1
|
Chen XF, Kroke B, Ni J, Munoz C, Appleman M, Jacobs B, Tran T, Nguyen KV, Qiu C, Stonestreet BS, Marshall J. Novel peptidomimetic compounds attenuate hypoxic-ischemic brain injury in neonatal rats. Exp Neurol 2025; 386:115151. [PMID: 39832663 PMCID: PMC11895808 DOI: 10.1016/j.expneurol.2025.115151] [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: 10/21/2024] [Revised: 01/04/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Hypoxic-ischemic (HI) brain injury is a common neurological problem in neonates. The postsynaptic density protein-95 (PSD-95) is an excitatory synaptic scaffolding protein that regulates synaptic function, and represents a potential therapeutic target to attenuate HI brain injury. Syn3 and d-Syn3 are novel high affinity cyclic peptides that bind the PDZ3 domain of PSD-95. We investigated the neuroprotective efficacy of Syn3 and d-Syn3 after exposure to HI in neonatal rodents. Postnatal (P) day-7 rats were treated with Syn3 and d-Syn3 at zero, 24, and 48-h after carotid artery ligation and 90-min of 8 % oxygen. Hemispheric volume atrophy and Iba-1 positive microglia were quantified by cresyl violet and immunohistochemical staining. Treatment with Syn3 and d-Syn3 reduced tissue volume loss by 47.0 % and 41.0 % in the male plus female, and by 42.1 % and 65.0 % in the male groups, respectively. Syn3 reduced tissue loss by 52.3 % in females. D-Syn3 reduced Iba-1 positive microglia/DAPI ratios in the pooled group, males, and females. Syn3 effects were observed in the pooled group and females. Changes in Iba-1 positive microglia/DAPI cellular ratios correlated directly with reduced hemispheric volume loss, suggesting that Syn3 and d-Syn3 provide neuroprotection in part by their effects on Iba-1 positive microglia. The pathogenic cis phosphorylated Thr231 in Tau (cis P-tau) is a marker of neuronal injury. Cis P-tau was induced in cortical cells of the placebo-treated pooled group, males and females after HI, and reduced by treatment with d-Syn3. Therefore, treatment with these peptidomimetic agents exert neuroprotective effects after exposure of neonatal subjects to HI related brain injury.
Collapse
Affiliation(s)
- Xiaodi F Chen
- Department of Pediatrics, Women & Infants Hospital of RI, The Alpert Medical School of Brown University, Providence, RI, USA.
| | - Brynn Kroke
- Department of Pediatrics, Women & Infants Hospital of RI, The Alpert Medical School of Brown University, Providence, RI, USA; Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Jun Ni
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Christian Munoz
- Department of Pediatrics, Women & Infants Hospital of RI, The Alpert Medical School of Brown University, Providence, RI, USA; Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Mark Appleman
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Bryce Jacobs
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Tuong Tran
- Department of Pediatrics, Women & Infants Hospital of RI, The Alpert Medical School of Brown University, Providence, RI, USA
| | - Kevin V Nguyen
- Department of Pediatrics, Women & Infants Hospital of RI, The Alpert Medical School of Brown University, Providence, RI, USA
| | - Chenxi Qiu
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Barbara S Stonestreet
- Department of Pediatrics, Women & Infants Hospital of RI, The Alpert Medical School of Brown University, Providence, RI, USA; Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA; Emerita, Department of Pediatrics, Women & Infants Hospital of RI, The Alpert Medical School of Brown University, Providence, RI, USA
| | - John Marshall
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA.
| |
Collapse
|
2
|
Koehn LM, Nguyen KV, Tucker R, Lim YP, Chen X, Stonestreet BS. Inter-alpha Inhibitor Proteins Modulate Microvascular Endothelial Components and Cytokines After Exposure to Hypoxia-Ischemia in Neonatal Rats. Mol Neurobiol 2025; 62:5057-5072. [PMID: 39505805 DOI: 10.1007/s12035-024-04594-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 10/25/2024] [Indexed: 11/08/2024]
Abstract
Inter-alpha inhibitor proteins (IAIPs) are neuroprotective and attenuate lipopolysaccharide (LPS)-mediated blood-brain barrier (BBB) disruption in neonatal rodents. We investigated some mechanism(s) fundamental to neuroprotection by IAIPs including changes in cerebral endothelial components and inflammation. Postnatal day-7 rats exposed to sham surgery and placebo or carotid ligation plus 8% FiO2 (90 min) were given IAIPs (30 or 60 mg/kg) or placebo and were killed 6, 12, 24, or 36 h after hypoxia-ischemia (HI). Proteins regulating BBB permeability to leukocytes (vascular cell adhesion molecule 1, VCAM-1), lipid-soluble (P-glycoprotein, PGP), and lipid-insoluble molecules (zonula occludens-1, ZO-1) were measured by immunoblot, and cytokines were measured in serum and cortex. HI resulted in reductions in ZO-1 and increases in VCAM-1, PGP, interferon-γ (IFN-γ), interleukin-12 (IL-12), vascular endothelial growth factor (VEGF), IL-α, and macrophage colony-stimulating factor (M-CSF) in cortex and increases in IL-4, IL-5, IL-10, and granulocyte colony-stimulating factor (G-CSF) in serum. IAIPs attenuated the reductions in ZO-1 and delayed increases in VCAM-1 and PGP in cortex and attenuated increases in cytokines in serum (IL-4, IL-5, IL-10, IFN-γ, G-CSF) and cortex (IL-1α, IL-12, IFN-γ, VEGF, M-CSF) after HI. We conclude that vascular endothelial proteins and cytokines exhibit sequential changes after HI and IAIPs modulate some of these HI-related changes in neonatal rats.
Collapse
Affiliation(s)
- Liam M Koehn
- Department of Pediatrics, Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
- Present Address: Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Kevin V Nguyen
- Department of Pediatrics, Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Richard Tucker
- Department of Pediatrics, Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Yow-Pin Lim
- ProThera Biologics Inc, Providence, RI, USA
- Department of Pathology and Laboratory Medicine, The Alpert Medical School of Brown University, Providence, RI, USA
| | - Xiaodi Chen
- Department of Pediatrics, Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Barbara S Stonestreet
- Department of Pediatrics, Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA.
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA.
| |
Collapse
|
3
|
Hollmén C, Parkkola R, Vorobyev V, Saunavaara J, Laitio R, Arola O, Hynninen M, Bäcklund M, Martola J, Ylikoski E, Roine RO, Tiainen M, Scheinin H, Maze M, Vahlberg T, Laitio TT. Neuroprotective Effects of Inhaled Xenon Gas on Brain Structural Gray Matter Changes After Out-of-Hospital Cardiac Arrest Evaluated by Morphometric Analysis: A Substudy of the Randomized Xe-Hypotheca Trial. Neurocrit Care 2025; 42:131-141. [PMID: 38982000 DOI: 10.1007/s12028-024-02053-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 06/14/2024] [Indexed: 07/11/2024]
Abstract
BACKGROUND We have earlier reported that inhaled xenon combined with hypothermia attenuates brain white matter injury in comatose survivors of out-of-hospital cardiac arrest (OHCA). A predefined secondary objective was to assess the effect of inhaled xenon on the structural changes in gray matter in comatose survivors after OHCA. METHODS Patients were randomly assigned to receive either inhaled xenon combined with target temperature management (33 °C) for 24 h (n = 55, xenon group) or target temperature management alone (n = 55, control group). A change of brain gray matter volume was assessed with a voxel-based morphometry evaluation of high-resolution structural brain magnetic resonance imaging (MRI) data with Statistical Parametric Mapping. Patients were scheduled to undergo the first MRI between 36 and 52 h and a second MRI 10 days after OHCA. RESULTS Of the 110 randomly assigned patients in the Xe-Hypotheca trial, 66 patients completed both MRI scans. After all imaging-based exclusions, 21 patients in the control group and 24 patients in the xenon group had both scan 1 and scan 2 available for analyses with scans that fulfilled the quality criteria. Compared with the xenon group, the control group had a significant decrease in brain gray matter volume in several clusters in the second scan compared with the first. In a between-group analysis, significant reductions were found in the right amygdala/entorhinal cortex (p = 0.025), left amygdala (p = 0.043), left middle temporal gyrus (p = 0.042), left inferior temporal gyrus (p = 0.008), left parahippocampal gyrus (p = 0.042), left temporal pole (p = 0.042), and left cerebellar cortex (p = 0.005). In the remaining gray matter areas, there were no significant changes between the groups. CONCLUSIONS In comatose survivors of OHCA, inhaled xenon combined with targeted temperature management preserved gray matter better than hypothermia alone. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov: NCT00879892.
Collapse
Affiliation(s)
- Carita Hollmén
- Department of Radiology, Turku University Hospital, University of Turku, Turku, Finland
| | - Riitta Parkkola
- Department of Radiology, Turku University Hospital, University of Turku, Turku, Finland
| | - Victor Vorobyev
- Department of Radiology, Turku University Hospital, University of Turku, Turku, Finland
| | - Jani Saunavaara
- Department of Medical Physics, Turku University Hospital, University of Turku, Turku, Finland
| | - Ruut Laitio
- Division of Perioperative Services, Intensive Care Medicine and Pain Management, Turku University Hospital, University of Turku, POB 52, 20521, Turku, Finland
| | - Olli Arola
- Division of Perioperative Services, Intensive Care Medicine and Pain Management, Turku University Hospital, University of Turku, POB 52, 20521, Turku, Finland
| | - Marja Hynninen
- Division of Intensive Care Medicine, Department of Anesthesiology, Intensive Care and Pain Medicine, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Minna Bäcklund
- Division of Intensive Care Medicine, Department of Anesthesiology, Intensive Care and Pain Medicine, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Juha Martola
- Department of Radiology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Emmi Ylikoski
- Division of Intensive Care Medicine, Department of Anesthesiology, Intensive Care and Pain Medicine, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Risto O Roine
- Division of Clinical Neurosciences, Turku University Hospital, University of Turku, Turku, Finland
| | - Marjaana Tiainen
- Department of Neurology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Harry Scheinin
- Division of Perioperative Services, Intensive Care Medicine and Pain Management, Turku University Hospital, University of Turku, POB 52, 20521, Turku, Finland
| | - Mervyn Maze
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Tero Vahlberg
- Department of Biostatistics, University of Turku and Turku University Hospital, Turku, Finland
| | - Timo T Laitio
- Division of Perioperative Services, Intensive Care Medicine and Pain Management, Turku University Hospital, University of Turku, POB 52, 20521, Turku, Finland.
| |
Collapse
|
4
|
Hu L, Geli S, Long F, Nie L, Wu J, Zhou J, Wang M, Chen Y. The 100 most-cited articles in hypothermic brain protection journals: a bibliometric and visualized analysis. Front Neurol 2024; 15:1433025. [PMID: 39563775 PMCID: PMC11575058 DOI: 10.3389/fneur.2024.1433025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/15/2024] [Indexed: 11/21/2024] Open
Abstract
Introduction A bibliometric analysis is used to assess the impact of research in a particular field. However, a specialized bibliometric analysis focused on hypothermic brain protection has not yet been conducted. This study aimed to identify the 100 most-cited articles published in the field of hypothermic brain protection and analyze their bibliometric characteristics. Methods After screening articles from the Web of Science citation database, complete bibliographic records were imported into Python for data extraction. The following parameters were analyzed: title, author's name and affiliation, country, publication year, publication date, first author, corresponding author, study design, language, number of citations, journal impact factors, keywords, Keywords Plus®, and research topic. Results The 100 articles were published between 1990 and 2016. The citation frequency for each publication ranged from 86 to 470. Among the 100 articles, 73 were original articles, 18 were review articles, 8 were clinical articles, and 1 was editorial material. These papers were published in 37 journals, with the Journal of Cerebral Blood Flow and Metabolism being the most prolific with 15 papers. Eighteen countries contributed to the 100 publications, 51 of which were from United States institutions. In addition, the keywords in the Sankey plot indicated that research in the field of hypothermic brain protection is growing deeper and overlapping with other disciplines. Discussion The results provide an overview of research on hypothermic brain protection, which may help researchers better understand classical research, historical developments, and new discoveries, as well as providing ideas for future research.
Collapse
Affiliation(s)
- Liren Hu
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Sichuan Province, China
| | - Sirui Geli
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Sichuan Province, China
| | - Feiyu Long
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Sichuan Province, China
| | - Liang Nie
- Department of Anesthesiology, Fushun County People's Hospital, Zigong, Sichuan Province, China
| | - Jiali Wu
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Sichuan Province, China
| | - Jun Zhou
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Sichuan Province, China
| | - Maohua Wang
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Sichuan Province, China
| | - Yingxu Chen
- Department of Anesthesiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- Anesthesiology and Critical Care Medicine Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Sichuan Province, China
| |
Collapse
|
5
|
Barros M, Liang M, Iannucci N, Dickinson R. Xenon and Argon as Neuroprotective Treatments for Perinatal Hypoxic-Ischemic Brain Injury: A Preclinical Systematic Review and Meta-Analysis. Anesth Analg 2024:00000539-990000000-01012. [PMID: 39453983 DOI: 10.1213/ane.0000000000007223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2024]
Abstract
Xenon and argon are currently being evaluated as potential neuroprotective treatments for acquired brain injuries. Xenon has been evaluated clinically as a treatment for brain ischemia with equivocal results in small trials, but argon has not yet undergone clinical evaluation. Several preclinical studies have investigated xenon or argon as treatments in animal models of perinatal hypoxic-ischemic encephalopathy (HIE). A systematic review of MEDLINE and Embase databases was performed. After screening of titles, abstracts, and full text, data were extracted from included studies. A pairwise meta-analysis of neuroprotective efficacy was performed using a random effects model. Heterogeneity was investigated using subgroup analysis, funnel plot asymmetry, and Egger's regression. The protocol was prospectively registered on PROSPERO (CRD42022301986). A total of 21 studies met the inclusion criteria. The data extracted included measurements from 1591 animals, involving models of HIE in mice, rats, and pigs. The meta-analysis found that both xenon and argon had significant (P < .0001) neuroprotective efficacies. The summary estimate for xenon was 39.7% (95% confidence interval [CI], 28.3%-51.1%) and for argon it was 70.3% (95% CI, 59.0%-81.7%). The summary effect for argon was significantly (P < .001) greater than that of xenon. Our results provide evidence supporting further investigation of xenon and argon as neuroprotective treatments for HIE.
Collapse
Affiliation(s)
- Mariana Barros
- From the Anaesthetics, Pain Medicine and Intensive Care Division, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Min Liang
- Anaesthesiology Research Institute, Department of Anaesthesiology, First Affiliated Hospital of Fujian Medical University, Binhai Campus, Fuzhou, China
| | - Noemi Iannucci
- From the Anaesthetics, Pain Medicine and Intensive Care Division, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Robert Dickinson
- From the Anaesthetics, Pain Medicine and Intensive Care Division, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
- Centre for Blast Injury Studies, Imperial College London, London, United Kingdom
| |
Collapse
|
6
|
Chen XF, Wu Y, Kim B, Nguyen KV, Chen A, Qiu J, Santoso AR, Disdier C, Lim YP, Stonestreet BS. Neuroprotective efficacy of hypothermia and Inter-alpha Inhibitor Proteins after hypoxic ischemic brain injury in neonatal rats. Neurotherapeutics 2024; 21:e00341. [PMID: 38453562 PMCID: PMC11070713 DOI: 10.1016/j.neurot.2024.e00341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 03/09/2024] Open
Abstract
Therapeutic hypothermia is the standard of care for hypoxic-ischemic (HI) encephalopathy. Inter-alpha Inhibitor Proteins (IAIPs) attenuate brain injury after HI in neonatal rats. Human (h) IAIPs (60 mg/kg) or placebo (PL) were given 15 min, 24 and 48 h to postnatal (P) day-7 rats after carotid ligation and 8% oxygen for 90 min with (30 °C) and without (36 °C) exposure to hypothermia 1.5 h after HI for 3 h. Hemispheric volume atrophy (P14) and neurobehavioral tests including righting reflex (P8-P10), small open field (P13-P14), and negative geotaxis (P14) were determined. Hemispheric volume atrophy in males was reduced (P < 0.05) by 41.9% in the normothermic-IAIP and 28.1% in the hypothermic-IAIP compared with the normothermic-PL group, and in females reduced (P < 0.05) by 30.3% in the normothermic-IAIP, 45.7% in hypothermic-PL, and 55.2% in hypothermic-IAIP compared with the normothermic-PL group after HI. Hypothermia improved (P < 0.05) the neuroprotective effects of hIAIPs in females. The neuroprotective efficacy of hIAIPs was comparable to hypothermia in female rats (P = 0.183). Treatment with hIAIPs, hypothermia, and hIAIPs with hypothermia decreased (P < 0.05) the latency to enter the peripheral zone in the small open field test in males. We conclude that hIAIPs provide neuroprotection from HI brain injury that is comparable to the protection by hypothermia, hypothermia increases the effects of hIAIPs in females, and hIAIPs and hypothermia exhibit some sex-related differential effects.
Collapse
Affiliation(s)
- Xiaodi F Chen
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, USA; The Alpert Medical School of Brown University, USA
| | - Yuqi Wu
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, USA; The Alpert Medical School of Brown University, USA
| | - Boram Kim
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, USA; The Alpert Medical School of Brown University, USA
| | - Kevin V Nguyen
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, USA; The Alpert Medical School of Brown University, USA
| | - Ainuo Chen
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, USA; The Alpert Medical School of Brown University, USA
| | - Joseph Qiu
- ProThera Biologics, Inc., Providence, RI, USA
| | | | - Clemence Disdier
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, USA; The Alpert Medical School of Brown University, USA
| | - Yow-Pin Lim
- ProThera Biologics, Inc., Providence, RI, USA; The Alpert Medical School of Brown University, Department of Pathology and Laboratory Medicine, Providence, RI, USA
| | - Barbara S Stonestreet
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, USA; The Alpert Medical School of Brown University, USA; Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA.
| |
Collapse
|
7
|
Kelly LA, Branagan A, Semova G, Molloy EJ. Sex differences in neonatal brain injury and inflammation. Front Immunol 2023; 14:1243364. [PMID: 37954620 PMCID: PMC10634351 DOI: 10.3389/fimmu.2023.1243364] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 10/17/2023] [Indexed: 11/14/2023] Open
Abstract
Neonatal brain injury and associated inflammation is more common in males. There is a well-recognised difference in incidence and outcome of neonatal encephalopathy according to sex with a pronounced male disadvantage. Neurodevelopmental differences manifest from an early age in infancy with females having a lower incidence of developmental delay and learning difficulties in comparison with males and male sex has consistently been identified as a risk factor for cerebral palsy in epidemiological studies. Important neurobiological differences exist between the sexes with respect to neuronal injury which are especially pronounced in preterm neonates. There are many potential reasons for these sex differences including genetic, immunological and hormonal differences but there are limited studies of neonatal immune response. Animal models with induced neonatal hypoxia have shown various sex differences including an upregulated immune response and increased microglial activation in males. Male sex is recognized to be a risk factor for neonatal hypoxic ischemic encephalopathy (HIE) during the perinatal period and this review discusses in detail the sex differences in brain injury in preterm and term neonates and some of the potential new therapies with possible sex affects.
Collapse
Affiliation(s)
- Lynne A. Kelly
- Discipline of Paediatrics, Trinity College Dublin, Dublin, Ireland
- Paediatrics, Trinity Translational Medicine Institute (TTMI), Dublin, Ireland
- Department of Medicine, Trinity Centre for Health Sciences, Trinity Research in Childhood Centre (TRiCC), Dublin, Ireland
| | - Aoife Branagan
- Discipline of Paediatrics, Trinity College Dublin, Dublin, Ireland
- Paediatrics, Trinity Translational Medicine Institute (TTMI), Dublin, Ireland
- Department of Medicine, Trinity Centre for Health Sciences, Trinity Research in Childhood Centre (TRiCC), Dublin, Ireland
- Coombe Women and Infants University Hospital Dublin, Dublin, Ireland
| | - Gergana Semova
- Discipline of Paediatrics, Trinity College Dublin, Dublin, Ireland
- Paediatrics, Trinity Translational Medicine Institute (TTMI), Dublin, Ireland
- Department of Medicine, Trinity Centre for Health Sciences, Trinity Research in Childhood Centre (TRiCC), Dublin, Ireland
| | - Eleanor J. Molloy
- Discipline of Paediatrics, Trinity College Dublin, Dublin, Ireland
- Paediatrics, Trinity Translational Medicine Institute (TTMI), Dublin, Ireland
- Department of Medicine, Trinity Centre for Health Sciences, Trinity Research in Childhood Centre (TRiCC), Dublin, Ireland
- Coombe Women and Infants University Hospital Dublin, Dublin, Ireland
- Neonatology, Children’s Health Ireland (CHI) at Crumlin, Dublin, Ireland
- Neonatology and Neurodisability, Children’s Health Ireland (CHI) at Tallaght, Dublin, Ireland
| |
Collapse
|
8
|
Ranjan AK, Gulati A. Advances in Therapies to Treat Neonatal Hypoxic-Ischemic Encephalopathy. J Clin Med 2023; 12:6653. [PMID: 37892791 PMCID: PMC10607511 DOI: 10.3390/jcm12206653] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is a condition that results in brain damage in newborns due to insufficient blood and oxygen supply during or after birth. HIE is a major cause of neurological disability and mortality in newborns, with over one million neonatal deaths occurring annually worldwide. The severity of brain injury and the outcome of HIE depend on several factors, including the cause of oxygen deprivation, brain maturity, regional blood flow, and maternal health conditions. HIE is classified into mild, moderate, and severe categories based on the extent of brain damage and resulting neurological issues. The pathophysiology of HIE involves different phases, including the primary phase, latent phase, secondary phase, and tertiary phase. The primary and secondary phases are characterized by episodes of energy and cell metabolism failures, increased cytotoxicity and apoptosis, and activated microglia and inflammation in the brain. A tertiary phase occurs if the brain injury persists, characterized by reduced neural plasticity and neuronal loss. Understanding the cellular and molecular aspects of the different phases of HIE is crucial for developing new interventions and therapeutics. This review aims to discuss the pathophysiology of HIE, therapeutic hypothermia (TH), the only approved therapy for HIE, ongoing developments of adjuvants for TH, and potential future drugs for HIE.
Collapse
Affiliation(s)
- Amaresh K Ranjan
- Research and Development, Pharmazz Inc., Willowbrook, IL 60527, USA
| | - Anil Gulati
- Research and Development, Pharmazz Inc., Willowbrook, IL 60527, USA
- Department of Bioengineering, The University of Illinois at Chicago, Chicago, IL 60607, USA
- College of Pharmacy, Midwestern University, Downers Grove, IL 60515, USA
| |
Collapse
|
9
|
Girolamo F, Lim YP, Virgintino D, Stonestreet BS, Chen XF. Inter-Alpha Inhibitor Proteins Modify the Microvasculature after Exposure to Hypoxia-Ischemia and Hypoxia in Neonatal Rats. Int J Mol Sci 2023; 24:6743. [PMID: 37047713 PMCID: PMC10094872 DOI: 10.3390/ijms24076743] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/21/2023] [Accepted: 03/29/2023] [Indexed: 04/08/2023] Open
Abstract
Microvasculature develops during early brain development. Hypoxia-ischemia (HI) and hypoxia (H) predispose to brain injury in neonates. Inter-alpha inhibitor proteins (IAIPs) attenuate injury to the neonatal brain after exposure to HI. However, the effects of IAIPs on the brain microvasculature after exposure to HI have not been examined in neonates. Postnatal day-7 rats were exposed to sham treatment or right carotid artery ligation and 8% oxygen for 90 min. HI comprises hypoxia (H) and ischemia to the right hemisphere (HI-right) and hypoxia to the whole body, including the left hemisphere (H-left). Human IAIPs (hIAIPs, 30 mg/kg) or placebo were injected immediately, 24 and 48 h after HI/H. The brains were analyzed 72 h after HI/H to determine the effects of hIAIPs on the microvasculature by laminin immunohistochemistry and calculation of (1) the percentage area stained by laminin, (2) cumulative microvessel length, and (3) density of tunneling nanotubes (TNTs), which are sensitive indicators of the earliest phases of neo-vascularization/collateralization. hIAIPs mainly affected the percent of the laminin-stained area after HI/H, cumulative vessel length after H but not HI, and TNT density in females but not males. hIAIPs modify the effects of HI/H on the microvasculature after brain injury in neonatal rats and exhibit sex-related differential effects. Our findings suggest that treatment with hIAIPs after exposure to H and HI in neonatal rats affects the laminin content of the vessel basal lamina and angiogenic responses in a sex-related fashion.
Collapse
Affiliation(s)
- Francesco Girolamo
- Department of Translational Biomedicines and Neuroscience (DiBraiN), University of Bari School of Medicine, 70124 Bari, Italy
| | - Yow-Pin Lim
- ProThera Biologics, Inc., Providence, RI 02905, USA
- Department of Pathology and Laboratory Medicine, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Daniela Virgintino
- Department of Translational Biomedicines and Neuroscience (DiBraiN), University of Bari School of Medicine, 70124 Bari, Italy
| | - Barbara S. Stonestreet
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Xiaodi F. Chen
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| |
Collapse
|
10
|
Yin H, Chen Z, Zhao H, Huang H, Liu W. Noble gas and neuroprotection: From bench to bedside. Front Pharmacol 2022; 13:1028688. [PMID: 36532733 PMCID: PMC9750501 DOI: 10.3389/fphar.2022.1028688] [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] [Received: 08/26/2022] [Accepted: 11/03/2022] [Indexed: 07/26/2023] Open
Abstract
In recent years, inert gases such as helium, argon, and xenon have gained considerable attention for their medical value. Noble gases present an intriguing scientific paradox: although extremely chemically inert, they display a remarkable spectrum of clinically useful biological properties. Despite a relative paucity of knowledge about their mechanisms of action, some noble gases have been used successfully in clinical practice. The neuroprotection elicited by these noble gases has been investigated in experimental animal models of various types of brain injuries, such as traumatic brain injury, stroke, subarachnoid hemorrhage, cerebral ischemic/reperfusion injury, and neurodegenerative diseases. Collectively, these central nervous system injuries are a leading cause of morbidity and mortality every year worldwide. Treatment options are presently limited to thrombolytic drugs and clot removal for ischemic stroke, or therapeutic cooling for other brain injuries before the application of noble gas. Currently, there is increasing interest in noble gases as novel treatments for various brain injuries. In recent years, neuroprotection elicited by particular noble gases, xenon, for example, has been reported under different conditions. In this article, we have reviewed the latest in vitro and in vivo experimental and clinical studies of the actions of xenon, argon, and helium, and discuss their potential use as neuroprotective agents.
Collapse
Affiliation(s)
- Haiying Yin
- Department of Anesthesiology and Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zijun Chen
- Department of Anesthesiology and Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Hailin Zhao
- Division of Anesthetics, Department of Surgery and Cancer, Pain Medicine and Intensive Care, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Han Huang
- Department of Anesthesiology and Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Wenwen Liu
- Department of Anesthesia Nursing, West China Second University Hospital, Sichuan University/West China School of Nursing, Ministry of Education, Sichuan University and Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Chengdu, China
| |
Collapse
|
11
|
Koehn LM, Nguyen K, Chen X, Santoso A, Tucker R, Lim YP, Stonestreet BS. Effects of Three Different Doses of Inter-Alpha Inhibitor Proteins on Severe Hypoxia-Ischemia-Related Brain Injury in Neonatal Rats. Int J Mol Sci 2022; 23:13473. [PMID: 36362257 PMCID: PMC9655902 DOI: 10.3390/ijms232113473] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 11/30/2024] Open
Abstract
Hypoxia-ischemia (HI)-related brain injury is an important cause of morbidity and long-standing disability in newborns. We have previously shown that human plasma-derived inter-alpha inhibitor proteins (hIAIPs) attenuate HI-related brain injury in neonatal rats. The optimal dose of hIAIPs for their neuroprotective effects and improvement in behavioral outcomes remains to be determined. We examined the efficacy of 30, 60, or 90 mg/kg of hIAIPs administered to neonatal rats after exposure to HI for 2 h. Postnatal day 7 (P7) Wistar rats were exposed to either sham-surgery or unilateral HI (right carotid artery ligation, 2 h of 8% O2) brain injury. A placebo, 30, 60, or 90 mg/kg of hIAIPs were injected intraperitoneally at 0, 24 and 48 h after HI (n = 9-10/sex). We carried out the following behavioral analyses: P8 (righting reflex), P9 (negative geotaxis) and P10 (open-field task). Rats were humanely killed on P10 and their brains were stained with cresyl violet. Male extension/contraction responses and female righting reflex times were higher in the HI placebo groups than the sham groups. Female open-field exploration was lower in the HI placebo group than the sham group. hIAIPs attenuated these behavioral deficits. However, the magnitude of the responses did not vary by hIAIP dose. hIAIPs reduced male brain infarct volumes in a manner that correlated with improved behavioral outcomes. Increasing the hIAIP dose from 30 to 90 mg/kg did not further accentuate the hIAIP-related decreases in infarct volumes. We conclude that larger doses of hIAIPs did not provide additional benefits over the 30 mg/kg dose for behavior tasks or reductions in infarct volumes in neonatal rats after exposure to severe HI.
Collapse
Affiliation(s)
- Liam M. Koehn
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Kevin Nguyen
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Xiaodi Chen
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | | | - Richard Tucker
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Yow-Pin Lim
- ProThera Biologics, Inc., Providence, RI 02905, USA
| | - Barbara S. Stonestreet
- Women & Infants Hospital of Rhode Island, Alpert Medical School of Brown University, Providence, RI 02905, USA
| |
Collapse
|
12
|
Fabres RB, Nunes RR, de Medeiros de Mattos M, Andrade MKG, Martini APR, Tassinari ID, Sanches EF, de Fraga LS, Netto CA. Therapeutic hypothermia for the treatment of neonatal hypoxia-ischemia: sex-dependent modulation of reactive astrogliosis. Metab Brain Dis 2022; 37:2315-2329. [PMID: 35778625 DOI: 10.1007/s11011-022-01030-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/09/2022] [Indexed: 11/26/2022]
Abstract
Therapeutic hypothermia (TH) is the standard treatment for neonatal hypoxia-ischemia (HI) with a time window limited up to 6 h post injury. However, influence of sexual dimorphism in the therapeutic window for TH has not yet been elucidated in animal models of HI. Therefore, the aim of this study was to investigate the most effective time window to start TH in male and female rats submitted to neonatal HI. Wistar rats (P7) were divided into the following groups: NAÏVE and SHAM (control groups), HI (submitted to HI) and TH (submitted to HI and TH; 32ºC for 5 h). TH was started at 2 h (TH-2 h group), 4 h (TH-4 h group), or 6 h (TH-6 h group) after HI. At P14, animals were subjected to behavioural tests, volume of lesion and reactive astrogliosis assessments. Male and female rats from the TH-2 h group showed reduction in the latency of behavioral tests, and decrease in volume of lesion and intensity of GFAP immunofluorescence. TH-2 h females also showed reduction of degenerative cells and morphological changes in astrocytes. Interestingly, females from the TH-6 h group showed an increase in volume of lesion and in number of degenerative hippocampal cells, associated with worse behavioral performance. Together, these results indicate that TH neuroprotection is time- and sex-dependent. Moreover, TH started later (6 h) can worsen volume of brain lesion in females. These data indicate the need to develop specific therapeutic protocols for each sex and reinforce the importance of early onset of the hypothermic treatment.
Collapse
Affiliation(s)
- Rafael Bandeira Fabres
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil.
- Postgraduate Programme in Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil.
- ICBS/UFRGS - Campus Centro, Rua Sarmento Leite, 500 - 2º Andar, 90050170, Porto Alegre, RS, Brazil.
| | - Ricardo Ribeiro Nunes
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil
- Postgraduate Programme in Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil
| | - Marcel de Medeiros de Mattos
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, 90035-003, Porto Alegre, Brazil
| | - Mirella Kielek Galvan Andrade
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil
| | - Ana Paula Rodrigues Martini
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, 90035-003, Porto Alegre, Brazil
- Postgraduate Programme in Neuroscience, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil
| | - Isadora D'Ávila Tassinari
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil
- Postgraduate Programme in Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil
| | - Eduardo Farias Sanches
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, 90035-003, Porto Alegre, Brazil
- Postgraduate Programme in Neuroscience, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil
| | - Luciano Stürmer de Fraga
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil
- Postgraduate Programme in Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil
| | - Carlos Alexandre Netto
- Postgraduate Programme in Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Ramiro Barcelos, 2600, 90035-003, Porto Alegre, Brazil
- Postgraduate Programme in Neuroscience, Universidade Federal do Rio Grande do Sul (UFRGS), Sarmento Leite, 500, 90050-170, Porto Alegre, Brazil
| |
Collapse
|
13
|
Chen X, Zhang J, Wu Y, Tucker R, Baird GL, Domonoske R, Barrios-Anderson A, Lim YP, Bath K, Walsh EG, Stonestreet BS. Inter-alpha Inhibitor Proteins Ameliorate Brain Injury and Improve Behavioral Outcomes in a Sex-Dependent Manner After Exposure to Neonatal Hypoxia Ischemia in Newborn and Young Adult Rats. Neurotherapeutics 2022; 19:528-549. [PMID: 35290609 PMCID: PMC9226254 DOI: 10.1007/s13311-022-01217-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2022] [Indexed: 12/16/2022] Open
Abstract
Hypoxic-ischemic (HI) brain injury is a major contributor to neurodevelopmental morbidities. Inter-alpha inhibitor proteins (IAIPs) have neuroprotective effects on HI-related brain injury in neonatal rats. However, the effects of treatment with IAIPs on sequential behavioral, MRI, and histopathological abnormalities in the young adult brain after treatment with IAIPs in neonates remain to be determined. The objective of this study was to examine the neuroprotective effects of IAIPs at different neurodevelopmental stages from newborn to young adults after exposure of neonates to HI injury. IAIPs were given as 11-sequential 30-mg/kg doses to postnatal (P) day 7-21 rats after right common carotid artery ligation and exposure to 90 min of 8% oxygen. The resulting brain edema and injury were examined by T2-weighted magnetic resonance imaging (MRI) and cresyl violet staining, respectively. The mean T2 values of the ipsilateral hemisphere from MRI slices 6 to 10 were reduced in IAIP-treated HI males + females on P8, P9, and P10 and females on P8, P9, P10, and P14. IAIP treatment reduced hemispheric volume atrophy by 44.5 ± 29.7% in adult male + female P42 rats and improved general locomotor abilities measured by the righting reflex over time at P7.5, P8, and P9 in males + females and males and muscle strength/endurance measured by wire hang on P16 in males + females and females. IAIPs provided beneficial effects during the learning phase of the Morris water maze with females exhibiting beneficial effects. IAIPs confer neuroprotection from HI-related brain injury in neonates and even in adult rats and beneficial MRI and behavioral benefits in a sex-dependent manner.
Collapse
Affiliation(s)
- Xiaodi Chen
- Department of Pediatrics, Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Jiyong Zhang
- Department of Pediatrics, Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Yuqi Wu
- Department of Pediatrics, Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Richard Tucker
- Department of Pediatrics, Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Grayson L Baird
- Department of Diagnostic Imaging, Biostatistics Core Lifespan Hospital System, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Rose Domonoske
- Department of Pediatrics, Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Adriel Barrios-Anderson
- Department of Pediatrics, Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA
| | - Yow-Pin Lim
- ProThera Biologics, Inc, Providence, RI, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Kevin Bath
- Division of Developmental Neuroscience, New York State Psychiatric Institute, New York, NY, USA
- Department of Psychiatry, Columbia University Irving Medical College, New York, NY, USA
| | - Edward G Walsh
- Department of Neuroscience, Brown University, Providence, RI, USA
| | - Barbara S Stonestreet
- Department of Pediatrics, Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Women &101 Dudley Street, Providence, RI, 02905-2499, USA.
| |
Collapse
|
14
|
Chakkarapani AA, Aly H, Benders M, Cotten CM, El-Dib M, Gressens P, Hagberg H, Sabir H, Wintermark P, Robertson NJ. Therapies for neonatal encephalopathy: Targeting the latent, secondary and tertiary phases of evolving brain injury. Semin Fetal Neonatal Med 2021; 26:101256. [PMID: 34154945 DOI: 10.1016/j.siny.2021.101256] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In term and near-term neonates with neonatal encephalopathy, therapeutic hypothermia protocols are well established. The current focus is on how to improve outcomes further and the challenge is to find safe and complementary therapies that confer additional protection, regeneration or repair in addition to cooling. Following hypoxia-ischemia, brain injury evolves over three main phases (latent, secondary and tertiary), each with a different brain energy, perfusion, neurochemical and inflammatory milieu. While therapeutic hypothermia has targeted the latent and secondary phase, we now need therapies that cover the continuum of brain injury that spans hours, days, weeks and months after the initial event. Most agents have several therapeutic actions but can be broadly classified under a predominant action (e.g., free radical scavenging, anti-apoptotic, anti-inflammatory, neuroregeneration, and vascular effects). Promising early/secondary phase therapies include Allopurinol, Azithromycin, Exendin-4, Magnesium, Melatonin, Noble gases and Sildenafil. Tertiary phase agents include Erythropoietin, Stem cells and others. We review a selection of promising therapeutic agents on the translational pipeline and suggest a framework for neuroprotection and neurorestoration that targets the evolving injury.
Collapse
Affiliation(s)
| | - Hany Aly
- Cleveland Clinic Children's Hospital, Cleveland, OH, USA.
| | - Manon Benders
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.
| | - C Michael Cotten
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA.
| | - Mohamed El-Dib
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Pierre Gressens
- Université de Paris, NeuroDiderot, Inserm, Paris, France; Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, SE1 7EH, United Kingdom.
| | - Henrik Hagberg
- Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, SE1 7EH, United Kingdom; Centre of Perinatal Medicine & Health, Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Hemmen Sabir
- Department of Neonatology and Pediatric Intensive Care, Children's Hospital University of Bonn, Bonn, Germany; German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany.
| | - Pia Wintermark
- Department of Pediatrics, Division of Newborn Medicine, Montreal Children's Hospital, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
| | - Nicola J Robertson
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, Edinburgh BioQuarter, Edinburgh, United Kingdom; Institute for Women's Health, University College London, London, United Kingdom.
| | | |
Collapse
|
15
|
Nolan JP, Sandroni C, Böttiger BW, Cariou A, Cronberg T, Friberg H, Genbrugge C, Haywood K, Lilja G, Moulaert VRM, Nikolaou N, Olasveengen TM, Skrifvars MB, Taccone F, Soar J. Postreanimationsbehandlung. Notf Rett Med 2021. [DOI: 10.1007/s10049-021-00892-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
16
|
Zakharova NM, Tarahovsky YS, Komelina NP, Khrenov MO, Kovtun AL. Pharmacological torpor prolongs rat survival in lethal normobaric hypoxia. J Therm Biol 2021; 98:102906. [PMID: 34016333 DOI: 10.1016/j.jtherbio.2021.102906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 11/16/2022]
Abstract
Resistance to hypoxia is one of the most prominent features of natural hibernation and is expected to be present in the pharmacological torpor (PT) that simulates hibernation. We studied resistance to lethal hypoxia (3.5% oxygen content) in rats under PT. To initiate PT, we used the previously developed pharmacological composition (PC) which, after a single intravenous injection, can induce a daily decrease in Tb by 7 °C-8 °C at the environmental temperature of 22 °C-23 °C. Half-survival (median) time of rats in lethal hypoxia was found to increase from 5 ± 0.8 min in anesthetized control rats to 150 ± 12 min in rats injected with PC, which is a 30-fold increase. Behavioral tests after PT and hypoxia, including the traveling distance, the number of rearing and grooming episodes, revealed that animal responses are significantly restored within a week. It is assumed that the discovered unprecedented resistance of artificially torpid rats to lethal hypoxia may open up broad prospects for the therapeutic use of PT for preconditioning to various damaging factors, treatment of diseases, and extend the so-called "golden hour" for lifesaving interventions.
Collapse
Affiliation(s)
| | - Yury S Tarahovsky
- Institute of Cell Biophysics, RAS, Pushchino, Moscow Region 142290, Russia; Institute of Theoretical and Experimental Biophysics, RAS, Pushchino, Moscow Region 142290, Russia.
| | - Natalia P Komelina
- Institute of Cell Biophysics, RAS, Pushchino, Moscow Region 142290, Russia
| | - Maxim O Khrenov
- Institute of Cell Biophysics, RAS, Pushchino, Moscow Region 142290, Russia
| | | |
Collapse
|
17
|
Nolan JP, Sandroni C, Böttiger BW, Cariou A, Cronberg T, Friberg H, Genbrugge C, Haywood K, Lilja G, Moulaert VRM, Nikolaou N, Olasveengen TM, Skrifvars MB, Taccone F, Soar J. European Resuscitation Council and European Society of Intensive Care Medicine guidelines 2021: post-resuscitation care. Intensive Care Med 2021; 47:369-421. [PMID: 33765189 PMCID: PMC7993077 DOI: 10.1007/s00134-021-06368-4] [Citation(s) in RCA: 564] [Impact Index Per Article: 141.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/08/2021] [Indexed: 12/13/2022]
Abstract
The European Resuscitation Council (ERC) and the European Society of Intensive Care Medicine (ESICM) have collaborated to produce these post-resuscitation care guidelines for adults, which are based on the 2020 International Consensus on Cardiopulmonary Resuscitation Science with Treatment Recommendations. The topics covered include the post-cardiac arrest syndrome, diagnosis of cause of cardiac arrest, control of oxygenation and ventilation, coronary reperfusion, haemodynamic monitoring and management, control of seizures, temperature control, general intensive care management, prognostication, long-term outcome, rehabilitation and organ donation.
Collapse
Affiliation(s)
- Jerry P. Nolan
- University of Warwick, Warwick Medical School, Coventry, CV4 7AL UK
- Royal United Hospital, Bath, BA1 3NG UK
| | - Claudio Sandroni
- Department of Intensive Care, Emergency Medicine and Anaesthesiology, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
- Institute of Anaesthesiology and Intensive Care Medicine, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Bernd W. Böttiger
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Alain Cariou
- Cochin University Hospital (APHP) and University of Paris (Medical School), Paris, France
| | - Tobias Cronberg
- Department of Clinical Sciences, Neurology, Lund University, Skane University Hospital, Lund, Sweden
| | - Hans Friberg
- Department of Clinical Sciences, Anaesthesia and Intensive Care Medicine, Lund University, Skane University Hospital, Lund, Sweden
| | - Cornelia Genbrugge
- Acute Medicine Research Pole, Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain, Brussels, Belgium
- Emergency Department, University Hospitals Saint-Luc, Brussels, Belgium
| | - Kirstie Haywood
- Warwick Research in Nursing, Division of Health Sciences, Warwick Medical School, University of Warwick, Room A108, Coventry, CV4 7AL UK
| | - Gisela Lilja
- Department of Clinical Sciences Lund, Neurology, Lund University, Skane University Hospital, Lund, Sweden
| | - Véronique R. M. Moulaert
- Department of Rehabilitation Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Nikolaos Nikolaou
- Cardiology Department, Konstantopouleio General Hospital, Athens, Greece
| | - Theresa Mariero Olasveengen
- Department of Anesthesiology, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Markus B. Skrifvars
- Department of Emergency Care and Services, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Fabio Taccone
- Department of Intensive Care, Hôpital Erasme, Université Libre de Bruxelles, Route de Lennik, 808, 1070 Brussels, Belgium
| | - Jasmeet Soar
- Southmead Hospital, North Bristol NHS Trust, Bristol, BS10 5NB UK
| |
Collapse
|
18
|
Nolan JP, Sandroni C, Böttiger BW, Cariou A, Cronberg T, Friberg H, Genbrugge C, Haywood K, Lilja G, Moulaert VRM, Nikolaou N, Mariero Olasveengen T, Skrifvars MB, Taccone F, Soar J. European Resuscitation Council and European Society of Intensive Care Medicine Guidelines 2021: Post-resuscitation care. Resuscitation 2021; 161:220-269. [PMID: 33773827 DOI: 10.1016/j.resuscitation.2021.02.012] [Citation(s) in RCA: 435] [Impact Index Per Article: 108.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The European Resuscitation Council (ERC) and the European Society of Intensive Care Medicine (ESICM) have collaborated to produce these post-resuscitation care guidelines for adults, which are based on the 2020 International Consensus on Cardiopulmonary Resuscitation Science with Treatment Recommendations. The topics covered include the post-cardiac arrest syndrome, diagnosis of cause of cardiac arrest, control of oxygenation and ventilation, coronary reperfusion, haemodynamic monitoring and management, control of seizures, temperature control, general intensive care management, prognostication, long-term outcome, rehabilitation, and organ donation.
Collapse
Affiliation(s)
- Jerry P Nolan
- University of Warwick, Warwick Medical School, Coventry CV4 7AL, UK; Royal United Hospital, Bath, BA1 3NG, UK.
| | - Claudio Sandroni
- Department of Intensive Care, Emergency Medicine and Anaesthesiology, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy; Institute of Anaesthesiology and Intensive Care Medicine, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Bernd W Böttiger
- University Hospital of Cologne, Kerpener Straße 62, D-50937 Cologne, Germany
| | - Alain Cariou
- Cochin University Hospital (APHP) and University of Paris (Medical School), Paris, France
| | - Tobias Cronberg
- Department of Clinical Sciences, Neurology, Lund University, Skane University Hospital, Lund, Sweden
| | - Hans Friberg
- Department of Clinical Sciences, Anaesthesia and Intensive Care Medicine, Lund University, Skane University Hospital, Lund, Sweden
| | - Cornelia Genbrugge
- Acute Medicine Research Pole, Institute of Experimental and Clinical Research (IREC) Université Catholique de Louvain, Brussels, Belgium; Emergency Department, University Hospitals Saint-Luc, Brussels, Belgium
| | - Kirstie Haywood
- Warwick Research in Nursing, Room A108, Division of Health Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Gisela Lilja
- Lund University, Skane University Hospital, Department of Clinical Sciences Lund, Neurology, Lund, Sweden
| | - Véronique R M Moulaert
- University of Groningen, University Medical Center Groningen, Department of Rehabilitation Medicine, Groningen, The Netherlands
| | - Nikolaos Nikolaou
- Cardiology Department, Konstantopouleio General Hospital, Athens, Greece
| | - Theresa Mariero Olasveengen
- Department of Anesthesiology, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Norway
| | - Markus B Skrifvars
- Department of Emergency Care and Services, University of Helsinki and Helsinki University Hospital, Finland
| | - Fabio Taccone
- Department of Intensive Care, Hôpital Erasme, Université Libre de Bruxelles, Route de Lennik, 808, 1070 Brussels, Belgium
| | - Jasmeet Soar
- Southmead Hospital, North Bristol NHS Trust, Bristol BS10 5NB, UK
| |
Collapse
|
19
|
Wortinger LA, Jørgensen KN, Barth C, Nerland S, Smelror RE, Vaskinn A, Ueland T, Andreassen OA, Agartz I. Significant association between intracranial volume and verbal intellectual abilities in patients with schizophrenia and a history of birth asphyxia. Psychol Med 2021; 52:1-10. [PMID: 33750510 PMCID: PMC9772907 DOI: 10.1017/s0033291721000489] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/20/2021] [Accepted: 02/04/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND The etiology of schizophrenia (SZ) is proposed to include an interplay between a genetic risk for disease development and the biological environment of pregnancy and birth, where early adversities may contribute to the poorer developmental outcome. We investigated whether a history of birth asphyxia (ASP) moderates the relationship between intracranial volume (ICV) and intelligence in SZ, bipolar disorder (BD) and healthy controls (HC). METHODS Two hundred seventy-nine adult patients (18-42 years) on the SZ and BD spectrums and 216 HC were evaluated for ASP based on information from the Medical Birth Registry of Norway. Participants underwent structural magnetic resonance imaging (MRI) to estimate ICV and intelligence quotient (IQ) assessment using the Wechsler Abbreviated Scale of Intelligence (WASI). Multiple linear regressions were used for analyses. RESULTS We found a significant three-way interaction (ICV × ASP × diagnosis) on the outcome variable, IQ, indicating that the correlation between ICV and IQ was stronger in patients with SZ who experienced ASP compared to SZ patients without ASP. This moderation by ASP was not found in BD or HC groups. In patients with SZ, the interaction between ICV and a history of the ASP was specifically related to the verbal subcomponent of IQ as measured by WASI. CONCLUSIONS The significant positive association between ICV and IQ in patients with SZ who had experienced ASP might indicate abnormal neurodevelopment. Our findings give support for ICV together with verbal intellectual abilities as clinically relevant markers that can be added to prediction tools to enhance evaluations of SZ risk.
Collapse
Affiliation(s)
- Laura Anne Wortinger
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kjetil Nordbø Jørgensen
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Claudia Barth
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Stener Nerland
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Runar Elle Smelror
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anja Vaskinn
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, NORMENT, Oslo University Hospital, Oslo, Norway
| | - Torill Ueland
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, NORMENT, Oslo University Hospital, Oslo, Norway
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Ole A. Andreassen
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, NORMENT, Oslo University Hospital, Oslo, Norway
| | - Ingrid Agartz
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
- NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Clinical Neuroscience, Centre for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
20
|
Htun Y, Nakamura S, Kusaka T. Hydrogen and therapeutic gases for neonatal hypoxic-ischemic encephalopathy: potential neuroprotective adjuncts in translational research. Pediatr Res 2021; 89:753-759. [PMID: 32505123 DOI: 10.1038/s41390-020-0998-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/08/2020] [Accepted: 05/12/2020] [Indexed: 11/09/2022]
Abstract
Numerous studies have examined the potential use of therapeutic gases for the treatment of various neurological disorders. Hydrogen gas, a promising neuroprotective agent, has been a focus of study due to its potent antioxidative properties. In translational research into adult diseases, hydrogen has been shown to be neuroprotective in disorders such as cerebral ischemia and traumatic brain injury, and in neurodegenerative diseases such as Alzheimer's disease. Animal and human studies have verified the safety and feasibility of molecular hydrogen. However, despite extensive research on its efficacy in adults, only a few studies have investigated its application in pediatric and neonatal medicine. Neonatal hypoxic-ischemic encephalopathy (HIE) is characterized by damage to neurons and other cells of the nervous system. One of the major contributing factors is excessive exposure to oxidative stress. Current research interest in HIE is shifting toward new neuroprotective agents, as single agents or as adjuncts to therapeutic hypothermia. Here, we review therapeutic gases, particularly hydrogen, and their potentials and limitations in the treatment of HIE in newborns. IMPACT: Translational animal models of neonatal HIE are a current focus of research into the therapeutic usefulness of various gases. Hydrogen ventilation as a single agent or in combination with therapeutic hypothermia shows short- and long-term neuroprotection in neonatal translational HIE models. The optimal target severity for therapeutic interventions should be well established to improve outcomes.
Collapse
Affiliation(s)
- Yinmon Htun
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan.,Graduate School of Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Shinji Nakamura
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Takashi Kusaka
- Department of Pediatrics, Faculty of Medicine, Kagawa University, Kagawa, Japan.
| |
Collapse
|
21
|
Chen X, Song D, Nakada S, Qiu J, Iwamoto K, Chen RH, Lim YP, Jusko WJ, Stonestreet BS. Pharmacokinetics of Inter-Alpha Inhibitor Proteins and Effects on Hemostasis After Hypoxic-Ischemic Brain Injury in Neonatal Rats. Curr Pharm Des 2021; 26:3997-4006. [PMID: 32316887 DOI: 10.2174/1381612826666200421123242] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 04/08/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Hypoxic-ischemic (HI) brain injury is a leading cause of long-term neurodevelopmental morbidities in neonates. Human plasma-derived Inter-Alpha Inhibitor Proteins (hIAIPs) are neuroprotective after HI brain injury in neonatal rats. The light chain (bikunin) of hIAIPs inhibits proteases involved in the coagulation of blood. Newborns exposed to HI can be at risk for significant bleeding in the brain and other organs. OBJECTIVE The objectives of the present study were to assess the pharmacokinetics (PK) and the duration of bleeding after intraperitoneal (IP) administration of hIAIPs in HI-exposed male and female neonatal rats. METHODS HI was induced with the Rice-Vannucci method in postnatal (P) day-7 rats. After the right common carotid artery ligation, rats were exposed to 90 min of 8% oxygen. hIAIPs (30 mg/kg, IP) were given immediately after Sham or HI exposure in the PK study and serum was collected 1, 6, 12, 24, or 36 h after the injections. Serum hIAIP concentrations were measured with a competitive ELISA. ADAPT5 software was used to fit the pooled PK data considering first-order absorption and disposition. hIAIPs (60 mg/kg, IP) were given in the bleeding time studies at 0, 24 and 48 h after HI with tail bleeding times measured 72 h after HI. RESULTS IP administration yielded significant systemic exposure to hIAIPs with PK being affected markedly including primarily faster absorption and reduced elimination as a result of HI and modestly of sex-related differences. hIAIP administration did not affect bleeding times after HI. CONCLUSION These results will help to inform hIAIP dosing regimen schedules in studies of neuroprotection in neonates exposed to HI.
Collapse
Affiliation(s)
- Xiaodi Chen
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Providence, RI, United States
| | - Dawei Song
- School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Sakura Nakada
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Providence, RI, United States
| | - Joseph Qiu
- ProThera Biologics, Inc., Providence, RI, United States
| | - Karin Iwamoto
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Providence, RI, United States
| | - Ray H Chen
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Providence, RI, United States
| | - Yow-Pin Lim
- ProThera Biologics, Inc., Providence, RI, United States
| | - William J Jusko
- School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Barbara S Stonestreet
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, Providence, RI, United States
| |
Collapse
|
22
|
Phillips T, Menassa DA, Grant S, Cohen N, Thoresen M. The effects of Xenon gas inhalation on neuropathology in a placental-induced brain injury model in neonates: A pilot study. Acta Paediatr 2021; 110:119-122. [PMID: 32681542 DOI: 10.1111/apa.15486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/10/2020] [Accepted: 07/14/2020] [Indexed: 11/29/2022]
Affiliation(s)
- Thomas Phillips
- Translational Health Sciences Bristol Medical School University of Bristol Bristol UK
- UK Dementia Research Institute Cardiff University Cardiff UK
| | - David A. Menassa
- Translational Health Sciences Bristol Medical School University of Bristol Bristol UK
- Biological Sciences Faculty of Environmental and Life Sciences University of Southampton Southampton UK
- Nuffield Department of Clinical Neurosciences University of Oxford Oxford UK
| | - Simon Grant
- Department of Obstetrics and Gynaecology Southmead Hospital Bristol UK
| | - Nicki Cohen
- Department of Medical Education King's College London London UK
| | - Marianne Thoresen
- Neonatal Neuroscience Translational Health Sciences Bristol Medical School University of Bristol Bristol UK
- Institute of Basic Medical Sciences Section for Physiology University of Oslo Oslo Norway
| |
Collapse
|
23
|
Zhang M, Cui Y, Zhu W, Yu J, Cheng Y, Wu X, Zhang J, Xin W, Yu Y, Sun H. Attenuation of the mutual elevation of iron accumulation and oxidative stress may contribute to the neuroprotective and anti-seizure effects of xenon in neonatal hypoxia-induced seizures. Free Radic Biol Med 2020; 161:212-223. [PMID: 33075502 DOI: 10.1016/j.freeradbiomed.2020.09.030] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/20/2020] [Accepted: 09/21/2020] [Indexed: 12/21/2022]
Abstract
Previous studies have suggested that xenon inhalation has neuroprotective and antiepileptic effects; however, the underlying mechanisms involved remain unclear. This study aimed to investigate the possible xenon inhalation mechanisms involved in the neuroprotection and antiepileptic effects. A neonatal hypoxic C57BL/6J mouse model was used for the experiments. Immediately after hypoxia treatment, the treatment group inhaled a xenon mixture (70% xenon/21% oxygen/9% nitrogen) for 60 min, while the hypoxia group inhaled a non-xenon mixture (21% oxygen/79% nitrogen) for 60 min. Seizure activity was recorded at designated time points using electroencephalography. Oxidative stress levels, iron levels, neuronal injury, and learning and memory functions were also studied. The results showed that hypoxia increased the levels of iron, oxidative stress, mitophagy, and neurodegeneration, which were accompanied by seizures and learning and memory disorders. In addition, our results confirmed that xenon treatment significantly attenuated the hypoxia-induced seizures and cognitive defects in neonatal C57 mice. Moreover, the increased levels of iron, oxidative stress, mitophagy, and neuronal injury were reduced in xenon-treated mice. This study confirms the significant protective effects of a xenon mixture on hypoxia-induced damage in neonatal mice. Furthermore, our results suggest that reducing oxidative stress levels and iron accumulation may be the underlying mechanisms of xenon activity. Studying the protective mechanisms of xenon will advance its applications in potential therapeutic strategies.
Collapse
Affiliation(s)
- Mengdi Zhang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Yaru Cui
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Wei Zhu
- Institute of Radiation Medicine, Shandong Academy of Medical Sciences, Shandong First Medical University, Jinan, 250062, China
| | - Jie Yu
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Yao Cheng
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Xiangdong Wu
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Jinjin Zhang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Wenyu Xin
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Yan Yu
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Hongliu Sun
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China.
| |
Collapse
|
24
|
Campos-Pires R, Onggradito H, Ujvari E, Karimi S, Valeo F, Aldhoun J, Edge CJ, Franks NP, Dickinson R. Xenon treatment after severe traumatic brain injury improves locomotor outcome, reduces acute neuronal loss and enhances early beneficial neuroinflammation: a randomized, blinded, controlled animal study. Crit Care 2020; 24:667. [PMID: 33246487 PMCID: PMC7691958 DOI: 10.1186/s13054-020-03373-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/04/2020] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a major cause of morbidity and mortality, but there are no clinically proven treatments that specifically target neuronal loss and secondary injury development following TBI. In this study, we evaluate the effect of xenon treatment on functional outcome, lesion volume, neuronal loss and neuroinflammation after severe TBI in rats. METHODS Young adult male Sprague Dawley rats were subjected to controlled cortical impact (CCI) brain trauma or sham surgery followed by treatment with either 50% xenon:25% oxygen balance nitrogen, or control gas 75% nitrogen:25% oxygen. Locomotor function was assessed using Catwalk-XT automated gait analysis at baseline and 24 h after injury. Histological outcomes were assessed following perfusion fixation at 15 min or 24 h after injury or sham procedure. RESULTS Xenon treatment reduced lesion volume, reduced early locomotor deficits, and attenuated neuronal loss in clinically relevant cortical and subcortical areas. Xenon treatment resulted in significant increases in Iba1-positive microglia and GFAP-positive reactive astrocytes that was associated with neuronal preservation. CONCLUSIONS Our findings demonstrate that xenon improves functional outcome and reduces neuronal loss after brain trauma in rats. Neuronal preservation was associated with a xenon-induced enhancement of microglial cell numbers and astrocyte activation, consistent with a role for early beneficial neuroinflammation in xenon's neuroprotective effect. These findings suggest that xenon may be a first-line clinical treatment for brain trauma.
Collapse
Affiliation(s)
- Rita Campos-Pires
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London, Sir Ernst Chain Building, South Kensington, London, SW7 2AZ, UK
- Royal British Legion Centre for Blast Injury Studies, Department of Bioengineering, Imperial College London, Bessemer Building, South Kensington, London, SW7 2AZ, UK
- Charing Cross Hospital Intensive Care Unit, Critical Care Directorate, Imperial College Healthcare NHS Trust, London, UK
| | - Haldis Onggradito
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London, Sir Ernst Chain Building, South Kensington, London, SW7 2AZ, UK
| | - Eszter Ujvari
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London, Sir Ernst Chain Building, South Kensington, London, SW7 2AZ, UK
| | - Shughoofa Karimi
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London, Sir Ernst Chain Building, South Kensington, London, SW7 2AZ, UK
| | - Flavia Valeo
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London, Sir Ernst Chain Building, South Kensington, London, SW7 2AZ, UK
| | - Jitka Aldhoun
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London, Sir Ernst Chain Building, South Kensington, London, SW7 2AZ, UK
| | - Christopher J Edge
- Department of Life Sciences, Imperial College London, Sir Ernst Chain Building, South Kensington, London, SW7 2AZ, UK
- Department of Anaesthetics, Royal Berkshire Hospital NHS Foundation Trust, London Road, Reading, RG1 5AN, UK
| | - Nicholas P Franks
- Department of Life Sciences, Imperial College London, Sir Ernst Chain Building, South Kensington, London, SW7 2AZ, UK
| | - Robert Dickinson
- Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London, Sir Ernst Chain Building, South Kensington, London, SW7 2AZ, UK.
- Royal British Legion Centre for Blast Injury Studies, Department of Bioengineering, Imperial College London, Bessemer Building, South Kensington, London, SW7 2AZ, UK.
| |
Collapse
|
25
|
Schuffels S, Nakada S, Wu Y, Lim YP, Chen X, Stonestreet BS. Effects of inter-alpha inhibitor proteins on brain injury after exposure of neonatal rats to severe hypoxia-ischemia. Exp Neurol 2020; 334:113442. [PMID: 32896573 DOI: 10.1016/j.expneurol.2020.113442] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/31/2020] [Accepted: 08/24/2020] [Indexed: 12/25/2022]
Abstract
Hypoxic-ischemic (HI) brain injury is one of the most common neurological problems occurring in premature and full-term infants after perinatal complications. Hypothermia is the only treatment approved for HI encephalopathy in newborns. However, this treatment is only partially protective, cannot be used to treat premature infants, and has limited efficacy to treat severe HI encephalopathy. Inflammation contributes to the evolution of HI brain injury in neonates. Inter-alpha Inhibitor Proteins (IAIPs) are immunomodulatory proteins that have neuroprotective properties after exposure to moderate HI in neonatal rats. The objective of the current study was to determine the neuroprotective efficacy of treatment with IAIPs starting immediately after or with a delay of one hour after exposure to severe HI of 120 min duration. One hundred and forty-six 7-day-old rat pups were randomized to sham control, HI and immediate treatment with IAIPs (60 mg/kg) or placebo (PL), and sham, HI and delayed treatment with IAIPs or PL. IAIPs or PL were given at zero, 24, and 48 h after HI or 1, 24 and 48 h after HI. Total brain infarct volume was determined 72 h after exposure to HI. Treatment with IAIPs immediately after HI decreased (P < 0.05) infarct volumes by 58.0% and 44.5% in male and female neonatal rats, respectively. Delayed treatment with IAIPs after HI decreased (P < 0.05) infarct volumes by 23.7% in male, but not in female rats. We conclude that IAIPs exert neuroprotective effects even after exposure to severe HI in neonatal rats and appear to exhibit some sex-related differential effects.
Collapse
Affiliation(s)
- Stephanie Schuffels
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, United States of America
| | - Sakura Nakada
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, United States of America
| | - Yuqi Wu
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, United States of America
| | - Yow-Pin Lim
- ProThera Biologics, Inc., Providence, RI, The Alpert Medical School of Brown University, Providence, RI, United States of America; Department of Pathology and Laboratory Medicine, The Alpert Medical School of Brown University, Providence, RI, United States of America
| | - Xiaodi Chen
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, United States of America.
| | - Barbara S Stonestreet
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, The Alpert Medical School of Brown University, United States of America.
| |
Collapse
|
26
|
Bruschettini M, Romantsik O, Moreira A, Ley D, Thébaud B. Stem cell-based interventions for the prevention of morbidity and mortality following hypoxic-ischaemic encephalopathy in newborn infants. Cochrane Database Syst Rev 2020; 8:CD013202. [PMID: 32813884 PMCID: PMC7438027 DOI: 10.1002/14651858.cd013202.pub2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Hypoxic-ischaemic encephalopathy (HIE) is a leading cause of mortality and long-term neurological sequelae, affecting thousands of children worldwide. Current therapies to treat HIE are limited to cooling. Stem cell-based therapies offer a potential therapeutic approach to repair or regenerate injured brain tissue. These preclinical findings have now culminated in ongoing human neonatal trials. OBJECTIVES To determine the efficacy and safety of stem cell-based interventions for the treatment of hypoxic-ischaemic encephalopathy (HIE) in newborn infants. SEARCH METHODS We used the standard search strategy of Cochrane Neonatal to search the Cochrane Central Register of Controlled Trials (CENTRAL; 2020, Issue 5), MEDLINE via PubMed (1966 to 8 June 2020), Embase (1980 to 8 June 2020), and CINAHL (1982 to 8 June 2020). We also searched clinical trials databases, conference proceedings, and the reference lists of retrieved articles for randomised controlled trials and quasi-randomised trials. SELECTION CRITERIA Randomised controlled trials, quasi-randomised controlled trials and cluster trials comparing 1) stem cell-based interventions (any type) compared to control (placebo or no treatment); 2) use of mesenchymal stem/stromal cells (MSCs) of type (e.g. number of doses or passages) or source (e.g. autologous versus allogeneic, or bone marrow versus cord) versus MSCs of other type or source; 3) use of stem cell-based interventions other than MSCs of type (e.g. mononuclear cells, oligodendrocyte progenitor cells, neural stem cells, hematopoietic stem cells, and inducible pluripotent stem cells) or source (e.g. autologous versus allogeneic, or bone marrow versus cord) versus stem cell-based interventions other than MSCs of other type or source; or 4) MSCs versus stem cell-based interventions other than MSCs. DATA COLLECTION AND ANALYSIS For each of the included trials, two authors independently planned to extract data (e.g. number of participants, birth weight, gestational age, type and source of MSCs or other stem cell-based interventions) and assess the risk of bias (e.g. adequacy of randomisation, blinding, completeness of follow-up). The primary outcomes considered in this review are all-cause neonatal mortality, major neurodevelopmental disability, death or major neurodevelopmental disability assessed at 18 to 24 months of age. We planned to use the GRADE approach to assess the quality of evidence. MAIN RESULTS Our search strategy yielded 616 references. Two review authors independently assessed all references for inclusion. We did not find any completed studies for inclusion. Fifteen RCTs are currently registered and ongoing. We describe the three studies we excluded. AUTHORS' CONCLUSIONS There is currently no evidence from randomised trials that assesses the benefit or harms of stem cell-based interventions for the prevention of morbidity and mortality following hypoxic-ischaemic encephalopathy in newborn infants.
Collapse
Affiliation(s)
- Matteo Bruschettini
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
- Cochrane Sweden, Lund University, Skåne University Hospital, Lund, Sweden
| | - Olga Romantsik
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
| | - Alvaro Moreira
- Pediatrics, Division of Neonatology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - David Ley
- Department of Clinical Sciences Lund, Paediatrics, Lund University, Skåne University Hospital, Lund, Sweden
| | - Bernard Thébaud
- Department of Pediatrics, Children's Hospital of Eastern Ontario, Ottawa, Canada
- Ottawa Hospital Research Institute, Sprott Centre for Stem Cell Research, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| |
Collapse
|
27
|
Variability and sex-dependence of hypothermic neuroprotection in a rat model of neonatal hypoxic-ischaemic brain injury: a single laboratory meta-analysis. Sci Rep 2020; 10:10833. [PMID: 32616806 PMCID: PMC7331720 DOI: 10.1038/s41598-020-67532-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 06/03/2020] [Indexed: 01/19/2023] Open
Abstract
Therapeutic hypothermia (HT) is standard care for term infants with hypoxic–ischaemic (HI) encephalopathy. However, the efficacy of HT in preclinical models, such as the Vannucci model of unilateral HI in the newborn rat, is often greater than that reported from clinical trials. Here, we report a meta-analysis of data from every experiment in a single laboratory, including pilot data, examining the effect of HT in the Vannucci model.
Across 21 experiments using 106 litters, median (95% CI) hemispheric area loss was 50.1% (46.0–51.9%; n = 305) in the normothermia group, and 41.3% (35.1–44.9%; n = 317) in the HT group, with a bimodal injury distribution. Median neuroprotection by HT was 17.6% (6.8–28.3%), including in severe injury, but was highly-variable across experiments. Neuroprotection was significant in females (p < 0.001), with a non-significant benefit in males (p = 0.07). Animals representing the median injury in each group within each litter (n = 277, 44.5%) were also analysed using formal neuropathology, which showed neuroprotection by HT throughout the brain, particularly in females. Our results suggest an inherent variability and sex-dependence of the neuroprotective response to HT, with the majority of studies in the Vannucci model vastly underpowered to detect true treatment effects due to the distribution of injury.
Collapse
|
28
|
Zhu Y, Mosko JJ, Chidekel A, Wolfson MR, Shaffer TH. Effects of xenon gas on human airway epithelial cells during hyperoxia and hypothermia. J Neonatal Perinatal Med 2020; 13:469-476. [PMID: 32444566 PMCID: PMC7836053 DOI: 10.3233/npm-190364] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Hypothermia with xenon gas has been used to reduce brain injury and disability rate after perinatal hypoxia-ischemia. We evaluated xenon gas therapy effects in an in vitro model with or without hypothermia on cultured human airway epithelial cells (Calu-3). METHODS Calu-3 monolayers were grown at an air-liquid interface and exposed to one of the following conditions: 1) 21% FiO2 at 37°C (control); 2) 45% FiO2 and 50% xenon at 37°C; 3) 21% FiO2 and 50% xenon at 32°C; 4) 45% FiO2 and 50% xenon at 32°C for 24 hours. Transepithelial resistance (TER) measurements were performed and apical surface fluids were collected and assayed for total protein, IL-6, and IL-8. Three monolayers were used for immunofluorescence localization of zonula occludens-1 (ZO-1). The data were analyzed by one-way ANOVA. RESULTS TER decreased at 24 hours in all treatment groups. Xenon with hyperoxia and hypothermia resulted in greatest decrease in TER compared with other groups. Immunofluorescence localization of ZO-1 (XY) showed reduced density of ZO-1 rings and incomplete ring-like staining in the 45% FiO2- 50% xenon group at 32°C compared with other groups. Secretion of total protein was not different among groups. Secretion of IL-6 in 21% FiO2 with xenon group at 32°C was less than that of the control group. The secretion of IL-8 in 45% FiO2 with xenon at 32°C was greater than that of other groups. CONCLUSION Hyperoxia and hypothermia result in detrimental epithelial cell function and inflammation over 24-hour exposure. Xenon gas did not affect cell function or reduce inflammation.
Collapse
Affiliation(s)
- Y Zhu
- Center for Pediatric Lung Research, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE
| | - J J Mosko
- Center for Pediatric Lung Research, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE
| | - A Chidekel
- Center for Pediatric Lung Research, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE.,Department of Pediatrics, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE
| | - M R Wolfson
- Departments of Physiology and Pediatrics, Department of Thoracic Medicine and Surgery, CENTRe: Collaborative for Environmental and Neonatal Therapeutics Research, Center for Inflammation and Translational Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - T H Shaffer
- Center for Pediatric Lung Research, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE.,Departments of Physiology and Pediatrics, Department of Thoracic Medicine and Surgery, CENTRe: Collaborative for Environmental and Neonatal Therapeutics Research, Center for Inflammation and Translational Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| |
Collapse
|
29
|
Zhang W, Zhu L, An C, Wang R, Yang L, Yu W, Li P, Gao Y. The blood brain barrier in cerebral ischemic injury – Disruption and repair. BRAIN HEMORRHAGES 2020. [DOI: 10.1016/j.hest.2019.12.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
30
|
Dingley J, Okano S, Lee-Kelland R, Scull-Brown E, Thoresen M, Chakkarapani E. Closed circuit xenon delivery for 72h in neonatal piglets following hypoxic insult using an ambient pressure automated control system: Development, technical evaluation and pulmonary effects. PLoS One 2020; 15:e0224447. [PMID: 31961878 PMCID: PMC6974042 DOI: 10.1371/journal.pone.0224447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 10/14/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Therapeutic hypothermia (TH) for 72h is the standard treatment following neonatal encephalopathy (NE). However, one-third do not benefit and adjunctive therapies are urgently needed. Xenon enhances neuroprotection with TH when administered at 50% concentration within 5hours of hypoxia in experimental studies. Delayed initiation (~10 hours of age) of 30% xenon for 24 hours during TH did not improve early adverse biomarkers in a clinical trial of Xenon+TH vs TH. After hypoxia-ischemia, excitotoxic injury via N-methyl-D-aspartate receptor overactivation lasts days. Since xenon partially inhibits this receptor, we hypothesised that giving 50% xenon throughout the entire 72h TH and rewarming periods would enhance neuroprotection. Xenon costs $30/litre, so a closed-circuit breathing system is desirable with automated fresh gas delivery. METHODS Seven mechanically ventilated newborn pigs were randomized to receive 50% inhaled xenon for 72h during hypothermia (rectal-temperature 35°C) and subsequent rewarming following a global hypoxic-ischemic insult (XeHT, N = 4) or under normothermia for 72h (rectal-temperature 38.5°C) following sham insult (XeNT, N = 3). An automated fresh gas delivery system injected oxygen/air/xenon boluses into a closed-circuit based on measured gas concentrations. RESULTS AND DISCUSSION Median (IQR) xenon consumption was 0.31 L/h (0.18, 0.50) and 0.34L/h (0.32, 0.49) for hypothermic and normothermic groups respectively, 0.34L/h (0.25, 0.53) overall. 92% of 9626 xenon and 69% of 9635 oxygen measurements were within 20% variation from targets. For xenon concentration, the median absolute performance errors for the XeHT and XeNT groups were 6.14% and 3.84% respectively and 4.31% overall. For oxygen these values were 13.42%, 15.05% and 12.4% respectively. There were no adverse pulmonary pathophysiology findings. Clinical problems over the total period included three related to sensors, seven breathing system leaks, ten partial and one complete tracheal tube occlusion episodes. CONCLUSION The automated controller functioned as intended maintaining an inhaled xenon concentration close to the 50% target for 72-78h at a xenon cost of $11.1/h.
Collapse
Affiliation(s)
- John Dingley
- Department of Anaesthetics ABM University Health Board, Swansea and College of Medicine, Swansea University, Swansea, Wales, United Kingdom
- * E-mail: ,
| | - Satomi Okano
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, England, United Kingdom
| | - Richard Lee-Kelland
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, England, United Kingdom
| | - Emma Scull-Brown
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, England, United Kingdom
| | - Marianne Thoresen
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, England, United Kingdom
- Department of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Ela Chakkarapani
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, England, United Kingdom
| |
Collapse
|
31
|
Le Nogue D, Lavaur J, Milet A, Ramirez-Gil JF, Katz I, Lemaire M, Farjot G, Hirsch EC, Michel PP. Neuroprotection of dopamine neurons by xenon against low-level excitotoxic insults is not reproduced by other noble gases. J Neural Transm (Vienna) 2020; 127:27-34. [PMID: 31807953 PMCID: PMC6942589 DOI: 10.1007/s00702-019-02112-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 11/27/2019] [Indexed: 12/19/2022]
Abstract
Using midbrain cultures, we previously demonstrated that the noble gas xenon is robustly protective for dopamine (DA) neurons exposed to L-trans-pyrrolidine-2,4-dicarboxylate (PDC), an inhibitor of glutamate uptake used to generate sustained, low-level excitotoxic insults. DA cell rescue was observed in conditions where the control atmosphere for cell culture was substituted with a gas mix, comprising the same amount of oxygen (20%) and carbon dioxide (5%) but 75% of xenon instead of nitrogen. In the present study, we first aimed to determine whether DA cell rescue against PDC remains detectable when concentrations of xenon are progressively reduced in the cell culture atmosphere. Besides, we also sought to compare the effect of xenon to that of other noble gases, including helium, neon and krypton. Our results show that the protective effect of xenon for DA neurons was concentration-dependent with an IC50 estimated at about 44%. We also established that none of the other noble gases tested in this study protected DA neurons from PDC-mediated insults. Xenon's effectiveness was most probably due to its unique capacity to block NMDA glutamate receptors. Besides, mathematical modeling of gas diffusion in the culture medium revealed that the concentration reached by xenon at the cell layer level is the highest of all noble gases when neurodegeneration is underway. Altogether, our data suggest that xenon may be of potential therapeutic value in Parkinson disease, a chronic neurodegenerative condition where DA neurons appear vulnerable to slow excitotoxicity.
Collapse
Affiliation(s)
- Déborah Le Nogue
- Sorbonne Université, Institut du Cerveau et de la Moelle Epinière (ICM), Inserm U 1127, CNRS, UMR 7225, Paris, France
| | - Jérémie Lavaur
- Sorbonne Université, Institut du Cerveau et de la Moelle Epinière (ICM), Inserm U 1127, CNRS, UMR 7225, Paris, France
| | - Aude Milet
- Air Liquide Santé International, Campus Innovation Paris, Jouy-en-Josas, France
| | | | - Ira Katz
- Air Liquide Santé International, Campus Innovation Paris, Jouy-en-Josas, France
| | - Marc Lemaire
- Air Liquide Santé International, Campus Innovation Paris, Jouy-en-Josas, France
| | - Géraldine Farjot
- Air Liquide Santé International, Campus Innovation Paris, Jouy-en-Josas, France
| | - Etienne C Hirsch
- Sorbonne Université, Institut du Cerveau et de la Moelle Epinière (ICM), Inserm U 1127, CNRS, UMR 7225, Paris, France
| | - Patrick Pierre Michel
- Sorbonne Université, Institut du Cerveau et de la Moelle Epinière (ICM), Inserm U 1127, CNRS, UMR 7225, Paris, France.
| |
Collapse
|
32
|
Liao R, Wood TR, Nance E. Nanotherapeutic modulation of excitotoxicity and oxidative stress in acute brain injury. Nanobiomedicine (Rij) 2020; 7:1849543520970819. [PMID: 35186151 PMCID: PMC8855450 DOI: 10.1177/1849543520970819] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 10/13/2020] [Indexed: 12/20/2022] Open
Abstract
Excitotoxicity is a primary pathological process that occurs during stroke, traumatic brain injury (TBI), and global brain ischemia such as perinatal asphyxia. Excitotoxicity is triggered by an overabundance of excitatory neurotransmitters within the synapse, causing a detrimental cascade of excessive sodium and calcium influx, generation of reactive oxygen species, mitochondrial damage, and ultimately cell death. There are multiple potential points of intervention to combat excitotoxicity and downstream oxidative stress, yet there are currently no therapeutics clinically approved for this specific purpose. For a therapeutic to be effective against excitotoxicity, the therapeutic must accumulate at the disease site at the appropriate concentration at the right time. Nanotechnology can provide benefits for therapeutic delivery, including overcoming physiological obstacles such as the blood-brain barrier, protect cargo from degradation, and provide controlled release of a drug. This review evaluates the use of nano-based therapeutics to combat excitotoxicity in stroke, TBI, and hypoxia-ischemia with an emphasis on mitigating oxidative stress, and consideration of the path forward toward clinical translation.
Collapse
Affiliation(s)
- Rick Liao
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
| | - Thomas R Wood
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, WA, USA
| | - Elizabeth Nance
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
- Department of Radiology, University of Washington, Seattle, WA, USA
- Center on Human Development and Disability, University of Washington, Seattle, WA, USA
| |
Collapse
|
33
|
Abstract
Perinatal brain injury is a major cause of neurological disability in both premature and term infants. In this review, we summarize the evidence behind some established neuroprotective practices such as administration of antenatal steroids, intrapartum magnesium for preterm delivery, and therapeutic hypothermia. In addition, we examine emerging practices such as delayed cord clamping, postnatal magnesium administration, recombinant erythropoietin, and non-steroidal anti-inflammatory agents and finally inform the reader about novel interventions, some of which are currently in trials, such as xenon, melatonin, topiramate, allopurinol, creatine, and autologous cord cell therapy.
Collapse
Affiliation(s)
- Samata Singhi
- Department of Neurology, Kennedy Krieger Institute, Baltimore, Maryland, 21205, USA
- Department of Pediatric Neurology, Johns Hopkins Medicine, Baltimore, MD, 21287, USA
| | - Michael Johnston
- Department of Neurology, Kennedy Krieger Institute, Baltimore, Maryland, 21205, USA
| |
Collapse
|
34
|
Abstract
Hypoxic-ischemic encephalopathy is a subtype of neonatal encephalopathy and a major contributor to global neonatal morbidity and mortality. Despite advances in obstetric and neonatal care there are still challenges in accurate determination of etiology of neonatal encephalopathy. Thus, identification of intrapartum risk factors and comprehensive evaluation of the neonate is important to determine the etiology and severity of neonatal encephalopathy. In developed countries, therapeutic hypothermia as a standard of care therapy for neonates with hypoxic-ischemic encephalopathy has proven to decrease incidence of death and neurodevelopmental disabilities, including cerebral palsy in surviving children. Advances in neuroimaging, brain monitoring modalities, and biomarkers of brain injury have improved the ability to diagnose, monitor, and treat newborns with encephalopathy. However, challenges remain in early identification of neonates at risk for hypoxic-ischemic brain injury, and determination of the timing and extent of brain injury. Using imaging studies such as Neonatal MRI and MR spectroscopy have proven to be most useful in predicting outcomes in infants with encephalopathy within the first week of life, although comprehensive neurodevelopmental assessments still remains the gold standard for determining long term outcomes. Future studies are needed to identify other newborns with encephalopathy that might benefit from therapeutic hypothermia and to determine the efficacy of other adjunctive neuroprotective strategies. This review focuses on newer evidence and advances in diagnoses and management of infants with neonatal encephalopathy, including novel therapies, as well as prognostication of outcomes to childhood.
Collapse
|
35
|
Abstract
Neonatal brain injury (NBI) remains a major contributor to neonatal mortality and long-term neurodevelopmental morbidity. Although therapeutic hypothermia is the only proven treatment to minimize brain injury caused by neonatal encephalopathy in term neonates, it provides incomplete neuroprotection. There are no specific drugs yet proven to prevent NBI in preterm neonates. This review discusses the scientific and emerging clinical trial data for several neuroprotective drugs in development, examining potential efficacy and safety concerns. Drugs with the highest likelihood of success and closest to clinical application include erythropoietin for term and preterm neonates and antenatal magnesium for preterm neonates.
Collapse
Affiliation(s)
- Melanie A McNally
- Department of Neurology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Janet S Soul
- Fetal-Neonatal Neurology Program, Department of Neurology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
36
|
Wood T, Moralejo D, Corry K, Snyder JM, Traudt C, Curtis C, Nance E, Parikh P, Juul SE. A Ferret Model of Encephalopathy of Prematurity. Dev Neurosci 2019; 40:475-489. [PMID: 31079096 PMCID: PMC6658350 DOI: 10.1159/000498968] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 02/18/2019] [Indexed: 12/14/2022] Open
Abstract
There is an ongoing need for relevant animal models in which to test therapeutic interventions for infants with neurological sequelae of prematurity. The ferret is an attractive model species as it has a gyrified brain with a white-to-gray matter ratio similar to that in the human brain. A model of encephalopathy of prematurity was developed in postnatal day 10 (P10) ferret kits, considered to be developmentally equivalent to infants of 24-26 weeks' gestation. Cross-fostered P10 ferret kits received 5 mg/kg of lipopolysaccharide (LPS) before undergoing consecutive hypoxia-hyperoxia-hypoxia (60 min at 9%, 120 min at 60%, and 30 min at 9%). Control animals received saline vehicle followed by normoxia. The development of basic reflexes (negative geotaxis, cliff aversion, and righting) as well as gait coordination on an automated catwalk were assessed between P28 and P70, followed by ex vivo magnetic resonance imaging (MRI) and immunohistochemical analysis. Compared to controls, injured animals had slower overall reflex development between P28 and P40, as well as smaller hind-paw areas consistent with "toe walking" at P42. Injured animals also displayed significantly greater lateral movement during CatWalk assessment as a result of reduced gait coordination. Ex vivo MRI showed widespread white-matter hyperintensity on T2-weighted imaging as well as altered connectivity patterns. This coincided with white-matter dysmaturation characterized by increased intensity of myelin basic protein staining, white-matter thinning, and loss of oligodendrocyte transcription factor 2 (OLIG2)-positive cells. These results suggest both pathological and motor deficits consistent with premature white-matter injury. This newborn ferret model can therefore provide an additional platform to assess potential therapies before translation to human clinical trials.
Collapse
Affiliation(s)
- Thomas Wood
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, Washington, USA,
| | - Daniel Moralejo
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Kylie Corry
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Jessica M Snyder
- Department of Comparative Medicine, University of Washington, Seattle, Washington, USA
| | - Christopher Traudt
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Chad Curtis
- Department of Chemical Engineering, University of Washington, Seattle, Washington, USA
| | - Elizabeth Nance
- Department of Chemical Engineering, University of Washington, Seattle, Washington, USA
| | - Pratik Parikh
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Sandra E Juul
- Division of Neonatology, Department of Pediatrics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
37
|
Tolaymat Y, Doré S, Griffin HW, Shih S, Edwards ME, Weiss MD. Inhaled Gases for Neuroprotection of Neonates: A Review. Front Pediatr 2019; 7:558. [PMID: 32047729 PMCID: PMC6996209 DOI: 10.3389/fped.2019.00558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 12/20/2019] [Indexed: 11/30/2022] Open
Abstract
Importance: Hypoxic-ischemic encephalopathy (HIE) is a significant cause of morbidity and mortality in neonates. The incidence of HIE is 1-8 per 1,000 live births in developed countries. Whole-body hypothermia reduces the risk of disability or death, but 7 infants needed to be treated to prevent death or major neurodevelopmental disability. Inhalational gases may be promising synergistic agents due to their rapid onset and easy titratability. Objective: To review current data on different inhaled gases with neuroprotective properties that may serve as adjunct therapies to hypothermia. Evidence review: Literature review was performed using the PubMed database, google scholar, and ClinicalTrials.Gov. Results focused on articles published from January 1, 2005, through December 31, 2017. Articles published earlier than 2005 were included when appropriate for historical perspective. Our review emphasized preclinical and clinical studies relevant to the use of inhaled agents for neuroprotection. Findings: Based on the relevance to our topic, 111 articles were selected pertaining to the incidence of HIE, pathophysiology of HIE, therapeutic hypothermia, and emerging therapies for hypoxic-ischemic encephalopathy in preclinical and clinical settings. Supplemental tables summarizes highly relevant 49 publications that were included in this review. The selected publications emphasize the emergence of promising inhaled gases that may improve neurologic survival and alleviate neurodevelopmental disability when combined with therapeutic hypothermia in the future. Conclusions: Many inhaled agents have neuroprotective properties and could serve as an adjunct therapy to whole-body hypothermia. Inhaled agents are ideal due to their easy administration, titrability, and rapid onset and offset.
Collapse
Affiliation(s)
- Youness Tolaymat
- Department of Pediatrics, University of Florida, Gainesville, FL, United States
| | - Sylvain Doré
- Departments of Neurology, Psychiatry, Pharmaceuticals and Neuroscience, University of Florida, Gainesville, FL, United States
| | - Hudson W Griffin
- Department of Anesthesiology, University of Florida, Gainesville, FL, United States
| | - Susana Shih
- Department of Anesthesiology, University of Florida, Gainesville, FL, United States
| | - Mary E Edwards
- Health Science Center Libraries, University of Florida, Gainesville, FL, United States
| | - Michael D Weiss
- Department of Pediatrics, University of Florida, Gainesville, FL, United States
| |
Collapse
|
38
|
Sabinina TS, Bagaev VG, Alekseev IF. Prospects for Applying Xenon Curative Properties in Pediatrics. PEDIATRIC PHARMACOLOGY 2018. [DOI: 10.15690/pf.v15i5.1961] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The review discusses experimental and clinical trials on applying noble gas Xenon to treat therapeutic conditions in adults, as well as the prospects for its applying in children. Xenon therapeutic effects on the body are based on the healing properties of a noble gas. Xenon is close to the ‘ideal anesthetic’ by its anesthetic properties; but in addition, it possesses organoand neuroprotective as well as anti-stress properties which have been proved in experiment and clinically. Xenon in pediatric practice is an attractive agent because it is non-toxic, effective for the treatment of posthypoxic and traumatic impairments of the central nervous system, pain syndromes and stress conditions at its therapeutic concentration up to 30%.
Collapse
Affiliation(s)
| | | | - Ilia F. Alekseev
- Research Institute of Emergency Pediatric Surgery and Traumatology
| |
Collapse
|
39
|
Koehler RC, Yang ZJ, Lee JK, Martin LJ. Perinatal hypoxic-ischemic brain injury in large animal models: Relevance to human neonatal encephalopathy. J Cereb Blood Flow Metab 2018; 38:2092-2111. [PMID: 30149778 PMCID: PMC6282216 DOI: 10.1177/0271678x18797328] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Perinatal hypoxia-ischemia resulting in death or lifelong disabilities remains a major clinical disorder. Neonatal models of hypoxia-ischemia in rodents have enhanced our understanding of cellular mechanisms of neural injury in developing brain, but have limitations in simulating the range, accuracy, and physiology of clinical hypoxia-ischemia and the relevant systems neuropathology that contribute to the human brain injury pattern. Large animal models of perinatal hypoxia-ischemia, such as partial or complete asphyxia at the time of delivery of fetal monkeys, umbilical cord occlusion and cerebral hypoperfusion at different stages of gestation in fetal sheep, and severe hypoxia and hypoperfusion in newborn piglets, have largely overcome these limitations. In monkey, complete asphyxia produces preferential injury to cerebellum and primary sensory nuclei in brainstem and thalamus, whereas partial asphyxia produces preferential injury to somatosensory and motor cortex, basal ganglia, and thalamus. Mid-gestational fetal sheep provide a valuable model for studying vulnerability of progenitor oligodendrocytes. Hypoxia followed by asphyxia in newborn piglets replicates the systems injury seen in term newborns. Efficacy of post-insult hypothermia in animal models led to the success of clinical trials in term human neonates. Large animal models are now being used to explore adjunct therapy to augment hypothermic neuroprotection.
Collapse
Affiliation(s)
- Raymond C Koehler
- 1 Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Zeng-Jin Yang
- 1 Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Jennifer K Lee
- 1 Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA.,2 The Pathobiology Graduate Training Program, Johns Hopkins University, Baltimore, MD, USA
| | - Lee J Martin
- 2 The Pathobiology Graduate Training Program, Johns Hopkins University, Baltimore, MD, USA.,3 Department of Pathology, Division of Neuropathology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
40
|
Bruschettini M, Romantsik O, Moreira A, Ley D, Thébaud B. Stem cell-based interventions for the prevention of morbidity and mortality following hypoxic-ischaemic encephalopathy in newborn infants. Hippokratia 2018. [DOI: 10.1002/14651858.cd013202] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Matteo Bruschettini
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
- Skåne University Hospital; Cochrane Sweden; Wigerthuset, Remissgatan 4, first floor room 11-221 Lund Sweden 22185
| | - Olga Romantsik
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
| | - Alvaro Moreira
- University of Texas Health Science Center at San Antonio; Pediatrics, Division of Neonatology; San Antonio Texas USA
| | - David Ley
- Lund University, Skåne University Hospital; Department of Paediatrics; Lund Sweden
| | - Bernard Thébaud
- Children's Hospital of Eastern Ontario; Department of Pediatrics; Ottawa ON Canada
- Ottawa Hospital Research Institute, Sprott Center for Stem Cell Research; Ottawa Canada
- University of Ottawa; Department of Cellular and Molecular Medicine; Ottawa Canada
| |
Collapse
|
41
|
Fazel Bakhsheshi M, Keenliside L, Lee TY. A novel selective cooling system for the brain: feasibility study in rabbits vs piglets. Intensive Care Med Exp 2018; 6:45. [PMID: 30387029 PMCID: PMC6212374 DOI: 10.1186/s40635-018-0211-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 10/21/2018] [Indexed: 01/09/2023] Open
Abstract
Background Selective brain cooling (SBC) methods could alleviate the complications associated with systemic hypothermia. The authors (MFB, LK, and T-YL) have developed a simple and an effective nasopharyngeal SBC method using a vortex tube. The primary focus of the study is to evaluate the effectiveness of this approach on rabbits and compare it with our previous published finding on piglets, which are mammals without and with a carotid rete, respectively. Methods Experiments were conducted on six rabbits. Body temperature was measured continuously using an esophageal temperature probe while brain temperature was measured with an implanted thermometer. Two successive experiments were performed on each animal. In the first experiment, brain cooling was initiated by blowing room temperature air from the hospital medical air outlet, at a flow rate of 14–15 L/min into both nostrils for 60 min. The second series of measurements and brain cooling was performed in the same manner as the first one but blowing cold air (− 7 °C) at the same flow rate. Results One hour post cooling with room temperature air at a flow rate of 14–15 L/min, the brain temperature was 34.2 ± 1.2 °C which resulted in mean brain cooling rates of 3.7 ± 0.9 °C/h. Brain temperature could be reduced more rapidly at mean rates of 5.2 ± 1.9 °C/h, while the body temperature as measured by the esophageal temperature probe was maintained above 36 °C during cooling and maintaining period. Conclusions We have demonstrated that using the vortex tube allows initial rapid and SBC in rabbits. Moreover, comparing results between piglets and rabbits demonstrates clearly that the lack of a carotid rete does not prevent specific cooling of the brain by means of the nasopharyngeal method.
Collapse
Affiliation(s)
- Mohammad Fazel Bakhsheshi
- Imaging Program, Lawson Health Research Institute, London, Ontario, Canada. .,Imaging Research Laboratories, Robarts Research Institute, 100 Perth Drive, P.O. Box 5015, London, Ontario, N6A 5k8, Canada.
| | - Lynn Keenliside
- Imaging Program, Lawson Health Research Institute, London, Ontario, Canada
| | - Ting-Yim Lee
- Imaging Program, Lawson Health Research Institute, London, Ontario, Canada.,Imaging Research Laboratories, Robarts Research Institute, 100 Perth Drive, P.O. Box 5015, London, Ontario, N6A 5k8, Canada.,Departments of Medical Imaging and Biophysics, The University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
42
|
Zhao CS, Li H, Wang Z, Chen G. Potential application value of xenon in stroke treatment. Med Gas Res 2018; 8:116-120. [PMID: 30319767 PMCID: PMC6178644 DOI: 10.4103/2045-9912.241077] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 08/03/2018] [Indexed: 11/04/2022] Open
Abstract
Stroke is an acute disease with extremely high mortality and disability, including ischemic stroke and hemorrhagic stroke. Currently only limited drugs and treatments have been shown to have neuroprotective effects in stroke. As a medical gas, xenon has been proven to have neuroprotective effect in considerable amount of previous study. Its unique properties are different from other neuroprotective agents, making it is promising to play a special therapeutic role in stroke, either alone or in combination with other treatments. In this article, we aim to review the role of xenon in the treatment of stroke, and summarize the mechanism of using xenon to produce therapeutic effects after stroke according to the existing research. Moreover, we intend to explore and demonstrate the feasibility and safety of xenon for clinical treatment of stroke. Despite the disadvantages of difficulty in obtaining and being expensive, as long as the use of reasonable methods, xenon can play an important role in the treatment of stroke.
Collapse
Affiliation(s)
- Chong-Shun Zhao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Hao Li
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
43
|
Rüegger CM, Davis PG, Cheong JL. Xenon as an adjuvant to therapeutic hypothermia in near-term and term newborns with hypoxic-ischaemic encephalopathy. Cochrane Database Syst Rev 2018; 8:CD012753. [PMID: 30123976 PMCID: PMC6513612 DOI: 10.1002/14651858.cd012753.pub2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Hypoxic-ischaemic encephalopathy (HIE) is a serious birth complication affecting term and late preterm newborns. Although therapeutic hypothermia (cooling) has been shown to be an effective therapy for neonatal HIE, many cooled infants have poor long-term neurodevelopmental outcomes. In animal models of neonatal encephalopathy, inhaled xenon combined with cooling has been shown to offer better neuroprotection than cooling alone. OBJECTIVES To determine the effects of xenon as an adjuvant to therapeutic hypothermia on mortality and neurodevelopmental morbidity, and to ascertain clinically important side effects of xenon plus therapeutic hypothermia in newborn infants with HIE. To assess early predictors of adverse outcomes and potential side effects of xenon. SEARCH METHODS We used the standard strategy of the Cochrane Neonatal Review Group to search the Cochrane Library (2017, Issue 8), MEDLINE (from 1966), Embase (from 1966), and PubMed (from 1966) for randomised controlled and quasi-randomised trials. We also searched conference proceedings and the reference lists of cited articles. We conducted our most recent search in August 2017. SELECTION CRITERIA We included all trials allocating term or late preterm encephalopathic newborns to cooling plus xenon or cooling alone, irrespective of timing (starting age and duration) and concentrations used for xenon administration. DATA COLLECTION AND ANALYSIS Two review authors independently assessed results of searches against predetermined criteria for inclusion, assessed risk of bias, and extracted data. We performed meta-analyses using risk ratios (RRs), risk differences (RDs), and number needed to treat for an additional beneficial outcome (NNTB) with 95% confidence intervals (CIs) for dichotomous outcomes and mean differences (MDs) for continuous data. MAIN RESULTS A single randomised controlled trial enrolling 92 participants was eligible for this review. Researchers have not reported long-term neurodevelopmental outcomes, including the primary outcome of this review - death or long-term major neurodevelopmental disability in infancy (18 months to three years of age). Cooling plus xenon was not associated with reduced mortality at latest follow-up, based upon low quality evidence. Investigators noted no substantial differences between groups for other secondary outcomes of this review, such as biomarkers of brain damage assessed with magnetic resonance imaging and occurrence of seizures during primary hospitalisation. Available data do not show an increased adverse event rate in the cooling plus xenon group compared with the cooling alone group. AUTHORS' CONCLUSIONS Current evidence from one small randomised controlled pilot trial is inadequate to show whether cooling plus xenon is safe or effective in near-term and term newborns with HIE. Further trials reporting long-term neurodevelopmental outcomes are needed.
Collapse
Affiliation(s)
- Christoph M Rüegger
- University Hospital and University of ZürichNewborn Research, Department of NeonatologyFrauenklinikstrasse 10ZürichZürichSwitzerland8091
- The Royal Women’s HospitalNewborn Research Centre and Neonatal ServicesMelbourneAustralia
| | - Peter G Davis
- The Royal Women’s HospitalNewborn Research Centre and Neonatal ServicesMelbourneAustralia
- Murdoch Children's Research InstituteMelbourneAustralia
- University of MelbourneDepartment of Obstetrics and GynecologyMelbourneAustralia
| | - Jeanie L Cheong
- The Royal Women’s HospitalNewborn Research Centre and Neonatal ServicesMelbourneAustralia
- Murdoch Children's Research InstituteMelbourneAustralia
- University of MelbourneDepartment of Obstetrics and GynecologyMelbourneAustralia
| | | |
Collapse
|
44
|
Amer AR, Oorschot DE. Xenon Combined With Hypothermia in Perinatal Hypoxic-Ischemic Encephalopathy: A Noble Gas, a Noble Mission. Pediatr Neurol 2018; 84:5-10. [PMID: 29887039 DOI: 10.1016/j.pediatrneurol.2018.02.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 02/21/2018] [Indexed: 12/14/2022]
Abstract
Perinatal hypoxia-ischemia is a major cause of neonatal morbidity. It generates primary neuronal damage of the neonatal brain and later secondary damage when reperfusion of the ischemic brain tissue causes a surge of oxygen free radicals and inflammation. This post-hypoxic-ischemic brain damage is a leading cause of motor and intellectual disabilities in survivors. Research worldwide has focused on mitigating this injury. Mild or moderate hypothermia is the standard treatment in many centers. However, its benefit is modest and the search for combinatorial effective neuroprotectants continues. This review focuses on xenon as one such agent. The use of mild to moderate hypothermia is reviewed first. Then promising results on the use of xenon to potentiate the effect of hypothermia in in vitro and in vivo animal experiments are discussed. In the first feasibility study on human neonates, researchers found a significant benefit of using 50% xenon for 18 hours in addition to 72 hours of hypothermia. Yet, this additional benefit of xenon was lacking in a larger cohort study, potentially because xenon was used beyond six hours of birth. The future of using xenon is promising, but further clinical studies are awaited to confirm the feasibility of its routine use and its optimal timing, concentration, and duration, for human neonatal hypoxia-ischemia.
Collapse
Affiliation(s)
- Ashraf R Amer
- Department of Anatomy, School of Biomedical Sciences and the Brain Health Research Center, University of Otago, Dunedin, New Zealand
| | - Dorothy E Oorschot
- Department of Anatomy, School of Biomedical Sciences and the Brain Health Research Center, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
45
|
Neurologic and cognitive outcomes associated with the clinical use of xenon: a systematic review and meta-analysis of randomized-controlled trials. Can J Anaesth 2018; 65:1041-1056. [PMID: 29858987 DOI: 10.1007/s12630-018-1163-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 04/18/2018] [Accepted: 05/25/2018] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Xenon has been shown to have positive neurologic effects in various pre-clinical models. This study systematically reviewed the randomized-controlled trials (RCTs) investigating neurologic and cognitive outcomes associated with the clinical use of xenon. METHODS We searched PubMed, CENTRAL, EMBASE, CINAHL, elibrary.ru (for Russian studies), Google Scholar (for Russian studies), and Wanfang (for Chinese studies) for appropriate RCTs comparing neurologic or cognitive outcomes after clinical use of xenon with control treatment or with other anesthetic agents. RESULTS Seventeen RCTs met the inclusion criteria. Two studies investigated the effects of xenon plus therapeutic hypothermia to treat neonatal asphyxia or out-of-hospital cardiac arrest. Compared with therapeutic hypothermia alone, xenon and therapeutic hypothermia reduced cerebral white matter abnormalities after cardiac arrest but had no effect on neurocognitive outcome and mortality. Xenon had no added value when used to treat neonatal asphyxia. Thirteen RCTs compared neurocognitive effects of xenon with other anesthetic agents in surgical patients. While xenon may be associated with improved short-term (< three hours) cognitive outcome, no medium-term (six hours to three months) advantage was observed, and longer-term data are lacking. No differences in biochemical (S-100β, neuron-specific enolase) and neuropsychologic (attentional performance) outcomes were found with xenon compared with other anesthetic drugs. Finally, two studies suggest that brief, intermittent administration of sub-anesthetic doses of xenon to patients during the acute phase of substance withdrawal may improve neurocognitive outcomes. CONCLUSIONS Despite promising pre-clinical results, the evidence for positive clinical neurologic and cognitive outcomes associated with xenon administration is modest. Nevertheless, there is some evidence to suggest that xenon may be associated with better neurologic outcomes compared with the standard of care therapy in certain specific clinical situations. More clinical trials are needed to determine any potential benefit linked to xenon administration.
Collapse
|
46
|
Campos-Pires R, Koziakova M, Yonis A, Pau A, Macdonald W, Harris K, Edge CJ, Franks NP, Mahoney PF, Dickinson R. Xenon Protects against Blast-Induced Traumatic Brain Injury in an In Vitro Model. J Neurotrauma 2018; 35:1037-1044. [PMID: 29285980 PMCID: PMC5899289 DOI: 10.1089/neu.2017.5360] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The aim of this study was to evaluate the neuroprotective efficacy of the inert gas xenon as a treatment for patients with blast-induced traumatic brain injury in an in vitro laboratory model. We developed a novel blast traumatic brain injury model using C57BL/6N mouse organotypic hippocampal brain-slice cultures exposed to a single shockwave, with the resulting injury quantified using propidium iodide fluorescence. A shock tube blast generator was used to simulate open field explosive blast shockwaves, modeled by the Friedlander waveform. Exposure to blast shockwave resulted in significant (p < 0.01) injury that increased with peak-overpressure and impulse of the shockwave, and which exhibited a secondary injury development up to 72 h after trauma. Blast-induced propidium iodide fluorescence overlapped with cleaved caspase-3 immunofluorescence, indicating that shock-wave–induced cell death involves apoptosis. Xenon (50% atm) applied 1 h after blast exposure reduced injury 24 h (p < 0.01), 48 h (p < 0.05), and 72 h (p < 0.001) later, compared with untreated control injury. Xenon-treated injured slices were not significantly different from uninjured sham slices at 24 h and 72 h. We demonstrate for the first time that xenon treatment after blast traumatic brain injury reduces initial injury and prevents subsequent injury development in vitro. Our findings support the idea that xenon may be a potential first-line treatment for those with blast-induced traumatic brain injury.
Collapse
Affiliation(s)
- Rita Campos-Pires
- 1 Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London , London, United Kingdom .,2 Royal British Legion Centre for Blast Injury Studies, Department of Bioengineering, Imperial College London , London, United Kingdom
| | - Mariia Koziakova
- 1 Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London , London, United Kingdom .,2 Royal British Legion Centre for Blast Injury Studies, Department of Bioengineering, Imperial College London , London, United Kingdom
| | - Amina Yonis
- 1 Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London , London, United Kingdom
| | - Ashni Pau
- 1 Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London , London, United Kingdom
| | - Warren Macdonald
- 2 Royal British Legion Centre for Blast Injury Studies, Department of Bioengineering, Imperial College London , London, United Kingdom .,3 Department of Bioengineering, Imperial College London , London, United Kingdom
| | - Katie Harris
- 1 Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London , London, United Kingdom
| | - Christopher J Edge
- 4 Department of Life Sciences, Imperial College London , London, United Kingdom .,5 Department of Anaesthetics, Royal Berkshire Hospital NHS Foundation Trust , Reading, United Kingdom
| | - Nicholas P Franks
- 4 Department of Life Sciences, Imperial College London , London, United Kingdom
| | - Peter F Mahoney
- 6 Royal Centre for Defence Medicine , Medical Directorate Joint Force Command, ICT Centre, Birmingham, United Kingdom
| | - Robert Dickinson
- 1 Anaesthetics, Pain Medicine and Intensive Care Section, Department of Surgery and Cancer, Imperial College London , London, United Kingdom .,2 Royal British Legion Centre for Blast Injury Studies, Department of Bioengineering, Imperial College London , London, United Kingdom
| |
Collapse
|
47
|
Yang YW, Wang YL, Lu JK, Tian L, Jin M, Cheng WP. Delayed xenon post-conditioning mitigates spinal cord ischemia/reperfusion injury in rabbits by regulating microglial activation and inflammatory factors. Neural Regen Res 2018; 13:510-517. [PMID: 29623938 PMCID: PMC5900516 DOI: 10.4103/1673-5374.228757] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The neuroprotective effect against spinal cord ischemia/reperfusion injury in rats exerted by delayed xenon post-conditioning is stronger than that produced by immediate xenon post-conditioning. However, the mechanisms underlying this process remain unclear. Activated microglia are the main inflammatory cell type in the nervous system. The release of pro-inflammatory factors following microglial activation can lead to spinal cord damage, and inhibition of microglial activation can relieve spinal cord ischemia/reperfusion injury. To investigate how xenon regulates microglial activation and the release of inflammatory factors, a rabbit model of spinal cord ischemia/reperfusion injury was induced by balloon occlusion of the infrarenal aorta. After establishment of the model, two interventions were given: (1) immediate xenon post-conditioning—after reperfusion, inhalation of 50% xenon for 1 hour, 50% N2/50%O2 for 2 hours; (2) delayed xenon post-conditioning—after reperfusion, inhalation of 50% N2/50%O2 for 2 hours, 50% xenon for 1 hour. At 4, 8, 24, 48 and 72 hours after reperfusion, hindlimb locomotor function was scored using the Jacobs locomotor scale. At 72 hours after reperfusion, interleukin 6 and interleukin 10 levels in the spinal cord of each group were measured using western blot assays. Iba1 levels were determined using immunohistochemistry and a western blot assay. The number of normal neurons at the injury site was quantified using hematoxylin-eosin staining. At 72 hours after reperfusion, delayed xenon post-conditioning remarkably enhanced hindlimb motor function, increased the number of normal neurons at the injury site, decreased Iba1 levels, and inhibited interleukin-6 and interleukin-10 levels in the spinal cord. Immediate xenon post-conditioning did not noticeably affect the above-mentioned indexes. These findings indicate that delayed xenon post-conditioning after spinal cord injury improves the recovery of neurological function by reducing microglial activation and the release of interleukin-6 and interleukin-10.
Collapse
Affiliation(s)
- Yan-Wei Yang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Yun-Lu Wang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Jia-Kai Lu
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Lei Tian
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Mu Jin
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| | - Wei-Ping Cheng
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China
| |
Collapse
|
48
|
Harriman T, Bradshaw WT, Blake SM. The Use of Whole Body Cooling in the Treatment of Hypoxic-Ischemic Encephalopathy. Neonatal Netw 2017; 36:273-279. [PMID: 28847350 DOI: 10.1891/0730-0832.36.5.273] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a major cause of morbidity and mortality in neonates. Hypoxic-ischemic encephalopathy occurs as a result of a perinatal hypoxic-ischemic event just prior to or during delivery. Therapeutic hypothermia using whole body cooling is the current treatment of choice to reduce brain injury and improve long-term neurodevelopmental outcomes for neonates with HIE. All English language articles published since 2005 in PubMed and the Cumulative Index to Nursing and Allied Health Literature (CINAHL) were analyzed for existing evidence-based methods for whole body cooling. Whole body cooling is effective in the treatment of HIE in term and near-term neonates. Further research is needed to investigate the use of adjunctive therapies in conjunction with whole body cooling for improved neuroprotection.
Collapse
|
49
|
Rüegger CM, Davis PG, Cheong JL. Xenon as an adjuvant to therapeutic hypothermia in near term and term newborns with hypoxic ischaemic encephalopathy. Hippokratia 2017. [DOI: 10.1002/14651858.cd012753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Christoph M Rüegger
- The Royal Women’s Hospital; Newborn Research Centre and Neonatal Services; Locked Bag 300 Grattan St & Flemington Road Melbourne Victoria Australia Parkville 3052
- University Hospital and University of Zürich; Newborn Research, Department of Neonatology; Frauenklinikstrasse 10 Zürich Zürich Switzerland 8091
| | - Peter G Davis
- The Royal Women’s Hospital; Newborn Research Centre and Neonatal Services; Locked Bag 300 Grattan St & Flemington Road Melbourne Victoria Australia Parkville 3052
- Murdoch Childrens Research Institute; Melbourne Australia
- University of Melbourne; Department of Obstetrics and Gynecology; Melbourne Australia
| | - Jeanie L Cheong
- The Royal Women’s Hospital; Newborn Research Centre and Neonatal Services; Locked Bag 300 Grattan St & Flemington Road Melbourne Victoria Australia Parkville 3052
- Murdoch Childrens Research Institute; Melbourne Australia
- University of Melbourne; Department of Obstetrics and Gynecology; Melbourne Australia
| |
Collapse
|
50
|
Effects of Poly(ADP-Ribose) Polymerase-1 Inhibition in a Neonatal Rodent Model of Hypoxic-Ischemic Injury. BIOMED RESEARCH INTERNATIONAL 2017; 2017:2924848. [PMID: 28698869 PMCID: PMC5494065 DOI: 10.1155/2017/2924848] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 04/11/2017] [Accepted: 04/26/2017] [Indexed: 12/31/2022]
Abstract
Background Hypoxia ischemia (HI) to the developing brain occurs in 1–6 in 1000 live births. Large numbers of survivors have neurological long-term sequelae. However, mechanisms of recovery after HI are not understood and preventive measures or clinical treatments are not effective. Poly(ADP-ribose) polymerase-1 is overactivated in response to ischemia. In neonatal mice HI activates PARP-1 but its role in perinatal brain injury remains uncertain. Objective Aim of this study was to explore the effect of TES448 (PARP-1-inhibitor) and hypothermia after an ischemic insult. Design and Methods 10-day-old Wistar rats underwent HI. TES448 was given 10 min, 3 hrs, and 6 hrs after hypoxia. Hypothermia was started 30 min after HI and brains were dissected at P12. Western blotting and histological staining were used to evaluate for degree of injury. Results Protein expression of PARP-1 levels was diminished after TES448 treatment. Cresyl violet and TUNEL staining revealed decreased injury in male rat pups following TES448 and combined treatment. Female rats showed increased numbers of TUNEL-positive cells after combined therapy. TES448 inhibited microglia activation after hypoxic-ischemic injury. A cellular response including NeuN, Olig2, and MBP was not affected by PARP-1-inhibition. Conclusions Inhibition of PARP-1 and hypothermia lead to an alteration of injury but this effect is sexually dimorphic.
Collapse
|