, USA
| |
Collapse
|
30
|
Vanderhasselt T, Zolfaghari R, Naeyaert M, Dudink J, Buls N, Allemeersch GJ, Raeymaekers H, Cools F, de Mey J. Synthetic MRI demonstrates prolonged regional relaxation times in the brain of preterm born neonates with severe postnatal morbidity. NEUROIMAGE-CLINICAL 2020; 29:102544. [PMID: 33385883 PMCID: PMC7786121 DOI: 10.1016/j.nicl.2020.102544] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/13/2020] [Accepted: 12/20/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND To identify preterm infants at risk for neurodevelopment impairment that might benefit from early neurorehabilitation, early prognostic biomarkers of future outcomes are needed. OBJECTIVE To determine whether synthetic MRI is sensitive to age-related changes in regional tissue relaxation times in the brain of preterm born neonates when scanned at term equivalent age (TEA, 37-42 weeks), and to investigate whether severe postnatal morbidity results in prolonged regional tissue relaxation times. MATERIALS AND METHODS This retrospective study included 70 very preterm born infants scanned with conventional and synthetic MRI between January 2017 and June 2019 at TEA. Infants with severe postnatal morbidity were allocated to a high-risk group (n = 22). All other neonates were allocated to a low-risk group (n = 48). Linear regression analysis was performed to determine the relationship between relaxation times and postmenstrual age (PMA) at scan. Analysis of covariance was used to evaluate the impact of severe postnatal morbidity in the high-risk group on T1 and T2 relaxation times. Receiver operating characteristic (ROC) curves were plotted and analysed with area under the ROC curve (AUC) to evaluate the accuracy of classifying high-risk patients based on regional relaxation times. RESULTS A linear age-related decrease of T1 and T2 relaxation times correlating with PMA at scan (between 37 and 42 weeks) was found in the deep gray matter, the cerebellum, the cortex, and the posterior limb of the internal capsule (PLIC) (p < .005 each), but not in the global, frontal, parietal, or central white matter. Analysis of covariance for both risk groups, adjusted for PMA, revealed significantly prolonged regional tissue relaxation times in neonates with severe postnatal morbidity, which was best illustrated in the central white matter of the centrum semiovale (T1 Δ = 11.5%, T2 Δ = 13.4%, p < .001) and in the PLIC (T1 Δ = 9.2%, T2 Δ = 6.9%, p < .001). The relaxation times in the PLIC and the central white matter predicted high-risk status with excellent accuracy (AUC range 0.82-0.86). CONCLUSION Synthetic MRI-based relaxometry in the brain of preterm born neonates is sensitive to age-related maturational changes close to TEA. Severe postnatal morbidity correlated with a significant delay in tissue relaxation. Synthetic MRI may provide early prognostic biomarkers for neurodevelopment impairment.
Collapse
Affiliation(s)
- Tim Vanderhasselt
- Department of Radiology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium.
| | - Roya Zolfaghari
- Department of Radiology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Maarten Naeyaert
- Department of Radiology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Jeroen Dudink
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands; Brain Center University Medical Center Utrecht, Utrecht, The Netherlands
| | - Nico Buls
- Department of Radiology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Gert-Jan Allemeersch
- Department of Radiology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Hubert Raeymaekers
- Department of Radiology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Filip Cools
- Department of Neonatology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Johan de Mey
- Department of Radiology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| |
Collapse
|
31
|
HIFα Regulates Developmental Myelination Independent of Autocrine Wnt Signaling. J Neurosci 2020; 41:251-268. [PMID: 33208471 DOI: 10.1523/jneurosci.0731-20.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 10/15/2020] [Accepted: 11/11/2020] [Indexed: 01/17/2023] Open
Abstract
The developing CNS is exposed to physiological hypoxia, under which hypoxia-inducible factor α (HIFα) is stabilized and plays a crucial role in regulating neural development. The cellular and molecular mechanisms of HIFα in developmental myelination remain incompletely understood. A previous concept proposes that HIFα regulates CNS developmental myelination by activating the autocrine Wnt/β-catenin signaling in oligodendrocyte progenitor cells (OPCs). Here, by analyzing a battery of genetic mice of both sexes, we presented in vivo evidence supporting an alternative understanding of oligodendroglial HIFα-regulated developmental myelination. At the cellular level, we found that HIFα was required for developmental myelination by transiently controlling upstream OPC differentiation but not downstream oligodendrocyte maturation and that HIFα dysregulation in OPCs but not oligodendrocytes disturbed normal developmental myelination. We demonstrated that HIFα played a minor, if any, role in regulating canonical Wnt signaling in the oligodendroglial lineage or in the CNS. At the molecular level, blocking autocrine Wnt signaling did not affect HIFα-regulated OPC differentiation and myelination. We further identified HIFα-Sox9 regulatory axis as an underlying molecular mechanism in HIFα-regulated OPC differentiation. Our findings support a concept shift in our mechanistic understanding of HIFα-regulated CNS myelination from the previous Wnt-dependent view to a Wnt-independent one and unveil a previously unappreciated HIFα-Sox9 pathway in regulating OPC differentiation.SIGNIFICANCE STATEMENT Promoting disturbed developmental myelination is a promising option in treating diffuse white matter injury, previously called periventricular leukomalacia, a major form of brain injury affecting premature infants. In the developing CNS, hypoxia-inducible factor α (HIFα) is a key regulator that adapts neural cells to physiological and pathologic hypoxic cues. The role and mechanism of HIFα in oligodendroglial myelination, which is severely disturbed in preterm infants affected with diffuse white matter injury, is incompletely understood. Our findings presented here represent a concept shift in our mechanistic understanding of HIFα-regulated developmental myelination and suggest the potential of intervening with an oligodendroglial HIFα-mediated signaling pathway to mitigate disturbed myelination in premature white matter injury.
