Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 35(2): e21338, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33428278

RESUMO

Pregnancy complications associated with prenatal hypoxia lead to increased placental oxidative stress. Previous studies suggest that prenatal hypoxia can reduce mitochondrial respiratory capacity and mitochondrial fusion, which could lead to placental dysfunction and impaired fetal development. We developed a placenta-targeted treatment strategy using a mitochondrial antioxidant, MitoQ, encapsulated into nanoparticles (nMitoQ) to reduce placental oxidative stress and (indirectly) improve fetal outcomes. We hypothesized that, in a rat model of prenatal hypoxia, nMitoQ improves placental mitochondrial function and promotes mitochondrial fusion in both male and female placentae. Pregnant rats were treated with saline or nMitoQ on gestational day (GD) 15 and exposed to normoxia (21% O2 ) or hypoxia (11% O2 ) from GD15-21. On GD21, male and female placental labyrinth zones were collected for mitochondrial respirometry assessments, mitochondrial content, and markers of mitochondrial biogenesis, fusion and fission. Prenatal hypoxia reduced complex IV activity and fusion in male placentae, while nMitoQ improved complex IV activity in hypoxic male placentae. In female placentae, prenatal hypoxia decreased respiration through the S-pathway (complex II) and increased N-pathway (complex I) respiration, while nMitoQ increased fusion in hypoxic female placentae. No changes in mitochondrial content, biogenesis or fission were found. In conclusion, nMitoQ improved placental mitochondrial function in male and female placentae from fetuses exposed to prenatal hypoxia, which may contribute to improved placental function. However, the mechanisms (ie, changes in mitochondrial respiratory capacity and mitochondrial fusion) were distinct between the sexes. Treatment strategies targeted against placental oxidative stress could improve placental mitochondrial function in complicated pregnancies.


Assuntos
Antioxidantes/uso terapêutico , Hipóxia Fetal/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Nanopartículas/química , Compostos Organofosforados/uso terapêutico , Placenta/efeitos dos fármacos , Ubiquinona/análogos & derivados , Animais , Antioxidantes/administração & dosagem , Antioxidantes/farmacologia , Respiração Celular , Feminino , Masculino , Mitocôndrias/metabolismo , Dinâmica Mitocondrial , Compostos Organofosforados/administração & dosagem , Compostos Organofosforados/farmacologia , Placenta/metabolismo , Gravidez , Ratos , Ratos Sprague-Dawley , Fatores Sexuais , Ubiquinona/administração & dosagem , Ubiquinona/farmacologia , Ubiquinona/uso terapêutico
2.
J Physiol ; 596(23): 6079-6092, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29572829

RESUMO

KEY POINTS: We evaluated the effect of magnesium sulphate (MgSO4 ) on seizures induced by asphyxia in preterm fetal sheep. MgSO4 did not prevent seizures, but significantly reduced the total duration, number of seizures, seizure amplitude and average seizure burden. Saline-asphyxia male fetuses had significantly more seizures than female fetuses, but male fetuses showed significantly greater reduction in seizures during MgSO4 infusion than female fetuses. A circadian profile of seizure activity was observed in all fetuses, with peak seizures seen around 04.00-06.00 h on the first and second days after the end of asphyxia. This study is the first to demonstrate that MgSO4 has utility as an anti-seizure agent after hypoxia-ischaemia. More information is needed about the mechanisms mediating the effect of MgSO4 on seizures and sexual dimorphism, and the influence of circadian rhythms on seizure expression. ABSTRACT: Seizures are common in newborns after asphyxia at birth and are often refractory to anti-seizure agents. Magnesium sulphate (MgSO4 ) has anticonvulsant effects and is increasingly given to women in preterm labour for potential neuroprotection. There is limited information on its effects on perinatal seizures. We examined the hypothesis that MgSO4 infusion would reduce fetal seizures after asphyxia in utero. Preterm fetal sheep at 0.7 gestation (104 days, term = 147 days) were given intravenous infusions of either saline (n = 14) or MgSO4 (n = 12, 160 mg bolus + 48 mg h-1 infusion over 48 h). Fetuses underwent umbilical cord occlusion (UCO) for 25 min, 24 h after the start of infusion. The start time for seizures did not differ between groups, but MgSO4 significantly reduced the total number of seizures (P < 0.001), peak seizure amplitude (P < 0.05) and seizure burden (P < 0.005). Within the saline-asphyxia group, male fetuses had significantly more seizures than females (P < 0.05). Within the MgSO4 -asphyxia group, although both sexes had fewer seizures than the saline-asphyxia group, the greatest effect of MgSO4 was on male fetuses, with reduced numbers of seizures (P < 0.001) and seizure burden (P < 0.005). Only 1 out of 6 MgSO4 males had seizures on the second day post-UCO compared to 5 out of 6 MgSO4 female fetuses (P = 0.08). Finally, seizures showed a circadian profile with peak seizures between 04.00 and 06.00 h on the first and second day post-UCO. Collectively, these results suggest that MgSO4 may have utility in treating perinatal seizures and has sexually dimorphic effects.


