Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 363
Filter
1.
Clin Perinatol ; 51(3): 617-628, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39095100

ABSTRACT

This article summarizes the current evidence regarding inflammatory biomarkers (placental and postnatal) and provides a comprehensive understanding of their roles: (1) diagnostic accuracy to predict the severity of hypoxic-ischemia encephalopathy (HIE), (2) value in assessing treatment responses, and (3) prediction of both short- and long-term neurodevelopmental outcomes. In the early critical stages of perinatal asphyxia, inflammatory biomarkers may guide clinical decision-making. Additional research is required to increase our understanding of the optimal utility of biomarkers to predict the severity, evolution, and developmental outcomes after exposure to HIE.


Subject(s)
Asphyxia Neonatorum , Biomarkers , Hypoxia-Ischemia, Brain , Humans , Asphyxia Neonatorum/metabolism , Biomarkers/metabolism , Infant, Newborn , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/diagnosis , Female , Pregnancy , Inflammation/metabolism , Placenta/metabolism
2.
Am J Physiol Renal Physiol ; 327(2): F314-F326, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38932694

ABSTRACT

Perinatal asphyxia (PA) poses a significant threat to multiple organs, particularly the kidneys. Diagnosing PA-associated kidney injury remains challenging, and treatment options are inadequate. Furthermore, there is a lack of long-term follow-up data regarding the renal implications of PA. In this study, 7-day-old male Wistar rats were exposed to PA using a gas mixture (4% O2; 20% CO2 in N2 for 15 min) to investigate molecular pathways linked to renal tubular damage, hypoxia, angiogenesis, heat shock response, inflammation, and fibrosis in the kidney. In a second experiment, adult rats with a history of PA were subjected to moderate renal ischemia-reperfusion (IR) injury to test the hypothesis that PA exacerbates renal susceptibility. Our results revealed an increased gene expression of renal injury markers (kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin), hypoxic and heat shock factors (hypoxia-inducible factor-1α, heat shock factor-1, and heat shock protein-27), proinflammatory cytokines (interleukin-1ß, interleukin-6, tumor necrosis factor-α, and monocyte chemoattractant protein-1), and fibrotic markers (transforming growth factor-ß, connective tissue growth factor, and fibronectin) promptly after PA. Moreover, a machine learning model was identified through random forest analysis, demonstrating an impressive classification accuracy (95.5%) for PA. Post-PA rats showed exacerbated functional decline and tubular injury and more intense hypoxic, heat shock, proinflammatory, and profibrotic response after renal IR injury compared with controls. In conclusion, PA leads to subclinical kidney injury, which may increase the susceptibility to subsequent renal damage later in life. In addition, the parameters identified through random forest analysis provide a robust foundation for future biomarker research in the context of PA.NEW & NOTEWORTHY This article demonstrates that perinatal asphyxia leads to subclinical kidney injury that permanently increases renal susceptibility to subsequent ischemic injury. We identified major molecular pathways involved in perinatal asphyxia-induced renal complications, highlighting potential targets of therapeutic approaches. In addition, random forest analysis revealed a model that classifies perinatal asphyxia with 95.5% accuracy that may provide a strong foundation for further biomarker research. These findings underscore the importance of multiorgan follow-up for perinatal asphyxia-affected patients.


Subject(s)
Acute Kidney Injury , Disease Models, Animal , Kidney , Rats, Wistar , Reperfusion Injury , Animals , Male , Acute Kidney Injury/pathology , Acute Kidney Injury/metabolism , Acute Kidney Injury/etiology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Kidney/pathology , Kidney/metabolism , Fibrosis , Asphyxia Neonatorum/metabolism , Asphyxia Neonatorum/complications , Asphyxia Neonatorum/pathology , Animals, Newborn , Rats , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Cytokines/metabolism , Age Factors , Inflammation Mediators/metabolism
3.
Neonatology ; 121(1): 25-33, 2024.
Article in English | MEDLINE | ID: mdl-37778335

ABSTRACT

OBJECTIVES: The aim of the study was to evaluate neuronal injury and immuno-inflammatory biomarkers in umbilical cord blood (UCB) at birth, in cases with perinatal asphyxia with or without hypoxic-ischemic encephalopathy (HIE), compared with healthy controls and to assess their ability to predict HIE. STUDY DESIGN: In this case-control study, term infants with perinatal asphyxia were recruited at birth. UCB was stored at delivery for batch analysis. HIE was diagnosed by clinical Sarnat staging at 24 h. Glial fibrillary acidic protein (GFAP), the neuronal biomarkers tau and neurofilament light protein (NFL), and a panel of cytokines were analyzed in a total of 150 term neonates: 50 with HIE, 50 with asphyxia without HIE (PA), and 50 controls. GFAP, tau, and NFL concentrations were measured using ultrasensitive single-molecule array (Simoa) assays, and a cytokine screening panel was applied to analyze the immuno-inflammatory and infectious markers. RESULTS: GFAP, tau, NFL, and several cytokines were significantly higher in newborns with moderate and severe HIE compared to a control group and provided moderate prediction of HIE II/III (AUC: 0.681-0.827). Furthermore, the levels of GFAP, tau, interleukin-6 (IL-6), and interleukin-8 (IL-8) were higher in HIE II/III cases compared with cases with PA/HIE I. IL-6 was also higher in HIE II/III compared with HIE I cases. CONCLUSIONS: Biomarkers of brain injury and inflammation were increased in umbilical blood in cases with asphyxia. Several biomarkers were higher in HIE II/III versus those with no HIE or HIE I, suggesting that they could assist in the prediction of HIE II/III.


Subject(s)
Asphyxia Neonatorum , Hypoxia-Ischemia, Brain , Infant , Humans , Infant, Newborn , Case-Control Studies , Interleukin-6 , Asphyxia , Hypoxia-Ischemia, Brain/metabolism , Fetal Blood/metabolism , Biomarkers , Cytokines/metabolism , Asphyxia Neonatorum/metabolism
4.
Pediatr Res ; 91(3): 598-605, 2022 02.
Article in English | MEDLINE | ID: mdl-33953355

ABSTRACT

BACKGROUND: Infants with moderate and severe neonatal encephalopathy (NE) frequently suffer from long-term adverse outcomes. We hypothesize that the urinary metabolome of newborns with NE reflects the evolution of injury patterns observed with magnetic resonance imaging (MRI). METHODS: Eligible patients were newborn infants with perinatal asphyxia evolving to NE and qualifying for therapeutic hypothermia (TH) included in the HYPOTOP trial. MRI was employed for characterizing brain injury. Urine samples of 55 infants were collected before, during, and after TH. Metabolic profiles of samples were recorded employing three complementary mass spectrometry-based assays, and the alteration of detected metabolic features between groups was assessed. RESULTS: The longitudinal assessment revealed significant perturbations of the urinary metabolome. After 24 h of TH, a stable disease pattern evolved characterized by the alterations of 4-8% of metabolic features related to lipid metabolism, metabolism of cofactors and vitamins, glycan biosynthesis and metabolism, amino acid metabolism, and nucleotide metabolism. Characteristic metabolomic fingerprints were observed for different MRI injury patterns. CONCLUSIONS: This study shows the potential of urinary metabolic profiles for the noninvasive monitoring of brain injury of infants with NE during TH. IMPACT: A comprehensive approach for the study of the urinary metabolome was employed involving a semi-targeted capillary electrophoresis-time-of-flight mass spectrometry (TOFMS) assay, an untargeted ultra-performance liquid chromatography (UPLC)-quadrupole TOFMS assay, and a targeted UPLC-tandem MS-based method for the quantification of amino acids. The longitudinal study of the urinary metabolome identified dynamic metabolic changes between birth and until 96 h after the initiation of TH. The identification of altered metabolic pathways in newborns with pathologic MRI outcomes might offer the possibility of developing noninvasive monitoring approaches for personalized adjustment of the treatment and for supporting early outcome prediction.


Subject(s)
Asphyxia Neonatorum , Brain Injuries , Hypothermia, Induced , Asphyxia Neonatorum/metabolism , Asphyxia Neonatorum/urine , Brain Diseases/metabolism , Brain Diseases/urine , Brain Injuries/metabolism , Brain Injuries/urine , Female , Humans , Infant , Infant, Newborn , Longitudinal Studies , Metabolome , Metabolomics/methods , Pregnancy
5.
Mol Cell Biochem ; 476(12): 4421-4434, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34472002

ABSTRACT

Perinatal asphyxia (PA)-induced brain injury may present as hypoxic-ischemic encephalopathy in the neonatal period, and long-term sequelae such as spastic motor deficits, intellectual disability, seizure disorders and learning disabilities. The brain injury is secondary to both the hypoxic-ischemic event and oxygenation-reperfusion following resuscitation. Following PA, a time-dependent progression of neuronal insult takes place in terms of transition of cell death from necrosis to apoptosis. This transition is the result of time-dependent progression of pathomechanisms which involve excitotoxicity, oxidative stress, and ultimately mitochondrial dysfunction in developing brain. More precisely mitochondrial respiration is suppressed and calcium signalling is dysregulated. Consequently, Bax-dependent mitochondrial permeabilization occurs leading to release of cytochrome c and activation of caspases leading to transition of cell death in developing brain. The therapeutic window lies within this transition process. At present, therapeutic hypothermia (TH) is the only clinical treatment available for treating moderate as well as severe asphyxia in new-born as it attenuates secondary loss of high-energy phosphates (ATP) (Solevåg et al. in Free Radic Biol Med 142:113-122, 2019; Gunn et al. in Pediatr Res 81:202-209, 2017), improving both short- and long-term outcomes. Mitoprotective therapies can offer a new avenue of intervention alone or in combination with therapeutic hypothermia for babies with birth asphyxia. This review will explore these mitochondrial pathways, and finally will summarize past and current efforts in targeting these pathways after PA, as a means of identifying new avenues of therapeutic intervention.


Subject(s)
Asphyxia Neonatorum/pathology , Asphyxia Neonatorum/therapy , Hypothermia, Induced/methods , Hypoxia-Ischemia, Brain/metabolism , Mitochondria/pathology , Neuroprotective Agents/pharmacology , Animals , Asphyxia Neonatorum/etiology , Asphyxia Neonatorum/metabolism , Female , Humans , Hypoxia-Ischemia, Brain/pathology , Infant, Newborn , Mitochondria/metabolism , Oxidative Stress , Pregnancy
6.
Int J Mol Sci ; 22(9)2021 May 01.
Article in English | MEDLINE | ID: mdl-34062911

ABSTRACT

Hypoxic-ischemic encephalopathy (HIE) remains to be a major cause of long-term neurodevelopmental deficits in term neonates. Hypothermia offers partial neuroprotection warranting research for additional therapies. Kynurenic acid (KYNA), an endogenous product of tryptophan metabolism, was previously shown to be beneficial in rat HIE models. We sought to determine if the KYNA analog SZR72 would afford neuroprotection in piglets. After severe asphyxia (pHa = 6.83 ± 0.02, ΔBE = -17.6 ± 1.2 mmol/L, mean ± SEM), anesthetized piglets were assigned to vehicle-treated (VEH), SZR72-treated (SZR72), or hypothermia-treated (HT) groups (n = 6, 6, 6; Tcore = 38.5, 38.5, 33.5 °C, respectively). Compared to VEH, serum KYNA levels were elevated, recovery of EEG was faster, and EEG power spectral density values were higher at 24 h in the SZR72 group. However, instantaneous entropy indicating EEG signal complexity, depression of the visual evoked potential (VEP), and the significant neuronal damage observed in the neocortex, the putamen, and the CA1 hippocampal field were similar in these groups. In the caudate nucleus and the CA3 hippocampal field, neuronal damage was even more severe in the SZR72 group. The HT group showed the best preservation of EEG complexity, VEP, and neuronal integrity in all examined brain regions. In summary, SZR72 appears to enhance neuronal activity after asphyxia but does not ameliorate early neuronal damage in this HIE model.


Subject(s)
Asphyxia Neonatorum/drug therapy , Brain Ischemia/drug therapy , Kynurenic Acid/analogs & derivatives , Neurons/metabolism , Animals , Asphyxia Neonatorum/metabolism , Asphyxia Neonatorum/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , CA1 Region, Hippocampal/diagnostic imaging , CA1 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/diagnostic imaging , CA3 Region, Hippocampal/drug effects , Disease Models, Animal , Electroencephalography , Evoked Potentials, Visual/drug effects , Humans , Kynurenic Acid/pharmacology , Neurons/drug effects , Neurons/pathology , Rats , Translational Research, Biomedical
7.
PLoS One ; 16(2): e0247229, 2021.
Article in English | MEDLINE | ID: mdl-33630892

ABSTRACT

BACKGROUND: Hypoxic perinatal brain injury is caused by lack of oxygen to baby's brain and can lead to death or permanent brain damage. However, the effectiveness of therapeutic hypothermia in birth asphyxiated infants with encephalopathy is uncertain. This systematic review and meta-analysis was aimed to estimate the pooled relative risk of mortality among birth asphyxiated neonates with hypoxic-ischemic encephalopathy in a global context. METHODS: We used the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) guidelines to search randomized control trials from electronic databases (PubMed, Cochrane library, Google Scholar, MEDLINE, Embase, Scopus, Web of Science, Cochrane Central Register of Controlled Trials (CENTRAL), and meta register of Current Controlled Trials (mCRT)). The authors extracted the author's name, year of publication, country, method of cooling, the severity of encephalopathy, the sample size in the hypothermic, and non-hypothermic groups, and the number of deaths in the intervention and control groups. A weighted inverse variance fixed-effects model was used to estimate the pooled relative risk of mortality. The subgroup analysis was done by economic classification of countries, methods of cooling, and cooling devices. Publication bias was assessed with a funnel plot and Eggers test. A sensitivity analysis was also done. RESULTS: A total of 28 randomized control trials with a total sample of 35, 92 (1832 hypothermic 1760 non-hypothermic) patients with hypoxic-ischemic encephalopathy were used for the analysis. The pooled relative risk of mortality after implementation of therapeutic hypothermia was found to be 0.74 (95%CI; 0.67, 0.80; I2 = 0.0%; p<0.996). The subgroup analysis revealed that the pooled relative risk of mortality in low, low middle, upper-middle and high income countries was 0.32 (95%CI; -0.95, 1.60; I2 = 0.0%; p<0.813), 0.5 (95%CI; 0.14, 0.86; I2 = 0.0%; p<0.998), 0.62 (95%CI; 0.41-0.83; I2 = 0.0%; p<0.634) and 0.76 (95%CI; 0.69-0.83; I2 = 0.0%; p<0.975) respectively. The relative risk of mortality was the same in selective head cooling and whole-body cooling method which was 0.74. Regarding the cooling device, the pooled relative risk of mortality is the same between the cooling cap and cooling blanket (0.74). However, it is slightly lower (0.73) in a cold gel pack. CONCLUSIONS: Therapeutic hypothermia reduces the risk of death in neonates with moderate to severe hypoxic-ischemic encephalopathy. Both selective head cooling and whole-body cooling method are effective in reducing the mortality of infants with this condition. Moreover, low income countries benefit the most from the therapy. Therefore, health professionals should consider offering therapeutic hypothermia as part of routine clinical care to newborns with hypoxic-ischemic encephalopathy especially in low-income countries.


Subject(s)
Asphyxia Neonatorum/metabolism , Hypoxia-Ischemia, Brain/metabolism , Animals , Asphyxia Neonatorum/therapy , Humans , Hypothermia, Induced , Hypoxia-Ischemia, Brain/therapy , Randomized Controlled Trials as Topic
8.
Acta Neurol Belg ; 121(3): 707-714, 2021 Jun.
Article in English | MEDLINE | ID: mdl-32533551

ABSTRACT

Neonatal hypoxic-ischemic encephalopathy (HIE) is a disorder mainly due to asphyxia during the perinatal period, and late diagnosis leads to high mortality. In this study, the expression of microRNA-199a (miR-199a) in HIE newborns was investigated, as well as its clinical significance in HIE diagnosis and prognosis. Circulating levels of S100B and NSE in HIE newborns were measured using enzyme-linked immunosorbent assay, and the expression of miR-199a was analyzed using quantitative real-time PCR. The diagnostic value of miR-199a, S100B and NSE was evaluated using the receiver operating characteristic (ROC) analysis, and their prognostic value was assessed by the evaluation of Gesell intellectual development of the HIE newborns. HIE newborns possessed significantly increased levels of S100B and NSE and decreased miR-199a (all P < 0.01). The Neonatal Behavioral Neurological Assessment (NBNA) score of HIE newborns was negatively correlated with S100B and NSE, while was positively correlated miR-199a. The ROC analysis results showed the diagnostic value of serum miR-199a, and the combined detection of miR-199a, S100B and NSE could obtained the highest diagnostic accuracy in HIE newborns. miR-199a expression was lowest in newborns with severe HIE, and it had diagnostic potential to distinguish HIE cases with different severity. Regarding the prognosis of neonatal HIE, the correlation of miR-199a, S100B, NSE with Gesell intellectual development was found in HIE newborns. The decreased miR-199a in HIE newborns serves as a potential diagnostic biomarker and may help to improve the diagnostic and prognostic value of S100B and NSE in neonatal HIE.


Subject(s)
Asphyxia Neonatorum/diagnosis , Hypoxia-Ischemia, Brain/diagnosis , MicroRNAs/metabolism , Phosphopyruvate Hydratase/blood , S100 Calcium Binding Protein beta Subunit/blood , Asphyxia Neonatorum/genetics , Asphyxia Neonatorum/metabolism , Biomarkers/blood , Female , Humans , Hypoxia-Ischemia, Brain/genetics , Hypoxia-Ischemia, Brain/metabolism , Infant, Newborn , Male , MicroRNAs/genetics , Prognosis
9.
Sci Rep ; 10(1): 16443, 2020 10 05.
Article in English | MEDLINE | ID: mdl-33020561

ABSTRACT

Hypoxic-ischaemia renders the neonatal brain susceptible to early secondary injury from oxidative stress and impaired autoregulation. We aimed to describe cerebral oxygen kinetics and haemodynamics immediately following return of spontaneous circulation (ROSC) and evaluate non-invasive parameters to facilitate bedside monitoring. Near-term sheep fetuses [139 ± 2 (SD) days gestation, n = 16] were instrumented to measure carotid artery (CA) flow, pressure, right brachial arterial and jugular venous saturation (SaO2 and SvO2, respectively). Cerebral oxygenation (crSO2) was measured using near-infrared spectroscopy (NIRS). Following induction of severe asphyxia, lambs received cardiopulmonary resuscitation using 100% oxygen until ROSC, with oxygen subsequently weaned according to saturation nomograms as per current guidelines. We found that oxygen consumption did not rise following ROSC, but oxygen delivery was markedly elevated until 15 min after ROSC. CrSO2 and heart rate each correlated with oxygen delivery. SaO2 remained > 90% and was less useful for identifying trends in oxygen delivery. CrSO2 correlated inversely with cerebral fractional oxygen extraction. In conclusion, ROSC from perinatal asphyxia is characterised by excess oxygen delivery that is driven by rapid increases in cerebrovascular pressure, flow, and oxygen saturation, and may be monitored non-invasively. Further work to describe and limit injury mediated by oxygen toxicity following ROSC is warranted.


Subject(s)
Asphyxia/metabolism , Brain/metabolism , Oxygen/metabolism , Return of Spontaneous Circulation/physiology , Animals , Animals, Newborn , Asphyxia/physiopathology , Asphyxia Neonatorum/metabolism , Asphyxia Neonatorum/physiopathology , Brachial Artery/metabolism , Brachial Artery/physiopathology , Brain/physiopathology , Cardiopulmonary Resuscitation/methods , Carotid Arteries/metabolism , Carotid Arteries/physiopathology , Cerebrovascular Circulation/physiology , Female , Hemodynamics/physiology , Hypoxia/metabolism , Hypoxia/physiopathology , Oxygen Consumption/physiology , Pregnancy , Sheep
10.
Exp Neurol ; 334: 113457, 2020 12.
Article in English | MEDLINE | ID: mdl-32889009

ABSTRACT

Neonatal hypoxia-ischemia and resulting encephalopathies are of significant concern. Intrapartum asphyxia is a leading cause of neonatal death globally. Among surviving infants, there remains a high incidence of hypoxic-ischemic encephalopathy due to neonatal hypoxic-ischemic brain injury, manifesting as mild conditions including attention deficit hyperactivity disorder, and debilitating disorders such as cerebral palsy. Various animal models of neonatal hypoxic brain injury have been implemented to explore cellular and molecular mechanisms, assess the potential of novel therapeutic strategies, and characterize the functional and behavioural correlates of injury. Each of the animal models has individual advantages and limitations. The present review looks at several widely-used and alternative rodent models of neonatal hypoxia and hypoxia-ischemia; it highlights their strengths and limitations, and their potential for continued and improved use.


Subject(s)
Asphyxia Neonatorum/metabolism , Disease Models, Animal , Hypoxia-Ischemia, Brain/metabolism , Animals , Asphyxia Neonatorum/complications , Asphyxia Neonatorum/pathology , Cerebral Palsy/etiology , Cerebral Palsy/metabolism , Cerebral Palsy/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Ischemia, Brain/etiology , Hypoxia-Ischemia, Brain/pathology , Infant, Newborn , Inflammation Mediators/metabolism , Rodentia
11.
PLoS One ; 15(5): e0233851, 2020.
Article in English | MEDLINE | ID: mdl-32470084

ABSTRACT

Brain interstitial pH (pHbrain) alterations play an important role in the mechanisms of neuronal injury in neonatal hypoxic-ischemic encephalopathy (HIE) induced by perinatal asphyxia. The newborn pig is an established large animal model to study HIE, however, only limited information on pHbrain alterations is available in this species and it is restricted to experimental perinatal asphyxia (PA) and the immediate reventilation. Therefore, we sought to determine pHbrain over the first 24h of HIE development in piglets. Anaesthetized, ventilated newborn pigs (n = 16) were instrumented to control major physiological parameters. pHbrain was determined in the parietal cortex using a pH-selective microelectrode. PA was induced by ventilation with a gas mixture containing 6%O2-20%CO2 for 20 min, followed by reventilation with air for 24h, then the brains were processed for histopathology assessment. The core temperature was maintained unchanged during PA (38.4±0.1 vs 38.3±0.1°C, at baseline versus the end of PA, respectively; mean±SEM). In the arterial blood, PA resulted in severe hypoxia (PaO2: 65±4 vs 23±1*mmHg, *p<0.05) as well as acidosis (pHa: 7.53±0.03 vs 6.79±0.02*) that is consistent with the observed hypercapnia (PaCO2: 37±3 vs 160±6*mmHg) and lactacidemia (1.6±0.3 vs 10.3±0.7*mmol/L). Meanwhile, pHbrain decreased progressively from 7.21±0.03 to 5.94±0.11*. Reventilation restored pHa, blood gases and metabolites within 4 hours except for PaCO2 that remained slightly elevated. pHbrain returned to 7.0 in 29.4±5.5 min and then recovered to its baseline level without showing secondary alterations during the 24 h observation period. Neuropathological assessment also confirmed neuronal injury. In conclusion, in spite of the severe acidosis and alterations in blood gases during experimental PA, pHbrain recovered rapidly and notably, there was no post-asphyxia hypocapnia that is commonly observed in many HIE babies. Thus, the neuronal injury in our piglet model is not associated with abnormal pHbrain or low PaCO2 over the first 24 h after PA.


Subject(s)
Brain/metabolism , Hypoxia-Ischemia, Brain/metabolism , Acidosis/blood , Acidosis/complications , Acidosis/metabolism , Acidosis/physiopathology , Animals , Animals, Newborn , Asphyxia Neonatorum/blood , Asphyxia Neonatorum/metabolism , Asphyxia Neonatorum/physiopathology , Brain/pathology , Brain/physiopathology , Hemodynamics , Hydrogen-Ion Concentration , Hypercapnia/blood , Hypercapnia/complications , Hypercapnia/metabolism , Hypercapnia/physiopathology , Hypoxia-Ischemia, Brain/blood , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/physiopathology , Male , Neurons/pathology , Oxygen/metabolism , Swine
12.
Int J Mol Sci ; 21(9)2020 Apr 25.
Article in English | MEDLINE | ID: mdl-32344930

ABSTRACT

High-dose human recombinant erythropoietin (rEPO) is a promising potential neuroprotective treatment in preterm and full-term neonates with hypoxic-ischemic encephalopathy (HIE). There are limited data on the pharmacokinetics of high-dose rEPO in neonates. We examined the effects of body weight, gestation age, global asphyxia, cerebral ischemia, hypothermia and exogenous rEPO on the pharmacokinetics of high-dose rEPO in fetal sheep. Near-term fetal sheep on gestation day 129 (0.87 gestation) (full term 147 days) received sham-ischemia (n = 5) or cerebral ischemia for 30 min followed by treatment with vehicle (n = 4), rEPO (n = 8) or combined treatment with rEPO and hypothermia (n = 8). Preterm fetal sheep on gestation day 104 (0.7 gestation) received sham-asphyxia (n = 1) or complete umbilical cord occlusion for 25 min followed by i.v. infusion of vehicle (n = 8) or rEPO (n = 27) treatment. rEPO was given as a loading bolus, followed by a prolonged continuous infusion for 66 to 71.5 h in preterm and near-term fetuses. A further group of preterm fetal sheep received repeated bolus injections of rEPO (n = 8). The plasma concentrations of rEPO were best described by a pharmacokinetic model that included first-order and mixed-order elimination with linear maturation of elimination with gestation age. There were no detectable effects of therapeutic hypothermia, cerebral ischemia, global asphyxia or exogenous treatment on rEPO pharmacokinetics. The increase in rEPO elimination with gestation age suggests that to maintain target exposure levels during prolonged treatment, the dose of rEPO may have to be adjusted to match the increase in size and growth. These results are important for designing and understanding future studies of neuroprotection with high-dose rEPO.


Subject(s)
Erythropoietin/pharmacokinetics , Hypothermia, Induced , Hypoxia-Ischemia, Brain/drug therapy , Neuroprotective Agents/pharmacokinetics , Animals , Asphyxia Neonatorum/drug therapy , Asphyxia Neonatorum/metabolism , Asphyxia Neonatorum/therapy , Birth Weight , Body Weight , Combined Modality Therapy , Dose-Response Relationship, Drug , Embryonic Development/drug effects , Erythropoietin/administration & dosage , Female , Fetus/drug effects , Gestational Age , Humans , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/therapy , Infant, Newborn , Infusions, Intravenous , Injections, Intravenous , Models, Biological , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/blood , Recombinant Proteins/administration & dosage , Recombinant Proteins/blood , Recombinant Proteins/pharmacokinetics , Sheep/embryology , Species Specificity
13.
J Perinat Med ; 48(5): 514-515, 2020 Jun 25.
Article in English | MEDLINE | ID: mdl-32286248

ABSTRACT

Background Metabolic acidosis, measured in arterial umbilical cord blood at birth, is the most accepted definition of birth asphyxia. The aim of the study was to investigate the rates of metabolic acidosis across the entire range of Apgar score values (0-10) at 1, 5, and 10 min in term infants. Methods In a population-based Swedish cohort of births between 2008 and 2013, we included 85,076 term (≥37 weeks) non-malformed infants with information from umbilical arterial blood gas analyses and complete information on Apgar scores (0-10) at 1, 5, and 10 min. Results Rates of metabolic acidosis generally decreased with increasing Apgar score values. For Apgar score at 1 min, this decrease was consistent from Apgar score 0 (35%) to Apgar score 10 (0%). For Apgar scores at 5 and 10 min, the decrease was consistent for Apgar score values from 6 to 10. Conclusion Although there is a close association between Apgar score values and rates of metabolic acidosis, Apgar score is not and should not be used as a measure of birth asphyxia.


Subject(s)
Acidosis , Apgar Score , Asphyxia Neonatorum , Fetal Blood/chemistry , Acidosis/blood , Acidosis/diagnosis , Acidosis/etiology , Asphyxia Neonatorum/complications , Asphyxia Neonatorum/diagnosis , Asphyxia Neonatorum/metabolism , Blood Gas Analysis/methods , Cohort Studies , Female , Humans , Hydrogen-Ion Concentration , Infant, Newborn , Male , Research Design/standards , Research Design/statistics & numerical data , Sweden , Time Factors
14.
J Neuroinflammation ; 17(1): 26, 2020 Jan 18.
Article in English | MEDLINE | ID: mdl-31954397

ABSTRACT

BACKGROUND: Recent findings describe microglia as modulators of neurogenesis in the subventricular zone (SVZ). SVZ microglia in the adult rat are thought to adopt a neurotrophic phenotype after ischemic stroke. Early postnatal microglia are endogenously activated and may therefore exhibit an increased sensitivity to neonatal hypoxia-ischemia (HI). The goal of this study was to investigate the impact of cortico-striatal HI on the microglial phenotype, function, and gene expression in the early postnatal SVZ. METHODS: Postnatal day (P)7 rats underwent sham or right-hemispheric HI surgery. Microglia in the SVZ, the uninjured cortex, and corpus callosum were immunohistochemically analyzed at P10, P20, and P40. The transcriptome of microdissected SVZ and cortical microglia was analyzed at P10 and P20, and the effect of P10 SVZ microglia on neurosphere generation in vitro was studied. RESULTS: The microglial response to HI was region-specific. In the SVZ, a microglial accumulation, prolonged activation and phagocytosis was noted that was not observed in the cortex and corpus callosum. The transcriptome of SVZ microglia and cortical microglia were distinct, and after HI, SVZ microglia concurrently upregulated pro- and anti-inflammatory as well as neurotrophic genes. In vitro, microglia isolated from the SVZ supported neurosphere generation in a concentration-dependent manner. CONCLUSIONS: Microglia are an inherent cellular component of the early postnatal SVZ and undergo developmental changes that are affected on many aspects by neonatal HI injury. Our results demonstrate that early postnatal SVZ microglia are sensitive to HI injury and display a long-lasting region-specific response including neurotrophic features.


Subject(s)
Hypoxia-Ischemia, Brain/pathology , Lateral Ventricles/pathology , Microglia/pathology , Neurogenesis/physiology , Animals , Animals, Newborn , Asphyxia Neonatorum/metabolism , Asphyxia Neonatorum/pathology , Hypoxia-Ischemia, Brain/metabolism , Lateral Ventricles/metabolism , Microglia/metabolism , Phenotype , Rats , Rats, Sprague-Dawley
15.
Neuroscience ; 425: 235-250, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31785355

ABSTRACT

Thioredoxin family proteins are key modulators of cellular redox regulation and have been linked to several physiological functions, including the cellular response to hypoxia-ischemia. During perinatal hypoxia-ischemia (PHI), the central nervous system is subjected to a fast decrease in O2 and nutrients with a subsequent reoxygenation that ultimately leads to the production of reactive species impairing physiological redox signaling. Particularly, the retina is one of the most affected tissues, due to its high oxygen consumption and exposure to light. One of the main consequences of PHI is retinopathy of prematurity, comprising changes in retinal neural and vascular development, with further compensatory mechanisms that can ultimately lead to retinal detachment and blindness. In this study, we have analyzed long-term changes that occur in the retina using two well established in vivo rat PHI models (perinatal asphyxia and carotid ligation model), as well as the ARPE-19 cell line that was exposed to hypoxia and reoxygenation. We observed significant changes in the protein levels of the cytosolic oxidoreductase thioredoxin 1 (Trx1) in both animal models and a cell model. Knock-down of Trx1 in ARPE-19 cells affected cell morphology, proliferation and the levels of specific differentiation markers. Administration of recombinant Trx1 decreased astrogliosis and improved delayed neurodevelopment in animals exposed to PHI. Taken together, our results suggest therapeutical implications for Trx1 in retinal damage induced by hypoxia-ischemia during birth.


Subject(s)
Asphyxia Neonatorum/metabolism , Central Nervous System/metabolism , Hypoxia/metabolism , Retina/metabolism , Thioredoxins/metabolism , Animals , Disease Models, Animal , Female , Oxidative Stress/physiology , Pregnancy , Rats, Sprague-Dawley , Retinal Diseases/metabolism
16.
EBioMedicine ; 47: 484-491, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31451436

ABSTRACT

BACKGROUND: The TOBY-Xe proof of concept randomised trial found no effect of xenon combined with hypothermia after birth asphyxia on the lactate to N-acetyl aspartate ratio (Lac/NAA) in the thalamus and fractional anisotropy (FA) in white matter tracts measured within 15 days of birth. To confirm that these biomarkers are qualified to predict long-term outcome after neural rescue therapy we assessed surviving participants at 2-3 years of age. METHODS: Of the 92 infants in TOBY-Xe, one was omitted from the study, 69 survived and we assessed 62 participants, 32 in the hypothermia and xenon and 30 in the hypothermia only groups. We examined the relation between Lac/NAA and FA and the scores of the Bayley Scales of Infant and Toddler Development III and calculated their predictive accuracy for moderate or severe disability or death. RESULTS: Fifteen of 62 participants (24%) developed moderate/severe disability, and 22/92 (24%) died. The Lac/NAA ratio (difference in medians 0.628, 95% CI -0.392 to 4.684) and FA (difference in means -0.055, 95% CI -0.033 to -0.077) differed significantly between participants with or without moderate or severe disability or death and were significantly related with development scores in both groups. Adverse outcomes were correctly identified in 95.65% of cases by Lac/NAA and 78.79% by FA, with adequate mean calibration of the model. INTERPRETATION: The results confirm the qualification of the cerebral magnetic resonance biomarkers employed in the TOBY-Xe study as predictors of outcome after neuroprotective therapy. FUND: The Centre for the Developing Brain, King's College London, UK.


Subject(s)
Asphyxia Neonatorum/metabolism , Asphyxia Neonatorum/therapy , Biomarkers , Cerebral Cortex/metabolism , Hypothermia, Induced , Xenon/therapeutic use , Asphyxia Neonatorum/etiology , Combined Modality Therapy , Humans , Hypothermia, Induced/methods , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/adverse effects , Neuroprotective Agents/therapeutic use , ROC Curve , Reproducibility of Results , Treatment Outcome , Xenon/administration & dosage , Xenon/adverse effects
17.
J Neurosci Res ; 97(10): 1266-1277, 2019 10.
Article in English | MEDLINE | ID: mdl-31257630

ABSTRACT

Anoxia during delivery is a complication that can disturb infant brain development leading to various types of neurological disorders. Our studies have shown that increased body temperature of newborn rats of both sexes intensifies the postanoxic oxidative stress and prevents triggering the endogenous adaptive response such as HIF-1α activation. Currently, brain-derived neurotrophic factor-BDNF is considered to be a modulator of neuronal plasticity. In the developing brain, mature BDNF and its precursor exhibit prosurvival action through the TrkB receptor and proapoptotic functions binding to p75NTR , respectively. The aim of our experiments was to check the effects of body temperature on the postanoxic level of BDNF and on the expression of its receptors as well as on the marker of apoptosis-caspase-3 in the rat brain. Two-day-old Wistar Han rats (male/female ratio, 1:1) were exposed to anoxia in 100% nitrogen atmosphere for 10 min in different thermal conditions, which allowed them to regulate their rectal temperature at the following levels: normothermic-33°C; hyperthermic-37°C; and extremely hyperthermic-39°C. Thermal conditions during neonatal anoxia affected the level of proBDNF, BDNF as well as their receptors and caspase-3 in the forebrain. The increased BDNF protein level followed by decreased caspase-3 protein level was probably dependent on body temperature under anoxic conditions and was observed only in rats maintaining decreased body temperature. The positive effect of BDNF was not observed under hyperthermic conditions. Moreover, BDNF level changes correlated with body temperature probably affected the learning and spatial memory in juvenile rats.


Subject(s)
Body Temperature/physiology , Brain-Derived Neurotrophic Factor/metabolism , Brain/metabolism , Hypoxia, Brain/metabolism , Animals , Animals, Newborn , Asphyxia Neonatorum/metabolism , Caspase 3/metabolism , Female , Male , Rats , Rats, Wistar , Receptor, trkB/metabolism
18.
PLoS One ; 14(7): e0219211, 2019.
Article in English | MEDLINE | ID: mdl-31276543

ABSTRACT

OBJECTIVE: Asphyxia of newborns is a severe and frequent challenge of the peri- and postnatal period. The purpose of this study was to study early morphological, immunological and structural alterations in lung tissue after asphyxia and hemorrhage (AH). METHODS: 44 neonatal piglets (age 32 hrs) underwent asphyxia and hemorrhage (AH) and were treated according to the international liaison committee of resuscitation (ILCOR) guidelines. For this study, 15 piglets (blood transfusion (RBC) n = 9; NaCl n = 6, mean age 31 hrs) were randomly picked. 4 hours after ROSC (return of spontaneous circulation), lung tissue and blood samples were collected. RESULTS: An elevation of myeloperoxidase (MPO) activity was observed 4 hrs after AH accompanied by an increase of surfactant D after RBC treatment. After AH tight junction proteins Claudin 18 and junctional adhesion molecule 1 (JAM1) were down-regulated, whereas Occludin was increased. Furthermore, after AH and RBC treatment dephosphorylated active form of Connexin 43 was increased. CONCLUSIONS: AH in neonatal pigs is associated with early lung injury, inflammation and alterations of tight junctions (Claudin, Occludin, JAM-1) and gap junctions (Connexin 43) in lung tissue, which contributes to the development of lung edema and impaired function.


Subject(s)
Asphyxia Neonatorum/physiopathology , Lung Injury/physiopathology , Lung/physiopathology , Animals , Animals, Newborn/metabolism , Asphyxia/physiopathology , Asphyxia Neonatorum/metabolism , Cell Adhesion Molecules/metabolism , Claudins/metabolism , Connexin 43/metabolism , Disease Models, Animal , Gap Junctions , Lung Injury/metabolism , Occludin/metabolism , Peroxidase/analysis , Pulmonary Surfactant-Associated Protein D/analysis , Shock, Hemorrhagic/immunology , Shock, Hemorrhagic/metabolism , Swine , Tight Junctions/metabolism
19.
Neurotox Res ; 36(3): 472-490, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31187430

ABSTRACT

We have recently reported that global perinatal asphyxia (PA) induces a regionally sustained increase in oxidized glutathione (GSSG) levels and GSSG/GSH ratio, a decrease in tissue-reducing capacity, a decrease in catalase activity, and an increase in apoptotic caspase-3-dependent cell death in rat neonatal brain up to 14 postnatal days, indicating a long-term impairment in redox homeostasis. In the present study, we evaluated whether the increase in GSSG/GSH ratio observed in hippocampus involves changes in glutathione reductase (GR) and glutathione peroxidase (GPx) activity, the enzymes reducing glutathione disulfide (GSSG) and hydroperoxides, respectively, as well as catalase, the enzyme protecting against peroxidation. The study also evaluated whether there is a shift in the metabolism towards the penthose phosphate pathway (PPP), by measuring TIGAR, the TP53-inducible glycolysis and apoptosis regulator, associated with delayed cell death, further monitoring calpain activity, involved in bax-dependent cell death, and XRCC1, a scaffolding protein interacting with genome sentinel proteins. Global PA was induced by immersing fetus-containing uterine horns removed by a cesarean section from on term rat dams into a water bath at 37 °C for 21 min. Asphyxia-exposed and sibling cesarean-delivered fetuses were manually resuscitated and nurtured by surrogate dams. Animals were euthanized at postnatal (P) days 1 or 14, dissecting samples from hippocampus to be assayed for glutathione, GR, GPx (all by spectrophotometry), catalase (Western blots and ELISA), TIGAR (Western blots), calpain (fluorescence), and XRCC1 (Western blots). One hour after delivery, asphyxia-exposed and control neonates were injected with either 100 µl saline or 0.8 mmol/kg nicotinamide, i.p., shown to protect from the short- and long-term consequences of PA. It was found that global PA produced (i) a sustained increase of GSSG levels and GSSG/GSH ratio at P1 and P14; (ii) a decrease of GR, GPx, and catalase activity at P1 and P14; (iii) a decrease at P1, followed by an increase at P14 of TIGAR levels; (iv) an increase of calpain activity at P14; and (v) an increase of XRCC1 levels, but only at P1. (vi) Nicotinamide prevented the effect of PA on GSSG levels and GSSG/GSH ratio, and on GR, GPx, and catalase activity, also on increased TIGAR levels and calpain activity observed at P14. The present study demonstrates that the long-term impaired redox homeostasis observed in the hippocampus of rats subjected to global PA implies changes in GR, GPx, and catalase, and a shift towards PPP, as indicated by an increase of TIGAR levels at P14.


Subject(s)
Asphyxia Neonatorum/complications , Glutathione/metabolism , Hippocampus/metabolism , Niacinamide/pharmacology , Oxidative Stress , Pentose Phosphate Pathway , Animals , Asphyxia Neonatorum/metabolism , Catalase/metabolism , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Hippocampus/drug effects , Hippocampus/enzymology , Homeostasis/drug effects , Metabolic Networks and Pathways , Oxidative Stress/drug effects , Pentose Phosphate Pathway/drug effects , Phosphoric Monoester Hydrolases/metabolism , Rats , Rats, Wistar
20.
Free Radic Biol Med ; 142: 3-15, 2019 10.
Article in English | MEDLINE | ID: mdl-31226400

ABSTRACT

The brain is one of the main organs affected by hypoxia and reoxygenation in the neonatal period and one of the most vulnerable to oxidative stress. Hypoxia/ischemia and reoxygenation leads to impairment of neurogenesis, disruption of cortical migration, mitochondrial damage and neuroinflammation. The extent of the injury depends on the clinical manifestation in the affected regions. Preterm newborns are highly vulnerable, and they exhibit severe clinical manifestations such as intraventricular hemorrhage (IVH), retinopathy of prematurity (ROP) and diffuse white matter injury (DWMI) among others. In the neonatal period, the accumulation of high levels of reactive oxygen species exacerbated by the immature antioxidant defense systems in represents cellular threats that, if they exceed or bypass physiological counteracting mechanisms, are responsible of significant neuronal damage. Several experimental models in mice mimic the consequences of perinatal asphyxia and the use of oxygen in the reanimation process that produce brain injury. The aim of this review is to highlight brain damage associated with oxidative stress in different murine models of hypoxia/ischemia and reoxygenation.


Subject(s)
Asphyxia Neonatorum/pathology , Brain Ischemia/pathology , Brain/pathology , Cerebral Intraventricular Hemorrhage/pathology , Hypoxia/pathology , Retinopathy of Prematurity/pathology , Animals , Asphyxia Neonatorum/metabolism , Brain/metabolism , Brain Ischemia/metabolism , Cerebral Intraventricular Hemorrhage/metabolism , Disease Models, Animal , Humans , Hypoxia/metabolism , Infant, Newborn , Mice , Oxidation-Reduction , Oxidative Stress , Oxygen/metabolism , Reactive Oxygen Species/metabolism , Retinopathy of Prematurity/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL