Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(13)2021 Jul 01.
Article in English | MEDLINE | ID: mdl-34281174

ABSTRACT

Seizures are common in newborn infants with hypoxic-ischemic encephalopathy and are highly associated with adverse neurodevelopmental outcomes. The impact of seizure activity on the developing brain and the most effective way to manage these seizures remain surprisingly poorly understood, particularly in the era of therapeutic hypothermia. Critically, the extent to which seizures exacerbate brain injury or merely reflect the underlying evolution of injury is unclear. Current anticonvulsants, such as phenobarbital and phenytoin have poor efficacy and preclinical studies suggest that most anticonvulsants are associated with adverse effects on the developing brain. Levetiracetam seems to have less potential neurotoxic effects than other anticonvulsants but may not be more effective. Given that therapeutic hypothermia itself has significant anticonvulsant effects, randomized controlled trials of anticonvulsants combined with therapeutic hypothermia, are required to properly determine the safety and efficacy of these drugs. Small clinical studies suggest that prophylactic phenobarbital administration may improve neurodevelopmental outcomes compared to delayed administration; however, larger high-quality studies are required to confirm this. In conclusion, there is a distinct lack of high-quality evidence for whether and to what extent neonatal seizures exacerbate brain damage after hypoxia-ischemia and how best to manage them in the era of therapeutic hypothermia.


Subject(s)
Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/therapy , Seizures/therapy , Animals , Anticonvulsants/therapeutic use , Brain/drug effects , Brain/growth & development , Humans , Hypothermia, Induced/methods , Hypothermia, Induced/trends , Phenobarbital/therapeutic use , Randomized Controlled Trials as Topic , Seizures/physiopathology
2.
J Physiol ; 598(20): 4523-4536, 2020 10.
Article in English | MEDLINE | ID: mdl-32705685

ABSTRACT

KEY POINTS: The majority of intrapartum decelerations are widely believed to be mediated by the baroreflex secondary to brief umbilical cord occlusions (UCOs) but this remains unproven. We examined the responses to brief-UCOs in fetal sheep and compared these to a phenylephrine-stimulated baroreflex in a separate cohort. A further cohort was instrumented with near-infrared spectroscopy to measure cerebral oxygenation during UCO. The first 3-4 s of the brief-UCOs were consistent with a baroreflex, and associated with a minor fall in fetal heart rate (FHR). Thereafter, the remainder of the FHR decelerations were highly consistent with the peripheral chemoreflex. The baroreflex is not sufficient to produce deep, rapid decelerations characteristic of variable decelerations and it is therefore likely to be a minor contributor to intrapartum decelerations. ABSTRACT: Fetal heart rate (FHR) monitoring is widely used to assess fetal wellbeing during labour, yet the physiology underlying FHR patterns remains incompletely understood. The baroreflex is widely believed to mediate brief intrapartum decelerations, but evidence supporting this theory is lacking. We therefore investigated the physiological changes in near-term fetal sheep during brief repeated umbilical cord occlusions (brief-UCOs, n = 15). We compared this to separate cohorts that underwent a phenylephrine challenge to stimulate the baroreflex (n = 9) or were instrumented with near-infrared spectroscopy and underwent prolonged 15-min complete UCO (prolonged-UCO, n = 9). The first 3-4 s of brief-UCOs were associated with hypertension (P = 0.000), a fall in FHR by 9.7-16.9 bpm (P = 0.002). The FHR/MAP relationship during this time was consistent with that observed during a phenylephrine-induced baroreflex. At 4-5 s, the FHR/MAP relationship began to deviate from the phenylephrine baroreflex curve as FHR fell independently of MAP until its nadir in association with intense peripheral vasoconstriction (P = 0.000). During prolonged-UCO, cerebral oxygenation remained steady until 4 s after the start of prolonged-UCO, and then began to fall (P = 0.000). FHR and cerebral oxygenation then fell in parallel until the FHR nadir. In conclusion, the baroreflex has a minor role in mediating the first 3-4 s of FHR decelerations during complete UCO, but thereafter the peripheral chemoreflex is the dominant mediator. Overall, the baroreflex is neither necessary nor sufficient to produce deep, rapid decelerations characteristic of variable decelerations; it is therefore likely to be a minor contributor to intrapartum decelerations.


Subject(s)
Baroreflex , Heart Rate, Fetal , Animals , Deceleration , Female , Fetus , Pregnancy , Sheep , Umbilical Cord
3.
Pediatr Res ; 88(6): 857-864, 2020 12.
Article in English | MEDLINE | ID: mdl-32179873

