1
|
Bhorat I, Buchmann E, Frank K, Soma-Pillay P, Nicolaou E, Pistorius L, Smuts I. Clinical practice. S Afr Med J 2023; 113:22-24. [PMID: 37882127 DOI: 10.7196/samj.2023.v113i9.1063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Indexed: 10/27/2023] Open
Abstract
Basal ganglia and thalamus (BGT) hypoxic-ischaemic brain injury is currently the most contentious issue in cerebral palsy (CP) litigation in South Africa (SA), and merits a consensus response based on the current available international literature. BGT pattern injury is strongly associated with a preceding perinatal sentinel event (PSE), which has a sudden onset and is typically unforeseen and unpreventable. Antepartum pathologies may result in fetal priming, leading to vulnerability to BGT injury by relatively mild hypoxic insults. BGT injury may uncommonly follow a gradual-onset fetal heart rate deterioration pattern, of duration ≥1 hour. To prevent BGT injury in a clinical setting, the interval from onset of PSE to delivery must be short, as little as 10 - 20 minutes. This is difficult to achieve in any circumstances in SA. Each case needs holistic, multidisciplinary, unbiased review of all available antepartum, intrapartum and postpartum and childhood information, aiming at fair resolution without waste of time and resources.
Collapse
Affiliation(s)
- I Bhorat
- Subdepartment of Fetal Medicine, Department of Obstetrics and Gynaecology, Faculty of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| | - E Buchmann
- Department of Obstetrics and Gynaecology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - K Frank
- Department of Obstetrics and Gynaecology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - P Soma-Pillay
- Department of Obstetrics and Gynaecology, Faculty of Health Sciences, University of Pretoria, South Africa.
| | - E Nicolaou
- Division of Fetal Medicine, Department of Obstetrics and Gynaecology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - L Pistorius
- Division of Fetal Medicine, Department of Obstetrics and Gynaecology, Stellenbosch University, Cape Town, South Africa.
| | - I Smuts
- Department of Paediatric Neurology, Faculty of Health Sciences, University of Pretoria, South Africa.
| |
Collapse
|
2
|
Guan Y, Zhou H, Luo B, Hussain S, Xiong L. Research progress of neonatal hypoxic-ischemic encephalopathy in nonhuman primate models. IBRAIN 2023; 9:183-194. [PMID: 37786551 PMCID: PMC10528769 DOI: 10.1002/ibra.12097] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/25/2023] [Accepted: 03/02/2023] [Indexed: 10/04/2023]
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is one of the important complications of neonatal asphyxia, which not only leads to neurological disability but also seriously threatens the life of neonates. Over the years, animal models of HIE have been a research hotspot to find ways to cope with HIE and thereby reduce the risk of neonatal death or disability in moderate-to-severe HIE. By reviewing the literature related to HIE over the years, it was found that nonhuman primates share a high degree of homology with human gross neural anatomy. The basic data on nonhuman primates are not yet complete, so it is urgent to mine and develop new nonhuman primate model data. In recent years, the research on nonhuman primate HIE models has been gradually enriched and the content is more novel. Therefore, the purpose of this review is to further summarize the methods for establishing the nonhuman primate HIE model and to better elucidate the relevance of the nonhuman primate model to humans by observing the behavioral manifestations, neuropathology, and a series of biomarkers of HIE in primates HIE. Finally, the most popular and desirable treatments studied in nonhuman primate models in the past 5 years are summarized.