Collapse
|
32
|
Wong FY, Gogos A, Hale N, Ingelse SA, Brew N, Shepherd KL, van den Buuse M, Walker DW. Impact of hypoxia-ischemia and dopamine treatment on dopamine receptor binding density in the preterm fetal sheep brain. J Appl Physiol (1985) 2020; 129:1431-1438. [PMID: 33054660 DOI: 10.1152/japplphysiol.00677.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Dopamine is often used to treat hypotension in preterm infants who are at risk of hypoxic-ischemic (HI) brain injury due to cerebral hypoperfusion and impaired autoregulation. There is evidence that systemically administered dopamine crosses the preterm blood-brain barrier. However, the effects of exogenous dopamine and cerebral HI on dopaminergic signaling in the immature brain are unknown. We determined the effect of HI and dopamine on D1 and D2 receptor binding and expressions of dopamine transporter (DAT) and tyrosine hydroxylase (TH) in the striatum of the preterm fetal sheep. Fetal sheep (99 days of gestation, term = 147days) were unoperated controls (n = 6) or exposed to severe HI using umbilical cord occlusion and saline infusion (UCO + saline, n = 8) or to HI with dopamine infusion (UCO + dopamine, 10 µg/kg/min, n = 7) for 74 h. D1 and D2 receptor densities were measured by autoradiography in vitro. DAT, TH, and cell death were measured using immunohistochemistry. HI resulted in cell death in the caudate nucleus and putamen, and dopamine infusion started before HI did not exacerbate or ameliorate these effects. HI led to reduced D1 and D2 receptor densities in the caudate nucleus and reduction in DAT protein expression in the caudate and putamen. Fetal brains exposed to dopamine in addition to HI were not different from those exposed to HI alone in these changes in dopaminergic parameters. We conclude that dopamine infusion does not alter the striatal cell death or the reductions in D1 and D2 receptor densities and DAT protein expression induced by HI in the preterm brain.NEW & NOTEWORTHY This is the first study on the effects of hypoxia-ischemia and dopamine treatment on the dopaminergic pathway in the preterm brain. In the striatum of fetal sheep (equivalent to ∼26-28 wk of human gestation), we demonstrate that hypoxia-ischemia leads to cell death, reduces D1 and D2 receptors, and reduces dopamine transporter. Intravenous dopamine infusion at clinical dosage used in preterm human infants does not alter the striatal cell death, D1 and D2 receptor density levels, and DAT protein expressions after hypoxia-ischemia in the preterm brain.
Collapse
Affiliation(s)
- F Y Wong
- The Ritchie Centre, The Hudson Institute of Medical Research, Melbourne, Australia.,Department of Paediatrics, Monash University, Melbourne, Australia.,Monash Newborn, Monash Medical Centre, Melbourne, Australia
| | - A Gogos
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Australia
| | - N Hale
- The Ritchie Centre, The Hudson Institute of Medical Research, Melbourne, Australia
| | - S A Ingelse
- The Ritchie Centre, The Hudson Institute of Medical Research, Melbourne, Australia
| | - N Brew
- The Ritchie Centre, The Hudson Institute of Medical Research, Melbourne, Australia
| | - K L Shepherd
- The Ritchie Centre, The Hudson Institute of Medical Research, Melbourne, Australia.,Department of Paediatrics, Monash University, Melbourne, Australia
| | - M van den Buuse
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Australia.,School of Psychology and Public Health, La Trobe University, Melbourne, Australia
| | - D W Walker
- The Ritchie Centre, The Hudson Institute of Medical Research, Melbourne, Australia.,School of Health & Biomedical Sciences, RMIT University, Melbourne, Australia
| |
Collapse
|
33
|
The vulnerability of the immature brain. HANDBOOK OF CLINICAL NEUROLOGY 2020. [PMID: 32958197 DOI: 10.1016/b978-0-444-64150-2.00010-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
The concept of vulnerability of the immature brain is multifactorial by definition. Newer scientific work in this area has shifted and enlarged the concept from theoretical frameworks to the multiple levels (molecular, cellular, anatomic, network, behavioral) of the organization of the growing brain. The concept of vulnerability was first introduced by Donald O. Hebb in the 1950s and referred to the inability of the immature brain to completely recover normal development after a brain insult. The concept of vulnerability was further extended to the limitations of the brain in the development of specific skills in neuronal substrates originally used for other functions. We present an overview of some neurodevelopmental processes that characterize the immature brain and that can predict vulnerability in the case of disturbances: Hebb's principle, synaptic homeostasis, selective vulnerability of immature cells in mammals, and inherited constraint networks. A better understanding of the vulnerability mechanisms may help in early detection and prevention and further proposed individualized therapeutic approaches to enhance children's developmental outcomes.
Collapse
|
34
|
Zhang N, Guan T, Shafiq K, Xing Y, Sun B, Huang Q, Kong J. Compromised Lactate Efflux Renders Vulnerability of Oligodendrocyte Precursor Cells to Metabolic Stresses. ACS Chem Neurosci 2020; 11:2717-2727. [PMID: 32667776 DOI: 10.1021/acschemneuro.0c00353] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Among the brain cells, oligodendrocyte progenitor cells (OPCs) are the most vulnerable in response to hypoxic and ischemic insults, of which the mechanism remains unknown. Brain cells are known to import or export lactate via differentially expressed monocarboxylate transporters (MCTs) to maintain energy metabolism and pH homeostasis. The present study aims to determine the role of MCT1 in the high vulnerability of OPCs. Here we show that a mild ischemic condition equivalent to ischemic preconditioning caused detectable loss of OPCs. MCT1, which is primarily expressed in oligodendrocyte lineage cells including OPCs, was up-regulated immediately under oxygen-glucose deprivation (OGD) conditions. However, persistent hypoxia, but not hypoglycemia, inhibited the function of MCT1, leading to an intracellular lactate accumulation and acidosis in OPCs. Neurons, which express primarily MCT2, were able to export lactate and maintain an intracellular pH homeostasis under similar conditions. The results support that compromised lactate efflux resulting from hypoxia-induced dysfunction of MCT1 contributes to the high vulnerability of OPCs.
Collapse
Affiliation(s)
- Nan Zhang
- Mental Health Center, Shantou University Medical College, 243 Daxue Road Shantou, Guangdong 515063, China
- Department of Human Anatomy and Cell Science, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, Manitoba R3E 0J9, Canada
- Department of Neurology and Central Laboratory, First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Teng Guan
- Department of Human Anatomy and Cell Science, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, Manitoba R3E 0J9, Canada
| | - Kashfia Shafiq
- Department of Human Anatomy and Cell Science, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, Manitoba R3E 0J9, Canada
| | - Yuan Xing
- Department of Neurology and Central Laboratory, First Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | - Baoliang Sun
- Key Lab of Cerebral Microcirculation, Shandong First Medical University, Tai'an, Shandong 271016, China
| | - Qingjun Huang
- Mental Health Center, Shantou University Medical College, 243 Daxue Road Shantou, Guangdong 515063, China
| | - Jiming Kong
- Mental Health Center, Shantou University Medical College, 243 Daxue Road Shantou, Guangdong 515063, China
- Department of Human Anatomy and Cell Science, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, Manitoba R3E 0J9, Canada
| |
Collapse
|
35
|
Park S, Chen L, Townsend J, Lee H, Feinberg DA. Simultaneous Multi-VENC and Simultaneous Multi-Slice Phase Contrast Magnetic Resonance Imaging. IEEE TRANSACTIONS ON MEDICAL IMAGING 2020; 39:742-752. [PMID: 31403409 PMCID: PMC7138512 DOI: 10.1109/tmi.2019.2934422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
This work develops a novel, simultaneous multi-VENC and simultaneous multi-slice (SMV+SMS) imaging in a single acquisition for robust phase contrast (PC) MRI. To this end, the pulse sequence was designed to permit concurrent acquisition of multiple VENCs as well as multiple slices on a shared frequency encoding gradient, in which each effective echo time for multiple VENCs was controlled by adjusting net gradient area while multiple slices were simultaneously excited by employing multiband resonance frequency (RF) pulses. For VENC and slice separation, RF phase cycling and gradient blip were applied to create both inter-VENC and inter-slice shifts along phase encoding direction, respectively. With an alternating RF phase cycling that generates oscillating steady-state with low and high signal amplitude, the acquired multi-VENC k-space was reformulated into 3D undersampled k-space by generating a virtual dimension along VENC direction for modulation induced artifact reduction. In vivo studies were conducted to validate the feasibility of the proposed method in comparison with conventional PC MRI. The proposed method shows comparable performance to the conventional method in delineating both low and high flow velocities across cardiac phases with high spatial coverage without apparent artifacts. In the presence of high flow velocity that is above the VENC value, the proposed method exhibits clear depiction of flow signals over conventional method, thereby leading to high VNR image with improved velocity dynamic range.