ABSTRACT

BACKGROUND: Sensitive biomarkers are needed to rapidly identify high-risk infants after hypoxia-ischemia for neuroprotective treatment. Hypotension is a key determinant of hypoxic-ischemic neural injury, and a potent stimulus of humoral pressors including angiotensin-II and arginine vasopressin. We therefore aimed to quantify the relationship between vasopressin and angiotensin-II levels in the latent phase after hypoxia-ischemia induced by umbilical cord occlusion (UCO) with both the severity of preceding hypotension and subsequent neuronal injury. METHODS: Chronically instrumented near-term fetal sheep underwent sham-UCO or UCO for either 15 min or until mean arterial pressure was <8 mmHg. Neuronal injury was assessed after 72 h recovery. RESULTS: Umbilical cord occlusion was associated with severe hypotension that recovered after UCO; two fetuses developed profound secondary hypotension within 6 h and died. Vasopressin levels but not angiotensin-II were significantly elevated 1-3 h after UCO and were closely associated with the severity of hypotension during UCO and the subsequent severity of neuronal loss in the parasagittal and lateral cortex, caudate nucleus and putamen. The Youden cut-point for vasopressin at 1 h was 180.0 pmol/L, with sensitivity 100% and specificity 92.3% for severe neuronal injury or death. CONCLUSION: Vasopressin levels shortly after moderate-severe hypoxia-ischemia may be a useful early biomarker to guide the timely implementation of neuroprotective treatment. IMPACT: It can be difficuIt to rapidly identify infants who might benefit from therapeutic hypothermia. We investigated whether increases in plasma pressor hormones early after hypoxia-ischemia were biomarkers for neonatal hypoxic-ischemic encephalopathy using near-term fetal sheep. Arginine vasopressin levels were elevated at 1-3 h after hypoxia-ischemia and were predictive of the severity of preceding hypotension and subsequent risk of severe neuronal injury or death after hypoxia-ischemia. Arginine vasopressin may help identify neonates at high risk of hypoxic-ischemic encephalopathy early within the therapeutic window for hypothermia.


Subject(s)
Biomarkers/blood , Hypotension/embryology , Hypoxia-Ischemia, Brain/embryology , Nervous System Diseases/embryology , Sheep/embryology , Vasopressins/blood , Angiotensin II/metabolism , Animals , Arginine Vasopressin/metabolism , Blood Gas Analysis , Disease Models, Animal , Female , Fetal Hypoxia , Hydrogen-Ion Concentration , Male , Neurons , Umbilical Cord/pathology
4.
J Physiol ; 597(23): 5535-5548, 2019 12.
Article in English | MEDLINE | ID: mdl-31529698

ABSTRACT

KEY POINTS: •Therapeutic hypothermia needs to be started as early as possible in the first 6 h after acute injury caused by hypoxia-ischaemia (HI), but the severity and timing of HI are often unclear. In this study we evaluated whether measures of heart rate variability (HRV) might provide early biomarkers of HI. •The duration but not magnitude of suppression of HRV power and conversely increased sample entropy of the heart rate were associated with severity of HI, such that changes in the first 3 h did not discriminate between groups. •Relative changes in HRV power bands showed different patterns between groups and therefore may have the potential to evaluate the severity of HI. •Aberrant fetal heart rate patterns and increased arginine vasopressin levels in the first hour after moderate and severe HI were correlated with loss of EEG power after 3 days' recovery, suggesting potential utility as early biomarkers of outcome. ABSTRACT: Therapeutic hypothermia is partially neuroprotective after acute injury caused by hypoxia-ischaemia (HI), likely because the timing and severity of HI are often unclear, making timely recruitment for treatment challenging. We evaluated the utility of changes in heart rate variability (HRV) after HI as biomarkers of the timing and severity of acute HI. Chronically instrumented fetal sheep at 0.85 gestational age were exposed to different durations of umbilical cord occlusion to produce mild (n = 6), moderate (n = 8) or severe HI (n = 8) or to sham occlusion (n = 5). Heart rate (HR) and HRV indices were assessed until 72 h after HI. All HI groups showed suppressed very low frequency HRV power and elevated sample entropy for the first 3 h; more prolonged changes were associated with greater severity of HI. Analysis of relative changes in spectral power showed that the moderate and severe groups showed a shift towards higher HRV frequencies, which was most marked after severe HI. This shift was associated with abnormal rhythmic HR patterns including sinusoidal patterns in the first hour after HI, and with elevated plasma levels of arginine vasopressin, which were correlated with subsequent loss of EEG power by day 3. In conclusion, absolute changes in HRV power in the first 3 h after acute HI were not significantly related to the severity of HI. The intriguing relative shift in spectral power towards higher frequencies likely reflects greater autonomic dysfunction after severe HI. However, sinusoidal HR patterns and elevated vasopressin levels may have utility as biomarkers of severe HI.


Subject(s)
Heart Rate, Fetal , Hypoxia-Ischemia, Brain/physiopathology , Animals , Arterial Pressure , Electroencephalography , Hypothermia, Induced , Hypoxia-Ischemia, Brain/therapy , Sheep
5.
Sci Rep ; 8(1): 16312, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30397231

ABSTRACT

The timing of hypoxia-ischemia (HI) in preterm infants is often uncertain and there are few biomarkers to determine whether infants are in a treatable stage of injury. We evaluated whether epileptiform sharp waves recorded from the parietal cortex could provide early prediction of neuronal loss after HI. Preterm fetal sheep (0.7 gestation) underwent acute HI induced by complete umbilical cord occlusion for 25 minutes (n = 6) or sham occlusion (control, n = 6). Neuronal survival was assessed 7 days after HI by immunohistochemistry. Sharp waves were quantified manually and using a wavelet-type-2-fuzzy-logic-system during the first 4 hours of recovery. HI resulted in significant subcortical neuronal loss. Sharp waves counted by the automated classifier in the first 30 minutes after HI were associated with greater neuronal survival in the caudate nucleus (r = 0.80), whereas sharp waves between 2-4 hours after HI were associated with reduced neuronal survival (r = -0.83). Manual and automated counts were closely correlated. This study suggests that automated quantification of sharp waves may be useful for early assessment of HI injury in preterm infants. However, the pattern of evolution of sharp waves after HI was markedly affected by the severity of neuronal loss, and therefore early, continuous monitoring is essential.


Subject(s)
Electroencephalography , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/physiopathology , Neurons/pathology , Signal Processing, Computer-Assisted , Animals , Biomarkers/metabolism , Cell Count , Cell Survival , Fuzzy Logic , Hemodynamics , Hypoxia-Ischemia, Brain/metabolism , Infant, Premature , Sheep , Survival Analysis , Wavelet Analysis
6.
J Cereb Blood Flow Metab ; 38(4): 706-718, 2018 04.
Article in English | MEDLINE | ID: mdl-28387144

ABSTRACT

Antenatal glucocorticoid therapy significantly improves the short-term systemic outcomes of prematurely born infants, but there is limited information available on their impact on neurodevelopmental outcomes in at-risk preterm babies exposed to perinatal asphyxia. Preterm fetal sheep (0.7 of gestation) were exposed to a maternal injection of 12 mg dexamethasone or saline followed 4 h later by asphyxia induced by 25 min of complete umbilical cord occlusion. In a subsequent study, fetuses received titrated glucose infusions followed 4 h later by asphyxia to examine the hypothesis that hyperglycemia mediated the effects of dexamethasone. Post-mortems were performed 7 days after asphyxia for cerebral histology. Maternal dexamethasone before asphyxia was associated with severe, cystic brain injury compared to diffuse injury after saline injection, with increased numbers of seizures, worse recovery of brain activity, and increased arterial glucose levels before, during, and after asphyxia. Glucose infusions before asphyxia replicated these adverse outcomes, with a strong correlation between greater increases in glucose before asphyxia and greater neural injury. These findings strongly suggest that dexamethasone exposure and hyperglycemia can transform diffuse injury into cystic brain injury after asphyxia in preterm fetal sheep.


Subject(s)
Asphyxia Neonatorum/pathology , Dexamethasone/toxicity , Hyperglycemia/chemically induced , Hyperglycemia/pathology , Leukomalacia, Periventricular/chemically induced , Leukomalacia, Periventricular/pathology , Neurons/pathology , Animals , Brain/pathology , Electroencephalography , Female , Fetus/metabolism , Glucose/metabolism , Glucose/pharmacology , Obstetric Labor, Premature , Pregnancy , Seizures/etiology , Sheep , Umbilical Cord
7.
J Cereb Blood Flow Metab ; 37(4): 1362-1373, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27317658

ABSTRACT

Magnesium sulfate is now widely recommended for neuroprotection for preterm birth; however, this has been controversial because there is little evidence that magnesium sulfate is neuroprotective. Preterm fetal sheep (104 days gestation; term is 147 days) were randomly assigned to receive sham occlusion (n = 7), i.v. magnesium sulfate (n = 10) or saline (n = 8) starting 24 h before asphyxia until 24 h after asphyxia. Sheep were killed 72 h after asphyxia. Magnesium sulfate infusion reduced electroencephalograph power and fetal movements before asphyxia. Magnesium sulfate infusion did not affect electroencephalograph power during recovery, but was associated with marked reduction of the post-asphyxial seizure burden (mean ± SD: 34 ± 18 min vs. 107 ± 74 min, P < 0.05). Magnesium sulfate infusion did not affect subcortical neuronal loss. In the intragyral and periventricular white matter, magnesium sulfate was associated with reduced numbers of all (Olig-2+ve) oligodendrocytes in the intragyral (125 ± 23 vs. 163 ± 38 cells/field) and periventricular white matter (162 ± 39 vs. 209 ± 44 cells/field) compared to saline-treated controls ( P < 0.05), but no effect on microglial induction or astrogliosis. In conclusion, a clinically comparable dose of magnesium sulfate showed significant anticonvulsant effects after asphyxia in preterm fetal sheep, but did not reduce asphyxia-induced brain injury and exacerbated loss of oligodendrocytes.


Subject(s)
Brain/drug effects , Electroencephalography/drug effects , Fetal Hypoxia/drug therapy , Magnesium Sulfate/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Brain/embryology , Brain/pathology , Disease Models, Animal , Fetal Hypoxia/embryology , Fetal Hypoxia/pathology , Gestational Age , Magnesium Sulfate/administration & dosage , Magnesium Sulfate/blood , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/blood , Sheep
8.
J Physiol ; 594(5): 1281-93, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26077461

ABSTRACT

Magnesium sulphate is a standard therapy for eclampsia in pregnancy and is widely recommended for perinatal neuroprotection during threatened preterm labour. MgSO4 is a vasodilator and negative inotrope. Therefore the aim of this study was to investigate the effect of MgSO4 on the cardiovascular and cerebrovascular responses of the preterm fetus to asphyxia. Fetal sheep were instrumented at 98 ± 1 days of gestation (term = 147 days). At 104 days, unanaesthetised fetuses were randomly assigned to receive an intravenous infusion of MgSO4 (n = 6) or saline (n = 9). At 105 days all fetuses underwent umbilical cord occlusion for 25 min. Before occlusion, MgSO4 treatment reduced heart rate and increased femoral blood flow (FBF) and vascular conductance compared to controls. During occlusion, carotid and femoral arterial conductance and blood flows were higher in MgSO4-treated fetuses than controls. After occlusion, fetal heart rate was lower and carotid and femoral arterial conductance and blood flows were higher in MgSO4-treated fetuses than controls. Femoral arterial waveform height and width were increased during MgSO4 infusion, consistent with increased stroke volume. MgSO4 did not alter the fetal neurophysiological or nuchal electromyographic responses to asphyxia. These data demonstrate that a clinically comparable dose of MgSO4 increased FBF and stroke volume without impairing mean arterial pressure (MAP) or carotid blood flow (CaBF) during and immediately after profound asphyxia. Thus, MgSO4 may increase perfusion of peripheral vascular beds during adverse perinatal events.


Subject(s)
Adaptation, Physiological , Cerebrovascular Circulation , Coronary Circulation , Fetal Heart/physiopathology , Fetal Hypoxia/drug therapy , Magnesium Sulfate/blood , Animals , Female , Fetal Hypoxia/blood , Fetal Hypoxia/physiopathology , Hemodynamics , Magnesium Sulfate/therapeutic use , Pregnancy , Sheep
9.
Sci Rep ; 5: 16201, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26537688

ABSTRACT

Subclinical (shallow) heart rate decelerations occur during neonatal sepsis, but there is limited information on their relationship with hypotension or whether they occur before birth. We examined whether subclinical decelerations, a fall in fetal heart rate (FHR) that remained above 100 bpm, were associated with hypotension in preterm fetal sheep exposed to lipopolysaccharide (LPS). Chronically-instrumented fetal sheep at 0.7 gestation received continuous low-dose LPS infusions (n = 15, 100 ng/kg over 24 h, followed by 250 ng/kg/24 h for 96 h) or saline (n = 8). Boluses of 1 µg LPS or saline were given at 48 and 72 h. FHR variability (FHRV) was calculated, and sample asymmetry was used to assess the severity and frequency of decelerations. Low-dose LPS infusion did not affect FHR. After the first LPS bolus, 7 fetuses remained normotensive, while 8 developed hypotension (a fall in mean arterial blood pressure of ≥5 mmHg). Developing hypotension was associated with subclinical decelerations, with a corresponding increase in sample asymmetry and FHRV (p < 0.05). The second LPS bolus was associated with similar but attenuated changes in FHR and blood pressure (p < 0.05). In conclusion, subclinical decelerations are not consistently seen during prenatal exposure to LPS, but may be a useful marker of developing inflammation-related hypotension before birth.


Subject(s)
Fetus/drug effects , Fetus/physiopathology , Heart Rate, Fetal/drug effects , Hypotension/physiopathology , Lipopolysaccharides/pharmacology , Sheep/physiology , Animals , Blood Pressure/drug effects , Deceleration , Female , Pregnancy , Sepsis/physiopathology
10.
J Physiol ; 592(24): 5493-505, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25384775

ABSTRACT

Exposure to clinical doses of the glucocorticoid dexamethasone increases brain activity and causes seizures in normoxic preterm fetal sheep without causing brain injury. In contrast, the same treatment after asphyxia increased brain injury. We hypothesised that increased injury was in part mediated by a mismatch between oxygen demand and oxygen supply. In preterm fetal sheep at 0.7 gestation we measured cerebral oxygenation using near-infrared spectroscopy, electroencephalographic (EEG) activity, and carotid blood flow (CaBF) from 24 h before until 72 h after asphyxia induced by 25 min of umbilical cord occlusion. Ewes received dexamethasone intramuscularly (12 mg 3 ml(-1)) or saline 15 min after the end of asphyxia. Fetuses were studied for 3 days after occlusion. During the first 6 h of recovery after asphyxia, dexamethasone treatment was associated with a significantly greater fall in CaBF (P < 0.05), increased carotid vascular resistance (P < 0.001) and a greater fall in cerebral oxygenation as measured by the difference between oxygenated and deoxygenated haemoglobin (delta haemoglobin; P < 0.05). EEG activity was similarly suppressed in both groups. From 6 to 10 h onward, dexamethasone treatment was associated with a return of CaBF to saline control levels, increased EEG power (P < 0.005), greater epileptiform transient activity (P < 0.001), increased oxidised cytochrome oxidase (P < 0.05) and an attenuated increase in [delta haemoglobin] (P < 0.05). In conclusion, dexamethasone treatment after asphyxia is associated with greater hypoperfusion in the critical latent phase, leading to impaired intracerebral oxygenation that may exacerbate neural injury after asphyxia.


Subject(s)
Anti-Inflammatory Agents/adverse effects , Brain/drug effects , Dexamethasone/adverse effects , Fetal Hypoxia/drug therapy , Oxygen Consumption , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Brain/metabolism , Brain/physiopathology , Brain Waves , Cerebrovascular Circulation , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Female , Fetal Hypoxia/metabolism , Fetal Hypoxia/physiopathology , Pregnancy , Sheep
11.
Am J Physiol Regul Integr Comp Physiol ; 307(4): R387-95, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24944248

ABSTRACT

Perinatal exposure to infection is highly associated with adverse outcomes. Experimentally, acute, severe exposure to gram-negative bacterial lipopolysaccharide (LPS) is associated with increased fetal heart rate variability (FHRV). It is unknown whether FHRV is affected by subclinical infection with or without acute exacerbations. We therefore tested the hypothesis that FHRV would be associated with hypotension after acute on chronic exposure to LPS. Chronically instrumented fetal sheep at 0.7 gestation were exposed to a continuous low-dose LPS infusion (n = 12, 100 ng/kg over 24 h, followed by 250 ng·kg(-1)·24 h(-1) for a further 96 h) or the same volume of saline (n = 10). Boluses of either 1 µg LPS or saline were given at 48, 72, and 96 h. Low-dose infusion was not associated with hemodynamic or FHRV changes. The first LPS bolus was associated with tachycardia and suppression of nuchal electromyographic activity in all fetuses. Seven of twelve fetuses developed hypotension (a fall in mean arterial blood pressure ≥5 mmHg). FHRV was transiently increased only at the onset of hypotension, in association with increased cytokine induction and electroencephalogram suppression. FHRV then fell before the nadir of hypotension, with transient suppression of short-term FHRV. After the second LPS bolus, the hypotension group showed a biphasic pattern of a transient increase in FHRV followed by more prolonged suppression. These findings suggest that infection-related hypotension in the preterm fetus mediates the transient increase in FHRV and that repeated exposure to LPS leads to progressive loss of FHRV.


Subject(s)
Bradycardia/physiopathology , Fetal Heart/physiopathology , Heart Rate, Fetal , Hypotension/physiopathology , Lipopolysaccharides , Sepsis/physiopathology , Tachycardia/physiopathology , Animals , Arterial Pressure , Biomarkers/blood , Blood Gas Analysis , Blood Glucose/metabolism , Bradycardia/chemically induced , Bradycardia/diagnosis , Circadian Rhythm , Disease Models, Animal , Electrocardiography , Electroencephalography , Electromyography , Female , Fetal Blood/metabolism , Fetal Monitoring/methods , Gestational Age , Hydrogen-Ion Concentration , Hypotension/chemically induced , Hypotension/diagnosis , Lactic Acid/blood , Pregnancy , Sepsis/chemically induced , Sepsis/diagnosis , Sheep , Tachycardia/chemically induced , Tachycardia/diagnosis , Time Factors
12.
PLoS One ; 9(5): e96558, 2014.
Article in English | MEDLINE | ID: mdl-24865217

ABSTRACT

Asphyxia around the time of preterm birth is associated with neurodevelopmental disability. In this study, we tested the hypothesis that blockade of connexin hemichannels would improve recovery of brain activity and reduce cell loss after asphyxia in preterm fetal sheep. Asphyxia was induced by 25 min of complete umbilical cord occlusion in preterm fetal sheep (103-104 d gestational age). Connexin hemichannels were blocked by intracerebroventricular infusion of mimetic peptide starting 90 min after asphyxia at a concentration of 50 µM/h for one hour followed by 50 µM/24 hour for 24 hours (occlusion-peptide group, n = 6) or vehicle infusion for controls (occlusion-vehicle group, n = 7). Peptide infusion was associated with earlier recovery of electroencephalographic power after asphyxia compared to occlusion-vehicle (p<0.05), with reduced neuronal loss in the caudate and putamen (p<0.05), but not in the hippocampus. In the intragyral and periventricular white matter, peptide administration was associated with an increase in total oligodendrocyte numbers (p<0.05) and immature/mature oligodendrocytes compared to occlusion-vehicle (p<0.05), with a significant increase in proliferation (p<0.05). Connexin hemichannel blockade was neuroprotective and reduced oligodendrocyte death and improved recovery of oligodendrocyte maturation in preterm fetuses after asphyxia.


Subject(s)
Asphyxia/complications , Connexins/antagonists & inhibitors , Fetal Hypoxia/complications , Fetus/physiopathology , Hypoxia-Ischemia, Brain/drug therapy , Neuroprotective Agents/therapeutic use , Oligopeptides/therapeutic use , Premature Birth/physiopathology , Animals , Asphyxia/physiopathology , Electroencephalography , Female , Fetal Hypoxia/physiopathology , Fetus/surgery , Gestational Age , Hypoxia-Ischemia, Brain/etiology , Hypoxia-Ischemia, Brain/metabolism , Immunoenzyme Techniques , Pregnancy , Sheep
13.
PLoS One ; 9(5): e96530, 2014.
Article in English | MEDLINE | ID: mdl-24797081

ABSTRACT

The majority of pre-clinical studies of hypoxic-ischemic encephalopathy at term-equivalent have focused on either relatively mild insults, or on functional paradigms of cerebral ischemia or hypoxia-ischemia/hypotension. There is surprisingly little information on the responses to single, severe 'physiological' insults. In this study we examined the evolution and pattern of neural injury after prolonged umbilical cord occlusion (UCO). 36 chronically instrumented fetal sheep at 125-129 days gestational age (term = 147 days) were subjected to either UCO until mean arterial pressure was < = 8 mmHg (n = 29), or sham occlusion (n = 7). Surviving fetuses were killed after 72 hours for histopathologic assessment with acid-fuchsin thionine. After UCO, 11 fetuses died with intractable hypotension and 5 ewes entered labor and were euthanized. The remaining 13 fetuses showed marked EEG suppression followed by evolving seizures starting at 5.8 (6.8) hours (median (interquartile range)). 6 of 13 developed status epilepticus, which was associated with a transient secondary increase in cortical impedance (a measure of cytotoxic edema, p<0.05). All fetuses showed moderate to severe neuronal loss in the hippocampus and the basal ganglia but mild cortical cell loss (p<0.05 vs sham occlusion). Status epilepticus was associated with more severe terminal hypotension (p<0.05) and subsequently, greater neuronal loss (p<0.05). In conclusion, profound UCO in term-equivalent fetal sheep was associated with delayed seizures, secondary cytotoxic edema, and subcortical injury, consistent with the predominant pattern after peripartum sentinel events at term. It is unclear whether status epilepticus exacerbated cortical injury or was simply a reflection of a longer duration of asphyxia.


Subject(s)
Neurons/pathology , Status Epilepticus/pathology , Umbilical Cord/pathology , Animals , Female , Fetal Hypoxia/physiopathology , Hippocampus/pathology , Hypotension/pathology , Hypoxia-Ischemia, Brain/pathology , Seizures , Sheep
14.
Neuropharmacology ; 83: 62-70, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24726307

ABSTRACT

Basal ganglia injury after hypoxia-ischemia remains common in preterm infants, and is closely associated with later cerebral palsy. In the present study we tested the hypothesis that a highly selective neuronal nitric oxide synthase (nNOS) inhibitor, JI-10, would improve survival of striatal phenotypic neurons after profound asphyxia, and that the subsequent seizure burden and recovery of EEG are associated with neural outcome. 24 chronically instrumented preterm fetal sheep were randomized to either JI-10 (3 ml of 0.022 mg/ml, n = 8) or saline (n = 8) infusion 15 min before 25 min complete umbilical cord occlusion, or saline plus sham-occlusion (n = 8). Umbilical cord occlusion was associated with reduced numbers of calbindin-28k-, GAD-, NPY-, PV-, Calretinin- and nNOS-positive striatal neurons (p < 0.05 vs. sham occlusion) but not ChAT-positive neurons. JI-10 was associated with increased numbers of calbindin-28k-, GAD-, nNOS-, NPY-, PV-, Calretinin- and ChAT-positive striatal neurons (p < 0.05 vs. saline + occlusion). Seizure burden was strongly associated with loss of calbindin-positive cells (p < 0.05), greater seizure amplitude was associated with loss of GAD-positive cells (p < 0.05), and with more activated microglia in the white matter tracts (p < 0.05). There was no relationship between EEG power after 7 days recovery and total striatal cell loss, but better survival of NPY-positive neurons was associated with lower EEG power. In summary, these findings suggest that selective nNOS inhibition during asphyxia is associated with protection of phenotypic striatal projection neurons and has potential to help reduce basal ganglia injury in some premature babies.


Subject(s)
Aminopyridines/therapeutic use , Asphyxia/complications , Corpus Striatum/drug effects , Enzyme Inhibitors/therapeutic use , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Nitric Oxide Synthase Type I/antagonists & inhibitors , Seizures/drug therapy , Animals , Asphyxia/physiopathology , Corpus Striatum/pathology , Corpus Striatum/physiopathology , Female , Fetal Hypoxia/physiopathology , Fetus , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/physiopathology , Neurons/pathology , Phenotype , Pregnancy , Premature Birth , Seizures/physiopathology , Seizures/radiotherapy , Sheep
15.
Obstet Gynecol Int ; 2014: 314159, 2014.
Article in English | MEDLINE | ID: mdl-24693290

ABSTRACT

T/QRS ratio monitoring is used to help identify fetal asphyxia. However, immature animals have greater capacity to maintain blood pressure during severe asphyxia, raising the possibility that they may show an attenuated T/QRS increase during asphyxia. Chronically instrumented fetal sheep at 0.6 of gestation (0.6 GA; n = 12), 0.7 GA (n = 12), and 0.8 GA (n = 8) underwent complete umbilical cord occlusion for 30 min, 25 min, or 15 min, respectively. Cord occlusion was associated with progressive metabolic acidosis and initial hypertension followed by severe hypotension, with a more rapid fall in mean arterial blood pressure (MAP) and carotid blood flow (CaBF) with advancing gestation. T/QRS ratio rose after occlusion more rapidly at 0.8 GA than in immature fetuses, to a similar final peak at all ages, followed by a progressive fall that was slower at 0.8 GA than in the immature fetuses. The increase in T/QRS ratio correlated with initial hypertension at 0.8 GA (P < 0.05, R (2) = 0.38), and conversely, its fall correlated closely with falling MAP in all gestational groups (P < 0.01, R (2) = 0.67). In conclusion, elevation of the T/QRS ratio is an index of onset of severe asphyxia in the last third of gestation, but not of fetal compromise.

16.
Front Neurosci ; 8: 40, 2014.
Article in English | MEDLINE | ID: mdl-24578682

ABSTRACT

Acute post-asphyxial encephalopathy occurring around the time of birth remains a major cause of death and disability. The recent seminal insight that allows active neuroprotective treatment is that even after profound asphyxia (the "primary" phase), many brain cells show initial recovery from the insult during a short "latent" phase, typically lasting approximately 6 h, only to die hours to days later after a "secondary" deterioration characterized by seizures, cytotoxic edema, and progressive failure of cerebral oxidative metabolism. Although many of these secondary processes are potentially injurious, they appear to be primarily epiphenomena of the "execution" phase of cell death. Animal and human studies designed around this conceptual framework have shown that moderate cerebral hypothermia initiated as early as possible but before the onset of secondary deterioration, and continued for a sufficient duration to allow the secondary deterioration to resolve, has been associated with potent, long-lasting neuroprotection. Recent clinical trials show that while therapeutic hypothermia significantly reduces morbidity and mortality, many babies still die or survive with disabilities. The challenge for the future is to find ways of improving the effectiveness of treatment. In this review, we will dissect the known mechanisms of hypoxic-ischemic brain injury in relation to the known effects of hypothermic neuroprotection.

17.
Clin Perinatol ; 41(1): 161-75, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24524453

ABSTRACT

Prolonged, moderate cerebral hypothermia initiated within a few hours after severe hypoxia-ischemia and continued until resolution of the acute phase of delayed cell death can reduce acute brain injury and improve long-term behavioral recovery in term infants and in adults after cardiac arrest. The specific mechanisms of hypothermic neuroprotection remain unclear, in part because hypothermia suppresses a broad range of potential injurious factors. This article examines proposed mechanisms in relation to the known window of opportunity for effective protection with hypothermia. Knowledge of the mechanisms of hypothermia will help guide the rational development of future combination treatments to augment neuroprotection with hypothermia and identify those most likely to benefit.


Subject(s)
Hypothermia, Induced/methods , Hypoxia-Ischemia, Brain/therapy , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/physiopathology , Disease Models, Animal , Excitatory Amino Acids/metabolism , Free Radicals/metabolism , Humans , Hypothermia/metabolism , Hypoxia-Ischemia, Brain/metabolism , Infant, Newborn , Time-to-Treatment
18.
J Cereb Blood Flow Metab ; 34(1): 126-35, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24103904

ABSTRACT

Melatonin is a naturally occurring indolamine with mild antioxidant properties that is neuroprotective in perinatal animals. There is limited information on its effects on preterm brain injury. In this study, 23 chronically instrumented fetal sheep received 25 minutes of complete umbilical cord occlusion at 101 to 104 days gestation (term is 147 days). Melatonin was administered to the ewe 15 minutes before occlusion (0.1 mg/kg bolus followed by 0.1 mg/kg per hour for 6 hours, n=8), or the equivalent volume of vehicle (2% ethanol, n=7), or saline (n=8), or maternal saline plus sham occlusion (n=8). Sheep were killed after 7 days recovery in utero. Fetal blood pressure, heart rate, nuchal activity, and temperature were similar between groups. Vehicle infusion was associated with improved neuronal survival in the caudate nucleus, but greater neuronal loss in the regions of the hippocampus, with reduced proliferation and increased ameboid microglia in the white matter (P<0.05). Maternal melatonin infusion was associated with faster recovery of fetal EEG, prolonged reduction in carotid blood flow, similar neuronal survival to vehicle, improved numbers of mature oligodendrocytes, and reduced microglial activation in the white matter (P<0.05). Prophylactic maternal melatonin treatment is partially protective but its effects may be partly confounded by ethanol used to dissolve melatonin.


Subject(s)
Asphyxia Neonatorum/prevention & control , Brain/drug effects , Fetal Hypoxia/pathology , Melatonin/therapeutic use , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Animals , Brain/embryology , Brain/pathology , Dose-Response Relationship, Drug , Drug Carriers/adverse effects , Drug Carriers/chemistry , Ethanol/adverse effects , Ethanol/chemistry , Female , Gestational Age , Maternal-Fetal Exchange , Melatonin/administration & dosage , Melatonin/pharmacokinetics , Neurons/pathology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacokinetics , Pregnancy , Premedication , Sheep/embryology
19.
PLoS One ; 8(10): e77480, 2013.
Article in English | MEDLINE | ID: mdl-24204840

ABSTRACT

BACKGROUND AND PURPOSE: Maternal glucocorticoid treatment for threatened premature delivery dramatically improves neonatal survival and short-term morbidity; however, its effects on neurodevelopmental outcome are variable. We investigated the effect of maternal glucocorticoid exposure after acute asphyxia on injury in the preterm brain. METHODS: Chronically instrumented singleton fetal sheep at 0.7 of gestation received asphyxia induced by complete umbilical cord occlusion for 25 minutes. 15 minutes after release of occlusion, ewes received a 3 ml i.m. injection of either dexamethasone (12 mg, n = 10) or saline (n = 10). Sheep were killed after 7 days recovery; survival of neurons in the hippocampus and basal ganglia, and oligodendrocytes in periventricular white matter were assessed using an unbiased stereological approach. RESULTS: Maternal dexamethasone after asphyxia was associated with more severe loss of neurons in the hippocampus (CA3 regions, 290 ± 76 vs 484 ± 98 neurons/mm(2), mean ± SEM, P<0.05) and basal ganglia (putamen, 538 ± 112 vs 814 ± 34 neurons/mm(2), P<0.05) compared to asphyxia-saline, and with greater loss of both total (913 ± 77 vs 1201 ± 75/mm(2), P<0.05) and immature/mature myelinating oligodendrocytes in periventricular white matter (66 ± 8 vs 114 ± 12/mm(2), P<0.05, vs sham controls 165 ± 10/mm(2), P<0.001). This was associated with transient hyperglycemia (peak 3.5 ± 0.2 vs. 1.4 ± 0.2 mmol/L at 6 h, P<0.05) and reduced suppression of EEG power in the first 24 h after occlusion (maximum -1.5 ± 1.2 dB vs. -5.0 ± 1.4 dB in saline controls, P<0.01), but later onset and fewer overt seizures. CONCLUSIONS: In preterm fetal sheep, exposure to maternal dexamethasone during recovery from asphyxia exacerbated brain damage.


Subject(s)
Anti-Inflammatory Agents/adverse effects , Asphyxia/pathology , Dexamethasone/adverse effects , Neurons/drug effects , Oligodendroglia/drug effects , Animals , Asphyxia/drug therapy , Basal Ganglia/drug effects , Basal Ganglia/pathology , Cell Count , Cell Death , Cerebral Ventricles/drug effects , Cerebral Ventricles/pathology , Electroencephalography , Female , Fetus , Hippocampus/drug effects , Hippocampus/pathology , Injections, Intramuscular , Neurons/pathology , Oligodendroglia/pathology , Pregnancy , Premature Birth/pathology , Sheep , Sheep, Domestic , Umbilical Cord/pathology
20.
Exp Neurol ; 250: 282-92, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24120436

ABSTRACT

Preterm brain injury is partly associated with hypoxia-ischemia starting before birth. Excessive nitric oxide production during HI may cause nitrosative stress, leading to cell membrane and mitochondrial damage. We therefore tested the hypothesis that therapy with a new, selective neuronal nitric oxide synthase (nNOS) inhibitor, JI-10 (0.022mg/kg bolus, n=8), given 30min before 25min of complete umbilical cord occlusion was protective in preterm fetal sheep at 101-104day gestation (term is 147days), compared to saline (n=8). JI-10 had no effect on fetal blood pressure, heart rate, carotid and femoral blood flow, total EEG power, nuchal activity, temperature or intracerebral oxygenation on near-infrared spectroscopy during or after occlusion. JI-10 was associated with later onset of post-asphyxial seizures compared with saline (p<0.05), and attenuation of the subsequent progressive loss of cytochrome oxidase (p<0.05). After 7days recovery, JI-10 was associated with improved neuronal survival in the caudate nucleus (p<0.05), but not the putamen or hippocampus, and more CNPase positive oligodendrocytes in the periventricular white matter (p<0.05). In conclusion, prophylactic nNOS inhibition before profound asphyxia was associated with delayed onset of seizures, slower decline of cytochrome oxidase and partial white and gray matter protection, consistent with protection of mitochondrial function.


Subject(s)
Enzyme Inhibitors/pharmacology , Fetal Hypoxia/complications , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type I/antagonists & inhibitors , Prenatal Exposure Delayed Effects/prevention & control , Animals , Cerebral Palsy/etiology , Cerebral Palsy/prevention & control , Disease Models, Animal , Female , Fetal Hypoxia/enzymology , Fetus , Hypoxia-Ischemia, Brain/physiopathology , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Seizures/etiology , Seizures/prevention & control , Sheep
SELECTION OF CITATIONS
SEARCH DETAIL
...