Assuntos
Hipóxia Fetal/tratamento farmacológico , Sulfato de Magnésio/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Convulsões/tratamento farmacológico , Animais , Asfixia/tratamento farmacológico , Feminino , Feto/efeitos dos fármacos , Isquemia/tratamento farmacológico , Masculino , Fatores Sexuais , Ovinos , Fatores de Tempo , Cordão Umbilical/irrigação sanguínea
3.
Pediatr Res ; 84(3): 442-450, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29976968

RESUMO

BACKGROUND: Impaired cerebral autoregulation in preterm infants makes circulatory management important to avoid cerebral hypoxic-ischemic injury. Dobutamine is frequently used as inotropic treatment in preterm neonates, but its effects on the brain exposed to cerebral hypoxia are unknown. We hypothesized that dobutamine would protect the immature brain from cerebral hypoxic injury. METHODS: In preterm (0.6 gestation) fetal sheep, dobutamine (Dob, 10 µg/kg/min) or saline (Sal) was infused intravenously for 74 h. Two hours after the beginning of the infusion, umbilical cord occlusion (UCO) was performed to produce fetal asphyxia (Sal+UCO: n = 9, Dob+UCO: n = 7), or sham occlusion (Sal+sham: n = 7, Dob+sham: n = 6) was performed. Brains were collected 72 h later for neuropathology. RESULTS: Dobutamine did not induce cerebral changes in the sham UCO group. UCO increased apoptosis and microglia density in white matter, hippocampus, and caudate nucleus, and astrocyte density in the caudate nucleus. Dobutamine commenced before UCO reduced microglia infiltration in the white matter, and microglial and astrocyte density in the caudate. CONCLUSION: In preterm hypoxia-induced brain injury, dobutamine decreases neuroinflammation in the white matter and caudate, and reduces astrogliosis in the caudate. Early administration of dobutamine in preterm infants for cardiovascular stabilization appears safe and may be neuroprotective against unforeseeable cerebral hypoxic injury.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/embriologia , Dobutamina/uso terapêutico , Hipóxia Fetal/tratamento farmacológico , Hipóxia-Isquemia Encefálica/patologia , Inflamação/tratamento farmacológico , Animais , Asfixia Neonatal/patologia , Gasometria , Peso Corporal , Modelos Animais de Doenças , Dopamina/farmacologia , Eletrocardiografia , Feminino , Frequência Cardíaca , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Inflamação/patologia , Microglia , Neurônios , Tamanho do Órgão , Estresse Oxidativo , Gravidez , Prenhez , Ovinos
4.
Pharmacol Res ; 134: 332-342, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29778808

RESUMO

Intrauterine growth restriction, a common consequence of prenatal hypoxia, is a leading cause of fetal morbidity and mortality with a significant impact on population health. Hypoxia may increase placental oxidative stress and lead to an abnormal release of placental-derived factors, which are emerging as potential contributors to developmental programming. Nanoparticle-linked drugs are emerging as a novel method to deliver therapeutics targeted to the placenta and avoid risking direct exposure to the fetus. We hypothesize that placental treatment with antioxidant MitoQ loaded onto nanoparticles (nMitoQ) will prevent the development of cardiovascular disease in offspring exposed to prenatal hypoxia. Pregnant rats were intravenously injected with saline or nMitoQ (125 µM) on gestational day (GD) 15 and exposed to either normoxia (21% O2) or hypoxia (11% O2) from GD15-21 (term: 22 days). In one set of animals, rats were euthanized on GD 21 to assess fetal body weight, placental weight and placental oxidative stress. In another set of animals, dams were allowed to give birth under normal atmospheric conditions (term: GD 22) and male and female offspring were assessed at 7 and 13 months of age for in vivo cardiac function (echocardiography) and vascular function (wire myography, mesenteric artery). Hypoxia increased oxidative stress in placentas of male and female fetuses, which was prevented by nMitoQ. 7-month-old male and female offspring exposed to prenatal hypoxia demonstrated cardiac diastolic dysfunction, of which nMitoQ improved only in 7-month-old female offspring. Vascular sensitivity to methacholine was reduced in 13-month-old female offspring exposed to prenatal hypoxia, while nMitoQ treatment improved vasorelaxation in both control and hypoxia exposed female offspring. Male 13-month-old offspring exposed to hypoxia showed an age-related decrease in vascular sensitivity to phenylephrine, which was prevented by nMitoQ. In summary, placental-targeted MitoQ treatment in utero has beneficial sex- and age-dependent effects on adult offspring cardiovascular function.