Collapse
Affiliation(s)
| | - Liyong Chen
- Advanced MRI Technologies, Sebastopol, CA, 95472, USA
| | - Jennifer Townsend
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720, USA and Advanced MRI Technologies, Sebastopol, CA, 95472, USA
| | - Hyunyeol Lee
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - David A. Feinberg
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720, USA and Advanced MRI Technologies, Sebastopol, CA, 95472, USA
| |
Collapse
|
36
|
Hirose-Ikeda M, Chu B, Zhao P, Akil O, Escalante E, Vergnes L, Cepeda C, Espinosa-Jeffrey A. Trophic factors are essential for the survival of grafted oligodendrocyte progenitors and for neuroprotection after perinatal excitotoxicity. Neural Regen Res 2020; 15:557-568. [PMID: 31571668 PMCID: PMC6921341 DOI: 10.4103/1673-5374.266066] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 07/26/2019] [Indexed: 12/29/2022] Open
Abstract
The consequences of neonatal white matter injury are devastating and represent a major societal problem as currently there is no cure. Prematurity, low weight birth and maternal pre-natal infection are the most frequent causes of acquired myelin deficiency in the human neonate leading to cerebral palsy and cognitive impairment. In the developing brain, oligodendrocyte (OL) maturation occurs perinatally, and immature OLs are particularly vulnerable. Cell replacement therapy is often considered a viable option to replace progenitors that die due to glutamate excitotoxicity. We previously reported directed specification and mobilization of endogenous committed and uncommitted neural progenitors by the combination of transferrin and insulin growth factor 1 (TSC1). Here, considering cell replacement and integration as therapeutic goals, we examined if OL progenitors (OLPs) grafted into the brain parenchyma of mice that were subjected to an excitotoxic insult could rescue white matter injury. For that purpose, we used a well-established model of glutamate excitotoxic injury. Four-day-old mice received a single intraparenchymal injection of the glutamate receptor agonist N-methyl-D-aspartate alone or in conjunction with TSC1 in the presence or absence of OLPs grafted into the brain parenchyma. Energetics and expression of stress proteins and OL developmental specific markers were examined. A comparison of the proteomic profile per treatment was also ascertained. We found that OLPs did not survive in the excitotoxic environment when grafted alone. In contrast, when combined with TSC1, survival and integration of grafted OLPs was observed. Further, energy metabolism in OLPs was significantly increased by N-methyl-D-aspartate and modulated by TSC1. The proteomic profile after the various treatments showed elevated ubiquitination and stress/heat shock protein 90 in response to N-methyl-D-aspartate. These changes were reversed in the presence of TSC1 and ubiquitination was decreased. The results obtained in this pre-clinical study indicate that the use of a combinatorial intervention including both trophic support and healthy OLPs constitutes a promising approach for long-term survival and successful graft integration. We established optimal conditioning of the host brain environment to promote long-term survival and integration of grafted OLPs into an inflamed neonate host brain. Experimental procedures were performed under the United States Public Health Service Guide for the Care and Use of Laboratory Animals and were approved by the Institutional Animal Care Committee at (UCLA) (ARC #1992-034-61) on July 1, 2010.
Collapse
Affiliation(s)
- Megumi Hirose-Ikeda
- Intellectual and Developmental Disabilities Research Center, Jane and Terry Semel Institute for Neuroscience and Human Behavior, Departments of Neurobiology, Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Brian Chu
- Intellectual and Developmental Disabilities Research Center, Jane and Terry Semel Institute for Neuroscience and Human Behavior, Departments of Neurobiology, Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Paul Zhao
- Intellectual and Developmental Disabilities Research Center, Jane and Terry Semel Institute for Neuroscience and Human Behavior, Departments of Neurobiology, Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Omar Akil
- Intellectual and Developmental Disabilities Research Center, Jane and Terry Semel Institute for Neuroscience and Human Behavior, Departments of Neurobiology, Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Elida Escalante
- Intellectual and Developmental Disabilities Research Center, Jane and Terry Semel Institute for Neuroscience and Human Behavior, Departments of Neurobiology, Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Laurent Vergnes
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Carlos Cepeda
- Intellectual and Developmental Disabilities Research Center, Jane and Terry Semel Institute for Neuroscience and Human Behavior, Departments of Neurobiology, Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Araceli Espinosa-Jeffrey
- Intellectual and Developmental Disabilities Research Center, Jane and Terry Semel Institute for Neuroscience and Human Behavior, Departments of Neurobiology, Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| |
Collapse
|
37
|
Abbasi H, Unsworth CP. Applications of advanced signal processing and machine learning in the neonatal hypoxic-ischemic electroencephalogram. Neural Regen Res 2020; 15:222-231. [PMID: 31552887 PMCID: PMC6905345 DOI: 10.4103/1673-5374.265542] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/24/2019] [Indexed: 01/15/2023] Open
Abstract
Perinatal hypoxic-ischemic-encephalopathy significantly contributes to neonatal death and life-long disability such as cerebral palsy. Advances in signal processing and machine learning have provided the research community with an opportunity to develop automated real-time identification techniques to detect the signs of hypoxic-ischemic-encephalopathy in larger electroencephalography/amplitude-integrated electroencephalography data sets more easily. This review details the recent achievements, performed by a number of prominent research groups across the world, in the automatic identification and classification of hypoxic-ischemic epileptiform neonatal seizures using advanced signal processing and machine learning techniques. This review also addresses the clinical challenges that current automated techniques face in order to be fully utilized by clinicians, and highlights the importance of upgrading the current clinical bedside sampling frequencies to higher sampling rates in order to provide better hypoxic-ischemic biomarker detection frameworks. Additionally, the article highlights that current clinical automated epileptiform detection strategies for human neonates have been only concerned with seizure detection after the therapeutic latent phase of injury. Whereas recent animal studies have demonstrated that the latent phase of opportunity is critically important for early diagnosis of hypoxic-ischemic-encephalopathy electroencephalography biomarkers and although difficult, detection strategies could utilize biomarkers in the latent phase to also predict the onset of future seizures.
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
|
38
|
Juul SE, Comstock BA, Wadhawan R, Mayock DE, Courtney SE, Robinson T, Ahmad KA, Bendel-Stenzel E, Baserga M, LaGamma EF, Downey LC, Rao R, Fahim N, Lampland A, Frantz ID, Khan JY, Weiss M, Gilmore MM, Ohls RK, Srinivasan N, Perez JE, McKay V, Vu PT, Lowe J, Kuban K, O'Shea TM, Hartman AL, Heagerty PJ. A Randomized Trial of Erythropoietin for Neuroprotection in Preterm Infants. N Engl J Med 2020; 382:233-243. [PMID: 31940698 PMCID: PMC7060076 DOI: 10.1056/nejmoa1907423] [Citation(s) in RCA: 187] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND High-dose erythropoietin has been shown to have a neuroprotective effect in preclinical models of neonatal brain injury, and phase 2 trials have suggested possible efficacy; however, the benefits and safety of this therapy in extremely preterm infants have not been established. METHODS In this multicenter, randomized, double-blind trial of high-dose erythropoietin, we assigned 941 infants who were born at 24 weeks 0 days to 27 weeks 6 days of gestation to receive erythropoietin or placebo within 24 hours after birth. Erythropoietin was administered intravenously at a dose of 1000 U per kilogram of body weight every 48 hours for a total of six doses, followed by a maintenance dose of 400 U per kilogram three times per week by subcutaneous injection through 32 completed weeks of postmenstrual age. Placebo was administered as intravenous saline followed by sham injections. The primary outcome was death or severe neurodevelopmental impairment at 22 to 26 months of postmenstrual age. Severe neurodevelopmental impairment was defined as severe cerebral palsy or a composite motor or composite cognitive score of less than 70 (which corresponds to 2 SD below the mean, with higher scores indicating better performance) on the Bayley Scales of Infant and Toddler Development, third edition. RESULTS A total of 741 infants were included in the per-protocol efficacy analysis: 376 received erythropoietin and 365 received placebo. There was no significant difference between the erythropoietin group and the placebo group in the incidence of death or severe neurodevelopmental impairment at 2 years of age (97 children [26%] vs. 94 children [26%]; relative risk, 1.03; 95% confidence interval, 0.81 to 1.32; P = 0.80). There were no significant differences between the groups in the rates of retinopathy of prematurity, intracranial hemorrhage, sepsis, necrotizing enterocolitis, bronchopulmonary dysplasia, or death or in the frequency of serious adverse events. CONCLUSIONS High-dose erythropoietin treatment administered to extremely preterm infants from 24 hours after birth through 32 weeks of postmenstrual age did not result in a lower risk of severe neurodevelopmental impairment or death at 2 years of age. (Funded by the National Institute of Neurological Disorders and Stroke; PENUT ClinicalTrials.gov number, NCT01378273.).
Collapse
Affiliation(s)
- Sandra E Juul
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Bryan A Comstock
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Rajan Wadhawan
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Dennis E Mayock
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Sherry E Courtney
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Tonya Robinson
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Kaashif A Ahmad
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Ellen Bendel-Stenzel
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Mariana Baserga
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Edmund F LaGamma
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - L Corbin Downey
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Raghavendra Rao
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Nancy Fahim
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Andrea Lampland
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Ivan D Frantz
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Janine Y Khan
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Michael Weiss
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Maureen M Gilmore
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Robin K Ohls
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Nishant Srinivasan
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Jorge E Perez
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Victor McKay
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Phuong T Vu
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Jean Lowe
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Karl Kuban
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - T Michael O'Shea
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Adam L Hartman
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| | - Patrick J Heagerty
- From the University of Washington, Seattle (S.E.J., B.A.C., D.E.M., P.T.V., P.J.H.); Florida Hospital Orlando, Orlando (R.W.), the University of Florida, Gainesville (M.W.), South Miami Hospital, South Miami (J.E.P.), and Johns Hopkins All Children's Hospital, St. Petersburg (V.M.) - all in Florida; the University of Arkansas for Medical Sciences, Little Rock (S.E.C.); the University of Louisville, Louisville, KY (T.R.); Methodist Children's Hospital, San Antonio, TX (K.A.A.); Children's Hospital and Clinics of Minnesota (E.B.-S.) and University of Minnesota Masonic Children's Hospital (R.R., N.F.), Minneapolis, and Children's Minnesota, St. Paul (A.L.) - all in Minnesota; the University of Utah, Salt Lake City (M.B.); Maria Fareri Children's Hospital at Westchester Medical Center, Valhalla, NY (E.F.L.); Wake Forest School of Medicine, Winston-Salem (L.C.D.), and the University of North Carolina, Chapel Hill (T.M.O.) - both in North Carolina; Beth Israel Deaconess Medical Center (I.D.F.) and Boston University (K.K.) - both in Boston; Prentice Women's Hospital (J.Y.K.) and Children's Hospital of the University of Illinois (N.S.) - both in Chicago; Johns Hopkins University, Baltimore (M.M.G.), and the National Institute of Neurological Disorders and Stroke, Bethesda (A.L.H.) - both in Maryland; and the University of New Mexico, Albuquerque (R.K.O., J.L.)
| |
Collapse
|
39
|
Hong HS, Kim SS, Park GY. MRI Findings to Predict Neurodevelopmental Outcomes in Preterm Infants Near Term-Equivalent Age. ACTA ACUST UNITED AC 2020. [DOI: 10.13104/imri.2020.24.1.30] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Affiliation(s)
- Hyun Sook Hong
- Department of Radiology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Sung Shin Kim
- Department of Pediatrics, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Ga Young Park
- Department of Pediatrics, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| |
Collapse
|
40
|
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: 2.3] [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
|
41
|
Kasala S, Briyal S, Prazad P, Ranjan AK, Stefanov G, Donovan R, Gulati A. Exposure to Morphine and Caffeine Induces Apoptosis and Mitochondrial Dysfunction in a Neonatal Rat Brain. Front Pediatr 2020; 8:593. [PMID: 33042927 PMCID: PMC7530195 DOI: 10.3389/fped.2020.00593] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/11/2020] [Indexed: 11/23/2022] Open
Abstract
Background: Preterm infants experience rapid brain growth during early post-natal life making them vulnerable to drugs acting on central nervous system. Morphine is administered to premature neonates for pain control and caffeine for apnea of prematurity. Simultaneous use of morphine and caffeine is common in the neonatal intensive care unit. Prior studies have shown acute neurotoxicity with this combination, however, little information is available on the mechanisms mediating the neurotoxic effects. The objective of this study was to determine the effects of morphine and caffeine, independently and in combination on mitochondrial dysfunction (Drp1 and Mfn2), neural apoptosis (Bcl-2, Bax, and cell damage) and endothelin (ET) receptors (ETA and ETB) in neonatal rat brain. Methods: Male and female rat pups were grouped separately and were divided into four different subgroups on the basis of treatments-saline (Control), morphine (MOR), caffeine (CAFF), and morphine + caffeine (M+C) treatment. Pups in MOR group were injected with 2 mg/kg morphine, CAFF group received 100 mg/kg caffeine, and M+C group received both morphine (2 mg/kg) and caffeine (100 mg/kg), subcutaneously on postnatal days (PND) 3-6. Pups were euthanized at PND 7, 14, or 28. Brains were isolated and analyzed for mitochondrial dysfunction, apoptosis markers, cell damage, and ET receptor expression via immunofluorescence and western blot analyses. Results: M+C showed a significantly higher expression of Bax compared to CAFF or MOR alone at PND 7, 14, 28 in female pups (p < 0.05) and at PND 7, 14 in male pups (p < 0.05). Significantly (p < 0.05) increased expression of Drp1, Bax, and suppressed expression of Mfn2, Bcl-2 at PND 7, 14, 28 in all the treatment groups compared to the control was observed in both genders. No significant difference in the expression of ETA and ETB receptors in male or female pups was seen at PND 7, 14, and 28. Conclusion: Concurrent use of morphine and caffeine during the first week of life increases apoptosis and cell damage in the developing brain compared to individual use of caffeine and morphine.
Collapse
Affiliation(s)
- Sweatha Kasala
- Division of Neonatology, Department of Pediatrics, Advocate Children's Hospital, Park Ridge, IL, United States
| | - Seema Briyal
- Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, United States
| | - Preetha Prazad
- Division of Neonatology, Department of Pediatrics, Advocate Children's Hospital, Park Ridge, IL, United States
| | - Amaresh K Ranjan
- Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, United States
| | - Gospodin Stefanov
- Division of Neonatology, Department of Pediatrics, Advocate Children's Hospital, Park Ridge, IL, United States
| | - Ramona Donovan
- Advocate Aurora Research Institute, Park Ridge, IL, United States
| | - Anil Gulati
- Chicago College of Pharmacy, Midwestern University, Downers Grove, IL, United States.,Pharmazz Inc. Research and Development, Willowbrook, IL, United States
| |
Collapse
|
42
|
Schiller RM, Allegaert K, Hunfeld M, van den Bosch GE, van den Anker J, Tibboel D. Analgesics and Sedatives in Critically Ill Newborns and Infants: The Impact on Long-Term Neurodevelopment. J Clin Pharmacol 2019; 58 Suppl 10:S140-S150. [PMID: 30248203 DOI: 10.1002/jcph.1139] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/23/2018] [Indexed: 12/12/2022]
Abstract
Inadequate pain and/or stress management in preterm- and term-born infants has been associated with increased morbidity and even mortality. However, exposure to analgosedatives during early infancy may also be one of the risk factors for subsequent neurodevelopmental impairment, at least in animal studies. Because infants admitted to neonatal or pediatric intensive care units may receive high amounts of these drugs for prolonged periods of time and the majority of these infants nowadays survive to discharge, this is of major concern. A balanced approach that incorporates the assessment and quantification of both wanted effects as well as unwanted side effects is therefore needed. In this article, the optimal dose determination of commonly used analgosedative drugs as well as their potential long-term effects on the developing human brain and neuropsychological functioning are reviewed.
Collapse
Affiliation(s)
- R M Schiller
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - K Allegaert
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - Maayke Hunfeld
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - G E van den Bosch
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands
| | - John van den Anker
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands.,Division of Pediatric Pharmacology and Pharmacometrics, University Children's Hospital Basel, Switzerland.,Division of Clinical Pharmacology, Children's National Health System, Washington, DC, USA
| | - D Tibboel
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, the Netherlands
| |
Collapse
|
43
|
Leeuwen L, Schiller RM, Rietman AB, van Rosmalen J, Wildschut ED, Houmes RJM, Tibboel D, IJsselstijn H. Risk Factors of Impaired Neuropsychologic Outcome in School-Aged Survivors of Neonatal Critical Illness. Crit Care Med 2019; 46:401-410. [PMID: 29194146 DOI: 10.1097/ccm.0000000000002869] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Until now, long-term outcome studies have focused on general cognitive functioning and its risk factors following neonatal extracorporeal membrane oxygenation and/or congenital diaphragmatic hernia. However, it is currently unknown which neuropsychological domains are most affected in these patients and which clinical variables can be used to predict specific neuropsychological problems. This study aimed to identify affected neuropsychological domains and its clinical determinants in survivors of neonatal extracorporeal membrane oxygenation and/or congenital diaphragmatic hernia. DESIGN Prospective follow-up study. SETTING Tertiary university hospital. PATIENTS Sixty-five 8-year-old survivors of neonatal extracorporeal membrane oxygenation and/or congenital diaphragmatic hernia. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Intelligence, attention, memory, executive functioning and visuospatial processing were evaluated using validated tests and compared with Dutch reference data. Assessed risk factors of outcome were illness severity indicators, number of anesthetic procedures in the first year of life, and growth at 1 year. Patients had average intelligence (mean intelligence quotient ± SD, 95 ± 16), but significantly poorer sustained attention (mean z score ± SD, -2.73 ± 2.57), verbal (immediate, -1.09 ± 1.27; delayed, -1.14 ± 1.86), and visuospatial memory (immediate, -1.48 ± 1.02; delayed, -1.57 ± 1.01; recognition, -1.07 ± 3.10) than the norm. Extracorporeal membrane oxygenation-treated congenital diaphragmatic hernia patients had significantly lower mean intelligence quotient (84 ± 12) than other neonatal extracorporeal membrane oxygenation patients (94 ± 10) and congenital diaphragmatic hernia patients not treated with extracorporeal membrane oxygenation (100 ± 20). Maximum vasoactive-inotropic score was negatively associated with delayed verbal (B = -0.02; 95% CI, -0.03 to -0.002; p = 0.026) and visuospatial memory (B = -0.01; 95% CI, -0.02 to -0.001; p = 0.024). CONCLUSIONS We found memory and attention deficits in 8-year-old neonatal extracorporeal membrane oxygenation and congenital diaphragmatic hernia survivors. The maximum dose of vasoactive medication was negatively associated with verbal and visuospatial memory, which may suggest an effect of early cerebral hypoperfusion in determining these abnormalities.
Collapse
Affiliation(s)
- Lisette Leeuwen
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Raisa M Schiller
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - André B Rietman
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | | | - Enno D Wildschut
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Robert Jan M Houmes
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Dick Tibboel
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Hanneke IJsselstijn
- Intensive Care and Department of Pediatric Surgery, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| |
Collapse
|
44
|
Stolp HB, Fleiss B, Arai Y, Supramaniam V, Vontell R, Birtles S, Yates AG, Baburamani AA, Thornton C, Rutherford M, Edwards AD, Gressens P. Interneuron Development Is Disrupted in Preterm Brains With Diffuse White Matter Injury: Observations in Mouse and Human. Front Physiol 2019; 10:955. [PMID: 31417418 PMCID: PMC6683859 DOI: 10.3389/fphys.2019.00955] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 07/09/2019] [Indexed: 12/18/2022] Open
Abstract
Preterm brain injury, occurring in approximately 30% of infants born <32 weeks gestational age, is associated with an increased risk of neurodevelopmental disorders, such as autism spectrum disorder (ASD) and attention deficit hyperactivity disorder (ADHD). The mechanism of gray matter injury in preterm born children is unclear and likely to be multifactorial; however, inflammation, a high predictor of poor outcome in preterm infants, has been associated with disrupted interneuron maturation in a number of animal models. Interneurons are important for regulating normal brain development, and disruption in interneuron development, and the downstream effects of this, has been implicated in the etiology of neurodevelopmental disorders. Here, we utilize postmortem tissue from human preterm cases with or without diffuse white matter injury (WMI; PMA range: 23+2 to 28+1 for non-WMI group, 26+6 to 30+0 for WMI group, p = 0.002) and a model of inflammation-induced preterm diffuse white matter injury (i.p. IL-1β, b.d., 10 μg/kg/injection in male CD1 mice from P1–5). Data from human preterm infants show deficits in interneuron numbers in the cortex and delayed growth of neuronal arbors at this early stage of development. In the mouse, significant reduction in the number of parvalbumin-positive interneurons was observed from postnatal day (P) 10. This decrease in parvalbumin neuron number was largely rectified by P40, though there was a significantly smaller number of parvalbumin positive cells associated with perineuronal nets in the upper cortical layers. Together, these data suggest that inflammation in the preterm brain may be a contributor to injury of specific interneuron in the cortical gray matter. This may represent a potential target for postnatal therapy to reduce the incidence and/or severity of neurodevelopmental disorders in preterm infants.
Collapse
Affiliation(s)
- Helen B Stolp
- Department for Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom.,Department of Perinatal Imaging & Health, Centre for the Developing Brain, School of Biomedical Engineering and Imaging Science, King's College London, London, United Kingdom
| | - Bobbi Fleiss
- Department of Perinatal Imaging & Health, Centre for the Developing Brain, School of Biomedical Engineering and Imaging Science, King's College London, London, United Kingdom.,Université de Paris, NeuroDiderot, Inserm, Paris, France.,School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, Australia
| | - Yoko Arai
- Université de Paris, NeuroDiderot, Inserm, Paris, France
| | - Veena Supramaniam
- Department of Perinatal Imaging & Health, Centre for the Developing Brain, School of Biomedical Engineering and Imaging Science, King's College London, London, United Kingdom
| | - Regina Vontell
- Department of Perinatal Imaging & Health, Centre for the Developing Brain, School of Biomedical Engineering and Imaging Science, King's College London, London, United Kingdom.,Department of Neurology, University of Miami, Miller School of Medicine, Miami, FL, United States
| | - Sebastian Birtles
- Department of Perinatal Imaging & Health, Centre for the Developing Brain, School of Biomedical Engineering and Imaging Science, King's College London, London, United Kingdom
| | - Abi G Yates
- Department of Perinatal Imaging & Health, Centre for the Developing Brain, School of Biomedical Engineering and Imaging Science, King's College London, London, United Kingdom.,Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Ana A Baburamani
- Department of Perinatal Imaging & Health, Centre for the Developing Brain, School of Biomedical Engineering and Imaging Science, King's College London, London, United Kingdom
| | - Claire Thornton
- Department for Comparative Biomedical Sciences, Royal Veterinary College, London, United Kingdom.,Department of Perinatal Imaging & Health, Centre for the Developing Brain, School of Biomedical Engineering and Imaging Science, King's College London, London, United Kingdom
| | - Mary Rutherford
- Department of Perinatal Imaging & Health, Centre for the Developing Brain, School of Biomedical Engineering and Imaging Science, King's College London, London, United Kingdom
| | - A David Edwards
- Department of Perinatal Imaging & Health, Centre for the Developing Brain, School of Biomedical Engineering and Imaging Science, King's College London, London, United Kingdom
| | - Pierre Gressens
- Department of Perinatal Imaging & Health, Centre for the Developing Brain, School of Biomedical Engineering and Imaging Science, King's College London, London, United Kingdom.,Université de Paris, NeuroDiderot, Inserm, Paris, France
| |
Collapse
|
45
|
Usuda H, Watanabe S, Saito M, Sato S, Musk GC, Fee ME, Carter S, Kumagai Y, Takahashi T, Kawamura MS, Hanita T, Kure S, Yaegashi N, Newnham JP, Kemp MW. Successful use of an artificial placenta to support extremely preterm ovine fetuses at the border of viability. Am J Obstet Gynecol 2019; 221:69.e1-69.e17. [PMID: 30853365 DOI: 10.1016/j.ajog.2019.03.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/26/2019] [Accepted: 03/04/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Ex vivo uterine environment therapy is an experimental life support platform designed to reduce the risk of morbidity and mortality for extremely preterm infants born at the border of viability (21-24 weeks' gestation). To spare the functionally immature lung, this platform performs gas exchange via a membranous oxygenator connected to the umbilical vessels, and the fetus is submerged in a protective bath of artificial amniotic fluid. We and others have demonstrated the feasibility of extended survival with ex vivo uterine environment therapy therapy in late preterm fetuses; however, there is presently no evidence to show that the use of such a platform can support extremely preterm fetuses, the eventual translational target for therapy of this nature. OBJECTIVE The objective of the study was to use our ex vivo uterine environment therapy platform to support the healthy maintenance of 600-700 g/95 days gestational age (equivalent to 24 weeks of human gestation) sheep fetuses. Primary outcome measures were as follows: (1) maintenance of key physiological variables; (2) absence of infection; (3) absence of brain injury; and (4) growth and cardiovascular function patterns matching that of noninstrumented, age-matched in utero controls. STUDY DESIGN Singleton fetuses from 8 ewes underwent surgical delivery at 95 days' gestation (term, 150 days). Fetuses were adapted to ex vivo uterine environment therapy and maintained for 120 hours with real-time monitoring of key physiological variables. Umbilical artery blood samples were regularly collected to assess blood gas data, differential counts, inflammation, and microbial load to exclude infection. Brain injury was evaluated by gross anatomical and histopathological approaches after euthanasia. Nine pregnant control animals were euthanized at 100 days' gestation to allow comparative postmortem analyses. Data were tested for mean differences with an analysis of variance. RESULTS Seven of 8 ex vivo uterine environment group fetuses (87.5%) completed 120 hours of therapy with key parameters maintained in a normal physiological range. There were no significant intergroup differences (P > .05) in final weight, crown-rump length, and body weight-normalized lung and brain weights at euthanasia compared with controls. There were no biologically significant differences in hematological parameters (total or differential leucocyte counts and plasma concentration of tumor necrosis factor-α and monocyte chemoattractant protein 1) (P > .05). Daily blood cultures were negative for aerobic and anaerobic growth in all ex vivo uterine environment animals. There was no difference in airspace consolidation between control and ex vivo uterine environment animals, and there was no increase in the number of lung cells staining positive for the T-cell marker CD3. There were no increases in interleukin-1, interleukin-6, interleukin-8, tumor necrosis factor-α, and monocyte chemoattractant protein 1 mRNA expression in lung tissues compared with the control group. No cases of intraventricular hemorrhage were observed, and white matter injury was identified in only 1 ex vivo uterine environment fetus. CONCLUSION For several decades, there has been little improvement in outcomes of extremely preterm infants born at the border of viability. In the present study, we report the use of artificial placenta technology to support, for the first time, extremely preterm ovine fetuses (equivalent to 24 weeks of human gestation) in a stable, growth-normal state for 120 hours. With additional refinement, the data generated by this study may inform a treatment option to improve outcomes for extremely preterm infants.
Collapse
|
46
|
Li Y, Meng DH, Wei QF, Pan XN, Liang WH, Huang HY, Zhen H, Zhang SY, Wei Y, Wu CB, Wei YC, Zhou JX, Lu GX. Neurodevelopmental outcomes of extremely preterm infants in southern China: A multicenter study. Early Hum Dev 2019; 133:5-10. [PMID: 30991237 DOI: 10.1016/j.earlhumdev.2019.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 02/11/2019] [Accepted: 04/01/2019] [Indexed: 10/27/2022]
Abstract
OBJECTIVE To assess treatment outcomes and associated factors of extremely preterm infants (EPIs) in GuangXi, China. METHODS This was a retrospective study consisting of 131 eligible cases with gestational age (GA) between 22 and 28 weeks, and infants were followed until 18-24 months. Data including clinical characteristics, perinatal factors and after-birth conditions were collected from the neonatal intensive care unit in 10 hospitals in Guangxi from January 1st 2010 until May 31st 2016. RESULTS During that period, 307 EPIs were born in the hospitals. 137 infants died in hospital after their parents decided to withdraw clinical treatment, and 11 infants died despite full resuscitation was provided. Of the 159 surviving infants, 28 infants were lost to follow-up. In total, 131 infants who survived and were presented to follow-up at 18-24 months of age were enrolled into this study. Of the 131 infants evaluated at 18-24 months follow-up, 47 (35.9%) were diagnosed with neurodevelopmental disability (ND), and 84 (64%) demonstrated on tract motor and language skills. The incidence of chorioamnionitis, early onset sepsis (EOS), bronchopulmonary dysplasia (BPD) were all higher in the group of infants who were diagnosed with ND compared to those with normal motor language development (NML), the duration of mechanical ventilation (MV) was longer in ND group, and the higher incidence of ND was seen in the smaller GA babies (p < 0.05). Adjusted the BPD severity, GA was a protective factor of neurodevelopmental outcome (combined OR = 0.338, 95% CI: 0.145-0.791). In EPIs with moderate BPD and severe BPD, chorioamnionitis was a risk factor of ND (OR = 10.313 and 5.778,respectively, 95% CI: 1.389-6.486 and 1.444-23.119, respectively). The Logistic regression analysis showed that GA (OR = 0.207, 95%CI = 0.047-0.917) was a protective factor for ND, and chorioamnionitis (OR = 6.010, 95%CI: 1.331-27.138), moderate-to-severe BPD (OR = 4.285, 95%CI: 1.495-12.287), the duration of MV (OR = 3.508, 95%CI: 2.077-5.926) were independent risk factors for ND in EPIs. CONCLUSIONS Chorioamnionitis, moderate-to-severe BPD, and the duration of MV were associated with neurodevelopmental disability in EPIs. The smaller the GA, the higher incidence of neurodevelopmental disability.
Collapse
Affiliation(s)
| | - Yan Li
- Department of Neonatology, Guangxi Maternal and Child Health Hospital, Nanning, 530003, Guangxi, China
| | - Dan-Hua Meng
- Department of Neonatology, Guangxi Maternal and Child Health Hospital, Nanning, 530003, Guangxi, China
| | - Qiu-Fen Wei
- Department of Neonatology, Guangxi Maternal and Child Health Hospital, Nanning, 530003, Guangxi, China
| | - Xin-Nian Pan
- Department of Neonatology, Guangxi Maternal and Child Health Hospital, Nanning, 530003, Guangxi, China.
| | - Wu-Hua Liang
- Department of Neonatology, Yulin Maternity and Child Health Care Hospital of Guangxi, Yulin, China
| | - Hai-Yan Huang
- Department of Neonatology, Qinzhou Maternity and Child Health Care Hospital of Guangxi, Qinzhou, China
| | - Hong Zhen
- Department of Neonatology, The People Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Shu-Ying Zhang
- Department of Neonatology, Nanning Maternity and Child Health Care Hospital, Nanning, China
| | - Yi Wei
- Department of Neonatology, Guilin Maternity and Child Health Care Hospital of Guangxi, Guilin, China
| | - Chao-Bo Wu
- Department of Neonatology, Minzu Hospital of Guangxi Zhuang autonomous region,Nanning, China
| | - Yan-Cheng Wei
- Department of Neonatology, The People Hospital of Hechi, Hechi, Guangxi, China
| | - Jun-Xin Zhou
- Department of Neonatology, Bobai People Hospital of Yulin, Yulin, Guangxi, China
| | - Guo-Xiu Lu
- Department of Neonatology, The First People's Hospital of Hechi, Hechi, Guangxi, China
| |
Collapse
|
47
|
Bachnas MA, Akbar MIA, Dachlan EG, Dekker G. The role of magnesium sulfate (MgSO 4) in fetal neuroprotection. J Matern Fetal Neonatal Med 2019; 34:966-978. [PMID: 31092073 DOI: 10.1080/14767058.2019.1619688] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Prevention of neurologic disability associated with preterm birth is one of the major challenges in current perinatal medicine. Magnesium sulfate (MgSO4), the focus of this review has been proposed as major step forward for that matter. MgSO4 is easily accessible, cheap, and has been proposed as a mandatory part of the management of inevitable preterm birth. The results of the various RCT's on the use of MgSO4 for neuroprotection has been the subject of many systematic reviews, other studies focused on dosing schedules, side effects and only a few focused on exploring magnesium's mechanism of action. Meanwhile, many guidelines worldwide have plugged MgSO4 as an essential ingredient of daily best practice when managing inevitable preterm birth because it has been shown to reduce the risk of severe neurologic deficit, in particular, cerebral palsy in appropriately selected patients. The more premature, the greater benefit associated with the use of antenatal MgSO4. The dose of 4 g given intravenously 15 min continued by 1 g/h until maximum 24 h and minimum for 4 h is the standard regiment proposed in most guidelines. It should be noted, however, that a recent study found that a total dose of 64 g was associated with the maximum protective effect. Only the protocol used by the largest RCT, the BEAM trial, with a loading dose of 6 g initially followed by a 2-g/h maintenance dose, if continued for 24 h would give a total dose over 50 g. Other studies report on an increased risk of neonatal death with these high doses. Several studies expressed concerns about the risk of serious side effects for both mother and neonate. The results from the systematic review showed that the most commonly used dosage, 4 g bolus continued by 1 g/h maintenance, did not increase neonatal mortality and other suspected neonatal complication such as neonatal asphyxia, spontaneous intestinal perforation, necrotizing enterocolitis, and feeding intolerance. Giving a single bolus injection of 4 g MgSO4 for stimulating BDNF production in highly "suspicious" preterm labor, and 4 g again when preterm birth become inevitable may be best from a safety perspective and also appears to have a stronger rationale.