Collapse
Affiliation(s)
- Yi‐Huan Guan
- School of AnesthesiologyZunyi Medical UniversityZunyiChina
| | - Hong‐Su Zhou
- Department of Experimental AnimalsKunming Medical UniversityKunmingChina
| | - Bo‐Yan Luo
- School of PharmacyZunyi Medical UniversityZunyiChina
| | - Sajid Hussain
- NUTECH School of Applied Sciences and HumanitiesNational University of TechnologyIslamabadPakistan
| | - Liu‐Lin Xiong
- School of Pharmacy and Medical Sciences, Faculty of Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| |
Collapse
|
3
|
Pregnolato S, Sabir H, Luyt K, Rienecker KDA, Isles AR, Chakkarapani E. Regulation of glutamate transport and neuroinflammation in a term newborn rat model of hypoxic–ischaemic brain injury. Brain Neurosci Adv 2022; 6:23982128221097568. [PMID: 35615059 PMCID: PMC9125068 DOI: 10.1177/23982128221097568] [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: 05/28/2021] [Accepted: 04/12/2022] [Indexed: 11/17/2022] Open
Abstract
In the newborn brain, moderate-severe hypoxia–ischaemia induces glutamate excitotoxicity and inflammation, possibly via dysregulation of candidate astrocytic glutamate transporter ( Glt1) and pro-inflammatory cytokines (e.g. Tnfα, Il1β, Il6). Epigenetic mechanisms may mediate dysregulation. Hypotheses: (1) hypoxia–ischaemia dysregulates mRNA expression of these candidate genes; (2) expression changes in Glt1 are mediated by DNA methylation changes; and (3) methylation values in brain and blood are correlated. Seven-day-old rat pups ( n = 42) were assigned to nine groups based on treatment (for each timepoint: naïve ( n = 3), sham ( n = 3), hypoxia–ischaemia ( n = 8) and timepoint for tissue collection (6, 12 and 24 h post-hypoxia). Moderate hypoxic–ischemic brain injury was induced via ligation of the left common carotid artery followed by 100 min hypoxia (8% O2, 36°C). mRNA was quantified in cortex and hippocampus for the candidate genes, myelin ( Mbp), astrocytic ( Gfap) and neuronal ( Map2) markers (qPCR). DNA methylation was measured for Glt1 in cortex and blood (bisulphite pyrosequencing). Hypoxia–ischaemia induced pro-inflammatory cytokine upregulation in both brain regions at 6 h. This was accompanied by gene expression changes potentially indicating onset of astrogliosis and myelin injury. There were no significant changes in expression or promoter DNA methylation of Glt1. This pilot study supports accumulating evidence that hypoxia–ischaemia causes neuroinflammation in the newborn brain and prioritises further expression and DNA methylation analyses focusing on this pathway. Epigenetic blood biomarkers may facilitate identification of high-risk newborns at birth, maximising chances of neuroprotective interventions.
Collapse
Affiliation(s)
- Silvia Pregnolato
- Department of Neonatal Neurology, Bristol Medical School, University of Bristol, Bristol, UK
| | - Hemmen Sabir
- Department of Neonatology and Pediatric Intensive Care, Children’s Hospital, University of Bonn, Bonn, Germany
- Department of Pediatrics I/Neonatology, University Hospital Essen, University Duisburg Essen, Essen, Germany
| | - Karen Luyt
- Department of Neonatal Neurology, Bristol Medical School, University of Bristol, Bristol, UK
| | - Kira DA Rienecker
- Department of Physical Therapy and Rehabilitation Science, University of California San Francisco, San Francisco, CA, USA
- Behavioural Genetics Group, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | - Anthony R Isles
- Behavioural Genetics Group, MRC Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, UK
| | | |
Collapse
|
4
|
Mike JK, Wu KY, White Y, Pathipati P, Ndjamen B, Hutchings RS, Losser C, Vento C, Arellano K, Vanhatalo O, Ostrin S, Windsor C, Ha J, Alhassen Z, Goudy BD, Vali P, Lakshminrusimha S, Gobburu JVS, Long-Boyle J, Chen P, Wu YW, Fineman JR, Ferriero DM, Maltepe E. Defining longer term outcomes in an ovine model of moderate perinatal hypoxia-ischemia. Dev Neurosci 2022; 44:277-294. [PMID: 35588703 DOI: 10.1159/000525150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 04/06/2022] [Indexed: 11/19/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is the leading cause of neonatal morbidity and mortality worldwide. Approximately 1 million infants born with HIE each year survive with cerebral palsy (CP) and/or serious cognitive disabilities. While infants born with mild and severe HIE frequently result in predictable outcomes, infants born with moderate HIE exhibit variable outcomes that are highly unpredictable. Here, we describe an umbilical cord occlusion (UCO) model of moderate HIE with a 6-day follow-up. Near term lambs (n=27) are resuscitated after the induction of 5 minutes of asystole. Following recovery, lambs are assessed to define neurodevelopmental outcomes. At the end of this period, lambs are euthanized, and brains harvested for histological analysis. Compared with prior models that typically follow lambs for 3 days, the observation of neurobehavioral outcomes for 6 days enables identification of animals that recover significant neurological function. Approximately 35 % of lambs exhibited severe motor deficits throughout the entirety of the 6-day course and, in the most severely affected lambs, developed spastic diparesis similar to that observed in infants who survive severe neonatal HIE (severe, UCOs). Importantly, and similar to outcomes in human neonates, while initially developing significant acidosis and encephalopathy, the remainder of the lambs in this model recovered normal motor activity and exhibited normal neurodevelopmental outcomes by 6 days of life (improved, UCOi). The UCOs group exhibited gliosis and inflammation in both white and gray matter, oligodendrocyte loss, and neuronal loss and cellular death in the hippocampus and cingulate cortex. While the UCOi group exhibited more cellular death and gliosis in the parasagittal cortex and demonstrated more preserved white matter markers, along with reduced markers of inflammation and lower cellular death and neuronal loss in Ca3 of the hippocampus compared with UCOs lambs. Our large animal model of moderate HIE with prolonged follow-up will help further define pathophysiologic drivers of brain injury while enabling identification of predictive biomarkers that correlate with disease outcomes and ultimately help support development of therapeutic approaches to this challenging clinical scenario.
Collapse
Affiliation(s)
- Jana Krystofova Mike
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Katherine Y Wu
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Yasmine White
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Praneeti Pathipati
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Blaise Ndjamen
- Histology and Microscopy Core, Gladstone Institutes University of California San Francisco, San Francisco, California, USA
| | - Rachel S Hutchings
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Courtney Losser
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Christian Vento
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Kimberly Arellano
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Oona Vanhatalo
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Samuel Ostrin
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Christine Windsor
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Janica Ha
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Ziad Alhassen
- Department of Pediatrics, University of California Davis, Davis, California, USA
| | - Brian D Goudy
- Department of Pediatrics, University of California Davis, Davis, California, USA
| | - Payam Vali
- Department of Pediatrics, University of California Davis, Davis, California, USA
| | | | - Jogarao V S Gobburu
- School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
- Initiative for Pediatric Drug and Device Development, San Francisco, California, USA
| | - Janel Long-Boyle
- Initiative for Pediatric Drug and Device Development, San Francisco, California, USA
- School of Pharmacy, University of California San Francisco, San Francisco, California, USA
| | - Peggy Chen
- Department of Pediatrics, University of California Davis, Davis, California, USA
| | - Yvonne W Wu
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
- Initiative for Pediatric Drug and Device Development, San Francisco, California, USA
| | - Donna M Ferriero
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Emin Maltepe
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
- Initiative for Pediatric Drug and Device Development, San Francisco, California, USA
- Department of Biomedical Sciences, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
5
|
Mota-Rojas D, Villanueva-García D, Solimano A, Muns R, Ibarra-Ríos D, Mota-Reyes A. Pathophysiology of Perinatal Asphyxia in Humans and Animal Models. Biomedicines 2022; 10:347. [PMID: 35203556 PMCID: PMC8961792 DOI: 10.3390/biomedicines10020347] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 12/16/2022] Open
Abstract
Perinatal asphyxia is caused by lack of oxygen delivery (hypoxia) to end organs due to an hypoxemic or ischemic insult occurring in temporal proximity to labor (peripartum) or delivery (intrapartum). Hypoxic-ischemic encephalopathy is the clinical manifestation of hypoxic injury to the brain and is usually graded as mild, moderate, or severe. The search for useful biomarkers to precisely predict the severity of lesions in perinatal asphyxia and hypoxic-ischemic encephalopathy (HIE) is a field of increasing interest. As pathophysiology is not fully comprehended, the gold standard for treatment remains an active area of research. Hypothermia has proven to be an effective neuroprotective strategy and has been implemented in clinical routine. Current studies are exploring various add-on therapies, including erythropoietin, xenon, topiramate, melatonin, and stem cells. This review aims to perform an updated integration of the pathophysiological processes after perinatal asphyxia in humans and animal models to allow us to answer some questions and provide an interim update on progress in this field.
Collapse
Affiliation(s)
- Daniel Mota-Rojas
- Neurophysiology, Behavior and Animal Welfare Assessment, Universidad Autónoma Metropolitana (UAM), Mexico City 04960, Mexico
| | - Dina Villanueva-García
- Division of Neonatology, National Institute of Health Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico;
| | - Alfonso Solimano
- Department of Pediatrics, University of British Columbia, Vancouver, BC V6H 3V4, Canada;
| | - Ramon Muns
- Livestock Production Sciences Unit, Agri-Food and Biosciences Institute, Hillsborough BT26 6DR, UK;
| | - Daniel Ibarra-Ríos
- Division of Neonatology, National Institute of Health Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico;
| | - Andrea Mota-Reyes
- School of Medicine and Health Sciences, TecSalud, Instituto Tecnológico y de Estudios Superiores de Monterrey (ITESM), Monterrey 64849, Mexico;
| |
Collapse
|
6
|
Wisnowski JL, Wintermark P, Bonifacio SL, Smyser CD, Barkovich AJ, Edwards AD, de Vries LS, Inder TE, Chau V. Neuroimaging in the term newborn with neonatal encephalopathy. Semin Fetal Neonatal Med 2021; 26:101304. [PMID: 34736808 PMCID: PMC9135955 DOI: 10.1016/j.siny.2021.101304] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Neuroimaging is widely used to aid in the diagnosis and clinical management of neonates with neonatal encephalopathy (NE). Yet, despite widespread use clinically, there are few published guidelines on neuroimaging for neonates with NE. This review outlines the primary patterns of brain injury associated with hypoxic-ischemic injury in neonates with NE and their frequency, associated neuropathological features, and risk factors. In addition, it provides an overview of neuroimaging methods, including the most widely used scoring systems used to characterize brain injury in these neonates and their utility as predictive biomarkers. Last, recommendations for neuroimaging in neonates with NE are presented.
Collapse
Affiliation(s)
- Jessica L Wisnowski
- Departments of Radiology and Pediatrics (Neonatology), Children's Hospital Los Angeles, 4650 Sunset Blvd. MS #81, Los Angeles CA 90027, USA.
| | - Pia Wintermark
- Department of Pediatrics (Neonatology), McGill University/Montreal Children's Hospital, Division of Newborn Medicine, Research Institute of the McGill University Health Centre, 1001 boul. Décarie, Site Glen Block E, EM0.3244, Montréal, QC H4A 3J1, Canada.
| | - Sonia L Bonifacio
- Division of Neonatal and Developmental Medicine, Department of Pediatrics (Neonatology), Lucile Packard Children's Hospital, Stanford University School of Medicine, 750 Welch Road, Suite 315, Palo Alto, CA 94304, USA.
| | - Christopher D Smyser
- Departments of Neurology, Radiology, and Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8111, St. Louis, MO 63110-1093, USA.
| | - A James Barkovich
- Department of Radiology, UCSF Benioff Children's Hospital, University of California San Francisco, 505 Parnassus Avenue, M-391, San Francisco, CA 94143-0628, USA.
| | - A David Edwards
- Evelina London Children's Hospital, Centre for Developing Brain, King's College London, Westminster Bridge Road, London, SE1 7EH, United Kingdom.
| | - Linda S de Vries
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Lundlaan 6, 3584 EA, Utrecht, the Netherlands.
| | - Terrie E Inder
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Vann Chau
- Department of Pediatrics (Neurology), The Hospital for Sick Children, University of Toronto, 555 University Avenue, Room 6513, Toronto, ON M5G 1X8, Canada.
| |
Collapse
|
7
|
Wisnowski JL, Bluml S, Panigrahy A, Mathur AM, Berman J, Chen PSK, Dix J, Flynn T, Fricke S, Friedman SD, Head HW, Ho CY, Kline-Fath B, Oveson M, Patterson R, Pruthi S, Rollins N, Ramos YM, Rampton J, Rusin J, Shaw DW, Smith M, Tkach J, Vasanawala S, Vossough A, Whitehead MT, Xu D, Yeom K, Comstock B, Heagerty PJ, Juul SE, Wu YW, McKinstry RC. Integrating neuroimaging biomarkers into the multicentre, high-dose erythropoietin for asphyxia and encephalopathy (HEAL) trial: rationale, protocol and harmonisation. BMJ Open 2021; 11:e043852. [PMID: 33888528 PMCID: PMC8070884 DOI: 10.1136/bmjopen-2020-043852] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
INTRODUCTION MRI and MR spectroscopy (MRS) provide early biomarkers of brain injury and treatment response in neonates with hypoxic-ischaemic encephalopathy). Still, there are challenges to incorporating neuroimaging biomarkers into multisite randomised controlled trials. In this paper, we provide the rationale for incorporating MRI and MRS biomarkers into the multisite, phase III high-dose erythropoietin for asphyxia and encephalopathy (HEAL) Trial, the MRI/S protocol and describe the strategies used for harmonisation across multiple MRI platforms. METHODS AND ANALYSIS Neonates with moderate or severe encephalopathy enrolled in the multisite HEAL trial undergo MRI and MRS between 96 and 144 hours of age using standardised neuroimaging protocols. MRI and MRS data are processed centrally and used to determine a brain injury score and quantitative measures of lactate and n-acetylaspartate. Harmonisation is achieved through standardisation-thereby reducing intrasite and intersite variance, real-time quality assurance monitoring and phantom scans. ETHICS AND DISSEMINATION IRB approval was obtained at each participating site and written consent obtained from parents prior to participation in HEAL. Additional oversight is provided by an National Institutes of Health-appointed data safety monitoring board and medical monitor. TRIAL REGISTRATION NUMBER NCT02811263; Pre-result.
Collapse
Affiliation(s)
- Jessica L Wisnowski
- Radiology, Children's Hospital of Los Angeles, Los Angeles, California, USA
- Pediatrics, Children's Hospital Los Angeles Division of Neonatology, Los Angeles, California, USA
| | - Stefan Bluml
- Radiology, Children's Hospital of Los Angeles, Los Angeles, California, USA
| | - Ashok Panigrahy
- Radiology, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Amit M Mathur
- Pediatrics, Division of Neonatal-Perinatal Medicine, SSM Health Cardinal Glennon Children's Hospital, Saint Louis, Missouri, USA
- Pediatrics, Division of Neonatal-Perinatal Medicine, Saint Louis University, Saint Louis, Missouri, USA
| | - Jeffrey Berman
- Radiology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | - James Dix
- Radiology, Methodist Children's Hospital, San Antonio, Texas, USA
| | - Trevor Flynn
- Radiology, University of California San Francisco, San Francisco, California, USA
| | - Stanley Fricke
- Radiology, Children's National Medical Center, Washington, District of Columbia, USA
- Radiology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Seth D Friedman
- Radiology, Seattle Children's Hospital, Seattle, Washington, USA
| | - Hayden W Head
- Radiology, Cook Children's Medical Center, Fort Worth, Texas, USA
| | - Chang Y Ho
- Radiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Beth Kline-Fath
- Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Michael Oveson
- Radiology, Primary Children's Hospital, Salt Lake City, Utah, USA
| | - Richard Patterson
- Radiology, Children's Hospitals and Clinics of Minnesota, Minneapolis, Minnesota, USA
| | - Sumit Pruthi
- Radiology, Vanderbilt University, Nashville, Tennessee, USA
| | - Nancy Rollins
- Radiology, University of Texas Southwestern Medical School, Dallas, Texas, USA
| | - Yanerys M Ramos
- Radiology, Children's Hospitals and Clinics of Minnesota, Minneapolis, Minnesota, USA
| | - John Rampton
- Radiology, Primary Children's Hospital, Salt Lake City, Utah, USA
| | - Jerome Rusin
- Radiology, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Dennis W Shaw
- Radiology, Seattle Children's Hospital, Seattle, Washington, USA
| | - Mark Smith
- Radiology, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Jean Tkach
- Radiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | | | - Arastoo Vossough
- Radiology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Matthew T Whitehead
- Radiology, Children's National Medical Center, Washington, District of Columbia, USA
| | - Duan Xu
- Radiology, University of California San Francisco, San Francisco, California, USA
| | - Kristen Yeom
- Radiology, Stanford University, Stanford, California, USA
| | - Bryan Comstock
- Biostatistics, University of Washington, Seattle, Washington, USA
| | - Patrick J Heagerty
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Sandra E Juul
- Pediatrics, Division of Neonatology, University of Washington, Seattle, Washington, USA
| | - Yvonne W Wu
- Neurology, University of California San Francisco, San Francisco, California, USA
| | - Robert C McKinstry
- Radiology, St. Louis Children's Hospital and Washington University, Saint Louis, Missouri, USA
| |
Collapse
|
8
|
Debuf MJ, Carkeek K, Piersigilli F. A Metabolomic Approach in Search of Neurobiomarkers of Perinatal Asphyxia: A Review of the Current Literature. Front Pediatr 2021; 9:674585. [PMID: 34249811 PMCID: PMC8267248 DOI: 10.3389/fped.2021.674585] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/24/2021] [Indexed: 11/13/2022] Open
Abstract
Perinatal asphyxia and the possible sequelae of hypoxic-ischemic encephalopathy (HIE), are associated with high morbidity and mortality rates. The use of therapeutic hypothermia (TH) commencing within the first 6 h of life-currently the only treatment validated for the management of HIE-has been proven to reduce the mortality rate and disability seen at follow up at 18 months. Although there have been attempts to identify neurobiomarkers assessing the severity levels in HIE; none have been validated in clinical use to date, and the lack thereof limits the optimal treatment for these vulnerable infants. Metabolomics is a promising field of the "omics technologies" that may: identify neurobiomarkers, help improve diagnosis, identify patients prone to developing HIE, and potentially improve targeted neuroprotection interventions. This review focuses on the current evidence of metabolomics, a novel tool which may prove to be a useful in the diagnosis, management and treatment options for this multifactorial complex disease. Some of the most promising metabolites analyzed are the group of acylcarnitines: Hydroxybutyrylcarnitine (Malonylcarnitine) [C3-DC (C4-OH)], Tetradecanoylcarnitine [C14], L-Palmitoylcarnitine [C16], Hexadecenoylcarnitine [C16:1], Stearoylcarnitine [C18], and Oleoylcarnitine [C18:1]. A metabolomic "fingerprint" or "index," made up of 4 metabolites (succinate × glycerol/(β-hydroxybutyrate × O-phosphocholine)), seems promising in identifying neonates at risk of developing severe HIE.
Collapse
Affiliation(s)
- Marie Julie Debuf
- Division of Neonatology, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Bruxelles, Belgium
| | - Katherine Carkeek
- Division of Neonatology, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Bruxelles, Belgium
| | - Fiammetta Piersigilli
- Division of Neonatology, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Bruxelles, Belgium
| |
Collapse
|
9
|
Tanaka E, Ogawa Y, Fujii R, Shimonaka T, Sato Y, Hamazaki T, Nagamura-Inoue T, Shintaku H, Tsuji M. Metabolomic analysis and mass spectrometry imaging after neonatal stroke and cell therapies in mouse brains. Sci Rep 2020; 10:21881. [PMID: 33318553 PMCID: PMC7736587 DOI: 10.1038/s41598-020-78930-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
Ischemic brain injury provokes complex, time-dependent downstream pathways that ultimately lead to cell death. We aimed to demonstrate the levels of a wide range of metabolites in brain lysates and their on-tissue distribution following neonatal stroke and cell therapies. Postnatal day 12 mice underwent middle cerebral artery occlusion (MCAO) and were administered 1 × 105 cells after 48 h. Metabolomic analysis of the injured hemisphere demonstrated that a variety of amino acids were significantly increased and that tricarboxylic acid cycle intermediates and some related amino acids, such as glutamate, were decreased. With the exception of the changes in citric acid, neither mesenchymal stem/stromal cells nor CD34+ cells ameliorated these changes. On-tissue visualization with matrix-assisted laser desorption/ionization-mass spectrometry (MALDI-MS) imaging revealed that the signal intensity of glutamate was significantly decreased in the infarct area, consistent with the metabolomic analysis, while its intensity was significantly increased in the peri-infarct area after MCAO. Although cell therapies did not ameliorate the changes in metabolites in the infarct area, mesenchymal stem cells ameliorated the increased levels of glutamate and carnitine in the peri-infarct area. MALDI-MS imaging showed the location-specific effect of cell therapies even in this subacute setting after MCAO. These methodologies may be useful for further investigation of possible treatments for ischemic brain injury.
Collapse
Affiliation(s)
- Emi Tanaka
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yuko Ogawa
- Institute of Biomedical Research and Innovation, Kobe, Japan
| | - Ritsuko Fujii
- Division of Bioenergetics, Research Center for Artificial Photosynthesis, Osaka City University, Osaka, Japan.,Division of Molecular Materials Science, Graduate School of Science, Osaka City University, Osaka, Japan
| | - Tomomi Shimonaka
- Analysis Division, Research Center for Artificial Photosynthesis, Osaka City University, Osaka, Japan
| | - Yoshiaki Sato
- Division of Neonatology, Center for Maternal-Neonatal Care, Nagoya University Hospital, Nagoya, Japan
| | - Takashi Hamazaki
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Haruo Shintaku
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masahiro Tsuji
- Department of Food and Nutrition, Kyoto Women's University, 35 Kitahiyoshi-cho, Imakumano, Higashiyama-ku, Kyoto, 605-8501, Japan. .,Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Suita, Japan.
| |
Collapse
|
10
|
Exploring Perinatal Asphyxia by Metabolomics. Metabolites 2020; 10:metabo10040141. [PMID: 32260446 PMCID: PMC7240960 DOI: 10.3390/metabo10040141] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/25/2020] [Accepted: 04/02/2020] [Indexed: 02/06/2023] Open
Abstract
Brain damage related to perinatal asphyxia is the second cause of neuro-disability worldwide. Its incidence was estimated in 2010 as 8.5 cases per 1000 live births worldwide, with no further recent improvement even in more industrialized countries. If so, hypoxic-ischemic encephalopathy is still an issue of global health concern. It is thought that a consistent number of cases may be avoided, and its sequelae may be preventable by a prompt and efficient physical and therapeutic treatment. The lack of early, reliable, and specific biomarkers has up to now hampered a more effective use of hypothermia, which represents the only validated therapy for this condition. The urge to unravel the biological modifications underlying perinatal asphyxia and hypoxic-ischemic encephalopathy needs new diagnostic and therapeutic tools. Metabolomics for its own features is a powerful approach that may help for the identification of specific metabolic profiles related to the pathological mechanism and foreseeable outcome. The metabolomic profiles of animal and human infants exposed to perinatal asphyxia or developing hypoxic-ischemic encephalopathy have so far been investigated by means of 1H nuclear magnetic resonance spectroscopy and mass spectrometry coupled with gas or liquid chromatography, leading to the identification of promising metabolomic signatures. In this work, an extensive review of the relevant literature was performed.
Collapse
|
11
|
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
|
12
|
Torres-Cuevas I, Corral-Debrinski M, Gressens P. Brain oxidative damage in murine models of neonatal hypoxia/ischemia and reoxygenation. Free Radic Biol Med 2019; 142:3-15. [PMID: 31226400 DOI: 10.1016/j.freeradbiomed.2019.06.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/26/2019] [Accepted: 06/10/2019] [Indexed: 02/08/2023]
Abstract
The brain is one of the main organs affected by hypoxia and reoxygenation in the neonatal period and one of the most vulnerable to oxidative stress. Hypoxia/ischemia and reoxygenation leads to impairment of neurogenesis, disruption of cortical migration, mitochondrial damage and neuroinflammation. The extent of the injury depends on the clinical manifestation in the affected regions. Preterm newborns are highly vulnerable, and they exhibit severe clinical manifestations such as intraventricular hemorrhage (IVH), retinopathy of prematurity (ROP) and diffuse white matter injury (DWMI) among others. In the neonatal period, the accumulation of high levels of reactive oxygen species exacerbated by the immature antioxidant defense systems in represents cellular threats that, if they exceed or bypass physiological counteracting mechanisms, are responsible of significant neuronal damage. Several experimental models in mice mimic the consequences of perinatal asphyxia and the use of oxygen in the reanimation process that produce brain injury. The aim of this review is to highlight brain damage associated with oxidative stress in different murine models of hypoxia/ischemia and reoxygenation.
Collapse
Affiliation(s)
| | | | - Pierre Gressens
- INSERM UMR1141, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
13
|
Montaldo P, Kaforou M, Pollara G, Hervás-Marín D, Calabria I, Panadero J, Pedrola L, Lally PJ, Oliveira V, Kage A, Atreja G, Mendoza J, Soe A, Pattnayak S, Shankaran S, Vento M, Herberg J, Thayyil S. Whole Blood Gene Expression Reveals Specific Transcriptome Changes in Neonatal Encephalopathy. Neonatology 2019; 115:68-76. [PMID: 30304723 PMCID: PMC6425817 DOI: 10.1159/000492420] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 07/24/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Variable responses to hypothermic neuroprotection are related to the clinical heterogeneity of encephalopathic babies; hence better disease stratification may facilitate the development of individualized neuroprotective therapies. OBJECTIVES We examined if whole blood gene expression analysis can identify specific transcriptome profiles in neonatal encephalopathy. MATERIAL AND METHODS We performed next-generation sequencing on whole blood RNA from 12 babies with neonatal encephalopathy and 6 time-matched healthy term babies. Genes significantly differentially expressed between encephalopathic and control babies were identified. This set of genes was then compared to the host RNA response in septic neonates and subjected to pathway analysis. RESULTS We identified 950 statistically significant genes discriminating perfectly between healthy controls and neonatal encephalopathy. The major pathways in neonatal encephalopathy were axonal guidance signaling (p = 0.0009), granulocyte adhesion and diapedesis (p = 0.003), IL-12 signaling and production in macrophages (p = 0.003), and hypoxia-inducible factor 1α signaling (p = 0.004). There were only 137 genes in common between neonatal encephalopathy and bacterial sepsis sets. CONCLUSION Babies with neonatal encephalopathy have striking differences in gene expression profiles compared with healthy control and septic babies. Gene expression profiles may be useful for disease stratification and for developing personalized neuroprotective therapies.
Collapse
Affiliation(s)
- Paolo Montaldo
- Centre for Perinatal Neuroscience, Imperial College London, London, United .,Neonatal Unit, Università degli Studi della Campania "Luigi Vanvitelli,", Naples,
| | - Myrsini Kaforou
- Paediatric Infectious Diseases, Imperial College London, London, United Kingdom
| | - Gabriele Pollara
- Infection and Immunity, University College London, London, United Kingdom
| | | | | | | | - Laia Pedrola
- Health Research Institute La Fe, Valencia, Spain
| | - Peter J Lally
- Centre for Perinatal Neuroscience, Imperial College London, London, United Kingdom
| | - Vânia Oliveira
- Centre for Perinatal Neuroscience, Imperial College London, London, United Kingdom
| | - Anup Kage
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Gaurav Atreja
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Josephine Mendoza
- Centre for Perinatal Neuroscience, Imperial College London, London, United Kingdom
| | - Aung Soe
- Medway NHS Foundation Trust, Gillingham, United Kingdom
| | | | - Seetha Shankaran
- Neonatal-Perinatal Division, Wayne State University, Detroit, Michigan, USA
| | - Máximo Vento
- Health Research Institute La Fe, Valencia, Spain
| | - Jethro Herberg
- Paediatric Infectious Diseases, Imperial College London, London, United Kingdom
| | - Sudhin Thayyil
- Centre for Perinatal Neuroscience, Imperial College London, London, United Kingdom
| |
Collapse
|
14
|
Jisa KA, Clarey DD, Peeples ES. Magnetic Resonance Imaging Findings of Term and Preterm Hypoxic-Ischemic Encephalopathy: A Review of Relevant Animal Models and Correlation to Human Imaging. Open Neuroimag J 2018; 12:55-65. [PMID: 30450146 PMCID: PMC6198416 DOI: 10.2174/1874440001812010055] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 08/07/2018] [Accepted: 09/16/2018] [Indexed: 11/22/2022] Open
Abstract
Background: Neonatal hypoxic-ischemic encephalopathy is brain injury caused by decreased perfusion and oxygen delivery that most commonly occurs in the context of delivery complications such as umbilical cord compression or placental abruption. Imaging is a key component for guiding treatment and prediction of prognosis, and the most sensitive clinical imaging modality for the brain injury patterns seen in hypoxic-ischemic encephalopathy is magnetic resonance imaging. Objective: The goal of this review is to compare magnetic resonance imaging findings demonstrated in the available animal models of hypoxic-ischemic encephalopathy to those found in preterm (≤ 36 weeks) and term (>36 weeks) human neonates with hypoxic-ischemic encephalopathy, with special attention to the strengths and weaknesses of each model. Methods: A structured literature search was performed independently by two authors and the results of the searches were compiled. Animal model, human brain age equivalency, mechanism of injury, and area of brain injury were recorded for comparison to imaging findings in preterm and term human neonates with hypoxic-ischemic encephalopathy. Conclusion: Numerous animal models have been developed to better elicit the expected findings that occur after HIE by allowing investigators to control many of the clinical variables that result in injury. Although modeling the same disease process, magnetic resonance imaging findings in the animal models vary with the species and methods used to induce hypoxia and ischemia. The further development of animal models of HIE should include a focus on comparing imaging findings, and not just pathologic findings, to human studies.
Collapse
Affiliation(s)
- Kyle A Jisa
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, United States
| | - Dillon D Clarey
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, United States
| | - Eric S Peeples
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
15
|
Juul SE, Comstock BA, Heagerty PJ, Mayock DE, Goodman AM, Hauge S, Gonzalez F, Wu YW. High-Dose Erythropoietin for Asphyxia and Encephalopathy (HEAL): A Randomized Controlled Trial - Background, Aims, and Study Protocol. Neonatology 2018; 113. [PMID: 29514165 PMCID: PMC5980685 DOI: 10.1159/000486820] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Hypoxic-ischemic encephalopathy (HIE) remains an important cause of neonatal death and frequently leads to significant long-term disability in survivors. Therapeutic hypothermia, while beneficial, still leaves many treated infants with lifelong disabilities. Adjunctive therapies are needed, and erythropoietin (Epo) has the potential to provide additional neuroprotection. OBJECTIVES The aim of this study was to review the current incidence, mechanism of injury, and sequelae of HIE, and to describe a new phase III randomized, placebo-controlled trial of Epo neuroprotection in term and near-term infants with moderate to severe HIE treated with therapeutic hypothermia. METHODS This article presents an overview of HIE, neuroprotective functions of Epo, and the design of a double-blind, placebo-controlled, multicenter trial of high-dose Epo administration, enrolling 500 neonates ≥36 weeks of gestation with moderate or severe HIE diagnosed by clinical criteria. RESULTS AND CONCLUSIONS Epo has robust neuroprotective effects in preclinical studies, and phase I/II trials suggest that multiple high doses of Epo may provide neuroprotection against brain injury in term infants. The High Dose Erythropoietin for Asphyxia and Encephalopathy (HEAL) Trial will evaluate whether high-dose Epo reduces the combined outcome of death or neurodevelopmental disability when given in conjunction with hypothermia to newborns with moderate/severe HIE.
Collapse
Affiliation(s)
- Sandra E Juul
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, Washington, USA
| | - Bryan A Comstock
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Patrick J Heagerty
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Dennis E Mayock
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, Washington, USA
| | - Amy M Goodman
- Department of Neurology, Division of Child Neurology, University of California, San Francisco, California, USA
| | - Stephanie Hauge
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, Washington, USA
| | - Fernando Gonzalez
- Department of Neurology, Division of Child Neurology, University of California, San Francisco, California, USA
| | - Yvonne W Wu
- Department of Neurology, Division of Child Neurology, University of California, San Francisco, California, USA
| |
Collapse
|