Assuntos
Antioxidantes/administração & dosagem , Doenças Cardiovasculares/prevenção & controle , Hipóxia Fetal/tratamento farmacológico , Compostos Organofosforados/administração & dosagem , Estresse Oxidativo/efeitos dos fármacos , Placenta/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal , Ubiquinona/análogos & derivados , Fatores Etários , Animais , Doenças Cardiovasculares/metabolismo , Doenças Cardiovasculares/fisiopatologia , Modelos Animais de Doenças , Feminino , Hipóxia Fetal/metabolismo , Hipóxia Fetal/fisiopatologia , Idade Gestacional , Hemodinâmica/efeitos dos fármacos , Masculino , Exposição Materna , Contração Miocárdica/efeitos dos fármacos , Nanopartículas , Placenta/metabolismo , Placenta/fisiopatologia , Gravidez , Ratos Sprague-Dawley , Fatores Sexuais , Ubiquinona/administração & dosagem , Função Ventricular Esquerda/efeitos dos fármacos
5.
Bull Exp Biol Med ; 163(5): 594-598, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28948549

RESUMO

In 60-day-old male rats after antenatal hypoxia, the body weight and the absolute weight of the cerebral hemispheres were significantly lower than in the progeny of intact animals. Analysis of brain sections stained with silver nitrate revealed reduced number of nucleoli in neocortical layer II and hippocampal CA1 neurons and smaller area of neuronal nuclei in neocortical layer V and total area of nucleoli in neurons of all studied zones. The animals demonstrated increased locomotor activity in the elevated plus-maze test. Chemiluminescent analysis of brain homogenates revealed the presence of oxidative stress at the organ level. Neonatal administration of non-opioid analog of leu-enkephalin (peptide NALE) after antenatal hypoxia normalized body weight, neutralized morphometric changes in the nucleoli and nuclei of neurons in the neocortex and hippocampus, and improved oxidative status of the brain. In 30- and 60-day-old male rats subjected to antenatal hypoxia and receiving peptide NALE during the neonatal period, behavioral responses were partially normalized. Non-opioid analog of leu-enkephalin can be a promising drug for correction of cerebral consequences of antenatal hypoxia.


Assuntos
Encefalina Leucina/uso terapêutico , Hipóxia Fetal/tratamento farmacológico , Hipocampo/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Feminino , Hipóxia Fetal/metabolismo , Hipocampo/metabolismo , Masculino , Atividade Motora/efeitos dos fármacos , Neocórtex/efeitos dos fármacos , Neocórtex/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Gravidez , Ratos , Ratos Wistar
6.
J Physiol ; 594(5): 1281-93, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26077461

RESUMO

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.


Assuntos
Adaptação Fisiológica , Circulação Cerebrovascular , Circulação Coronária , Coração Fetal/fisiopatologia , Hipóxia Fetal/tratamento farmacológico , Sulfato de Magnésio/sangue , Animais , Feminino , Hipóxia Fetal/sangue , Hipóxia Fetal/fisiopatologia , Hemodinâmica , Sulfato de Magnésio/uso terapêutico , Gravidez , Ovinos
7.
J Physiol ; 594(5): 1295-310, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26497972

RESUMO

Acute fetal hypoxia is a form of fetal stress that stimulates renal vasoconstriction and ischaemia as a consequence of the physiological redistribution of combined ventricular output. Because of the potential ischaemia-reperfusion injury to the kidney, we hypothesized that it would respond to hypoxia with an increase in the expression of inflammatory genes, and that ketamine (an N-methyl-D-aspartate receptor antagonist) would reduce or block this response. Hypoxia was induced for 30 min in chronically catheterized fetal sheep (125 ± 3 days), with or without ketamine (3 mg kg(-1)) administered intravenously to the fetus 10 min prior to hypoxia. Gene expression in fetal kidney cortex collected 24 h after the onset of hypoxia was analysed using ovine Agilent 15.5k array and validated with qPCR and immunohistochemistry in four groups of ewes: normoxic control, normoxia + ketamine, hypoxic control and hypoxia + ketamine (n = 3-4 per group). Significant differences in gene expression between groups were determined with t-statistics using the limma package for R (P ≤ 0.05). Enriched biological processes for the 427 upregulated genes were immune and inflammatory responses and for the 946 downregulated genes were metabolic processes. Ketamine countered the effects of hypoxia on upregulated immune/inflammatory responses as well as the downregulated metabolic responses. We conclude that our transcriptomics modelling predicts that hypoxia activates inflammatory pathways and reduces metabolism in the fetal kidney cortex, and ketamine blocks or ameliorates this response. The results suggest that ketamine may have therapeutic potential for protection from ischaemic renal damage.