Collapse
Affiliation(s)
- Muhammad Adrianes Bachnas
- Maternal Fetal Medicine Division, Obstetrics and Gynecology Department, Faculty of Medicine Universitas Sebelas Maret, Dr. Moewardi General Hospital, Surakarta, Indonesia
| | - Muhammad Ilham Aldika Akbar
- Maternal Fetal Medicine Division, Obstetrics and Gynecology Department, Faculty of Medicine Universitas Airlangga, Dr.Soetomo Hospital, Universitas Airlangga Hospital, Surabaya, Surakarta, Indonesia
| | - Erry Gumilar Dachlan
- Maternal Fetal Medicine Division, Obstetrics and Gynecology Department, Faculty of Medicine Universitas Airlangga, Dr.Soetomo Hospital, Universitas Airlangga Hospital, Surabaya, Surakarta, Indonesia
| | - Gustaaf Dekker
- Obstetrics and Gynaecology Department, Lyell-McEwin Hospital, the University of Adelaide, Adelaide, Australia
| |
Collapse
|
48
|
Ceprian M, Fulton D. Glial Cell AMPA Receptors in Nervous System Health, Injury and Disease. Int J Mol Sci 2019; 20:E2450. [PMID: 31108947 PMCID: PMC6566241 DOI: 10.3390/ijms20102450] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/11/2019] [Accepted: 04/22/2019] [Indexed: 12/16/2022] Open
Abstract
Glia form a central component of the nervous system whose varied activities sustain an environment that is optimised for healthy development and neuronal function. Alpha-amino-3-hydroxy-5-methyl-4-isoxazole (AMPA)-type glutamate receptors (AMPAR) are a central mediator of glutamatergic excitatory synaptic transmission, yet they are also expressed in a wide range of glial cells where they influence a variety of important cellular functions. AMPAR enable glial cells to sense the activity of neighbouring axons and synapses, and as such many aspects of glial cell development and function are influenced by the activity of neural circuits. However, these AMPAR also render glia sensitive to elevations of the extracellular concentration of glutamate, which are associated with a broad range of pathological conditions. Excessive activation of AMPAR under these conditions may induce excitotoxic injury in glial cells, and trigger pathophysiological responses threatening other neural cells and amplifying ongoing disease processes. The aim of this review is to gather information on AMPAR function from across the broad diversity of glial cells, identify their contribution to pathophysiological processes, and highlight new areas of research whose progress may increase our understanding of nervous system dysfunction and disease.
Collapse
Affiliation(s)
- Maria Ceprian
- Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain.
- Departamento de Bioquímica y Biología Molecular, CIBERNED, IRICYS. Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - Daniel Fulton
- Neuroscience and Ophthalmology Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| |
Collapse
|
49
|
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: 22] [Impact Index Per Article: 4.4] [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
|
50
|
Bruckert L, Borchers LR, Dodson CK, Marchman VA, Travis KE, Ben-Shachar M, Feldman HM. White Matter Plasticity in Reading-Related Pathways Differs in Children Born Preterm and at Term: A Longitudinal Analysis. Front Hum Neurosci 2019; 13:139. [PMID: 31139064 PMCID: PMC6519445 DOI: 10.3389/fnhum.2019.00139] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 04/08/2019] [Indexed: 12/29/2022] Open
Abstract
Children born preterm (PT) are at risk for white matter injuries based on complications of prematurity. They learn to read but on average perform below peers born full term (FT). Studies have yet to establish whether properties of white matter pathways at the onset of learning to read are associated with individual variation later in reading development in PT children. Here, we asked whether fractional anisotropy (FA) at age 6 years is associated with reading outcome at age 8 years in PT children in the same pathways as previously demonstrated in a sample of FT children. PT (n = 34, mean gestational age = 29.5 weeks) and FT children (n = 37) completed diffusion MRI and standardized measures of non-verbal IQ, language, and phonological awareness at age 6 years. Reading skills were assessed at age 8 years. Mean tract-FA was extracted from pathways that predicted reading outcome in children born FT: left arcuate fasciculus (Arc), bilateral superior longitudinal fasciculus (SLF), and left inferior cerebellar peduncle (ICP). We explored associations in additional pathways in the PT children: bilateral inferior fronto-occipital fasciculus, inferior longitudinal fasciculus, and uncinate fasciculus. Linear regression models examined whether the prediction of reading outcome at age 8 years based on mean tract-FA at age 6 years was moderated by birth group. Children born PT and FT did not differ significantly in tract-FA at age 6 years or in reading at age 8 years. Sex, socioeconomic status, and non-verbal IQ at age 6 years were associated with reading outcome and were included as covariates in all models. Birth group status significantly moderated associations between reading outcome and mean tract-FA only in the left Arc, right SLF, and left ICP, before and after consideration of pre-literacy skills. Microstructural properties of these cerebral and cerebellar pathways predicted later reading outcome in FT but not in PT children. Children born PT may rely on alternative pathways to achieve fluent reading. These findings have implications for plasticity of neural organization after early white matter injury.
Collapse
Affiliation(s)
- Lisa Bruckert
- The Developmental-Behavioral Pediatrics Research Group, Division of Developmental-Behavioral Pediatrics, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, United States
| | - Lauren R Borchers
- The Developmental-Behavioral Pediatrics Research Group, Division of Developmental-Behavioral Pediatrics, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, United States
| | - Cory K Dodson
- The Developmental-Behavioral Pediatrics Research Group, Division of Developmental-Behavioral Pediatrics, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, United States
| | - Virginia A Marchman
- Language Learning Lab, Center for Infant Studies, Department of Psychology, Stanford University, Stanford, CA, United States
| | - Katherine E Travis
- The Developmental-Behavioral Pediatrics Research Group, Division of Developmental-Behavioral Pediatrics, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, United States
| | - Michal Ben-Shachar
- The Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, Israel.,Department of English Literature and Linguistics, Bar-Ilan University, Ramat Gan, Israel
| | - Heidi M Feldman
- The Developmental-Behavioral Pediatrics Research Group, Division of Developmental-Behavioral Pediatrics, Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|