1
|
Hernández AF, Romero-Molina D, Gonzalez-Alzaga B, López-Flores I, Lacasaña M. Changes in molecular biomarkers of neurotoxicity in newborns following prenatal exposure to organophosphate compounds. CHEMOSPHERE 2025; 374:144204. [PMID: 39938320 DOI: 10.1016/j.chemosphere.2025.144204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/15/2025] [Accepted: 02/05/2025] [Indexed: 02/14/2025]
Abstract
The rising incidence of neurodevelopmental disorders has been associated with early-life exposure to environmental factors, particularly neurotoxicants like organophosphate compounds (OPCs), which can impact brain development and function. This study examined the relationship between prenatal exposure to OPCs and molecular biomarkers of neurotoxicity in cord blood from 398 mother-child pairs in the GENEIDA birth cohort (Southeastern Spain). Urine samples were collected in the first and third trimesters and analysed for metabolites of organophosphate pesticides (dialkylphosphates, DAPs) and flame retardants (OPFRs) using LC-MS/MS. Six neurotoxicity biomarkers-brain-derived neurotrophic factor (BDNF), neurogranin (Ng), ubiquitin C-terminal hydrolase L1 (UCHL1), neurofilament heavy chain (NFH), glial fibrillary acidic protein (GFAP), and S100 calcium-binding protein B (S100B)-were measured in cord blood via multiplex analysis using Luminex xMAP technology. The results showed that higher levels of DAP metabolites in maternal urine, particularly those with dimethyl groups, were associated with increased GFAP levels in cord blood. OPFR metabolites correlated with elevated GFAP and UCHL1 levels. Sex-specific effects were observed: OPFR levels were linked to higher GFAP in boys, while higher DAP metabolites, particularly those with diethyl groups, were associated with elevated BDNF in girls. Additionally, higher levels of diphenyl phosphate, an OPFR metabolite, were linked to increased S100B in boys. These findings suggest sexually dimorphic effects of prenatal OPC exposure. These findings provide preliminary evidence of developmental neurotoxicity and suggest potential biomarkers in cord blood for early detection of neurodevelopmental deficits. Further studies, including neurobehavioral evaluations and brain imaging, are needed to better understand these molecular effects.
Collapse
Affiliation(s)
- Antonio F Hernández
- Department of Legal Medicine and Toxicology, University of Granada School of Medicine, Granada, Spain; Instituto de Investigación Biosanitaria, ibs.GRANADA, Granada, Spain; CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain.
| | - Desirée Romero-Molina
- Instituto de Investigación Biosanitaria, ibs.GRANADA, Granada, Spain; Statistics and Operations Research Department, Faculty of Sciences, University of Granada, Granada, Spain
| | - Beatriz Gonzalez-Alzaga
- Instituto de Investigación Biosanitaria, ibs.GRANADA, Granada, Spain; CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain; Andalusian School of Public Health, Granada, Spain
| | - Inmaculada López-Flores
- Instituto de Investigación Biosanitaria, ibs.GRANADA, Granada, Spain; Department of Genetics, Faculty of Sciences, University of Granada, Granada, Granada, Spain
| | - Marina Lacasaña
- Instituto de Investigación Biosanitaria, ibs.GRANADA, Granada, Spain; CIBER Epidemiology and Public Health (CIBERESP), Madrid, Spain; Andalusian School of Public Health, Granada, Spain; Andalusian Health and Environment Observatory (OSMAN), Granada, Spain.
| |
Collapse
|
2
|
Yang M, Wang K, Liu B, Shen Y, Liu G. Hypoxic-Ischemic Encephalopathy: Pathogenesis and Promising Therapies. Mol Neurobiol 2025; 62:2105-2122. [PMID: 39073530 DOI: 10.1007/s12035-024-04398-9] [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: 01/05/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a brain lesion caused by inadequate blood supply and oxygen deprivation, often occurring in neonates. It has emerged as a grave complication of neonatal asphyxia, leading to chronic neurological damage. Nevertheless, the precise pathophysiological mechanisms underlying HIE are not entirely understood. This paper aims to comprehensively elucidate the contributions of hypoxia-ischemia, reperfusion injury, inflammation, oxidative stress, mitochondrial dysfunction, excitotoxicity, ferroptosis, endoplasmic reticulum stress, and apoptosis to the onset and progression of HIE. Currently, hypothermia therapy stands as the sole standard treatment for neonatal HIE, albeit providing only partial neuroprotection. Drug therapy and stem cell therapy have been explored in the treatment of HIE, exhibiting certain neuroprotective effects. Employing drug therapy or stem cell therapy as adjunctive treatments to hypothermia therapy holds great significance. This article presents a systematic review of the pathogenesis and treatment strategies of HIE, with the goal of enhancing the effect of treatment and improving the quality of life for HIE patients.
Collapse
Affiliation(s)
- Mingming Yang
- Department of Pediatrics, Binhai County People's Hospital, Yancheng, Jiangsu Province, 224500, P. R. China
| | - Kexin Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Boya Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, 226001, P. R. China.
| | - Guangliang Liu
- Department of Pediatrics, Binhai County People's Hospital, Yancheng, Jiangsu Province, 224500, P. R. China.
| |
Collapse
|
3
|
Notarbartolo V, Badiane BA, Angileri VM, Piro E, Giuffrè M. Antioxidant Therapy in Neonatal Hypoxic Ischemic Encephalopathy: Adjuvant or Future Alternative to Therapeutic Hypothermia? Metabolites 2024; 14:630. [PMID: 39590867 PMCID: PMC11596076 DOI: 10.3390/metabo14110630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/05/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Oxidative stress-related diseases in newborns arise from pro-oxidant/antioxidant imbalance in both term and preterm neonates. Pro-oxidant/antioxidant imbalance has shown to be present in different pathological conditions such as hypoxic ischemic encephalopathy (HIE), retinopathy of prematurity (ROP), bronchopulmonary dysplasia (BPD), necrotizing enterocolitis (NEC), and patent ductus arteriosus (PDA). METHODS AND RESULTS We performed a narrative review according to the most recent available literature (2012-2024), using Scopus and PubMed as electronic databases. Many observational and experimental studies in vitro and in vivo have evaluated the effectiveness of antioxidant therapies such as melatonin, erythropoietin (EPO), allopurinol, N-acetylcisteine (NAS), and nitric oxide synthase (NOS) inhibitors in these diseases. Perinatal asphyxia is one of the most important causes of mortality and morbidity in term and near-term newborns. Therapeutic hypothermia (TH) is the gold standard treatment for neonates with moderate-severe perinatal asphyxia, resulting in a reduction in the mortality and neurodevelopmental disability rates. CONCLUSIONS According to the most recent literature and clinical trials, melatonin, allopurinol, NAS, NOS inhibitors, magnesium sulfate, and stem cells stand out as promising as both adjuvants and future probable alternatives to TH in the treatment of HIE.
Collapse
Affiliation(s)
- Veronica Notarbartolo
- Neonatology and Neonatal Intensive Care Unit, University Hospital “Paolo Giaccone”, 90127 Palermo, Italy
| | - Bintu Ayla Badiane
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy; (B.A.B.); (E.P.); (M.G.)
| | - Vita Maria Angileri
- Neonatal Intensive Care Unit with Neonatology, “G.F. Ingrassia” Hospital Unit, 90131 Palermo, Italy;
| | - Ettore Piro
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy; (B.A.B.); (E.P.); (M.G.)
| | - Mario Giuffrè
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy; (B.A.B.); (E.P.); (M.G.)
| |
Collapse
|
4
|
Sulyok E, Farkas B, Bodis J. Pathomechanisms of Prenatally Programmed Adult Diseases. Antioxidants (Basel) 2023; 12:1354. [PMID: 37507894 PMCID: PMC10376205 DOI: 10.3390/antiox12071354] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/22/2023] [Accepted: 06/25/2023] [Indexed: 07/30/2023] Open
Abstract
Based on epidemiological observations Barker et al. put forward the hypothesis/concept that an adverse intrauterine environment (involving an insufficient nutrient supply, chronic hypoxia, stress, and toxic substances) is an important risk factor for the development of chronic diseases later in life. The fetus responds to the unfavorable environment with adaptive reactions, which ensure survival in the short run, but at the expense of initiating pathological processes leading to adult diseases. In this review, the major mechanisms (including telomere dysfunction, epigenetic modifications, and cardiovascular-renal-endocrine-metabolic reactions) will be outlined, with a particular emphasis on the role of oxidative stress in the fetal origin of adult diseases.
Collapse
Affiliation(s)
- Endre Sulyok
- National Laboratory on Human Reproduction, University of Pécs, 7624 Pécs, Hungary
- Faculty of Health Sciences, Doctoral School of Health Sciences, University of Pécs, 7624 Pécs, Hungary
- MTA-PTE Human Reproduction Scientific Research Group, 7624 Pécs, Hungary
| | - Balint Farkas
- National Laboratory on Human Reproduction, University of Pécs, 7624 Pécs, Hungary
- MTA-PTE Human Reproduction Scientific Research Group, 7624 Pécs, Hungary
- Department of Obstetrics and Gynecology, School of Medicine, University of Pécs, 7624 Pécs, Hungary
| | - Jozsef Bodis
- National Laboratory on Human Reproduction, University of Pécs, 7624 Pécs, Hungary
- Faculty of Health Sciences, Doctoral School of Health Sciences, University of Pécs, 7624 Pécs, Hungary
- MTA-PTE Human Reproduction Scientific Research Group, 7624 Pécs, Hungary
- Department of Obstetrics and Gynecology, School of Medicine, University of Pécs, 7624 Pécs, Hungary
| |
Collapse
|
5
|
Ivkin AA, Balakhnin DG, Borisenko DV, Grigoriev EV. Opportunities of cerebroprotection in children in cardiac surgery (review of literature). MESSENGER OF ANESTHESIOLOGY AND RESUSCITATION 2023. [DOI: 10.24884/2078-5658-2023-20-1-89-96] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
Abstract
The analysis of the literature in the main search scientific systems was carried out to identify the current means of cerebroprotection. The assessment is given both to the familiar methods that have become «traditional» for cardiac surgery (hypothermia, etc.) and pharmacological approaches that are less common in clinical practice: the use of melatonin, ketamine. The characteristics of some drugs that are promising for solving this problem are also given.
Collapse
Affiliation(s)
- A. A. Ivkin
- Research Institute for Complex Problems of Cardiovascular Diseases
| | - D. G. Balakhnin
- Research Institute for Complex Problems of Cardiovascular Diseases
| | - D. V. Borisenko
- Research Institute for Complex Problems of Cardiovascular Diseases
| | - E. V. Grigoriev
- Research Institute for Complex Problems of Cardiovascular Diseases
| |
Collapse
|
6
|
Rodríguez-Rovira I, Arce C, De Rycke K, Pérez B, Carretero A, Arbonés M, Teixidò-Turà G, Gómez-Cabrera MC, Campuzano V, Jiménez-Altayó F, Egea G. Allopurinol blocks aortic aneurysm in a mouse model of Marfan syndrome via reducing aortic oxidative stress. Free Radic Biol Med 2022; 193:538-550. [PMID: 36347404 DOI: 10.1016/j.freeradbiomed.2022.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/28/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Increasing evidence indicates that redox stress participates in MFS aortopathy, though its mechanistic contribution is little known. We reported elevated reactive oxygen species (ROS) formation and NADPH oxidase NOX4 upregulation in MFS patients and mouse aortae. Here we address the contribution of xanthine oxidoreductase (XOR), which catabolizes purines into uric acid and ROS in MFS aortopathy. METHODS AND RESULTS In aortic samples from MFS patients, XOR protein expression, revealed by immunohistochemistry, increased in both the tunicae intima and media of the dilated zone. In MFS mice (Fbn1C1041G/+), aortic XOR mRNA transcripts and enzymatic activity of the oxidase form (XO) were augmented in the aorta of 3-month-old mice but not in older animals. The administration of the XOR inhibitor allopurinol (ALO) halted the progression of aortic root aneurysm in MFS mice. ALO administrated before the onset of the aneurysm prevented its subsequent development. ALO also inhibited MFS-associated endothelial dysfunction as well as elastic fiber fragmentation, nuclear translocation of pNRF2 and increased 3'-nitrotyrosine levels, and collagen maturation remodeling, all occurring in the tunica media. ALO reduced the MFS-associated large aortic production of H2O2, and NOX4 and MMP2 transcriptional overexpression. CONCLUSIONS Allopurinol interferes in aortic aneurysm progression acting as a potent antioxidant. This study strengthens the concept that redox stress is an important determinant of aortic aneurysm formation and progression in MFS and warrants the evaluation of ALO therapy in MFS patients.
Collapse
Affiliation(s)
- Isaac Rodríguez-Rovira
- Department of Biomedical Sciences, University of Barcelona School of Medicine and Health Sciences, 08036, Barcelona, Spain
| | - Cristina Arce
- Department of Biomedical Sciences, University of Barcelona School of Medicine and Health Sciences, 08036, Barcelona, Spain
| | - Karo De Rycke
- Department of Biomedical Sciences, University of Barcelona School of Medicine and Health Sciences, 08036, Barcelona, Spain
| | - Belén Pérez
- Department of Pharmacology, Toxicology and Therapeutics, Neuroscience Institute, School of Medicine, Autonomous University of Barcelona, 08193, Cerdanyola del Vallès, Spain
| | - Aitor Carretero
- Department of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, Valencia, Spain
| | - Marc Arbonés
- Department of Biomedical Sciences, University of Barcelona School of Medicine and Health Sciences, 08036, Barcelona, Spain
| | - Gisela Teixidò-Turà
- Department of Cardiology, Hospital Universitari Vall d'Hebron, Barcelona, Spain; CIBER-CV, Vall d'Hebrón Institut de Recerca (VHIR), Barcelona, Spain
| | - Mari Carmen Gómez-Cabrera
- Department of Physiology, Faculty of Medicine, University of Valencia, CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, Valencia, Spain
| | - Victoria Campuzano
- Department of Biomedical Sciences, University of Barcelona School of Medicine and Health Sciences, 08036, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Spain
| | - Francesc Jiménez-Altayó
- Department of Pharmacology, Toxicology and Therapeutics, Neuroscience Institute, School of Medicine, Autonomous University of Barcelona, 08193, Cerdanyola del Vallès, Spain
| | - Gustavo Egea
- Department of Biomedical Sciences, University of Barcelona School of Medicine and Health Sciences, 08036, Barcelona, Spain.
| |
Collapse
|
7
|
Stegeman R, Nijman M, Breur JMPJ, Groenendaal F, Haas F, Derks JB, Nijman J, van Beynum IM, Taverne YJHJ, Bogers AJJC, Helbing WA, de Boode WP, Bos AF, Berger RMF, Accord RE, Roes KCB, de Wit GA, Jansen NJG, Benders MJNL. CeRebrUm and CardIac Protection with ALlopurinol in Neonates with Critical Congenital Heart Disease Requiring Cardiac Surgery with Cardiopulmonary Bypass (CRUCIAL): study protocol of a phase III, randomized, quadruple-blinded, placebo-controlled, Dutch multicenter trial. Trials 2022; 23:174. [PMID: 35197082 PMCID: PMC8867620 DOI: 10.1186/s13063-022-06098-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 02/10/2022] [Indexed: 10/20/2024] Open
Abstract
Background Neonates with critical congenital heart disease (CCHD) undergoing cardiac surgery with cardiopulmonary bypass (CPB) are at risk of brain injury that may result in adverse neurodevelopment. To date, no therapy is available to improve long-term neurodevelopmental outcomes of CCHD neonates. Allopurinol, a xanthine oxidase inhibitor, prevents the formation of reactive oxygen and nitrogen species, thereby limiting cell damage during reperfusion and reoxygenation to the brain and heart. Animal and neonatal studies suggest that allopurinol reduces hypoxic-ischemic brain injury and is cardioprotective and safe. This trial aims to test the hypothesis that allopurinol administration in CCHD neonates will result in a 20% reduction in moderate to severe ischemic and hemorrhagic brain injury. Methods This is a phase III, randomized, quadruple-blinded, placebo-controlled, multicenter trial. Neonates with a prenatal or postnatal CCHD diagnosis requiring cardiac surgery with CPB in the first 4 weeks after birth are eligible to participate. Allopurinol or mannitol-placebo will be administered intravenously in 2 doses early postnatally in neonates diagnosed antenatally and 3 doses perioperatively of 20 mg/kg each in all neonates. The primary outcome is a composite endpoint of moderate/severe ischemic or hemorrhagic brain injury on early postoperative MRI, being too unstable for postoperative MRI, or mortality within 1 month following CPB. A total of 236 patients (n = 188 with prenatal diagnosis) is required to demonstrate a reduction of the primary outcome incidence by 20% in the prenatal group and by 9% in the postnatal group (power 80%; overall type 1 error controlled at 5%, two-sided), including 1 interim analysis at n = 118 (n = 94 with prenatal diagnosis) with the option to stop early for efficacy. Secondary outcomes include preoperative and postoperative brain injury severity, white matter injury volume (MRI), and cardiac function (echocardiography); postnatal and postoperative seizure activity (aEEG) and regional cerebral oxygen saturation (NIRS); neurodevelopment at 3 months (general movements); motor, cognitive, and language development and quality of life at 24 months; and safety and cost-effectiveness of allopurinol. Discussion This trial will investigate whether allopurinol administered directly after birth and around cardiac surgery reduces moderate/severe ischemic and hemorrhagic brain injury and improves cardiac function and neurodevelopmental outcome in CCHD neonates. Trial registration EudraCT 2017-004596-31. Registered on November 14, 2017. ClinicalTrials.gov NCT04217421. Registered on January 3, 2020 Supplementary Information The online version contains supplementary material available at 10.1186/s13063-022-06098-y.
Collapse
Affiliation(s)
- Raymond Stegeman
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center (UMC) Utrecht, Utrecht University, KE 04.123.1, PO Box 85909, 3508, AB, Utrecht, The Netherlands.,Department of Pediatric Cardiology, Wilhelmina Children's Hospital, UMC Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Pediatric Intensive Care, Wilhelmina Children's Hospital, UMC Utrecht, Utrecht University, Utrecht, The Netherlands.,Congenital Cardiothoracic Surgery, Wilhelmina Children's Hospital, UMC Utrecht, Utrecht University, Utrecht, The Netherlands.,Utrecht Brain Center, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Maaike Nijman
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center (UMC) Utrecht, Utrecht University, KE 04.123.1, PO Box 85909, 3508, AB, Utrecht, The Netherlands.,Department of Pediatric Cardiology, Wilhelmina Children's Hospital, UMC Utrecht, Utrecht University, Utrecht, The Netherlands.,Utrecht Brain Center, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Johannes M P J Breur
- Department of Pediatric Cardiology, Wilhelmina Children's Hospital, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Floris Groenendaal
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center (UMC) Utrecht, Utrecht University, KE 04.123.1, PO Box 85909, 3508, AB, Utrecht, The Netherlands.,Utrecht Brain Center, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Felix Haas
- Congenital Cardiothoracic Surgery, Wilhelmina Children's Hospital, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jan B Derks
- Department of Obstetrics, Wilhelmina Children's Hospital, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Joppe Nijman
- Department of Pediatric Intensive Care, Wilhelmina Children's Hospital, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Ingrid M van Beynum
- Department of Pediatrics, Division of Pediatric Cardiology, Academic Center for Congenital Heart Disease, Erasmus Medical Center (MC) - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Yannick J H J Taverne
- Department of Cardiothoracic Surgery, Erasmus MC, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Ad J J C Bogers
- Department of Cardiothoracic Surgery, Erasmus MC, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Willem A Helbing
- Department of Pediatrics, Division of Pediatric Cardiology, Academic Center for Congenital Heart Disease, Erasmus Medical Center (MC) - Sophia Children's Hospital, Rotterdam, The Netherlands.,Department of Pediatrics, Division of Pediatric Cardiology, Academic Center for Congenital Heart Disease, Radboudumc - Amalia Children's Hospital, Nijmegen, The Netherlands
| | - Willem P de Boode
- Department of Neonatology, Radboudumc, Radboud Institute for Health Sciences, Amalia Children's Hospital, Nijmegen, The Netherlands
| | - Arend F Bos
- Division of Neonatology, Beatrix Children's Hospital, UMC Groningen, University of Groningen, Groningen, The Netherlands
| | - Rolf M F Berger
- Center for Congenital Heart Diseases, Pediatric Cardiology, Beatrix Children's Hospital, UMC Groningen, University of Groningen, Groningen, The Netherlands
| | - Ryan E Accord
- Center for Congenital Heart Diseases, Department of Cardiothoracic Surgery, UMC Groningen, University of Groningen, Groningen, The Netherlands
| | - Kit C B Roes
- Department of Health Evidence, Section Biostatistics, Radboudumc, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - G Ardine de Wit
- Julius Center for Health Sciences and Primary Care, UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Nicolaas J G Jansen
- Department of Pediatric Intensive Care, Wilhelmina Children's Hospital, UMC Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Pediatrics, Beatrix Children's Hospital, UMC Groningen, University of Groningen, Groningen, The Netherlands
| | - Manon J N L Benders
- Department of Neonatology, Wilhelmina Children's Hospital, University Medical Center (UMC) Utrecht, Utrecht University, KE 04.123.1, PO Box 85909, 3508, AB, Utrecht, The Netherlands.
| | | |
Collapse
|
8
|
Martini S, Castellini L, Parladori R, Paoletti V, Aceti A, Corvaglia L. Free Radicals and Neonatal Brain Injury: From Underlying Pathophysiology to Antioxidant Treatment Perspectives. Antioxidants (Basel) 2021; 10:2012. [PMID: 34943115 PMCID: PMC8698308 DOI: 10.3390/antiox10122012] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 01/23/2023] Open
Abstract
Free radicals play a role of paramount importance in the development of neonatal brain injury. Depending on the pathophysiological mechanisms underlying free radical overproduction and upon specific neonatal characteristics, such as the GA-dependent maturation of antioxidant defenses and of cerebrovascular autoregulation, different profiles of injury have been identified. The growing evidence on the detrimental effects of free radicals on the brain tissue has led to discover not only potential biomarkers for oxidative damage, but also possible neuroprotective therapeutic approaches targeting oxidative stress. While a more extensive validation of free radical biomarkers is required before considering their use in routine neonatal practice, two important treatments endowed with antioxidant properties, such as therapeutic hypothermia and magnesium sulfate, have become part of the standard of care to reduce the risk of neonatal brain injury, and other promising therapeutic strategies are being tested in clinical trials. The implementation of currently available evidence is crucial to optimize neonatal neuroprotection and to develop individualized diagnostic and therapeutic approaches addressing oxidative brain injury, with the final aim of improving the neurological outcome of this population.
Collapse
Affiliation(s)
- Silvia Martini
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (A.A.); (L.C.)
- Neonatal Intensive Care Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Laura Castellini
- School of Medicine and Surgery, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy;
| | - Roberta Parladori
- Specialty School of Pediatrics, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy;
| | - Vittoria Paoletti
- Neonatal Intensive Care Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Arianna Aceti
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (A.A.); (L.C.)
- Neonatal Intensive Care Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Luigi Corvaglia
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (A.A.); (L.C.)
- Neonatal Intensive Care Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| |
Collapse
|
9
|
Martini S, Austin T, Aceti A, Faldella G, Corvaglia L. Free radicals and neonatal encephalopathy: mechanisms of injury, biomarkers, and antioxidant treatment perspectives. Pediatr Res 2020; 87:823-833. [PMID: 31655487 DOI: 10.1038/s41390-019-0639-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 09/19/2019] [Accepted: 09/22/2019] [Indexed: 12/11/2022]
Abstract
Neonatal encephalopathy (NE), most commonly a result of the disruption of cerebral oxygen delivery, is the leading cause of neurologic disability in term neonates. Given the key role of free radicals in brain injury development following hypoxia-ischemia-reperfusion, several oxidative biomarkers have been explored in preclinical and clinical models of NE. Among these, antioxidant enzyme activity, uric acid excretion, nitric oxide, malondialdehyde, and non-protein-bound iron have shown promising results as possible predictors of NE severity and outcome. Owing to high costs and technical complexity, however, their routine use in clinical practice is still limited. Several strategies aimed at reducing free radical production or upregulating physiological scavengers have been proposed for NE. Room-air resuscitation has proved to reduce oxidative stress following perinatal asphyxia and is now universally adopted. A number of medications endowed with antioxidant properties, such as melatonin, erythropoietin, allopurinol, or N-acetylcysteine, have also shown potential neuroprotective effects in perinatal asphyxia; nevertheless, further evidence is needed before these antioxidant approaches could be implemented as standard care.
Collapse
Affiliation(s)
- Silvia Martini
- Neonatology and Neonatal Intensive Care Unit, St. Orsola-Malpighi Hospital, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy.
| | - Topun Austin
- Neonatal Intensive Care Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Arianna Aceti
- Neonatology and Neonatal Intensive Care Unit, St. Orsola-Malpighi Hospital, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Giacomo Faldella
- Neonatology and Neonatal Intensive Care Unit, St. Orsola-Malpighi Hospital, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Luigi Corvaglia
- Neonatology and Neonatal Intensive Care Unit, St. Orsola-Malpighi Hospital, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
10
|
Cardinali DP. An Assessment of Melatonin's Therapeutic Value in the Hypoxic-Ischemic Encephalopathy of the Newborn. Front Synaptic Neurosci 2019; 11:34. [PMID: 31920617 PMCID: PMC6914689 DOI: 10.3389/fnsyn.2019.00034] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 11/26/2019] [Indexed: 12/17/2022] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is one of the most frequent causes of brain injury in the newborn. From a pathophysiological standpoint, a complex process takes place at the cellular and tissue level during the development of newborn brain damage in the absence of oxygen. Initially, the lesion is triggered by a deficit in the supply of oxygen to cells and tissues, causing a primary energy insufficiency. Subsequently, high energy phosphate levels recover transiently (the latent phase) that is followed by a secondary phase, in which many of the pathophysiological mechanisms involved in the development of neonatal brain damage ensue (i.e., excitotoxicity, massive influx of Ca2+, oxidative and nitrosative stress, inflammation). This leads to cell death by necrosis or apoptosis. Eventually, a tertiary phase occurs, characterized by the persistence of brain damage for months and even years after the HI insult. Hypothermia is the only therapeutic strategy against HIE that has been incorporated into neonatal intensive care units with limited success. Thus, there is an urgent need for agents with the capacity to curtail acute and chronic damage in HIE. Melatonin, a molecule of unusual phylogenetic conservation present in all known aerobic organisms, has a potential role as a neuroprotective agent both acutely and chronically in HIE. Melatonin displays a remarkable antioxidant and anti-inflammatory activity and is capable to cross the blood-brain barrier readily. Moreover, in many animal models of brain degeneration, melatonin was effective to impair chronic mechanisms of neuronal death. In animal models, and in a limited number of clinical studies, melatonin increased the level of protection developed by hypothermia in newborn asphyxia. This review article summarizes briefly the available therapeutic strategies in HIE and assesses the role of melatonin as a potentially relevant therapeutic tool to cover the hypoxia-ischemia phase and the secondary and tertiary phases following a hypoxic-ischemic insult.
Collapse
Affiliation(s)
- Daniel P. Cardinali
- Faculty of Medical Sciences, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| |
Collapse
|
11
|
|
12
|
Maiwald CA, Annink KV, Rüdiger M, Benders MJNL, van Bel F, Allegaert K, Naulaers G, Bassler D, Klebermaß-Schrehof K, Vento M, Guimarães H, Stiris T, Cattarossi L, Metsäranta M, Vanhatalo S, Mazela J, Metsvaht T, Jacobs Y, Franz AR. Effect of allopurinol in addition to hypothermia treatment in neonates for hypoxic-ischemic brain injury on neurocognitive outcome (ALBINO): study protocol of a blinded randomized placebo-controlled parallel group multicenter trial for superiority (phase III). BMC Pediatr 2019; 19:210. [PMID: 31248390 PMCID: PMC6595623 DOI: 10.1186/s12887-019-1566-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/31/2019] [Indexed: 02/07/2023] Open
Abstract
Background Perinatal asphyxia and resulting hypoxic-ischemic encephalopathy is a major cause of death and long-term disability in term born neonates. Up to 20,000 infants each year are affected by HIE in Europe and even more in regions with lower level of perinatal care. The only established therapy to improve outcome in these infants is therapeutic hypothermia. Allopurinol is a xanthine oxidase inhibitor that reduces the production of oxygen radicals as superoxide, which contributes to secondary energy failure and apoptosis in neurons and glial cells after reperfusion of hypoxic brain tissue and may further improve outcome if administered in addition to therapeutic hypothermia. Methods This study on the effects of ALlopurinol in addition to hypothermia treatment for hypoxic-ischemic Brain Injury on Neurocognitive Outcome (ALBINO), is a European double-blinded randomized placebo-controlled parallel group multicenter trial (Phase III) to evaluate the effect of postnatal allopurinol administered in addition to standard of care (including therapeutic hypothermia if indicated) on the incidence of death and severe neurodevelopmental impairment at 24 months of age in newborns with perinatal hypoxic-ischemic insult and signs of potentially evolving encephalopathy. Allopurinol or placebo will be given in addition to therapeutic hypothermia (where indicated) to infants with a gestational age ≥ 36 weeks and a birth weight ≥ 2500 g, with severe perinatal asphyxia and potentially evolving encephalopathy. The primary endpoint of this study will be death or severe neurodevelopmental impairment versus survival without severe neurodevelopmental impairment at the age of two years. Effects on brain injury by magnetic resonance imaging and cerebral ultrasound, electric brain activity, concentrations of peroxidation products and S100B, will also be studied along with effects on heart function and pharmacokinetics of allopurinol after iv-infusion. Discussion This trial will provide data to assess the efficacy and safety of early postnatal allopurinol in term infants with evolving hypoxic-ischemic encephalopathy. If proven efficacious and safe, allopurinol could become part of a neuroprotective pharmacological treatment strategy in addition to therapeutic hypothermia in children with perinatal asphyxia. Trial registration NCT03162653, www.ClinicalTrials.gov, May 22, 2017.
Collapse
Affiliation(s)
- Christian A Maiwald
- University Hospital Tuebingen, Calwerstr. 7, 72076, Tuebingen, Germany.,Center for Pediatric Clinical Studies (CPCS), University Hospital Tuebingen, Tuebingen, Germany
| | - Kim V Annink
- Universitair Medisch Centrum Utrecht, Utrecht, The Netherlands
| | - Mario Rüdiger
- Universitätsklinikum C. G. Carus - Medizinische Fakultät der TU Dresden, Dresden, Germany
| | | | - Frank van Bel
- Universitair Medisch Centrum Utrecht, Utrecht, The Netherlands
| | | | | | - Dirk Bassler
- UniversitaetsSpital Zuerich, Zuerich, Switzerland
| | | | - Maximo Vento
- Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | | | - Tom Stiris
- Oslo Universitetssykehus HF, Oslo, Norway
| | - Luigi Cattarossi
- Azienda sanitaria universitaria integrata di Udine, Udine, Italy
| | | | | | - Jan Mazela
- Poznan University of Medical Sciences - Department of Neonatology, Poznan, Poland
| | | | | | - Axel R Franz
- University Hospital Tuebingen, Calwerstr. 7, 72076, Tuebingen, Germany. .,Center for Pediatric Clinical Studies (CPCS), University Hospital Tuebingen, Tuebingen, Germany.
| | | |
Collapse
|
13
|
Cánovas-Ahedo M, Alonso-Alconada D. [Combined therapy in neonatal hypoxic-ischaemic encephalopathy]. An Pediatr (Barc) 2019; 91:59.e1-59.e7. [PMID: 31109785 DOI: 10.1016/j.anpedi.2019.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/08/2019] [Accepted: 04/12/2019] [Indexed: 01/01/2023] Open
Abstract
Neonatal hypoxic-ischaemic encephalopathy due to the lack of oxygen at birth can have severe neurological consequences, such as cerebral palsy, or even the death of the asphyxiated newborn. Hypothermia is currently the only therapy included in intensive care neonatal units. This shows a clinical benefit in neonates suffering from hypoxic-ischaemic encephalopathy, mainly because of its ability to decrease the accumulation of excitatory amino acids and its anti-inflammatory, antioxidant, and anti-apoptotic effects. However, hypothermia is not effective in half of the cases, making it necessary to search for new, or to optimize current therapies, with the aim on reducing asphyxia-derived neurological consequences, either as single treatments or in combination with cooling. Within current potential therapies, melatonin, allopurinol, and erythropoietin stand out among the others, with clinical trials on the way. While, stem cells, N-acetylcysteine and noble gases have obtained promising pre-clinical results. Melatonin produces a powerful antioxidant and anti-inflammatory effect, acting as free radical scavenger and regulating pro-inflammatory mediators. Through the inhibition of xanthine oxidase, allopurinol can decrease oxidative stress. Erythropoietin has cell death and neurogenesis as its main therapeutic targets. Keeping in mind the whole scenario of current therapies, management of neonates suffering from neonatal asphyxia could rely on the combination of one or some of these treatments, together with therapeutic hypothermia.
Collapse
Affiliation(s)
- María Cánovas-Ahedo
- Departamento de Biología Celular e Histología, Facultad de Medicina y Enfermería, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), Leioa, Bizkaia, España
| | - Daniel Alonso-Alconada
- Departamento de Biología Celular e Histología, Facultad de Medicina y Enfermería, Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), Leioa, Bizkaia, España.
| |
Collapse
|
14
|
Qin X, Cheng J, Zhong Y, Mahgoub OK, Akter F, Fan Y, Aldughaim M, Xie Q, Qin L, Gu L, Jian Z, Xiong X, Liu R. Mechanism and Treatment Related to Oxidative Stress in Neonatal Hypoxic-Ischemic Encephalopathy. Front Mol Neurosci 2019; 12:88. [PMID: 31031592 PMCID: PMC6470360 DOI: 10.3389/fnmol.2019.00088] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/21/2019] [Indexed: 12/24/2022] Open
Abstract
Hypoxic ischemic encephalopathy (HIE) is a type of neonatal brain injury, which occurs due to lack of supply and oxygen deprivation to the brain. It is associated with a high morbidity and mortality rate. There are several therapeutic strategies that can be used to improve outcomes in patients with HIE. These include cell therapies such as marrow mesenchymal stem cells (MSCs) and umbilical cord blood stem cells (UCBCs), which are being incorporated into the new protocols for the prevention of ischemic brain damage. The focus of this review is to discuss the mechanism of oxidative stress in HIE and summarize the current available treatments for HIE. We hope that a better understanding of the relationship between oxidative stress and HIE will provide new insights on the potential therapy of this devastating condition.
Collapse
Affiliation(s)
- Xingping Qin
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Neurosurgery, Harvard Medical School, Boston, MA, United States
| | - Jing Cheng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi Zhong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Omer Kamal Mahgoub
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Farhana Akter
- Department of Neurosurgery, Harvard Medical School, Boston, MA, United States.,Department of Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Yanqin Fan
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mohammed Aldughaim
- Department of Neurosurgery, Harvard Medical School, Boston, MA, United States
| | - Qiurong Xie
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lingxia Qin
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Renzhong Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
15
|
Klumper J, Kaandorp JJ, Schuit E, Groenendaal F, Koopman-Esseboom C, Mulder EJH, Van Bel F, Benders MJNL, Mol BWJ, van Elburg RM, Bos AF, Derks JB. Behavioral and neurodevelopmental outcome of children after maternal allopurinol administration during suspected fetal hypoxia: 5-year follow up of the ALLO-trial. PLoS One 2018; 13:e0201063. [PMID: 30138355 PMCID: PMC6107129 DOI: 10.1371/journal.pone.0201063] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 07/03/2018] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE To evaluate the long-term neurodevelopmental and behavioral outcome of antenatal allopurinol treatment during suspected fetal hypoxia. STUDY DESIGN We studied children born from women who participated in a randomized double-blind placebo controlled multicenter study (ALLO-trial). Labouring women in whom the fetus was suspected to have fetal hypoxia were randomly allocated to receive allopurinol or placebo. At 5 years of age, the children were assessed with 2 parent reported questionnaires, the Ages and Stages Questionnaire (ASQ) and the Child Behavior Checklist (CBCL). A child was marked abnormal for ASQ if it scored below 2 standard deviation under the normative mean of a reference population in at least one domain. For CBCL, a score above the cut-off value (95th percentile for narrowband scale, 85th percentile for broadband scale) in at least one scale was marked as abnormal. RESULTS We obtained data from 138 out of the original 222 mildly asphyxiated children included in the ALLO-trial (response rate 62%, allopurinol n = 73, placebo n = 65). At 5 years of age, the number of children that scored abnormal on the ASQ were 11 (15.1%) in the allopurinol group versus 11 (9.2%) in the placebo group (relative risk (RR) 1.64, 95% confidence interval (CI): 0.64 to 4.17, p = 0.30). On CBCL 21 children (30.4%) scored abnormal in de allopurinol group versus 12 children (20.0%) in the placebo group (RR 1.52, 95% CI: 0.82 to 2.83, p = 0.18). CONCLUSION We found no proof that allopurinol administered to labouring women with suspected fetal hypoxia improved long-term developmental and behavioral outcome. These findings are limited due to the fact that the study was potentially underpowered. TRIAL REGISTRATION NCT00189007 Dutch Trial Register NTR1383.
Collapse
Affiliation(s)
- Job Klumper
- Department of Perinatology, University Medical Center, Utrecht, The Netherlands
| | - Joepe J Kaandorp
- Department of Perinatology, University Medical Center, Utrecht, The Netherlands
| | - Ewoud Schuit
- Julius Center for Health Sciences and Primary Care, University Medical Center, Utrecht, The Netherlands
| | - Floris Groenendaal
- Department of Perinatology, University Medical Center, Utrecht, The Netherlands
| | | | - Eduard J H Mulder
- Department of Perinatology, University Medical Center, Utrecht, The Netherlands
| | - Frank Van Bel
- Department of Perinatology, University Medical Center, Utrecht, The Netherlands
| | - Manon J N L Benders
- Department of Perinatology, University Medical Center, Utrecht, The Netherlands
| | - Ben W J Mol
- Department of Perinatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Ruurd M van Elburg
- Department of Perinatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Arend F Bos
- Department of Perinatology, University Medical Center, Groningen, The Netherlands
| | - Jan B Derks
- Department of Perinatology, University Medical Center, Utrecht, The Netherlands
| |
Collapse
|
16
|
Simsek M, Opperman RCM, Mulder CJJ, Lambalk CB, de Boer NKH. The teratogenicity of allopurinol: A comprehensive review of animal and human studies. Reprod Toxicol 2018; 81:180-187. [PMID: 30125681 DOI: 10.1016/j.reprotox.2018.08.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/08/2018] [Accepted: 08/13/2018] [Indexed: 02/09/2023]
Abstract
Allopurinol is widely used in the management of multiple disorders including gout, kidney stones and inflammatory bowel disease. Despite of long-term experience, its safety in pregnancy has been debated due to reports on possible teratogenicity. We aimed to review the literature on the safety of allopurinol in pregnancy and offspring. In animals, allopurinol induced species-specific reproductive toxicity. In humans, a total of 53 allopurinol exposed infants were reported in the literature. Major congenital malformations were reported in two cases with a comparable pattern of multiple abnormalities. Five other infants had minor birth defects. In conclusion, the association between allopurinol and teratogenicity appears to be weak and limited to two reports with uncertain causality. However, the available data are insufficient to make a certain judgement, and as allopurinol treatment evolves, report and prospective follow-up of all exposed infants (i.e. deviant and normal cases) should be encouraged.
Collapse
Affiliation(s)
- Melek Simsek
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Gastroenterology and Hepatology, AG&M Research Institute, Amsterdam, The Netherlands.
| | - Roza C M Opperman
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Gastroenterology and Hepatology, AG&M Research Institute, Amsterdam, The Netherlands
| | - Chris J J Mulder
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Gastroenterology and Hepatology, AG&M Research Institute, Amsterdam, The Netherlands
| | - Cornelis B Lambalk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Reproductive Medicine, Obstetrics and Gynecology, Amsterdam, The Netherlands
| | - Nanne K H de Boer
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Gastroenterology and Hepatology, AG&M Research Institute, Amsterdam, The Netherlands
| |
Collapse
|
17
|
Nair J, Kumar VHS. Current and Emerging Therapies in the Management of Hypoxic Ischemic Encephalopathy in Neonates. CHILDREN (BASEL, SWITZERLAND) 2018; 5:E99. [PMID: 30029531 PMCID: PMC6069156 DOI: 10.3390/children5070099] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 07/09/2018] [Accepted: 07/16/2018] [Indexed: 01/01/2023]
Abstract
Neonatal hypoxic ischemic encephalopathy (HIE) presents a significant clinical burden with its high mortality and morbidity rates globally. Therapeutic hypothermia (TH) is now standard of care for infants with moderate to severe HIE, but has not definitively changed outcomes in severe HIE. In this review, we discuss newer promising markers that may help the clinician identify severity of HIE. Therapies that are beneficial and agents that hold promise for neuroprotection are described, both for use either alone or as adjuncts to TH. These include endogenous pathway modifiers such as erythropoietin and analogues, melatonin, and remote ischemic post conditioning. Stem cells have therapeutic potential in this condition, as in many other neonatal conditions. Of the agents listed, only erythropoietin and analogues are currently being evaluated in large randomized controlled trials (RCTs). Exogenous therapies such as argon and xenon, allopurinol, monosialogangliosides, and magnesium sulfate continue to be investigated. The recognition of tertiary mechanisms of brain damage has opened up new research into therapies not only to attenuate brain damage but also to promote cell repair and regeneration in a developmentally disorganized brain long after the perinatal insult. These alternative modalities may be especially important in mild HIE and in areas of the world where there is limited access to expensive hypothermia equipment and services.
Collapse
Affiliation(s)
- Jayasree Nair
- Division of Neonatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA.
| | - Vasantha H S Kumar
- Division of Neonatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA.
| |
Collapse
|
18
|
Stegeman R, Lamur KD, van den Hoogen A, Breur JMPJ, Groenendaal F, Jansen NJG, Benders MJNL. Neuroprotective Drugs in Infants With Severe Congenital Heart Disease: A Systematic Review. Front Neurol 2018; 9:521. [PMID: 30018590 PMCID: PMC6037764 DOI: 10.3389/fneur.2018.00521] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 06/13/2018] [Indexed: 12/20/2022] Open
Abstract
Background: Perinatal and perioperative brain injury is a fundamental problem in infants with severe congenital heart disease undergoing neonatal cardiac surgery with cardiopulmonary bypass. An impaired neuromotor and neurocognitive development is encountered and associated with a reduction in quality of life. New neuroprotective drugs during surgery are described to reduce brain injury and improve neurodevelopmental outcome. Therefore, our aim was to provide a systematic review and best-evidence synthesis on the effects of neuroprotective drugs on brain injury and neurodevelopmental outcome in congenital heart disease infants requiring cardiac surgery with cardiopulmonary bypass. Methods: A systematic search was performed in PubMed, Embase and the Cochrane Library (PRISMA statement). Search terms were “infants,” “congenital heart disease,” “cardiac surgery,” “cardiopulmonary bypass,” and “neuroprotective drug.” Data describing the effects on brain injury and neurodevelopmental outcome were extracted. Study quality was assessed with the Cochrane Risk of Bias Tool. Two reviewers independently screened sources, extracted data and scored bias. Disagreements were resolved by involving a third researcher. Results: The search identified 293 studies of which 6 were included. In total 527 patients with various congenital heart diseases participated with an average of 88 infants (13–318) per study. Allopurinol, sodium nitroprusside, erythropoietin, ketamine, dextromethorphan and phentolamine were administered around cardiac surgery with cardiopulmonary bypass. Allopurinol showed less seizures, coma, death and cardiac events in hypoplastic left heart syndrome (HLHS) infants (OR: 0.44; 95%-CI:0.21–0.91). Sodium nitroprusside resulted in lower post cardiopulmonary bypass levels of S100ß in infants with transposition of the great arteries after 24 (p < 0.01) and 48 (p = 0.04) h of treatment. Erytropoietin, ketamine and dextromethorphan showed no neuroprotective effects. Phentolamine led to higher S100ß-levels and cerebrovascular resistance after rewarming and at the end of surgery (both p < 0.01). Risk of bias varied between studies, including low (sodium nitroprusside, phentolamine), moderate (ketamine, dextromethorphan), and high (erytropoietin, allopurinol) quality. Conclusions: Allopurinol seems promising for future trials in congenital heart disease infants to reduce brain injury given the early neuroprotective effects in hypoplastic left heart syndrome infants. Larger well-designed trials are needed to assess the neuroprotective effects of sodium nitroprusside, erytropoietin, ketamine and dextromethorphan. Future neuroprotective studies in congenital heart disease infants should not only focus on the perioperative period, however also on the perinatal period, since significant brain injury already exists before surgery.
Collapse
Affiliation(s)
- Raymond Stegeman
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Wilhelmina Children's Hospital, Utrecht, Netherlands.,Department of Pediatric Cardiology, University Medical Center Utrecht, Utrecht University, Wilhelmina Children's Hospital, Utrecht, Netherlands.,Department of Pediatric Intensive Care, University Medical Center Utrecht, Utrecht University, Wilhelmina Children's Hospital, Utrecht, Netherlands
| | - Kaya D Lamur
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Wilhelmina Children's Hospital, Utrecht, Netherlands.,Department of Pediatric Cardiology, University Medical Center Utrecht, Utrecht University, Wilhelmina Children's Hospital, Utrecht, Netherlands.,Department of Pediatric Intensive Care, University Medical Center Utrecht, Utrecht University, Wilhelmina Children's Hospital, Utrecht, Netherlands
| | - Agnes van den Hoogen
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Wilhelmina Children's Hospital, Utrecht, Netherlands
| | - Johannes M P J Breur
- Department of Pediatric Cardiology, University Medical Center Utrecht, Utrecht University, Wilhelmina Children's Hospital, Utrecht, Netherlands
| | - Floris Groenendaal
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Wilhelmina Children's Hospital, Utrecht, Netherlands
| | - Nicolaas J G Jansen
- Department of Pediatric Intensive Care, University Medical Center Utrecht, Utrecht University, Wilhelmina Children's Hospital, Utrecht, Netherlands
| | - Manon J N L Benders
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Wilhelmina Children's Hospital, Utrecht, Netherlands
| |
Collapse
|
19
|
Nuñez A, Benavente I, Blanco D, Boix H, Cabañas F, Chaffanel M, Fernández-Colomer B, Fernández-Lorenzo JR, Loureiro B, Moral MT, Pavón A, Tofé I, Valverde E, Vento M. Oxidative stress in perinatal asphyxia and hypoxic-ischaemic encephalopathy. An Pediatr (Barc) 2018. [DOI: 10.1016/j.anpede.2017.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
20
|
Rodríguez-Fanjul J, Durán Fernández-Feijóo C, Lopez-Abad M, Lopez Ramos MG, Balada Caballé R, Alcántara-Horillo S, Camprubí Camprubí M. Neuroprotection with hypothermia and allopurinol in an animal model of hypoxic-ischemic injury: Is it a gender question? PLoS One 2017; 12:e0184643. [PMID: 28931035 PMCID: PMC5606927 DOI: 10.1371/journal.pone.0184643] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 08/28/2017] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Hypoxic-ischemic encephalopathy (HIE) is one of the most important causes of neonatal brain injury. Therapeutic hypothermia (TH) is the standard treatment for term newborns after perinatal hypoxic ischemic injury (HI). Despite this, TH does not provide complete neuroprotection. Allopurinol seems to be a good neuroprotector in several animal studies, but it has never been tested in combination with hypothermia. Clinical findings show that male infants with (HI) fare more poorly than matched females in cognitive outcomes. However, there are few studies about neuroprotection taking gender into account in the results. The aim of the present study was to evaluate the potential additive neuroprotective effect of allopurinol when administrated in association with TH in a rodent model of moderate HI. Gender differences in neuroprotection were also evaluated. METHODS P10 male and female rat pups were subjected to HI (Vannucci model) and randomized into five groups: sham intervention (Control), no treatment (HI), hypothermia (HIH), allopurinol (HIA), and dual therapy (hypothermia and allopurinol) (HIHA). To evaluate a treatment's neuroprotective efficiency, 24 hours after the HI event caspase3 activation was measured. Damaged area and hippocampal volume were also measured 72 hours after the HI event. Negative geotaxis test was performed to evaluate early neurobehavioral reflexes. Learning and spatial memory were assessed via Morris Water Maze (MWM) test at 25 days of life. RESULTS Damaged area and hippocampal volume were different among treatment groups (p = 0.001). The largest tissue lesion was observed in the HI group, followed by HIA. There were no differences between control, HIH, and HIHA. When learning process was analyzed, no differences were found. Females from the HIA group had similar results to the HIH and HIHA groups. Cleaved caspase 3 expression was increased in both HI and HIA. Despite this, in females cleaved caspase-3 was only differently increased in the HI group. All treated animals present an improvement in short-term (Negative geotaxis) and long-term (WMT) functional tests. Despite this, treated females present better long-term outcome. In short-term outcome no sex differences were observed. CONCLUSIONS Our results suggest that dual therapy confers great neuroprotection after an HI event. There were functional, histological, and molecular improvements in all treated groups. These differences were more important in females than in males. No statistically significant differences were found between HIHA and HIH; both of them present a great improvement. Our results support the idea of different regulation mechanisms and pathways of cell death, depending on gender.
Collapse
Affiliation(s)
| | | | - Míriam Lopez-Abad
- Department of Neonatology, BCNatal, Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
| | | | - Rafael Balada Caballé
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Biomedical Sciences, Bellvitge Campus, University of Barcelona, Barcelona, Spain
| | - Soledad Alcántara-Horillo
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Biomedical Sciences, Bellvitge Campus, University of Barcelona, Barcelona, Spain
| | | |
Collapse
|
21
|
Abrahams VM, Chamley LW, Salmon JE. Emerging Treatment Models in Rheumatology: Antiphospholipid Syndrome and Pregnancy: Pathogenesis to Translation. Arthritis Rheumatol 2017; 69:1710-1721. [PMID: 28445926 DOI: 10.1002/art.40136] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 04/20/2017] [Indexed: 02/06/2023]
Affiliation(s)
| | | | - Jane E Salmon
- Hospital for Special Surgery, Weill Cornell Medicine, New York, New York
| |
Collapse
|
22
|
Nuñez A, Benavente I, Blanco D, Boix H, Cabañas F, Chaffanel M, Fernández-Colomer B, Fernández-Lorenzo JR, Loureiro B, Moral MT, Pavón A, Tofé I, Valverde E, Vento M. [Oxidative stress in perinatal asphyxia and hypoxic-ischaemic encephalopathy]. An Pediatr (Barc) 2017. [PMID: 28648366 DOI: 10.1016/j.anpedi.2017.05.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Birth asphyxia is one of the principal causes of early neonatal death. In survivors it may evolve to hypoxic-ischaemic encephalopathy and major long-term neurological morbidity. Prolonged and intense asphyxia will lead to energy exhaustion in tissues exclusively dependent on aerobic metabolism, such as the central nervous system. Energy deficit leads to ATP-dependent pumps blockage, with the subsequent loss of neuronal transmembrane potential. The most sensitive areas of the brain will die due to necrosis. In more resistant areas, neuronal hyper-excitability, massive entrance of ionic calcium, activation of NO-synthase, free radical generation, and alteration in mitochondrial metabolism will lead to a secondary energy failure and programmed neuronal death by means of the activation of the caspase pathways. A third phase has recently been described that includes persistent inflammation and epigenetic changes that would lead to a blockage of oligodendrocyte maturation, alteration of neurogenesis, axonal maturation, and synaptogenesis. In this scenario, oxidative stress plays a critical role causing direct damage to the central nervous system and activating metabolic cascades leading to apoptosis and inflammation. Moderate whole body hypothermia to preserve energy stores and to reduce the formation of oxygen reactive species attenuates the mechanisms that lead to the amplification of cerebral damage upon resuscitation. The combination of hypothermia with coadjuvant therapies may contribute to improve the prognosis.
Collapse
Affiliation(s)
- Antonio Nuñez
- Hospital Universitario y Politécnico La Fe, Valencia, España
| | | | | | - Héctor Boix
- Hospital Universitario Vall d'Hebron, Barcelona, España
| | - Fernando Cabañas
- Hospital Universitario Quirónsalud Madrid, Pozuelo de Alarcón, Madrid, España
| | | | | | | | - Begoña Loureiro
- Hospital Universitario de Cruces, Barakaldo, Vizcaya, España
| | | | - Antonio Pavón
- Hospital Universitario Virgen del Rocío, Sevilla, España
| | - Inés Tofé
- Hospital Universitario Reina Sofía, Córdoba, España
| | | | - Máximo Vento
- Hospital Universitario y Politécnico La Fe, Valencia, España.
| |
Collapse
|
23
|
Arteaga O, Álvarez A, Revuelta M, Santaolalla F, Urtasun A, Hilario E. Role of Antioxidants in Neonatal Hypoxic-Ischemic Brain Injury: New Therapeutic Approaches. Int J Mol Sci 2017; 18:E265. [PMID: 28134843 PMCID: PMC5343801 DOI: 10.3390/ijms18020265] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Revised: 01/14/2017] [Accepted: 01/19/2017] [Indexed: 01/08/2023] Open
Abstract
Hypoxic-ischemic brain damage is an alarming health and economic problem in spite of the advances in neonatal care. It can cause mortality or detrimental neurological disorders such as cerebral palsy, motor impairment and cognitive deficits in neonates. When hypoxia-ischemia occurs, a multi-faceted cascade of events starts out, which can eventually cause cell death. Lower levels of oxygen due to reduced blood supply increase the production of reactive oxygen species, which leads to oxidative stress, a higher concentration of free cytosolic calcium and impaired mitochondrial function, triggering the activation of apoptotic pathways, DNA fragmentation and cell death. The high incidence of this type of lesion in newborns can be partly attributed to the fact that the developing brain is particularly vulnerable to oxidative stress. Since antioxidants can safely interact with free radicals and terminate that chain reaction before vital molecules are damaged, exogenous antioxidant therapy may have the potential to diminish cellular damage caused by hypoxia-ischemia. In this review, we focus on the neuroprotective effects of antioxidant treatments against perinatal hypoxic-ischemic brain injury, in the light of the most recent advances.
Collapse
Affiliation(s)
- Olatz Arteaga
- Department of Cell Biology & Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain.
| | - Antonia Álvarez
- Department of Cell Biology & Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain.
| | - Miren Revuelta
- Department of Cell Biology & Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain.
| | - Francisco Santaolalla
- Department of Otorhinolaryngology, Basurto University Hospital, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain.
| | - Andoni Urtasun
- Department of Neuroscience, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain.
- Neurogenomiks Laboratory, Achucarro Basque Center for Neuroscience, Bizkaia Science and Technology Park, 48170 Zamudio, Spain.
| | - Enrique Hilario
- Department of Cell Biology & Histology, School of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain.
| |
Collapse
|
24
|
Yıldız EP, Ekici B, Tatlı B. Neonatal hypoxic ischemic encephalopathy: an update on disease pathogenesis and treatment. Expert Rev Neurother 2016; 17:449-459. [PMID: 27830959 DOI: 10.1080/14737175.2017.1259567] [Citation(s) in RCA: 151] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Hypoxic ischemic encephalopathy (HIE) is the most important reason for morbidity and mortality in term-born infants. Understanding pathophysiology of the brain damage is essential for the early detection of patients with high risk for HIE and development of strategies for their treatments. Areas covered: This review discusses pathophysiology of the neonatal HIE and its treatment options, including hypothermia, melatonin, allopurinol, topiramate, erythropoietin, N-acetylcyctein, magnesium sulphate and xenon. Expert commentary: Several clinical studies have been performed in order to decrease the risk of brain injury due to difficulties in the early diagnosis and treatment, and to develop strategies for better long-term outcomes. Although currently standard treatment methods include therapeutic hypothermia for neonates with moderate to severe HIE, new supportive options are needed to enhance neuroprotective effects of the hypothermia, which should aim to reduce production of the free radicals and to have anti-inflammatory and anti-apoptotic actions.
Collapse
Affiliation(s)
| | - Barış Ekici
- b Department of Pediatric Neurology , Liv Hospital , Istanbul , Turkey
| | - Burak Tatlı
- a Department of Pediatric Neurology , Istanbul University , Istanbul , Turkey
| |
Collapse
|
25
|
Cabiddu G, Castellino S, Gernone G, Santoro D, Moroni G, Giannattasio M, Gregorini G, Giacchino F, Attini R, Loi V, Limardo M, Gammaro L, Todros T, Piccoli GB. A best practice position statement on pregnancy in chronic kidney disease: the Italian Study Group on Kidney and Pregnancy. J Nephrol 2016; 29:277-303. [PMID: 26988973 PMCID: PMC5487839 DOI: 10.1007/s40620-016-0285-6] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 02/08/2016] [Indexed: 01/09/2023]
Abstract
Pregnancy is increasingly undertaken in patients with chronic kidney disease (CKD) and, conversely, CKD is increasingly diagnosed in pregnancy: up to 3 % of pregnancies are estimated to be complicated by CKD. The heterogeneity of CKD (accounting for stage, hypertension and proteinuria) and the rarity of several kidney diseases make risk assessment difficult and therapeutic strategies are often based upon scattered experiences and small series. In this setting, the aim of this position statement of the Kidney and Pregnancy Study Group of the Italian Society of Nephrology is to review the literature, and discuss the experience in the clinical management of CKD in pregnancy. CKD is associated with an increased risk for adverse pregnancy-related outcomes since its early stage, also in the absence of hypertension and proteinuria, thus supporting the need for a multidisciplinary follow-up in all CKD patients. CKD stage, hypertension and proteinuria are interrelated, but they are also independent risk factors for adverse pregnancy-related outcomes. Among the different kidney diseases, patients with glomerulonephritis and immunologic diseases are at higher risk of developing or increasing proteinuria and hypertension, a picture often difficult to differentiate from preeclampsia. The risk is higher in active immunologic diseases, and in those cases that are detected or flare up during pregnancy. Referral to tertiary care centres for multidisciplinary follow-up and tailored approaches are warranted. The risk of maternal death is, almost exclusively, reported in systemic lupus erythematosus and vasculitis, which share with diabetic nephropathy an increased risk for perinatal death of the babies. Conversely, patients with kidney malformation, autosomal-dominant polycystic kidney disease, stone disease, and previous upper urinary tract infections are at higher risk for urinary tract infections, in turn associated with prematurity. No risk for malformations other than those related to familiar urinary tract malformations is reported in CKD patients, with the possible exception of diabetic nephropathy. Risks of worsening of the renal function are differently reported, but are higher in advanced CKD. Strict follow-up is needed, also to identify the best balance between maternal and foetal risks. The need for further multicentre studies is underlined.
Collapse
Affiliation(s)
| | | | | | | | - Gabriella Moroni
- Nephrology, Fondazione Ca' Granda Ospedale Maggiore, Milano, Italy
| | | | | | | | - Rossella Attini
- Obstetrics, Department of Surgery, University of Torino, Torino, Italy
| | - Valentina Loi
- Nephrology, Azienda Ospedaliera Brotzu, Cagliari, Italy
| | - Monica Limardo
- Nephrology, Azienda Ospedaliera della Provincia di Lecco, Lecco, Italy
| | - Linda Gammaro
- Nephrology, Ospedale Fracastoro, San Bonifacio, Italy
| | - Tullia Todros
- Obstetrics, Department of Surgery, University of Torino, Torino, Italy
| | - Giorgina Barbara Piccoli
- Nephrology, ASOU San Luigi, Department of Clinical and Biological Sciences, University of Torino, Torino, Italy.
- Nephrologie, Centre Hospitalier du Mans, Le Mans, France.
| |
Collapse
|
26
|
Abstract
An adverse outcome is still encountered in 45% of full-term neonates with perinatal asphyxia who are treated with moderate hypothermia. At present pharmacologic therapies are developed to be added to hypothermia. In the present article, these potential neuroprotective interventions are described based on the molecular pathways set in motion during fetal hypoxia and following reoxygenation and reperfusion after birth. These pathways include excessive production of excitotoxins with subsequent over-stimulation of NMDA receptors and calcium influx in neuronal cells, excessive production of reactive oxygen and nitrogen species, activation of inflammation leading to inappropriate apoptosis, and loss of neurotrophic factors. Possibilities for pharmacologic combination therapy, where each drug will be administered based on the optimal point of time in the cascade of destructive molecular reactions, may further reduce brain damage due to perinatal asphyxia.
Collapse
Affiliation(s)
- Frank van Bel
- Department of Neonatology, University Medical Center Utrecht, Wilhelmina Children's Hospital, Utrecht, The Netherlands.
| | - Floris Groenendaal
- Department of Neonatology, University Medical Center Utrecht, Wilhelmina Children's Hospital, Utrecht, The Netherlands
| |
Collapse
|
27
|
Sheikh M, Nelson-Piercy C, Duley J, Florin T, Ansari A. Successful Pregnancies with Thiopurine-Allopurinol Co-Therapy for Inflammatory Bowel Disease. J Crohns Colitis 2015; 9:680-4. [PMID: 25939351 DOI: 10.1093/ecco-jcc/jjv072] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 04/07/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND Thiopurines are an effective treatment for moderate to severe inflammatory bowel disease [IBD] and can be used safely in pregnancy. Combining allopurinol with a lower dose of thiopurine can improve clinical efficacy and bypass some adverse reactions associated with thiopurine monotherapy. Data on allopurinol in pregnancy are scarce. We report on a total of 13 cases where thiopurine and allopurinol co-therapy was used successfully to manage IBD during pregnancy without attributable adverse fetal effects. METHODS Patients were retrospectively identified at our two hospitals, one in the UK and one in Australia, using local IBD databases. Data regarding pregnancy and fetal outcomes including in utero fetal ultrasound scans, APGAR scores, fetal birthweights and neonate checks were collected from patient notes. RESULTS We identified 12 women with a total of 13 pregnancies treated with co-therapy before conception and for the duration of pregnancy. There were no miscarriages or spontaneous pre-term deliveries. There were 14 live births [seven vaginal deliveries; six caesarean sections]. Except for a primagravid twin pregnancy complicated by pre-eclampsia and twin-to-twin transfusion syndrome requiring caesarean section at 25 weeks, there were no low birthweight [< 2.5 kg] babies born and the APGAR scores of all babies were normal. No congenital malformations were identified. CONCLUSIONS Adverse pregnancy outcomes attributable to thiopurine and allopurinol co-therapy were not detected in our case series. Our study provides reassurance for clinicians and patients who wish to continue the thiopurine-allopurinol co-therapy combination before conception and during pregnancy to maintain remission of IBD.
Collapse
Affiliation(s)
- Mohammed Sheikh
- Department of Gastroenterology, East Surrey Hospital, Redhill, UK
| | | | - John Duley
- School of Pharmacy and Mater Research Institute, University of Queensland, Brisbane, QLD, Australia
| | - Timothy Florin
- Department of Gastroenterology, Mater Health Services, University of Queensland and School of Medicine, Brisbane, QLD, Australia
| | - Azhar Ansari
- Department of Gastroenterology, East Surrey Hospital, Redhill, UK
| |
Collapse
|
28
|
|
29
|
Kaandorp JJ, Benders MJNL, Schuit E, Rademaker CMA, Oudijk MA, Porath MM, Oetomo SB, Wouters MGAJ, van Elburg RM, Franssen MTM, Bos AF, de Haan TR, Boon J, de Boer IP, Rijnders RJP, Jacobs CJWFM, Scheepers LHCJ, Gavilanes DAW, Bloemenkamp KWM, Rijken M, van Meir CA, von Lindern JS, Huisjes AJM, Bakker SCMJER, Mol BWJ, Visser GHA, Van Bel F, Derks JB. Maternal allopurinol administration during suspected fetal hypoxia: a novel neuroprotective intervention? A multicentre randomised placebo controlled trial. Arch Dis Child Fetal Neonatal Ed 2015; 100:F216-23. [PMID: 25512466 DOI: 10.1136/archdischild-2014-306769] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 11/17/2014] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To determine whether maternal allopurinol treatment during suspected fetal hypoxia would reduce the release of biomarkers associated with neonatal brain damage. DESIGN A randomised double-blind placebo controlled multicentre trial. PATIENTS We studied women in labour at term with clinical indices of fetal hypoxia, prompting immediate delivery. SETTING Delivery rooms of 11 Dutch hospitals. INTERVENTION When immediate delivery was foreseen based on suspected fetal hypoxia, women were allocated to receive allopurinol 500 mg intravenous (ALLO) or placebo intravenous (CONT). MAIN OUTCOME MEASURES Primary endpoint was the difference in cord S100ß, a tissue-specific biomarker for brain damage. RESULTS 222 women were randomised to receive allopurinol (ALLO, n=111) or placebo (CONT, n=111). Cord S100ß was not significantly different between the two groups: 44.5 pg/mL (IQR 20.2-71.4) in the ALLO group versus 54.9 pg/mL (IQR 26.8-94.7) in the CONT group (difference in median -7.69 (95% CI -24.9 to 9.52)). Post hoc subgroup analysis showed a potential treatment effect of allopurinol on the proportion of infants with a cord S100ß value above the 75th percentile in girls (ALLO n=5 (12%) vs CONT n=10 (31%); risk ratio (RR) 0.37 (95% CI 0.14 to 0.99)) but not in boys (ALLO n=18 (32%) vs CONT n=15 (25%); RR 1.4 (95% CI 0.84 to 2.3)). Also, cord neuroketal levels were significantly lower in girls treated with allopurinol as compared with placebo treated girls: 18.0 pg/mL (95% CI 12.1 to 26.9) in the ALLO group versus 32.2 pg/mL (95% CI 22.7 to 45.7) in the CONT group (geometric mean difference -16.4 (95% CI -24.6 to -1.64)). CONCLUSIONS Maternal treatment with allopurinol during fetal hypoxia did not significantly lower neuronal damage markers in cord blood. Post hoc analysis revealed a potential beneficial treatment effect in girls. TRIAL REGISTRATION NUMBER NCT00189007, Dutch Trial Register NTR1383.
Collapse
Affiliation(s)
- Joepe J Kaandorp
- Department of Perinatology, University Medical Center, Utrecht, The Netherlands
| | - Manon J N L Benders
- Department of Perinatology, University Medical Center, Utrecht, The Netherlands
| | - Ewoud Schuit
- Julius Center for Health Sciences and Primary Care, University Medical Center, Utrecht, The Netherlands
| | - Carin M A Rademaker
- Department of Clinical Pharmacy, University Medical Center, Utrecht, The Netherlands
| | - Martijn A Oudijk
- Department of Perinatology, University Medical Center, Utrecht, The Netherlands
| | - Martina M Porath
- Department of Perinatology, Maxima Medical Center, Veldhoven, The Netherlands
| | | | | | - Ruurd M van Elburg
- Department of Perinatology, VU Medical Center, Amsterdam, The Netherlands Danone Research, Wageningen, The Netherlands
| | - Maureen T M Franssen
- Department of Perinatology, University Medical Center, Groningen, The Netherlands
| | - Arie F Bos
- Department of Perinatology, University Medical Center, Groningen, The Netherlands
| | - Timo R de Haan
- Department of Perinatology, Academic Medical Center, Amsterdam, The Netherlands
| | - Janine Boon
- Department of Perinatology, Diakonessenhuis, Utrecht, The Netherlands
| | - Inge P de Boer
- Department of Perinatology, Diakonessenhuis, Utrecht, The Netherlands
| | - Robbert J P Rijnders
- Department of Perinatology, Jeroen Bosch Medical Center, Den Bosch, The Netherlands
| | | | | | - Danilo A W Gavilanes
- Department of Perinatology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Kitty W M Bloemenkamp
- Department of Perinatology, Leids University Medical Center, Leiden, The Netherlands
| | - Monique Rijken
- Department of Perinatology, Leids University Medical Center, Leiden, The Netherlands
| | - Claudia A van Meir
- Department of Perinatology, Groene Hart Hospital, Gouda, The Netherlands
| | | | | | | | - Ben W J Mol
- Department of Perinatology, University Medical Center, Groningen, The Netherlands
| | - Gerard H A Visser
- Department of Perinatology, University Medical Center, Utrecht, The Netherlands
| | - Frank Van Bel
- Department of Perinatology, University Medical Center, Utrecht, The Netherlands
| | - Jan B Derks
- Department of Perinatology, University Medical Center, Utrecht, The Netherlands
| |
Collapse
|
30
|
New antioxidant drugs for neonatal brain injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:108251. [PMID: 25685254 PMCID: PMC4313724 DOI: 10.1155/2015/108251] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 12/19/2014] [Indexed: 11/17/2022]
Abstract
The brain injury concept covers a lot of heterogeneity in terms of aetiology involving multiple factors, genetic, hemodynamic, metabolic, nutritional, endocrinological, toxic, and infectious mechanisms, acting in antenatal or postnatal period. Increased vulnerability of the immature brain to oxidative stress is documented because of the limited capacity of antioxidant enzymes and the high free radicals (FRs) generation in rapidly growing tissue. FRs impair transmembrane enzyme Na+/K+-ATPase activity resulting in persistent membrane depolarization and excessive release of FR and excitatory aminoacid glutamate. Besides being neurotoxic, glutamate is also toxic to oligodendroglia, via FR effects. Neuronal cells die of oxidative stress. Excess of free iron and deficient iron/binding metabolising capacity are additional features favouring oxidative stress in newborn. Each step in the oxidative injury cascade has become a potential target for neuroprotective intervention. The administration of antioxidants for suspected or proven brain injury is still not accepted for clinical use due to uncertain beneficial effects when treatments are started after resuscitation of an asphyxiated newborn. The challenge for the future is the early identification of high-risk babies to target a safe and not toxic antioxidant therapy in combination with standard therapies to prevent brain injury and long-term neurodevelopmental impairment.
Collapse
|
31
|
Zhu H, Han X, Ji D, Lv G, Xu M. Estrogen inhibits lipid peroxidation after hypoxic-ischemic brain damage in neonatal rats. Neural Regen Res 2014; 7:2424-31. [PMID: 25337092 PMCID: PMC4200716 DOI: 10.3969/j.issn.1673-5374.2012.31.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 10/08/2012] [Indexed: 11/18/2022] Open
Abstract
Sprague-Dawley neonatal rats within 7 days after birth were used in this study. The left common carotid artery was occluded and rats were housed in an 8% O2 environment for 2 hours to establish a hypoxic-ischemic brain damage model. 17β-estradiol (1 × 10(-5) M) was injected into the rat abdominal cavity after the model was successfully established. The left hemisphere was obtained at 12, 24, 48, 72 hours after operation. Results showed that malondialdehyde content in the left brain of neonatal rats gradually increased as modeling time prolonged, while malondialdehyde content of 17β-estrodial-treated rats significantly declined by 24 hours, reached lowest levels at 48 hours, and then peaked at 72 hours after injury. Nicotinamide-adenine dinucleotide phosphate histochemical staining showed the nitric oxide synthase-positive cells and fibers dyed blue/violet and were mainly distributed in the cortex, hippocampus and medial septal nuclei. The number of nitric oxide synthase-positive cells peaked at 48 hours and significantly decreased after 17β-estrodial treatment. Our experimental findings indicate that estrogen plays a protective role following hypoxic-ischemic brain damage by alleviating lipid peroxidation through reducing the expression of nitric oxide synthase and the content of malondialdehyde.
Collapse
Affiliation(s)
- Hui Zhu
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Xiao Han
- Department of Human Anatomy, Institute of Neurobiology, Medical School of Nantong University, Jiangsu Key Laboratory of Neuroregeneration, Nantong 226001, Jiangsu Province, China
| | - Dafeng Ji
- Department of Human Anatomy, Institute of Neurobiology, Medical School of Nantong University, Jiangsu Key Laboratory of Neuroregeneration, Nantong 226001, Jiangsu Province, China
| | - Guangming Lv
- Department of Human Anatomy, Institute of Neurobiology, Medical School of Nantong University, Jiangsu Key Laboratory of Neuroregeneration, Nantong 226001, Jiangsu Province, China
| | - Meiyu Xu
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| |
Collapse
|
32
|
Kaandorp JJ, van den Broek MPH, Benders MJNL, Oudijk MA, Porath MM, Bambang Oetomo S, Wouters MGAJ, van Elburg R, Franssen MTM, Bos AF, Mol BWJ, Visser GHA, van Bel F, Rademaker CMA, Derks JB. Rapid target allopurinol concentrations in the hypoxic fetus after maternal administration during labour. Arch Dis Child Fetal Neonatal Ed 2014; 99:F144-8. [PMID: 24352085 DOI: 10.1136/archdischild-2013-304876] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Perinatal hypoxia-induced free radical formation is an important cause of hypoxic-ischaemic encephalopathy and subsequent neurodevelopmental disabilities. Allopurinol reduces the formation of free radicals, which potentially limits hypoxia-induced brain damage. We investigated placental transfer and safety of allopurinol after maternal allopurinol treatment during labour to evaluate its potential role as a neuroprotective agent in suspected fetal hypoxia. DESIGN We used data from a randomised, double-blind multicentre trial comparing maternal allopurinol versus placebo in case of imminent fetal hypoxia (NCT00189007). PATIENTS We studied 58 women in labour at term, with suspected fetal hypoxia prompting immediate delivery, in the intervention arm of the study. SETTING Delivery rooms of 11 Dutch hospitals. INTERVENTION 500 mg allopurinol, intravenously to the mother, immediately prior to delivery. MAIN OUTCOME MEASURES Drug disposition (maternal plasma concentrations, cord blood concentrations) and drug safety (maternal and fetal adverse events). RESULTS Within 5 min after the end of maternal allopurinol infusion, target plasma concentrations of allopurinol of ≥2 mg/L were present in cord blood. Of all analysed cord blood samples, 95% (52/55) had a target allopurinol plasma concentration at the moment of delivery. No adverse events were observed in the neonates. Two mothers had a red and/or painful arm during infusion. CONCLUSIONS A dose of 500 mg intravenous allopurinol rapidly crosses the placenta and provides target concentrations in 95% of the fetuses at the moment of delivery, which makes it potentially useful as a neuroprotective agent in perinatology with very little side effects. TRIAL REGISTRATION The study is registered in the Dutch Trial Register (NTR1383) and the Clinical Trials protocol registration system (NCT00189007).
Collapse
Affiliation(s)
- J J Kaandorp
- Department of Perinatology, University Medical Centre, , Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Chronic allopurinol treatment during the last trimester of pregnancy in sows: effects on low and normal birth weight offspring. PLoS One 2014; 9:e86396. [PMID: 24466072 PMCID: PMC3899238 DOI: 10.1371/journal.pone.0086396] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 12/10/2013] [Indexed: 11/19/2022] Open
Abstract
Low-birth-weight (LBW) children are born with several risk factors for disease, morbidity and neonatal mortality, even if carried to term. Placental insufficiency leading to hypoxemia and reduced nutritional supply is the main cause for LBW. Brain damage and poor neurological outcome can be the consequence. LBW after being carried to term gives better chances for survival, but these children are still at risk for poor health and the development of cognitive impairments. Preventive therapies are not yet available. We studied the risk/efficacy of chronic prenatal treatment with the anti-oxidative drug allopurinol, as putative preventive treatment in piglets. LBW piglets served as a natural model for LBW. A cognitive holeboard test was applied to study the learning and memory abilities of these allopurinol treated piglets after weaning. Preliminary analysis of the plasma concentrations in sows and their piglets suggested that a daily dose of 15 mg.kg(-1) resulted in effective plasma concentration of allopurinol in piglets. No adverse effects of chronic allopurinol treatment were found on farrowing, birth weight, open field behavior, learning abilities, relative brain, hippocampus and spleen weights. LBW piglets showed increased anxiety levels in an open field test, but cognitive performance was not affected by allopurinol treatment. LBW animals treated with allopurinol showed the largest postnatal compensatory body weight gain. In contrast to a previous study, no differences in learning abilities were found between LBW and normal-birth-weight piglets. This discrepancy might be attributable to experimental differences. Our results indicate that chronic prenatal allopurinol treatment during the third trimester of pregnancy is safe, as no adverse side effects were observed. Compensatory weight gain of treated piglets is a positive indication for the chronic prenatal use of allopurinol in these animals. Further studies are needed to assess the possible preventive effects of allopurinol on brain functions in LBW piglets.
Collapse
|
34
|
Kane AD, Camm EJ, Richter HG, Lusby C, Tijsseling D, Kaandorp JJ, Derks JB, Ozanne SE, Giussani DA. Maternal-to-fetal allopurinol transfer and xanthine oxidase suppression in the late gestation pregnant rat. PHYSICS REPORTS-REVIEW SECTION OF PHYSICS LETTERS 2013; 1:e00156. [PMID: 24400158 PMCID: PMC3871471 DOI: 10.1002/phy2.156] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 10/21/2013] [Indexed: 02/02/2023]
Abstract
Fetal brain hypoxic injury remains a concern in high-risk delivery. There is significant clinical interest in agents that may diminish neuronal damage during birth asphyxia, such as in allopurinol, an inhibitor of the prooxidant enzyme xanthine oxidase. Here, we established in a rodent model the capacity of allopurinol to be taken up by the mother, cross the placenta, rise to therapeutic levels, and suppress xanthine oxidase activity in the fetus. On day 20 of pregnancy, Wistar dams were given 30 or 100 mg kg(-1) allopurinol orally. Maternal and fetal plasma allopurinol and oxypurinol concentrations were measured, and xanthine oxidase activity in the placenta and maternal and fetal tissues determined. There were significant strong positive correlations between maternal and fetal plasma allopurinol (r = 0.97, P < 0.05) and oxypurinol (r = 0.88, P < 0.05) levels. Under baseline conditions, maternal heart (2.18 ± 0.62 mU mg(-1)), maternal liver (0.29 ± 0.08 mU mg(-1)), placenta (1.36 ± 0.42 mU mg(-1)), fetal heart (1.64 ± 0.59 mU mg(-1)), and fetal liver (0.14 ± 0.08 mU mg(-1)) samples all showed significant xanthine oxidase activity. This activity was suppressed in all tissues 2 h after allopurinol administration and remained suppressed 24 h later (P < 0.05), despite allopurinol and oxypurinol levels returning toward baseline. The data establish a mammalian model of xanthine oxidase inhibition in the mother, placenta, and fetus, allowing investigation of the role of xanthine oxidase-derived reactive oxygen species in the maternal, placental, and fetal physiology during healthy and complicated pregnancy.
Collapse
Affiliation(s)
- Andrew D Kane
- Department of Physiology, Development and Neuroscience, University of CambridgeCambridge, United Kingdom
| | - Emily J Camm
- Department of Physiology, Development and Neuroscience, University of CambridgeCambridge, United Kingdom
| | - Hans G Richter
- Department of Physiology, Development and Neuroscience, University of CambridgeCambridge, United Kingdom
| | - Ciara Lusby
- Department of Physiology, Development and Neuroscience, University of CambridgeCambridge, United Kingdom,University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrooke's HospitalCambridge, United Kingdom
| | | | - Joepe J Kaandorp
- Perinatal Center, University Medical CenterUtrecht, The Netherlands
| | - Jan B Derks
- Perinatal Center, University Medical CenterUtrecht, The Netherlands
| | - Susan E Ozanne
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrooke's HospitalCambridge, United Kingdom
| | - Dino A Giussani
- Department of Physiology, Development and Neuroscience, University of CambridgeCambridge, United Kingdom
| |
Collapse
|
35
|
Kaandorp JJ, Derks JB, Oudijk MA, Torrance HL, Harmsen MG, Nikkels PGJ, van Bel F, Visser GHA, Giussani DA. Antenatal allopurinol reduces hippocampal brain damage after acute birth asphyxia in late gestation fetal sheep. Reprod Sci 2013; 21:251-9. [PMID: 23793473 DOI: 10.1177/1933719113493516] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Free radical-induced reperfusion injury is a recognized cause of brain damage in the newborn after birth asphyxia. The xanthine oxidase inhibitor allopurinol reduces free radical synthesis and crosses the placenta easily. Therefore, allopurinol is a promising therapeutic candidate. This study tested the hypothesis that maternal treatment with allopurinol during fetal asphyxia limits ischemia-reperfusion (I/R) damage to the fetal brain in ovine pregnancy. The I/R challenge was induced by 5 repeated measured compressions of the umbilical cord, each lasting 10 minutes, in chronically instrumented fetal sheep at 0.8 of gestation. Relative to control fetal brains, the I/R challenge induced significant neuronal damage in the fetal hippocampal cornu ammonis zones 3 and 4. Maternal treatment with allopurinol during the I/R challenge restored the fetal neuronal damage toward control scores. Maternal treatment with allopurinol offers potential neuroprotection to the fetal brain in the clinical management of perinatal asphyxia.
Collapse
Affiliation(s)
- Joepe J Kaandorp
- 1Perinatal Center, University Medical Center, Utrecht, the Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Marks KA. Hypoxic–ischemic brain injury and neuroprotection in the newborn infant. FUTURE NEUROLOGY 2013. [DOI: 10.2217/fnl.13.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Recent clinical trials have confirmed that in term infants with moderate-to-severe hypoxic–ischemic encephalopathy, death and severe developmental disability can be reduced by early treatment with hypothermia. However, meta-analysis of these trials has confirmed that two-thirds of the survivors remain seriously impaired. The search for new neuroprotective interventions has therefore continued. Extensive research has identified the important biochemical pathways that result in neuronal loss, and the subsequent repair and regeneration processes. The most promising neuroprotective agents that limit the former, and promote the latter, are being tested in animal models of hypoxic–ischemic brain injury and are awaiting clinical trials. It is likely that a ‘cocktail’ of agents, affecting a number of pathways, will ultimately prove to be the most effective intervention. The latest additions to a long list of proposed substances are various stem cells that promote neurogenesis by releasing trophic substances into the injured brain. Future clinical trials are likely to employ early biomarkers, of which MRI and proton spectroscopy are probably the most predictive of long-term neurodevelopmental outcome. In conclusion, the exponential increase in knowledge in this field can be expected to provide many more neuroprotective agents within the next decade.
Collapse
Affiliation(s)
- Kyla-Anna Marks
- Department of Neonatal Medicine, Soroka University Medical Centre, PO Box 151, Beersheva, Israel
| |
Collapse
|
37
|
Cerio FGD, Lara-Celador I, Alvarez A, Hilario E. Neuroprotective therapies after perinatal hypoxic-ischemic brain injury. Brain Sci 2013; 3:191-214. [PMID: 24961314 PMCID: PMC4061821 DOI: 10.3390/brainsci3010191] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 02/13/2013] [Accepted: 02/22/2013] [Indexed: 12/29/2022] Open
Abstract
Hypoxic-ischemic (HI) brain injury is one of the main causes of disabilities in term-born infants. It is the result of a deprivation of oxygen and glucose in the neural tissue. As one of the most important causes of brain damage in the newborn period, the neonatal HI event is a devastating condition that can lead to long-term neurological deficits or even death. The pattern of this injury occurs in two phases, the first one is a primary energy failure related to the HI event and the second phase is an energy failure that takes place some hours later. Injuries that occur in response to these events are often manifested as severe cognitive and motor disturbances over time. Due to difficulties regarding the early diagnosis and treatment of HI injury, there is an increasing need to find effective therapies as new opportunities for the reduction of brain damage and its long term effects. Some of these therapies are focused on prevention of the production of reactive oxygen species, anti-inflammatory effects, anti-apoptotic interventions and in a later stage, the stimulation of neurotrophic properties in the neonatal brain which could be targeted to promote neuronal and oligodendrocyte regeneration.
Collapse
Affiliation(s)
- Felipe Goñi de Cerio
- Biotechnology Area, GAIKER Technology Centre, Parque Tecnológico de Zamudio Ed 202, 48170 Zamudio, Vizcaya, Spain.
| | - Idoia Lara-Celador
- Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, 48949 Leioa, Bizkaia, Spain.
| | - Antonia Alvarez
- Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, 48949 Leioa, Bizkaia, Spain.
| | - Enrique Hilario
- Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, 48949 Leioa, Bizkaia, Spain.
| |
Collapse
|
38
|
Kim HM. Pharmacological Approaches in Newborn Infants with Hypoxic Ischemic Encephalopathy. NEONATAL MEDICINE 2013. [DOI: 10.5385/nm.2013.20.3.335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Affiliation(s)
- Heng-mi Kim
- Department of Pediatrics, Kyungpook National University School of Medicine, Daegu, Korea
| |
Collapse
|
39
|
Pérez-Mazliah D, Albareda MC, Alvarez MG, Lococo B, Bertocchi GL, Petti M, Viotti RJ, Laucella SA. Allopurinol reduces antigen-specific and polyclonal activation of human T cells. Front Immunol 2012; 3:295. [PMID: 23049532 PMCID: PMC3448060 DOI: 10.3389/fimmu.2012.00295] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Accepted: 09/04/2012] [Indexed: 11/13/2022] Open
Abstract
Allopurinol is the most popular commercially available xanthine oxidase inhibitor and it is widely used for treatment of symptomatic hyperuricaemia, or gout. Although, several anti-inflammatory actions of allopurinol have been demonstrated in vivo and in vitro, there have been few studies on the action of allopurinol on T cells. In the current study, we have assessed the effect of allopurinol on antigen-specific and mitogen-driven activation and cytokine production in human T cells. Allopurinol markedly decreased the frequency of IFN-γ and IL-2-producing T cells, either after polyclonal or antigen-specific stimulation with Herpes Simplex virus 1, Influenza (Flu) virus, tetanus toxoid and Trypanosoma cruzi-derived antigens. Allopurinol attenuated CD69 upregulation after CD3 and CD28 engagement and significantly reduced the levels of spontaneous and mitogen-induced intracellular reactive oxygen species in T cells. The diminished T cell activation and cytokine production in the presence of allopurinol support a direct action of allopurinol on human T cells, offering a potential pharmacological tool for the management of cell-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Damián Pérez-Mazliah
- Instituto Nacional de Parasitología "Dr. Mario Fatala Chaben" Ciudad Autónoma de Buenos Aires, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Chaudhari T, McGuire W. Allopurinol for preventing mortality and morbidity in newborn infants with hypoxic-ischaemic encephalopathy. Cochrane Database Syst Rev 2012; 2012:CD006817. [PMID: 22786499 PMCID: PMC11260067 DOI: 10.1002/14651858.cd006817.pub3] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Delayed neuronal death following a perinatal hypoxic insult is due partly to xanthine oxidase-mediated production of cytotoxic free radicals. Evidence exists that allopurinol, a xanthine-oxidase inhibitor, reduces delayed cell death in experimental models of perinatal asphyxia and in people with organ reperfusion injury. OBJECTIVES To determine the effect of allopurinol on mortality and morbidity in newborn infants with hypoxic-ischaemic encephalopathy. SEARCH METHODS We used the standard search strategy of the Cochrane Neonatal Group. We searched the Cochrane Central Register of Controlled Trials (CENTRAL, The Cochrane Library, 2012, Issue 1), MEDLINE (1966 to March 2012), EMBASE (1980 to March 2012), CINAHL (1982 to March 2012), conference proceedings, and previous reviews. SELECTION CRITERIA Randomised or quasi-randomised controlled trials that compared allopurinol administration versus placebo or no drug in newborn infants with hypoxic-ischaemic encephalopathy. DATA COLLECTION AND ANALYSIS We extracted data using the standard methods of the Cochrane Neonatal Review Group with separate evaluation of trial quality and data extraction by two review authors. MAIN RESULTS We included three trials in which a total of 114 infants participated. In one trial, participants were exclusively infants with severe encephalopathy. The other trials also included infants with mild and moderately severe encephalopathy. These studies were generally of good methodological quality, but were too small to exclude clinically important effects of allopurinol on mortality and morbidity. Meta-analysis did not reveal a statistically significant difference in the risk of death (typical risk ratio 0.88; 95% confidence interval (95% CI) 0.56 to 1.38; risk difference -0.04; 95% CI -0.18 to 0.10) or a composite of death or severe neurodevelopmental disability (typical risk ratio 0.78; 95% CI 0.56 to 1.08; risk difference -0.14; 95% CI -0.31 to 0.04). AUTHORS' CONCLUSIONS The available data are not sufficient to determine whether allopurinol has clinically important benefits for newborn infants with hypoxic-ischaemic encephalopathy. Much larger trials are needed. Such trials could assess allopurinol as an adjunct to therapeutic hypothermia in infants with moderate and severe encephalopathy and should be designed to exclude important effects on mortality and adverse long-term neurodevelopmental outcomes.
Collapse
Affiliation(s)
| | - William McGuire
- Hull York Medical School & Centre for Reviews and Dissemination, University of YorkYorkY010 5DDUK
| | | |
Collapse
|
41
|
Robertson NJ, Tan S, Groenendaal F, van Bel F, Juul SE, Bennet L, Derrick M, Back SA, Valdez RC, Northington F, Gunn AJ, Mallard C. Which neuroprotective agents are ready for bench to bedside translation in the newborn infant? J Pediatr 2012; 160:544-552.e4. [PMID: 22325255 PMCID: PMC4048707 DOI: 10.1016/j.jpeds.2011.12.052] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 12/02/2011] [Accepted: 12/30/2011] [Indexed: 02/07/2023]
|
42
|
Beharier O, Kahn J, Shusterman E, Sheiner E. S100B - a potential biomarker for early detection of neonatal brain damage following asphyxia. J Matern Fetal Neonatal Med 2012; 25:1523-8. [PMID: 22348227 DOI: 10.3109/14767058.2012.664200] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Birth asphyxia results in a significant percentage of neonatal morbidity and mortality. A key factor in the management of this complication is the early and accurate detection of brain damage following asphyxia. Currently, reliable tools for such diagnosis are absent. Extensive research has focused on biomarkers in an attempt to solve this matter. Recent data marked serum and urine elevation of the S100B protein as an established peripheral biomarker for detection of brain injury including traumatic head injuries and brain damage following cardiac arrest and stroke. In the past decade, a substantial number of studies illustrated the potential use of S100B testing in order to detect brain damage in asphyxiated newborns. This review summarizes the available data regarding the use of S100B as a biomarker of brain damage following birth asphyxia.
Collapse
Affiliation(s)
- Ofer Beharier
- Department of Obstetrics and Gynecology, Faculty of Health Sciences, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | | | | | | |
Collapse
|
43
|
Perrone S, Stazzoni G, Tataranno ML, Buonocore G. New pharmacologic and therapeutic approaches for hypoxic-ischemic encephalopathy in the newborn. J Matern Fetal Neonatal Med 2012; 25 Suppl 1:83-8. [DOI: 10.3109/14767058.2012.663168] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
44
|
Herrera EA, Kane AD, Hansell JA, Thakor AS, Allison BJ, Niu Y, Giussani DA. A role for xanthine oxidase in the control of fetal cardiovascular function in late gestation sheep. J Physiol 2012; 590:1825-37. [PMID: 22331413 DOI: 10.1113/jphysiol.2011.224576] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Virtually nothing is known about the effects on fetal physiology of xanthine oxidase inhibition. This is despite maternal treatment with the xanthine oxidase inhibitor allopurinol being considered in human complicated pregnancy to protect the infant’s brain from excessive generation of ROS.We investigated the in vivo effects of maternal treatment with allopurinol on fetal cardiovascular function in ovine pregnancy in late gestation. Under anaesthesia, pregnant ewes and their singleton fetus were instrumented with vascular catheters and flow probes around an umbilical and a fetal femoral artery at 118±1 dGA (days of gestational age; termca. 145 days). Five days later, mothers were infused I.V. with either vehicle (n =11) or allopurinol (n =10). Fetal cardiovascular function was stimulated with increasing bolus doses of phenylephrine (PE) following maternal vehicle or allopurinol. The effects of maternal allopurinol on maternal and fetal cardiovascular function were also investigated following fetal NO blockade (n =6) or fetal β1-adrenergic antagonism (n =7). Maternal allopurinol led to significant increases in fetal heart rate, umbilical blood flow and umbilical vascular conductance, effects abolished by fetal β1-adrenergic antagonism but not by fetal NO blockade. Maternal allopurinol impaired fetal α1-adrenergic pressor and femoral vasopressor responses and enhanced the gain of the fetal cardiac baroreflex. These effects of maternal allopurinol were restored to control levels during fetal NO blockade. Maternal treatment with allopurinol induced maternal hypotension, tachycardia and acid–base disturbance. We conclude that maternal treatment with allopurinol alters in vivo maternal, umbilical and fetal vascular function via mechanisms involving NO and β1-adrenergic stimulation. The evidence suggests that the use of allopurinol in clinical practice should be approached with caution.
Collapse
Affiliation(s)
- E A Herrera
- Department of Physiology Development & Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | | | | | | | | | | | | |
Collapse
|
45
|
Miller SL, Wallace EM, Walker DW. Antioxidant therapies: a potential role in perinatal medicine. Neuroendocrinology 2012; 96:13-23. [PMID: 22377769 DOI: 10.1159/000336378] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 12/30/2011] [Indexed: 11/19/2022]
Abstract
Pregnancies complicated by impaired placentation, acute severe reductions in oxygen supply to the fetus, or intrauterine infection are associated with oxidative stress to the mother and developing baby. Such oxidative stress is characterized as an upregulation in the production of oxidative or nitrative free radicals and a concomitant decrease in the availability of antioxidant species, thereby creating a state of fetoplacental oxidative imbalance. Recently, there has been a good deal of interest in the potential for the use of antioxidant therapies in the perinatal period to protect the fetus, particularly the developing brain, against oxidative stress in complications of pregnancy and birth. This review will examine why the immature brain is particularly susceptible to oxidative imbalance and will provide discussion on antioxidant treatments currently receiving attention in the adult and perinatal literature - allopurinol, melatonin, α-lipoic acid, and vitamins C and E. In addition, we aim to address the interaction between oxidative stress and the fetal inflammatory response, an interaction that may be vital when proposing antioxidant or other neuroprotective strategies.
Collapse
Affiliation(s)
- S L Miller
- The Ritchie Centre, Monash Institute of Medical Research, Clayton, Vic., Australia
| | | | | |
Collapse
|
46
|
Reid SM, Carlin JB, Reddihough DS. Rates of cerebral palsy in Victoria, Australia, 1970 to 2004: has there been a change? Dev Med Child Neurol 2011; 53:907-12. [PMID: 21752018 DOI: 10.1111/j.1469-8749.2011.04039.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
AIM The aim of this study was to assess overall and gestational age-specific trends in the rate of cerebral palsy (CP) in Victoria, Australia, and to compare these findings with other population data. METHOD Individuals born in Victoria from 1970 to 2004 with non-postneonatally acquired CP were identified from a population register; 3491 were included in the study (1963 males, 1528 females). After a literature review, comparison data were extracted from publications using previously devised inclusion criteria. Rates were calculated per 1000 live births for all CP and by gestational age group: these were tabulated and plotted by year of birth. RESULTS Data from nine registries, including the Victorian register, showed an increase in the rates of CP over the 1970s and 1980s, consistently seen in extremely preterm (<28 wks) survivors but also in those born at term (≥37 wks). Since the early 1990s, CP rates either stabilized or decreased, particularly for children born extremely preterm. INTERPRETATION Increases in the rates of CP during the 1970s and 1980s are in part because of the increasing survival of extremely preterm infants that occurred without a concomitant improvement in neurological outcomes. Evidence from population samples now suggests that this trend has been reversed since the mid- to late 1990s.
Collapse
Affiliation(s)
- Susan M Reid
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia.
| | | | | |
Collapse
|
47
|
Fan X, Kavelaars A, Heijnen CJ, Groenendaal F, van Bel F. Pharmacological neuroprotection after perinatal hypoxic-ischemic brain injury. Curr Neuropharmacol 2011; 8:324-34. [PMID: 21629441 PMCID: PMC3080590 DOI: 10.2174/157015910793358150] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 03/31/2010] [Accepted: 04/08/2010] [Indexed: 11/22/2022] Open
Abstract
Perinatal hypoxia-ischemia (HI) is an important cause of neonatal brain injury. Recent progress in the search for neuroprotective compounds has provided us with several promising drugs to reduce perinatal HI-induced brain injury. In the early stage (first 6 hours after birth) therapies are concentrated on prevention of the production of reactive oxygen species or free radicals (xanthine-oxidase-, nitric oxide synthase-, and prostaglandin inhibition), anti-inflammatory effects (erythropoietin, melatonin, Xenon) and anti-apoptotic interventions (nuclear factor kappa B- and c-jun N-terminal kinase inhibition); in a later stage stimulation of neurotrophic properties in the neonatal brain (erythropoietin, growth factors) can be targeted to promote neuronal and oligodendrocyte regeneration. Combination of pharmacological means of treatment with moderate hypothermia, which is accepted now as a meaningful therapy, is probably the next step in clinical treatment to fight post-asphyxial brain damage. Further studies should be directed at a more rational use of therapies by determining the optimal time and dose to inhibit the different potentially destructive molecular pathways or to enhance endogenous repair while at the same time avoiding adverse effects of the drugs used.
Collapse
Affiliation(s)
- Xiyong Fan
- Department of Neonatology, University Medical Center, Utrecht, the Netherlands
| | | | | | | | | |
Collapse
|
48
|
Boda D. Results of and further prevention of hypoxic fetal brain damage by inhibition of xanthine oxidase enzyme with allopurinol. J Perinat Med 2011; 39:441-4. [PMID: 21410409 DOI: 10.1515/jpm.2011.025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Several experimental models on adult and newborn animals showed that in cerebral hypoxic-ischemic conditions similar to clinical states the main source of the excessive production of free oxygen radicals is the highly activated xanthine oxidase (XO) enzyme reaction. Long before this data were available, it became known that the main role of allopurinol (AP) is the inhibition of XO. On the basis of these results, many therapeutic trials with AP were performed both in experimental and clinical studies of ischemia and reperfusion. However, it has been shown that only preventive administration of AP has favorable effects. The explanation for the poor results of AP treatment in human fetal brain damage (FBD) cases is that the drug was applied postnatally. The clinical studies performed in healthy laboring mothers whose deliveries were complicated with FBD showed that placental transfer after prenatal administration of AP may be effective in protecting newborns at increased risk of hypoxic-ischemic cerebral damage. Further controlled trials are required to determine if the prophylactic use of the drug might prevent hypoxic-ischemic injuries when the drug is administered immediately prior to impending fetal hypoxia, or even in deliveries at risk of developing hypoxia.
Collapse
Affiliation(s)
- Domokos Boda
- Department of Pediatrics, University of Szeged, Szeged, Hungary.
| |
Collapse
|
49
|
Schlapbach LJ, Frey S, Bigler S, Manh-Nhi C, Aebi C, Nelle M, Nuoffer JM. Copeptin concentration in cord blood in infants with early-onset sepsis, chorioamnionitis and perinatal asphyxia. BMC Pediatr 2011; 11:38. [PMID: 21595972 PMCID: PMC3118890 DOI: 10.1186/1471-2431-11-38] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Accepted: 05/19/2011] [Indexed: 11/30/2022] Open
Abstract
Background Vasopressin is one of the most important physiological stress and shock hormones. Copeptin, a stable vasopressin precursor, is a promising sepsis marker in adults. In contrast, its involvement in neonatal diseases remains unknown. The aim of this study was to establish copeptin concentrations in neonates of different stress states such as sepsis, chorioamnionitis and asphyxia. Methods Copeptin cord blood concentration was determined using the BRAHMS kryptor assay. Neonates with early-onset sepsis (EOS, n = 30), chorioamnionitis (n = 33) and asphyxia (n = 25) were compared to a control group of preterm and term (n = 155) neonates. Results Median copeptin concentration in cord blood was 36 pmol/l ranging from undetectable to 5498 pmol/l (IQR 7 - 419). Copeptin cord blood concentrations were non-normally distributed and increased with gestational age (p < 0.0001). Neonates born after vaginal compared to cesarean delivery had elevated copeptin levels (p < 0.0001). Copeptin correlated strongly with umbilical artery pH (Spearman's Rho -0.50, p < 0.0001), umbilical artery base excess (Rho -0.67, p < 0.0001) and with lactate at NICU admission (Rho 0.54, p < 0.0001). No difference was found when comparing copeptin cord blood concentrations between neonates with EOS and controls (multivariate p = 0.30). The highest copeptin concentrations were found in neonates with asphyxia (median 993 pmol/l). Receiver-operating-characteristic curve analysis showed that copeptin cord blood concentrations were strongly associated with asphyxia: the area under the curve resulted at 0.91 (95%-CI 0.87-0.96, p < 0.0001). A cut-off of 400 pmol/l had a sensitivity of 92% and a specifity of 82% for asphyxia as defined in this study. Conclusions Copeptin concentrations were strongly related to factors associated with perinatal stress such as birth acidosis, asphyxia and vaginal delivery. In contrast, copeptin appears to be unsuitable for the diagnosis of EOS.
Collapse
Affiliation(s)
- Luregn J Schlapbach
- Neonatal and Pediatric Intensive Care Unit, Department of Pediatrics, University of Bern, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
50
|
Fan X, van Bel F. Pharmacological neuroprotection after perinatal asphyxia. J Matern Fetal Neonatal Med 2011; 23 Suppl 3:17-9. [PMID: 20695757 DOI: 10.3109/14767058.2010.505052] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Recent progress has provided us with several promising neuroprotective compounds to reduce perinatal hypoxic-ischemic (HI) brain injury. In the early post HI phase, therapies can be concentrated on ion channel blockage (Xenon), anti-oxidation (allopurinol, 2-iminobiotin, and indomethacin), anti-inflammation (erythropoietin [EPO], melatonin), and anti-apoptosis (nuclear factor kappa B [NF-κB]and c-jun N-terminal kinase [JNK] inhibitors); in the later phase, therapies should be targeted to promote neuronal regeneration by stimulation of neurotrophic properties of the neonatal brain (EPO, growth factors, stem cells transplantation). Combination of pharmacological interventions with moderate hypothermia, which is the only established therapy for post HI brain injury, is probably the next step to fight HI brain damage in the clinical setting. Further studies should be concentrated on more rational pharmacological strategies by determining the optimal time and dose to inhibit the various potentially destructive molecular pathways and/or to enhance endogenous repair meanwhile avoiding the adverse effects.
Collapse
Affiliation(s)
- Xiyong Fan
- Department of Neonatology, University Medical Center, Utrecht, The Netherlands
| | | |
Collapse
|