Assuntos
Antagonistas de Aminoácidos Excitatórios/uso terapêutico , Hipóxia Fetal/tratamento farmacológico , Ketamina/uso terapêutico , Rim/fisiopatologia , Animais , Quimiocinas/genética , Quimiocinas/metabolismo , Feminino , Inflamação/tratamento farmacológico , Interleucinas/genética , Interleucinas/metabolismo , Rim/irrigação sanguínea , Rim/metabolismo , Gravidez , Ovinos
8.
J Physiol ; 594(5): 1399-420, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26537782

RESUMO

Inhibition of hypoxia signalling leads to respiratory distress syndrome (RDS), whereas administration of vascular endothelial growth factor (VEGF), the most widely characterized hypoxia responsive factor, protects from RDS. In the lung of the chronically hypoxaemic placentally restricted (PR) fetus, there is altered regulation of hypoxia signalling. This leads to reduced surfactant maturation in late gestation and provides evidence for the increased risk of RDS in growth restricted neonates at birth. We evaluated the effect of recombinant human VEGF administration with respect to bypassing the endogenous regulation of hypoxia signalling in the lung of the normally grown and PR sheep fetus. There was no effect of VEGF administration on fetal blood pressure or fetal breathing movements. We examined the effect on the expression of genes regulating VEGF signalling (FLT1 and KDR), angiogenesis (ANGPT1, AQP1, ADM), alveolarization (MMP2, MMP9, TIMP1, COL1A1, ELN), proliferation (IGF1, IGF2, IGF1R, MKI67, PCNA), inflammation (CCL2, CCL4, IL1B, TNFA, TGFB1, IL10) and surfactant maturation (SFTP-A, SFTP-B, SFTP-C, SFTP-D, PCYT1A, LPCAT, LAMP3, ABCA3). Despite the effects of PR on the expression of genes regulating airway remodelling, inflammatory signalling and surfactant maturation, there were very few effects of VEGF administration on gene expression in the lung of both the normally grown and PR fetus. There were, however, positive effects of VEGF administration on percentage tissue, air space and numerical density of SFTP-B positive alveolar epithelial cells in fetal lung tissue. These results provide evidence for the stimulatory effects of VEGF administration on structural maturation in the lung of both the normally grown and PR fetus.


Assuntos
Hipóxia Fetal/tratamento farmacológico , Maturidade dos Órgãos Fetais , Pulmão/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Feminino , Hipóxia Fetal/metabolismo , Hipóxia Fetal/patologia , Pulmão/embriologia , Pulmão/metabolismo , Neovascularização Fisiológica , Gravidez , Síndrome do Desconforto Respiratório do Recém-Nascido/prevenção & controle , Ovinos , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/administração & dosagem , Fator A de Crescimento do Endotélio Vascular/uso terapêutico
9.
Neurobiol Dis ; 73: 118-29, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25258170

RESUMO

We have previously shown that increases in blood-brain barrier permeability represent an important component of ischemia-reperfusion related brain injury in the fetus. Pro-inflammatory cytokines could contribute to these abnormalities in blood-brain barrier function. We have generated pharmacological quantities of mouse anti-ovine interleukin-1ß monoclonal antibody and shown that this antibody has very high sensitivity and specificity for interleukin-1ß protein. This antibody also neutralizes the effects of interleukin-1ß protein in vitro. In the current study, we hypothesized that the neutralizing anti-interleukin-1ß monoclonal antibody attenuates ischemia-reperfusion related fetal blood-brain barrier dysfunction. Instrumented ovine fetuses at 127 days of gestation were studied after 30 min of carotid occlusion and 24h of reperfusion. Groups were sham operated placebo-control- (n=5), ischemia-placebo- (n=6), ischemia-anti-IL-1ß antibody- (n=7), and sham-control antibody- (n=2) treated animals. Systemic infusions of placebo (0.154M NaCl) or anti-interleukin-1ß monoclonal antibody (5.1±0.6 mg/kg) were given intravenously to the same sham or ischemic group of fetuses at 15 min and 4h after ischemia. Concentrations of interleukin-1ß protein and anti-interleukin-1ß monoclonal antibody were measured by ELISA in fetal plasma, cerebrospinal fluid, and parietal cerebral cortex. Blood-brain barrier permeability was quantified using the blood-to-brain transfer constant (Ki) with α-aminoisobutyric acid in multiple brain regions. Interleukin-1ß protein was also measured in parietal cerebral cortices and tight junction proteins in multiple brain regions by Western immunoblot. Cerebral cortical interleukin-1ß protein increased (P<0.001) after ischemia-reperfusion. After anti-interleukin-1ß monoclonal antibody infusions, plasma anti-interleukin-1ß monoclonal antibody was elevated (P<0.001), brain anti-interleukin-1ß monoclonal antibody levels were higher (P<0.03), and interleukin-1ß protein concentrations (P<0.03) and protein expressions (P<0.001) were lower in the monoclonal antibody-treated group than in placebo-treated-ischemia-reperfusion group. Monoclonal antibody infusions attenuated ischemia-reperfusion-related increases in Ki across the brain regions (P<0.04), and Ki showed an inverse linear correlation (r= -0.65, P<0.02) with anti-interleukin-1ß monoclonal antibody concentrations in the parietal cortex, but had little effect on tight junction protein expression. We conclude that systemic anti-interleukin-1ß monoclonal antibody infusions after ischemia result in brain anti-interleukin-1ß antibody uptake, and attenuate ischemia-reperfusion-related interleukin-1ß protein up-regulation and increases in blood-brain barrier permeability across brain regions in the fetus. The pro-inflammatory cytokine, interleukin-1ß, contributes to impaired blood-brain barrier function after ischemia in the fetus.


Assuntos
Anticorpos Neutralizantes/uso terapêutico , Barreira Hematoencefálica/efeitos dos fármacos , Hipóxia Fetal/tratamento farmacológico , Hipóxia Fetal/patologia , Interleucina-1beta/imunologia , Animais , Anticorpos Neutralizantes/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Barreira Hematoencefálica/fisiopatologia , Encéfalo/embriologia , Encéfalo/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Estenose das Carótidas/complicações , Citocinas/metabolismo , Modelos Animais de Doenças , Embrião de Mamíferos , Ensaio de Imunoadsorção Enzimática , Feminino , Hipóxia Fetal/etiologia , Frequência Cardíaca Fetal/efeitos dos fármacos , Interleucina-1beta/metabolismo , Camundongos , Gravidez , Fluxo Sanguíneo Regional/efeitos dos fármacos , Ovinos , Proteínas de Junções Íntimas/metabolismo
10.
J Physiol ; 592(24): 5493-505, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25384775

RESUMO

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.


Assuntos
Anti-Inflamatórios/efeitos adversos , Encéfalo/efeitos dos fármacos , Dexametasona/efeitos adversos , Hipóxia Fetal/tratamento farmacológico , Consumo de Oxigênio , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Ondas Encefálicas , Circulação Cerebrovascular , Dexametasona/farmacologia , Dexametasona/uso terapêutico , Feminino , Hipóxia Fetal/metabolismo , Hipóxia Fetal/fisiopatologia , Gravidez , Ovinos
11.
Ann Neurol ; 71(1): 121-32, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22275258

RESUMO

OBJECTIVE: Connexin hemichannels can open during ischemia, resulting in loss of membrane potential, calcium influx, and release of glutamate. In this study, we tested the hypothesis that opening of hemichannels after cerebral ischemia may contribute to delayed evolution of injury. METHODS: We infused a mimetic peptide that blocks connexin 43 hemichannels into the lateral ventricle of chronically instrumented fetal sheep in utero at 128 ± 1 days gestation (term is 147 days), starting 90 minutes after 30 minutes of severe ischemia induced by reversible bilateral carotid artery occlusion, for either 1 or 25 hours. Sheep were killed 7 days later. RESULTS: Peptide infusion was associated with a graded improvement in recovery of electroencephalographic power after 7 days recovery, from -13 ± 1.9 dB (n = 7) after ischemia-vehicle to -9 ± 1.6 dB (n = 7) after ischemia-short infusion and -5 ± 1.6 dB after ischemia-long infusion (n = 6, p < 0.05). Peptide infusion was associated with reduced seizure activity after ischemia, less frequent status epilepticus (p < 0.05), and earlier return of sleep state cycling (p < 0.05). Ischemia-long infusion (but not ischemia-short infusion) was associated with improved survival of oligodendrocytes in intragyral and periventricular white matter (p < 0.05) and increased brain weight (p < 0.05). Ischemia-long infusion was associated with an intermediate estimate of surviving neurons in the parasagittal cortex of 2.9 ± 0.8 × 10(6), in comparison to sham control (4.3 ± 0.9 × 10(6)) or ischemia-vehicle (1.5 ± 0.4 × 10(6); p < 0.05 vs sham control). INTERPRETATION: These data support the hypothesis that opening of connexin hemichannels is a significant mediator of postischemic white and gray matter dysfunction and injury.


Assuntos
Isquemia Encefálica/metabolismo , Isquemia Encefálica/prevenção & controle , Conexina 43/antagonistas & inibidores , Hipóxia Fetal/tratamento farmacológico , Fragmentos de Peptídeos/administração & dosagem , Animais , Isquemia Encefálica/fisiopatologia , Sobrevivência Celular/fisiologia , Conexina 43/fisiologia , Modelos Animais de Doenças , Eletroencefalografia , Feminino , Hipóxia Fetal/metabolismo , Hipóxia Fetal/patologia , Marcação de Genes , Masculino , Neurônios/patologia , Neurônios/fisiologia , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/uso terapêutico , Gravidez , Convulsões/prevenção & controle , Ovinos , Fases do Sono/fisiologia , Resultado do Tratamento
12.
Exp Physiol ; 98(3): 699-709, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23104935

RESUMO

Dopamine is commonly used for blood pressure support in the neonate, but has limited empirical evidence to support its use. We tested the hypothesis that after near-terminal asphyxia in utero, dopamine infusions would prevent secondary hypotension. Fetal sheep (122-129 days of gestation; term is 147 days) received umbilical cord occlusion for 15 min or sham occlusion (n = 5). If the mean arterial blood pressure fell below 90% of baseline within 6 h after occlusion, fetuses were randomized to either dopamine infusion starting at 4 µg kg(-1) min(-1) and titrated according to mean arterial blood pressure up to a maximum of 40 µg kg(-1) min(-1) (n = 5) or to the same volume of normal saline (n = 5). Dopamine infusion, initiated at a median of 180 min after occlusion (range 96-280 min), was associated with a marked but transient increase in mean arterial blood pressure and fall in femoral blood flow compared with saline. Terminal hypotension developed later in four of the five fetuses that received maximal dopamine infusions than in five of five receiving saline infusion [517 (range 240-715) versus 106 min (range 23-497) after the start of infusions, P < 0.05]. In conclusion, dopamine infusion delayed but did not prevent terminal hypotension after severe asphyxia.


Assuntos
Asfixia/tratamento farmacológico , Dopamina/uso terapêutico , Hipóxia Fetal/tratamento farmacológico , Feto/irrigação sanguínea , Hipotensão/prevenção & controle , Animais , Pressão Arterial , Asfixia/complicações , Artérias Carótidas/fisiologia , Feminino , Idade Gestacional , Frequência Cardíaca Fetal/fisiologia , Gravidez , Fluxo Sanguíneo Regional/fisiologia , Carneiro Doméstico , Cordão Umbilical/irrigação sanguínea
13.
Bull Exp Biol Med ; 154(6): 737-9, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23658911

RESUMO

We evaluated physical development and activity of erythropoiesis in the offspring of rats with experimentally impaired uteroplacental circulation as well as the effect of exogenous nitric oxide donator used during pregnancy, on offspring development. Exogenous NO producing an anti-hypoxic effects contributes to the increase in somatometric parameters of the offspring on postnatal days 15 and 30. The rates of erythropoiesis in the liver and bone marrow did not differ from the normal; hemopoietic organs were not overstrained, which prevented exhaustion and failure of functional reserves of the erythrocyte system.


Assuntos
Eritropoese/efeitos dos fármacos , Hipóxia Fetal/tratamento farmacológico , Nitroglicerina/farmacologia , Circulação Placentária/efeitos dos fármacos , Vasodilatadores/farmacologia , Animais , Animais não Endogâmicos , Peso Corporal , Feminino , Hipóxia Fetal/patologia , Hipóxia Fetal/fisiopatologia , Doadores de Óxido Nítrico/administração & dosagem , Doadores de Óxido Nítrico/farmacologia , Nitroglicerina/administração & dosagem , Tamanho do Órgão , Gravidez , Ratos , Vasodilatadores/administração & dosagem
14.
Int J Mol Sci ; 13(5): 6303-6319, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22754366

RESUMO

Hypoxic-ischaemic brain injury at birth is associated with 1-3/1000 cases of moderate to severe encephalopathy. Previously, we have shown that connexin 43 hemichannel blockade, with a specific mimetic peptide, reduced the occurrence of seizures, improved recovery of EEG power and sleep state cycling, and improved cell survival following global cerebral ischaemia. In the present study, we examined the dose response for intracerebroventricular mimetic peptide infusion (50 µmol/kg/h for 1 h, followed by 50 µmol/kg/24 h (low dose) or 50 µmol/kg/h for 25 h (high dose) or vehicle only (control group), starting 90 min after the end of ischaemia), following global cerebral ischaemia, induced by 30 min bilateral carotid artery occlusion, in near-term fetal sheep (128 ± 1 days gestation). Both peptide infusion groups were associated with a transient significant increase in EEG power between 2-12 h after ischaemia. The ischaemia-low dose group showed a significant recovery of EEG power from day five compared to the ischaemia-vehicle and -high dose groups. In contrast, the high dose infusion was associated with greater secondary increase in impedance (brain cell swelling), as well as a trend towards a greater increase in lactate concentration and mortality. These data suggest that higher doses of connexin mimetic peptide are not beneficial and may be associated with adverse outcomes, most likely attributable to uncoupling of connexin 43 gap junctions leading to dysfunction of the astrocytic syncytium.


Assuntos
Encéfalo/fisiopatologia , Conexina 43/metabolismo , Hipóxia Fetal/veterinária , Hipóxia-Isquemia Encefálica/veterinária , Oligopeptídeos/efeitos adversos , Doenças dos Ovinos/tratamento farmacológico , Animais , Biomimética , Encéfalo/efeitos dos fármacos , Encéfalo/embriologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Eletroencefalografia/efeitos dos fármacos , Feminino , Hipóxia Fetal/tratamento farmacológico , Hipóxia Fetal/fisiopatologia , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/fisiopatologia , Infusões Intraventriculares , Oligopeptídeos/administração & dosagem , Gravidez , Convulsões/tratamento farmacológico , Convulsões/fisiopatologia , Convulsões/veterinária , Ovinos , Doenças dos Ovinos/fisiopatologia
15.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 43(5): 687-9, 724, 2012 Sep.
Artigo em Zh | MEDLINE | ID: mdl-23230739

RESUMO

OBJECTIVE: To observe the permeability of recombinant human erythropoietin through placenta barrier and fetal blood-brain barrier after transient uteroplacental ischemia. METHODS: Rats on days 19 of pregnancy were divided into rhEPO treated group, ischemia-reperfusion group and sham operated group. Fetal ischemia in rhEPO treated group and ischemia-reperfusion group was induced by bilateral occlusion of the utero-ovarian artery for 20 minutes. Different dosage of 125I-rhEPO (2500 U/kg, 5000 U/kg, 7500 U/kg) was injected into the rats through caudal veins 30 min before injury in rhEPO treated group and sham-operated group. Saline was administered intravenously 30 min before the induction of hypoxic-ischemic injury in ischemia-reperfusion group. The amniotic fluid, placenta and fetal organs including brain, liver, heart, lung and kidney were collected to measure the radioactivity at 24h after injury. RESULTS: 125I-rhEPO was detected in amniotic fluid, placenta and fetal organs. The radioactivity of 125I-rhEPO in these tissues increased gradually with the increased dose injected in rhEPO treated group and sham-operated group. There were significant differences in the radioactivity of 125I-rhEPO between rhEPO treated group and sham-operated group (P < 0.05). CONCLUSION: The permeability of rhEPO through placental barrier and blood-brain barrier increased under the condition of fetal ischemia and hypoxia.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Eritropoetina/farmacocinética , Eritropoetina/uso terapêutico , Hipóxia Fetal/tratamento farmacológico , Troca Materno-Fetal/efeitos dos fármacos , Animais , Feminino , Permeabilidade , Gravidez , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/uso terapêutico , Traumatismo por Reperfusão/tratamento farmacológico
16.
Eksp Klin Farmakol ; 75(5): 21-3, 2012.
Artigo em Russo | MEDLINE | ID: mdl-22834124

RESUMO

The effect of exogenous nitric oxide donor deponit-10 (nitroglycerin) on red cell indices in the offspring of rats with experimental disturbances of uteroplacental circulation has been investigated. It is established that fetal hypoxia facilitates the mobilization of functional reserves of the red cell system in the prenatal and early days of postnatal life of offspring in white rats, which is manifested by the growing process of erythropoiesis. Hyperfunction of the erythrocyte system in the first lifedays of pups leads eventually to a depletion of its functional capacities. The administration of an exogenous nitric oxide donor on the background of damaged uteroplacental circulation prevents the depletion and disruption of the functional reserves of the blood red cell system.


Assuntos
Eritrócitos/metabolismo , Eritropoese/efeitos dos fármacos , Hipóxia Fetal/tratamento farmacológico , Óxido Nítrico/metabolismo , Nitroglicerina/farmacologia , Circulação Placentária/fisiologia , Animais , Animais Recém-Nascidos , Feminino , Hipóxia Fetal/fisiopatologia , Masculino , Doadores de Óxido Nítrico/farmacologia , Circulação Placentária/efeitos dos fármacos , Gravidez , Ratos , Vasodilatadores/farmacologia
17.
Reprod Fertil Dev ; 23(2): 346-52, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21211468

RESUMO

Perinatal morbidity and mortality are significantly higher in pregnancies complicated by chronic hypoxia and intrauterine growth restriction (IUGR). Clinically, placental insufficiency and IUGR are strongly associated with a fetoplacental inflammatory response. To explore this further, hypoxia was induced in one fetus in twin-bearing pregnant sheep (n=9) by performing single umbilical artery ligation (SUAL) at 110 days gestation. Five ewes were administered the anti-inflammatory drug sulfasalazine (SSZ) daily, beginning 24h before surgery. Fetal blood gases and inflammatory markers were examined. In both SSZ- and placebo-treated ewes, SUAL fetuses were hypoxic and growth-restricted at 1 week (P<0.05). A fetoplacental inflammatory response was observed in SUAL pregnancies, with elevated pro-inflammatory cytokines, activin A and prostaglandin E(2). SSZ did not mitigate this inflammatory response. It is concluded that SUAL induces fetal hypoxia and a fetoplacental inflammatory response and that SSZ does not improve oxygenation or reduce inflammation. Further studies to explore whether alternative anti-inflammatory treatments may improve IUGR outcomes are warranted.


Assuntos
Anti-Inflamatórios/administração & dosagem , Hipóxia Fetal/tratamento farmacológico , Hipóxia Fetal/etiologia , Artérias Umbilicais/cirurgia , Ativinas/análise , Ativinas/sangue , Líquido Amniótico/química , Animais , Dinoprostona/análise , Dinoprostona/sangue , Modelos Animais de Doenças , Feminino , Sangue Fetal/química , Concentração de Íons de Hidrogênio , Inflamação/prevenção & controle , Interleucina-6/análise , Ligadura , Oxigênio/sangue , Gravidez , Ovinos , Sulfassalazina/administração & dosagem , Fator de Necrose Tumoral alfa/análise
18.
J Perinat Med ; 39(4): 441-4, 2011 07.
Artigo em Inglês | MEDLINE | ID: mdl-21410409

RESUMO

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.


Assuntos
Alopurinol/uso terapêutico , Lesões Encefálicas/prevenção & controle , Hipóxia Fetal/tratamento farmacológico , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Xantina Oxidase/antagonistas & inibidores , Alopurinol/administração & dosagem , Animais , Animais Recém-Nascidos , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/enzimologia , Lesões Encefálicas/etiologia , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/uso terapêutico , Feminino , Hipóxia Fetal/complicações , Hipóxia Fetal/enzimologia , Humanos , Hipóxia-Isquemia Encefálica/complicações , Hipóxia-Isquemia Encefálica/enzimologia , Recém-Nascido , Gravidez , Traumatismo por Reperfusão/tratamento farmacológico
19.
Ann Neurol ; 66(3): 323-31, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19798726

RESUMO

OBJECTIVE: Tetrahydrobiopterin (BH(4)) deficiency is a cause of dystonia at birth. We hypothesized that BH(4) is a developmental factor determining vulnerability of the immature fetal brain to hypoxic-ischemic injury and subsequent motor deficits in newborns. METHODS: Pregnant rabbits were subjected to 40-minute uterine ischemia, and fetal brains were investigated for global and focal changes in BH(4). Newborn kits were assessed by neurobehavioral tests following vehicle and sepiapterin (BH(4) analog) treatment of dams. RESULTS: Naive fetal brains at 70% gestation (E22) were severely deficient for BH(4) compared with maternal and other fetal tissues. BH(4) concentration rapidly increased normally in the perinatal period, with the highest concentrations found in the thalamus compared with basal ganglia, frontal, occipital, hippocampus, and parietal cortex. Global sustained 40-minute hypoxia-ischemia depleted BH(4) in E22 thalamus and to a lesser extent in basal ganglia, but not in the frontal, occipital, and parietal regions. Maternal supplementation prior to hypoxia-ischemia with sepiapterin increased BH(4) in all brain regions and especially in the thalamus, but did not increase the intermediary metabolite, 7,8-BH(2). Sepiapterin treatment also reduced incidence of severe motor deficits and perinatal death following E22 hypoxia-ischemia. INTERPRETATION: We conclude that early developmental BH(4) deficiency plays a critical role in hypoxic-ischemic brain injury. Increasing brain BH(4) via maternal supplementation may be an effective strategy in preventing motor deficits from antenatal hypoxia-ischemia.


Assuntos
Biopterinas/análogos & derivados , Encéfalo/embriologia , Encéfalo/metabolismo , Distonia/prevenção & controle , Desenvolvimento Fetal/efeitos dos fármacos , Hipóxia Fetal/metabolismo , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/metabolismo , Hipertonia Muscular/prevenção & controle , Pterinas/uso terapêutico , Animais , Animais Recém-Nascidos , Biopterinas/análise , Biopterinas/deficiência , Biopterinas/uso terapêutico , Química Encefálica/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Desenvolvimento Fetal/fisiologia , Hipóxia Fetal/tratamento farmacológico , Idade Gestacional , Humanos , Troca Materno-Fetal/efeitos dos fármacos , Óxido Nítrico Sintase/análise , Óxido Nítrico Sintase/deficiência , Óxido Nítrico Sintase/uso terapêutico , Gravidez , Pterinas/farmacologia , Coelhos
20.
Stomatologiia (Mosk) ; 89(6): 7-9, 2010.
Artigo em Russo | MEDLINE | ID: mdl-21311435

RESUMO

Intrauterine hypoxia decreased the nucleolar organizator quantity in enameloblasts of newborn albino rats tooth germ. Influence the leu-encepgalin synthetic analog dalargin and non-opiate synthetic leu-encephalin on newborn rats from 2 to 6 day abolished the intrauterine hypoxia affect. The main mechanism of peptide antioxidant effect may be NO system stimulation.


Assuntos
Antioxidantes/administração & dosagem , Esmalte Dentário/efeitos dos fármacos , Leucina Encefalina-2-Alanina/análogos & derivados , Hipóxia Fetal/complicações , Hipóxia Fetal/tratamento farmacológico , Germe de Dente/efeitos dos fármacos , Animais , Esmalte Dentário/anormalidades , Esmalte Dentário/metabolismo , Leucina Encefalina-2-Alanina/administração & dosagem , Feminino , Óxido Nítrico/metabolismo , Ratos , Ratos Endogâmicos , Germe de Dente/anormalidades , Germe de Dente/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA