Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
1.
Sci Rep ; 14(1): 13852, 2024 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-38879681

RESUMO

Neurological and cardiac injuries are significant contributors to morbidity and mortality following pediatric in-hospital cardiac arrest (IHCA). Preservation of mitochondrial function may be critical for reducing these injuries. Dimethyl fumarate (DMF) has shown potential to enhance mitochondrial content and reduce oxidative damage. To investigate the efficacy of DMF in mitigating mitochondrial injury in a pediatric porcine model of IHCA, toddler-aged piglets were subjected to asphyxia-induced CA, followed by ventricular fibrillation, high-quality cardiopulmonary resuscitation, and random assignment to receive either DMF (30 mg/kg) or placebo for four days. Sham animals underwent similar anesthesia protocols without CA. After four days, tissues were analyzed for mitochondrial markers. In the brain, untreated CA animals exhibited a reduced expression of proteins of the oxidative phosphorylation system (CI, CIV, CV) and decreased mitochondrial respiration (p < 0.001). Despite alterations in mitochondrial content and morphology in the myocardium, as assessed per transmission electron microscopy, mitochondrial function was unchanged. DMF treatment counteracted 25% of the proteomic changes induced by CA in the brain, and preserved mitochondrial structure in the myocardium. DMF demonstrates a potential therapeutic benefit in preserving mitochondrial integrity following asphyxia-induced IHCA. Further investigation is warranted to fully elucidate DMF's protective mechanisms and optimize its therapeutic application in post-arrest care.


Assuntos
Asfixia , Fumarato de Dimetilo , Modelos Animais de Doenças , Parada Cardíaca , Mitocôndrias , Animais , Parada Cardíaca/metabolismo , Parada Cardíaca/tratamento farmacológico , Asfixia/metabolismo , Asfixia/tratamento farmacológico , Asfixia/complicações , Suínos , Fumarato de Dimetilo/farmacologia , Fumarato de Dimetilo/uso terapêutico , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Humanos , Miocárdio/metabolismo , Miocárdio/patologia , Fosforilação Oxidativa/efeitos dos fármacos
2.
Pediatr Neonatol ; 65(2): 117-122, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37659900

RESUMO

BACKGROUND: Although neonatal disseminated intravascular coagulation (DIC) is associated with high mortality and severe complications, few studies have reported its clinical course. We aimed to describe the characteristics, treatments, and outcomes of neonatal DIC by using a national inpatient database. METHODS: Using the Japanese Diagnosis Procedure Combination database, we identified 5533 patients with neonatal DIC who were admitted to neonatal intensive care units between July 2010 and March 2020. We categorized the patients into those with asphyxia (n = 2911) and those without asphyxia (n = 2622). We investigated the patient characteristics, treatments, and outcomes. We further categorized neonates with asphyxia according to its severity. RESULTS: The gestational age of neonates with asphyxia was significantly lower than that of neonates without asphyxia (P < 0.001). Antithrombin was most commonly used for DIC (40%). Neonates with asphyxia were more likely to receive antithrombin (43% vs. 38%; P < 0.001), recombinant human soluble thrombomodulin (28% vs. 20%; P < 0.001), and fresh frozen plasma transfusion (68% vs. 51%; P < 0.001) than those without asphyxia. Neonates with asphyxia had higher in-hospital mortality (17% vs. 10%; P < 0.001), severe bleeding (11% vs. 6.8%; P < 0.001), and hospitalization costs than those without asphyxia. Additionally, neonates with severe asphyxia were more likely to receive several DIC therapies (such as recombinant human soluble thrombomodulin [30% vs. 24%]) and had higher in-hospital mortality (19% vs. 11%) and hospitalization costs than those with mild asphyxia. CONCLUSIONS: In this large retrospective study of neonatal DIC, patients with asphyxia received several treatments and demonstrated unfavorable outcomes when compared to those without asphyxia.


Assuntos
Asfixia Neonatal , Coagulação Intravascular Disseminada , Doenças do Recém-Nascido , Recém-Nascido , Humanos , Trombomodulina/uso terapêutico , Japão , Estudos Retrospectivos , Asfixia/complicações , Asfixia/tratamento farmacológico , Coagulação Intravascular Disseminada/etiologia , Coagulação Intravascular Disseminada/terapia , Transfusão de Componentes Sanguíneos/efeitos adversos , Plasma , Antitrombinas/uso terapêutico , Asfixia Neonatal/complicações , Asfixia Neonatal/terapia
3.
Epilepsy Behav ; 142: 109189, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37037061

RESUMO

Birth asphyxia and the resulting hypoxic-ischemic encephalopathy (HIE) are highly associated with perinatal and neonatal death, neonatal seizures, and an adverse later-life outcome. Currently used drugs, including phenobarbital and midazolam, have limited efficacy to suppress neonatal seizures. There is a medical need to develop new therapies that not only suppress neonatal seizures but also prevent later-life consequences. We have previously shown that the loop diuretic bumetanide does not potentiate the effects of phenobarbital in a rat model of birth asphyxia. Here we compared the effects of bumetanide (0.3 or 10 mg/kg i.p.), midazolam (1 mg/kg i.p.), and a combination of bumetanide and midazolam on neonatal seizures and later-life outcomes in this model. While bumetanide at either dose was ineffective when administered alone, the higher dose of bumetanide markedly potentiated midazolam's effect on neonatal seizures. Median bumetanide brain levels (0.47-0.53 µM) obtained with the higher dose were in the range known to inhibit the Na-K-Cl-cotransporter NKCC1 but it remains to be determined whether brain NKCC1 inhibition was underlying the potentiation of midazolam. When behavioral and cognitive alterations were examined over three months after asphyxia, treatment with the bumetanide/midazolam combination, but not with bumetanide or midazolam alone, prevented impairment of learning and memory. Furthermore, the combination prevented the loss of neurons in the dentate hilus and aberrant mossy fiber sprouting in the CA3a area of the hippocampus. The molecular mechanisms that explain that bumetanide potentiates midazolam but not phenobarbital in the rat model of birth asphyxia remain to be determined.


Assuntos
Asfixia Neonatal , Epilepsia , Humanos , Recém-Nascido , Ratos , Animais , Bumetanida/uso terapêutico , Bumetanida/farmacologia , Midazolam/uso terapêutico , Anticonvulsivantes/uso terapêutico , Anticonvulsivantes/farmacologia , Asfixia/complicações , Asfixia/tratamento farmacológico , Nascimento a Termo , Membro 2 da Família 12 de Carreador de Soluto , Fenobarbital/uso terapêutico , Fenobarbital/farmacologia , Epilepsia/tratamento farmacológico , Asfixia Neonatal/complicações , Asfixia Neonatal/tratamento farmacológico , Convulsões/tratamento farmacológico , Convulsões/etiologia
4.
J Physiol ; 601(10): 1999-2016, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36999348

RESUMO

Maternal magnesium sulphate (MgSO4 ) treatment is widely recommended before preterm birth for neuroprotection. However, this is controversial because there is limited evidence that MgSO4 provides long-term neuroprotection. Preterm fetal sheep (104 days gestation; term is 147 days) were assigned randomly to receive sham occlusion with saline infusion (n = 6) or i.v. infusion with MgSO4 (n = 7) or vehicle (saline, n = 6) from 24 h before hypoxia-ischaemia induced by umbilical cord occlusion until 24 h after occlusion. Sheep were killed after 21 days of recovery, for fetal brain histology. Functionally, MgSO4 did not improve long-term EEG recovery. Histologically, in the premotor cortex and striatum, MgSO4 infusion attenuated post-occlusion astrocytosis (GFAP+ ) and microgliosis but did not affect numbers of amoeboid microglia or improve neuronal survival. In the periventricular and intragyral white matter, MgSO4 was associated with fewer total (Olig-2+ ) oligodendrocytes compared with vehicle + occlusion. Numbers of mature (CC1+ ) oligodendrocytes were reduced to a similar extent in both occlusion groups compared with sham occlusion. In contrast, MgSO4 was associated with an intermediate improvement in myelin density in the intragyral and periventricular white matter tracts. In conclusion, a clinically comparable dose of MgSO4 was associated with moderate improvements in white and grey matter gliosis and myelin density but did not improve EEG maturation or neuronal or oligodendrocyte survival. KEY POINTS: Magnesium sulphate is widely recommended before preterm birth for neuroprotection; however, there is limited evidence that magnesium sulphate provides long-term neuroprotection. In preterm fetal sheep exposed to hypoxia-ischaemia (HI), MgSO4 was associated with attenuated astrocytosis and microgliosis in the premotor cortex and striatum but did not improve neuronal survival after recovery to term-equivalent age, 21 days after HI. Magnesium sulphate was associated with loss of total oligodendrocytes in the periventricular and intragyral white matter tracts, whereas mature, myelinating oligodendrocytes were reduced to a similar extent in both occlusion groups. In the same regions, MgSO4 was associated with an intermediate improvement in myelin density. Functionally, MgSO4 did not improve long-term recovery of EEG power, frequency or sleep stage cycling. A clinically comparable dose of MgSO4 was associated with moderate improvements in white and grey matter gliosis and myelin density but did not improve EEG maturation or neuronal or oligodendrocyte survival.


Assuntos
Nascimento Prematuro , Substância Branca , Recém-Nascido , Humanos , Feminino , Ovinos , Animais , Substância Cinzenta , Asfixia/tratamento farmacológico , Sulfato de Magnésio/farmacologia , Sulfato de Magnésio/uso terapêutico , Gliose/tratamento farmacológico , Sobrevivência Celular , Eletroencefalografia , Isquemia/tratamento farmacológico , Hipóxia
5.
Ann Neurol ; 93(2): 226-243, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36054632

RESUMO

OBJECTIVE: Birth asphyxia (BA) is the most frequent cause of neonatal death as well as central nervous system (CNS) injury. BA is often associated with neonatal seizures, which only poorly respond to anti-seizure medications and may contribute to the adverse neurodevelopmental outcome. Using a non-invasive rat model of BA, we have recently reported that the potent benzodiazepine, midazolam, prevents neonatal seizures in ~50% of rat pups. In addition to its anti-seizure effect, midazolam exerts anti-inflammatory actions, which is highly relevant for therapeutic intervention following BA. The 2 major aims of the present study were to examine (1) whether midazolam reduces the adverse outcome of BA, and (2) whether this effect is different in rats that did or did not exhibit neonatal seizures after drug treatment. METHODS: Behavioral and cognitive tests were performed over 14 months after asphyxia, followed by immunohistochemical analyses. RESULTS: All vehicle-treated rats had seizures after asphyxia and developed behavioral and cognitive abnormalities, neuroinflammation in gray and white matter, neurodegeneration in the hippocampus and thalamus, and hippocampal mossy fiber sprouting in subsequent months. Administration of midazolam (1 mg/kg i.p.) directly after asphyxia prevented post-asphyctic seizures in ~50% of the rats and resulted in the prevention or decrease of neuroinflammation and the behavioral, cognitive, and neurodegenerative consequences of asphyxia. Except for neurodegeneration in the thalamus, seizures did not seem to contribute to the adverse outcome of asphyxia. INTERPRETATION: The disease-modifying effect of midazolam identified here strongly suggests that this drug provides a valuable option for improving the treatment and outcome of BA. ANN NEUROL 2023;93:226-243.


Assuntos
Asfixia Neonatal , Epilepsia , Humanos , Recém-Nascido , Ratos , Animais , Midazolam/farmacologia , Midazolam/uso terapêutico , Asfixia/complicações , Asfixia/tratamento farmacológico , Doenças Neuroinflamatórias , Benzodiazepinas/uso terapêutico , Epilepsia/complicações , Asfixia Neonatal/complicações , Asfixia Neonatal/tratamento farmacológico
6.
Minerva Pediatr (Torino) ; 74(2): 107-115, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-33107271

RESUMO

BACKGROUND: The current study uses a population modeling approach to evaluate and quantify the impact of severity of asphyxia and hypoxic-ischemic encephalopathy (HIE) on the pharmacokinetics of phenobarbital in asphyxiated newborns treated with therapeutic hypothermia. METHODS: Included newborns received phenobarbital (the TOBY trial protocol). 120 plasma samples were available from 50 newborns, median (IQR) weight 3.3 (2.8-3.5) kg and gestational age 39 (39-40) weeks. NONMEM® version 7.2 was used for the data analysis. Age, body weight, sex, concomitant medications, kidney and liver function markers, as well as severity parameters of asphyxia and HIE were tested as potential covariates of pharmacokinetics of phenobarbital. Severe asphyxia was defined as pH of arterial umbilical cord blood ≤7.1 and Apgar 5 ≤5, and severe HIE was defined as time to normalization of amplitude-integrated electroencephalography (aEEG) >24 h. RESULTS: Weight was found to be the only statistically significant covariate for the volume of distribution. At weight of 1 kg volume of distribution was 0.91 L and for every additional kg it increased in 0.91 L. Clearance was 0.00563 L/h. No covariates were statistically significant for the clearance of phenobarbital. CONCLUSIONS: Phenobarbital dose adjustments are not indicated in the studied population, irrespective of the severity of asphyxia or HIE.


Assuntos
Asfixia Neonatal , Hipotermia Induzida , Hipóxia-Isquemia Encefálica , Adulto , Asfixia/complicações , Asfixia/tratamento farmacológico , Asfixia Neonatal/complicações , Asfixia Neonatal/tratamento farmacológico , Humanos , Hipotermia Induzida/métodos , Hipóxia-Isquemia Encefálica/terapia , Recém-Nascido , Fenobarbital/farmacocinética , Fenobarbital/uso terapêutico
8.
Epilepsia ; 62(8): 1971-1984, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34180051

RESUMO

OBJECTIVE: Seizures are common in neonates recovering from birth asphyxia but there is general consensus that current pharmacotherapy is suboptimal and that novel antiseizure drugs are needed. We recently showed in a rat model of birth asphyxia that seizures are triggered by the post-asphyxia recovery of brain pH. Here our aim was to investigate whether carbonic anhydrase inhibitors (CAIs), which induce systemic acidosis, block the post-asphyxia seizures. METHODS: The CAIs acetazolamide (AZA), benzolamide (BZA), and ethoxzolamide (EZA) were administered intraperitoneally or intravenously to 11-day-old rats exposed to intermittent asphyxia (30 min; three 7+3 min cycles of 9% and 5% O2 at 20% CO2 ). Electrode measurements of intracortical pH, Po2 , and local field potentials (LFPs) were made under urethane anesthesia. Convulsive seizures and blood acid-base parameters were examined in freely behaving animals. RESULTS: The three CAIs decreased brain pH by 0.14-0.17 pH units and suppressed electrographic post-asphyxia seizures. AZA, BZA, and EZA differ greatly in their lipid solubility (EZA > AZA > BZA) and pharmacokinetics. However, there were only minor differences in the delay (range 0.8-3.7 min) from intraperitoneal application to their action on brain pH. The CAIs induced a modest post-asphyxia elevation of brain Po2 that had no effect on LFP activity. AZA was tested in freely behaving rats, in which it induced a respiratory acidosis and decreased the incidence of convulsive seizures from 9 of 20 to 2 of 17 animals. SIGNIFICANCE: AZA, BZA, and EZA effectively block post-asphyxia seizures. Despite the differences in their pharmacokinetics, they had similar effects on brain pH, which indicates that their antiseizure mode of action was based on respiratory (hypercapnic) acidosis resulting from inhibition of blood-borne and extracellular vascular carbonic anhydrases. AZA has been used for several indications in neonates, suggesting that it can be safely repurposed for the treatment of neonatal seizures as an add-on to the current treatment regimen.


Assuntos
Acidose , Asfixia Neonatal , Acetazolamida/uso terapêutico , Animais , Asfixia/complicações , Asfixia/tratamento farmacológico , Inibidores da Anidrase Carbônica , Humanos , Recém-Nascido , Ratos , Convulsões/tratamento farmacológico , Convulsões/etiologia
9.
PLoS One ; 16(6): e0253328, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34138955

RESUMO

It has been reported that valproic acid (VPA) combined with therapeutic hypothermia can improve survival and neurologic outcomes in a rat asphyxial cardiac arrest model. However, neuroprotective mechanisms of such combined treatment of valproic acid with hypothermia remains unclear. We hypothesized that epigenetic regulation of HSP70 by histone acetylation could increase HSP70-mediated neuroprotection suppressed under hypothermia. Male Sprague-Dawley rats that achieved return of spontaneous circulation (ROSC) from asphyxial cardiac arrest were randomized to four groups: normothermia (37°C ± 1°C), hypothermia (33°C ± 1°C), normothermia + VPA (300 mg/kg IV initiated 5 minutes post-ROSC and infused over 20 min), and hypothermia + VPA. Three hours after ROSC, acetyl-histone H3 was highly expressed in VPA-administered groups (normothermia + VPA, hypothermia + VPA). Four hours after ROSC, HSP70 mRNA expression levels were significantly higher in normothermic groups (normothermia, normothermia + VPA) than in hypothermic groups (hypothermia, hypothermia + VPA). The hypothermia + VPA group showed significantly higher HSP70 mRNA expression than the hypothermia group. Similarly, at five hours after ROSC, HSP70 protein levels were significantly higher in normothermic groups than in hypothermic groups. HSP70 levels were significantly higher in the hypothermia + VPA group than in the hypothermia group. Only the hypothermia + VPA group showed significantly attenuated cleaved caspase-9 levels than the normothermia group. Hypothermia can attenuate the expression of HSP70 at transcriptional level. However, VPA administration can induce hyperacetylation of histone H3, leading to epigenetic transcriptional activation of HSP70 even in a hypothermic status. Combining VPA treatment with hypothermia may compensate for reduced activation of HSP70-mediated anti-apoptotic pathway.


Assuntos
Asfixia/terapia , Proteínas de Choque Térmico HSP70/metabolismo , Parada Cardíaca/terapia , Hipotermia Induzida/métodos , Neuroproteção , Ácido Valproico/uso terapêutico , Acetilação , Animais , Asfixia/tratamento farmacológico , Asfixia/metabolismo , Terapia Combinada , Modelos Animais de Doenças , Parada Cardíaca/tratamento farmacológico , Parada Cardíaca/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley
10.
Neurochem Int ; 147: 105064, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33951501

RESUMO

Hypoxic-ischemic encephalopathy (HIE) causes mortality and long-term neurologic morbidities in newborns, affecting pathways related to energy failure, excitotoxicity and oxidative stress that often lead to cell death. The whole process of HIE injury is coupled to changes in the expression of a great array of proteins. A nanoliposomal preparation of the flavonoid quercetin has been shown to exert neuroprotective effects in perinatal asphyxia models. This study aimed to identify neonatal HIE markers and explore the effect of quercetin administration in two perinatal asphyxia models: newborn rats and piglets. In the rat model, nanoliposomal quercetin administration reduced mortality after asphyxia. In the piglet model, quercetin partially overrode the reduction of HIF-1α mRNA levels in the cortex induced by asphyxia. Quercetin administration also reduced increased level of HO-1 mRNA in asphyctic piglets. These results suggest that quercetin neuroprotection might be involved in the regulation of HIF-1α, HO-1 and their targets. A proteomic approach revealed that the glycolytic pathway is strongly regulated by quercetin in both species. We also identified a set of proteins differentially expressed that could be further considered as markers. In piglets, this set includes Acidic Leucine-rich nuclear phosphoprotein 32 (ANP32A), associated with nervous system differentiation, proteins related with death pathways and alpha-enolase which can be converted to neuron-specific enolase, a glycolytic enzyme that may promote neuroprotection. In newborn rats, other promising proteins associated with neurogenesis and neuroprotection emerged, such as dihydropyrimidinase-related proteins, catalytic and regulatory subunits of phosphatases and heterogeneous nuclear ribonucleoprotein K (hnRNPK). Our results show that a nanoliposomal preparation of quercetin, with protective effect in two HIE mammal models, modulates the expression of proteins involved in energy metabolism and other putative neuroprotective signals in the cortex. Identification of these signals could reveal potential molecular pathways involved in disease onset and the novel quercetin neuroprotective strategy.


Assuntos
Asfixia/tratamento farmacológico , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Neuroproteção/efeitos dos fármacos , Quercetina/farmacologia , Animais , Animais Recém-Nascidos , Asfixia/metabolismo , Modelos Animais de Doenças , Humanos , Hipóxia-Isquemia Encefálica/metabolismo , Recém-Nascido , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Ratos , Suínos
11.
Apoptosis ; 25(9-10): 747-762, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32816128

RESUMO

Each year, 1 million children die due to perinatal asphyxia; however, there are no effective drugs to protect the neonatal brain against hypoxic/ischemic damage. In this study, we demonstrated for the first time the neuroprotective capacity of 3,3'-diindolylmethane (DIM) in an in vivo model of rat perinatal asphyxia, which has translational value and corresponds to hypoxic/ischemic episodes in human newborns. Posttreatment with DIM restored the weight of the ipsilateral hemisphere and normalized cell number in the brain structures of rats exposed to perinatal asphyxia. DIM also downregulated the mRNA expression of HIF1A-regulated Bnip3 and Hif1a which is a hypoxic marker, and the expression of miR-181b which is an indicator of perinatal asphyxia. In addition, DIM inhibited apoptosis and oxidative stress accompanying perinatal asphyxia through: downregulation of FAS, CASP-3, CAPN1, GPx3 and SOD-1, attenuation of caspase-9 activity, and upregulation of anti-apoptotic Bcl2 mRNA. The protective effects of DIM were accompanied by the inhibition of the AhR and NMDA signaling pathways, as indicated by the reduced expression levels of AhR, ARNT, CYP1A1, GluN1 and GluN2B, which was correlated with enhanced global DNA methylation and the methylation of the Ahr and Grin2b genes. Because our study provided evidence that in rat brain undergoing perinatal asphyxia, DIM predominantly targets AhR and NMDA, we postulate that compounds that possess the ability to inhibit their signaling are promising therapeutic tools to prevent stroke.


Assuntos
Apoptose/genética , Asfixia/tratamento farmacológico , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Receptores de Hidrocarboneto Arílico/genética , Receptores de N-Metil-D-Aspartato/genética , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Asfixia/genética , Asfixia/patologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Metilação de DNA/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Indóis/farmacologia , Recém-Nascido , Masculino , Proteínas de Membrana/genética , MicroRNAs/genética , Proteínas Mitocondriais/genética , N-Metilaspartato/farmacologia , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Ratos
12.
Artigo em Inglês | MEDLINE | ID: mdl-32663656

RESUMO

Cardiac arrest causes neuronal damage and functional impairments that can result in learning/memory dysfunction after ischemia. We previously identified a saturated fatty acid (stearic acid methyl ester, SAME) that was released from the superior cervical ganglion (sympathetic ganglion). The function of stearic acid methyl ester is currently unknown. Here, we show that SAME can inhibit the detrimental effects of global cerebral ischemia (i.e. cardiac arrest). Treatment with SAME in the presence of asphyxial cardiac arrest (ACA) revived learning and working memory deficits. Similarly, SAME-treated hippocampal slices after oxygen-glucose deprivation inhibited neuronal cell death. Moreover, SAME afforded neuroprotection against ACA in the CA1 region of the hippocampus, reduced ionized calcium-binding adapter molecule 1 expression and inflammatory cytokines/chemokines, with restoration in mitochondria respiration. Altogether, we describe a unique and uncharted role of saturated fatty acids in the brain that may have important implications against cerebral ischemia.


Assuntos
Asfixia/tratamento farmacológico , Região CA1 Hipocampal/metabolismo , Parada Cardíaca/tratamento farmacológico , Neuroproteção/efeitos dos fármacos , Ácidos Esteáricos/farmacologia , Animais , Asfixia/metabolismo , Asfixia/fisiopatologia , Região CA1 Hipocampal/fisiopatologia , Modelos Animais de Doenças , Parada Cardíaca/metabolismo , Parada Cardíaca/fisiopatologia , Masculino , Ratos , Ratos Sprague-Dawley
14.
Sci Rep ; 10(1): 3898, 2020 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-32127612

RESUMO

With the current practice of therapeutic hypothermia for neonatal encephalopathy, disability rates and the severity spectrum of cerebral palsy are reduced. Nevertheless, safe and effective adjunct therapies are needed to optimize outcomes. This study's objective was to assess if 18 mg/kg melatonin given rapidly over 2 h at 1 h after hypoxia-ischemia with cooling from 1-13 h was safe, achieved therapeutic levels within 3 h and augmented hypothermic neuroprotection. Following hypoxia-ischemia, 20 newborn piglets were randomized to: (i) Cooling 1-13 h (HT; n = 6); (ii) HT+ 2.5% ethanol vehicle (HT+V; n = 7); (iii) HT + Melatonin (HT+M; n = 7). Intensive care was maintained for 48 h; aEEG was acquired throughout, brain MRS acquired at 24 and 48 h and cell death (TUNEL) evaluated at 48 h. There were no differences for insult severity. Core temperature was higher in HT group for first hour after HI. Comparing HT+M to HT, aEEG scores recovered more quickly by 19 h (p < 0.05); comparing HT+V to HT, aEEG recovered from 31 h (p < 0.05). Brain phosphocreatine/inorganic phosphate and NTP/exchangeable phosphate were higher at 48 h in HT+M versus HT (p = 0.036, p = 0.049 respectively). Including both 24 h and 48 h measurements, the rise in Lactate/N-acetyl aspartate was reduced in white (p = 0.030) and grey matter (p = 0.038) after HI. Reduced overall TUNEL positive cells were observed in HT+M (47.1 cells/mm2) compared to HT (123.8 cells/mm2) (p = 0.0003) and HT+V (97.5 cells/mm2) compared to HT (p = 0.012). Localized protection was seen in white matter for HT+M versus HT (p = 0.036) and internal capsule for HT+M compared to HT (p = 0.001) and HT+V versus HT (p = 0.006). Therapeutic melatonin levels (15-30mg/l) were achieved at 2 h and were neuroprotective following HI, but ethanol vehicle was partially protective.


Assuntos
Asfixia/terapia , Etanol/farmacologia , Hipotermia Induzida , Melatonina/farmacologia , Animais , Animais Recém-Nascidos , Asfixia/tratamento farmacológico , Asfixia/metabolismo , Asfixia/fisiopatologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Eletroencefalografia/efeitos dos fármacos , Melatonina/farmacocinética , Melatonina/uso terapêutico , Suínos , Distribuição Tecidual
15.
Paediatr Drugs ; 22(1): 95-104, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31960360

RESUMO

AIM: The main burden of hypoxic-ischemic encephalopathy falls in low-income countries. 2-Iminobiotin, a selective inhibitor of neuronal and inducible nitric oxide synthase, has been shown to be safe and effective in preclinical studies of birth asphyxia. Recently, safety and pharmacokinetics of 2-iminobiotin treatment on top of hypothermia has been described. Since logistics and the standard of medical care are very different in low-resource settings, the aim of this study was to investigate safety and pharmacokinetics of Two-IminoBiotin in the Democratic Republic of Congo (TIBC). METHODS: Near-term neonates, born in Kinshasa, Democratic Republic of Congo, with a Thompson score ≥ 7 were eligible for inclusion. Excluded were patients with (1) inability to insert an umbilical venous catheter for administration of the study drug; (2) major congenital or chromosomal abnormalities; (3) birth weight < 1800 g; (4) clear signs of infection; and (5) moribund patients. Neonates received six infusions of 2-iminobiotin 0.16 mg/kg started within 6 h after birth, with 4-h intervals, targeting an AUC0-4h of 365 ng*h/mL. Safety, defined as vital signs, the need for clinical intervention after administration of study drug, occurrence of (serious) adverse events, and pharmacokinetics were assessed. RESULTS: After parental consent, seven patients were included with a median Thompson score of 10 (range 8-16). No relevant changes in vital signs were observed over time. There was no need for clinical intervention due to administration of study drug. Three patients died, two after completing the study protocol, one was moribund at inclusion and should not have been included. Pharmacokinetic data of 2-iminobiotin were best described using a two-compartment model. Median AUC0-4h was 664 ng*h/mL (range 414-917). No safety issues attributed to the administration of 2-iminobiotin were found. CONCLUSION: The present dosing regimen resulted in higher AUCs than targeted, necessitating a change in the dose regimen in future efficacy trials. No adverse effects that could be attributed to the use of 2-iminobiotin were observed. EudraCT number 2015-003063-12.


Assuntos
Asfixia/tratamento farmacológico , Biotina/análogos & derivados , Adulto , Biotina/farmacologia , Biotina/uso terapêutico , Feminino , Humanos , Recém-Nascido , Masculino , Pobreza
16.
Am J Physiol Regul Integr Comp Physiol ; 315(2): R205-R217, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29561649

RESUMO

Magnesium sulfate (MgSO4) is recommended for preterm neuroprotection, preeclampsia, and preterm labor prophylaxis. There is an important, unmet need to carefully test clinical interventions in both sexes. Therefore, we aimed to investigate cardiovascular and neurophysiological adaptations to MgSO4 during normoxia and asphyxia in preterm male and female fetal sheep. Fetuses were instrumented at 98 ± 1 days of gestation (term = 147 days). At 104 days, unanesthetized fetuses were randomly assigned to intravenous MgSO4 ( n = 12 female, 10 male) or saline ( n = 13 female, 10 male). At 105 days fetuses underwent umbilical cord occlusion for up to 25 min. Occlusions were stopped early if mean arterial blood pressure (MAP) fell below 8 mmHg or asystole occurred for >20 s. During normoxia, MgSO4 was associated with similar reductions in fetal heart rate (FHR), EEG power, and movement in both sexes ( P < 0.05 vs. saline controls) and suppression of α- and ß-spectral band power in males ( P < 0.05 vs. saline controls). During occlusion, similar FHR and MAP responses occurred in MgSO4-treated males and females compared with saline controls. Recovery of FHR and MAP after release of occlusion was more prolonged in MgSO4-treated males ( P < 0.05 vs. saline controls). During and after occlusion, EEG power was lower in MgSO4-treated females ( P < 0.05 vs. saline controls). In conclusion, MgSO4 infusion was associated with subtle sex-specific effects on EEG spectral power and cardiac responses to asphyxia in utero, possibly reflecting sex-specific differences in interneuronal connectivity and regulation of cardiac output.


Assuntos
Asfixia/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Sistema Cardiovascular/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Sulfato de Magnésio/farmacologia , Fármacos Neuroprotetores/farmacologia , Adaptação Fisiológica , Animais , Pressão Arterial/efeitos dos fármacos , Asfixia/diagnóstico por imagem , Asfixia/fisiopatologia , Encéfalo/fisiopatologia , Ondas Encefálicas/efeitos dos fármacos , Sistema Cardiovascular/fisiopatologia , Modelos Animais de Doenças , Eletroencefalografia , Feminino , Movimento Fetal/efeitos dos fármacos , Idade Gestacional , Frequência Cardíaca Fetal/efeitos dos fármacos , Masculino , Diagnóstico Pré-Natal/métodos , Fatores Sexuais , Carneiro Doméstico
17.
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
18.
J Clin Pharm Ther ; 43(2): 196-201, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28940525

RESUMO

WHAT IS KNOWN AND OBJECTIVE: Phenobarbital is the first-line treatment of seizures in asphyxiated neonates; however, due to the high pharmacokinetic variability in this population, there is no consensus on the optimal dosage regimen. This study was conducted to identify variables that affect phenobarbital fate during routine clinical care and then to evaluate the dosage schedule that could be applied in term asphyxiated neonates with respect to achieving the target therapeutic range. METHODS: Phenobarbital pharmacokinetics was calculated based on serum concentrations measurements using one-compartmental model. Body weight, body surface area, gestational age, creatinine clearance, total bilirubin, alanine aminotransferase, aspartate aminotransferase, international normalized ratio, Apgar scores, umbilical cord arterial pH and base excess were explored as covariates in linear regression models. Based on this analysis, phenobarbital loading and maintenance dose regimen were projected. RESULTS AND DISCUSSION: In the whole study population (N = 36), phenobarbital volume of distribution, clearance and half-life median (interquartile range) values were 0.49 (0.38-0.59) L/kg, 0.0045 (0.0034-0.0055) L/h/kg and 75.1 (60.2-103.3) hours, respectively. The drug volume of distribution was associated with body weight, length and body surface area, whereas clearance was not in relationship with any explored features. Weight-normalized loading dose of 15 mg/kg and weight-normalized daily maintenance dose of 3 mg/kg proved to be optimal in our study population to reach phenobarbital therapeutic range. WHAT IS NEW AND CONCLUSIONS: This study presents basis for phenobarbital initial dosing in term asphyxiated neonates during first week of life. Phenobarbital weight-normalized loading dose of 15 mg/kg lead to simulated target peak concentrations in 72% of neonates, weight-normalized maintenance dose of 3 mg/kg lead to steady state within therapeutic window in the same proportion of patients.


Assuntos
Asfixia/tratamento farmacológico , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Fenobarbital/administração & dosagem , Fenobarbital/farmacocinética , Asfixia/metabolismo , Feminino , Idade Gestacional , Meia-Vida , Humanos , Hipóxia-Isquemia Encefálica/metabolismo , Recém-Nascido , Masculino , Estudos Retrospectivos , Distribuição Tecidual
19.
Neurotox Res ; 33(2): 461-473, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28844085

RESUMO

Perinatal asphyxia (PA) is a relevant cause of death at the time of labour, and when survival is stabilised, associated with short- and long-term developmental disabilities, requiring inordinate care by health systems and families. Its prevalence is high (1 to 10/1000 live births) worldwide. At present, there are few therapeutic options, apart from hypothermia, that regrettably provides only limited protection if applied shortly after the insult.PA implies a primary and a secondary insult. The primary insult relates to the lack of oxygen, and the secondary one to the oxidative stress triggered by re-oxygenation, formation of reactive oxygen (ROS) and reactive nitrogen (RNS) species, and overactivation of glutamate receptors and mitochondrial deficiencies. PA induces overactivation of a number of sentinel proteins, including hypoxia-induced factor-1α (HIF-1α) and the genome-protecting poly(ADP-ribose) polymerase-1 (PARP-1). Upon activation, PARP-1 consumes high amounts of ATP at a time when this metabolite is scarce, worsening in turn the energy crisis elicited by asphyxia. The energy crisis also impairs ATP-dependent transport, including glutamate re-uptake by astroglia. Nicotinamide, a PARP-1 inhibitor, protects against the metabolic cascade elicited by the primary stage, avoiding NAD+ exhaustion and the energetic crisis. Upon re-oxygenation, however, oxidative stress leads to nuclear translocation of the NF-κB subunit p65, overexpression of the pro-inflammatory cytokines IL-1ß and TNF-α, and glutamate-excitotoxicity, due to impairment of glial-glutamate transport, extracellular glutamate overflow, and overactivation of NMDA receptors, mainly of the extrasynaptic type. This leads to calcium influx, mitochondrial impairment, and inactivation of antioxidant enzymes, increasing further the activity of pro-oxidant enzymes, thereby making the surviving neonate vulnerable to recurrent metabolic insults whenever oxidative stress is involved. Here, we discuss evidence showing that (i) inhibition of PARP-1 overactivation by nicotinamide and (ii) inhibition of extrasynaptic NMDA receptor overactivation by memantine can prevent the short- and long-term consequences of PA. These hypotheses have been evaluated in a rat preclinical model of PA, aiming to identify the metabolic cascades responsible for the long-term consequences induced by the insult, also assessing postnatal vulnerability to recurrent oxidative insults. Thus, we present and discuss evidence demonstrating that PA induces long-term changes in metabolic pathways related to energy and oxidative stress, priming vulnerability of cells with both the neuronal and the glial phenotype. The effects induced by PA are region dependent, the substantia nigra being particularly prone to cell death. The issue of short- and long-term consequences of PA provides a framework for addressing a fundamental issue referred to plasticity of the CNS, since the perinatal insult triggers a domino-like sequence of events making the developing individual vulnerable to recurrent adverse conditions, decreasing his/her coping repertoire because of a relevant insult occurring at birth.


Assuntos
Asfixia/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Niacinamida/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Receptores de Glutamato/efeitos dos fármacos , Animais , Antioxidantes/farmacologia , Humanos
20.
J Am Heart Assoc ; 6(2)2017 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-28214793

RESUMO

BACKGROUND: Epinephrine administered by low umbilical venous catheter (UVC) or endotracheal tube (ETT) is indicated in neonates who fail to respond to positive pressure ventilation and chest compressions at birth. Pharmacokinetics of ETT epinephrine via fluid-filled lungs or UVC epinephrine in the presence of fetal shunts is unknown. We hypothesized that epinephrine administered by ETT or low UVC results in plasma epinephrine concentrations and rates of return of spontaneous circulation (ROSC) similar to right atrial (RA) epinephrine. METHODS AND RESULTS: Forty-four lambs were randomized into the following groups: RA epinephrine (0.03 mg/kg), low UVC epinephrine (0.03 mg/kg), postcompression ETT epinephrine (0.1 mg/kg), and precompression ETT epinephrine (0.1 mg/kg). Asystole was induced by umbilical cord occlusion. Resuscitation was initiated following 5 minutes of asystole. Thirty-eight of 44 lambs achieved ROSC (10/11, 9/11, and 12/22 in the RA, UVC, and ETT groups, respectively; subsequent RA epinephrine resulted in a total ROSC of 19/22 in the ETT groups). Median time (interquartile range) to achieve ROSC was significantly longer in the ETT group (including those that received RA epinephrine) compared to the intravenous group (4.5 [2.9-7.4] versus 2 [1.9-3] minutes; P=0.02). RA and low UVC epinephrine administration achieved comparable peak plasma epinephrine concentrations (470±250 versus 450±190 ng/mL) by 1 minute compared to ETT values of 130±60 ng/mL at 5 minutes; P=0.03. Following ROSC with ETT epinephrine alone, there was a delayed peak epinephrine concentration (652±240 ng/mL). CONCLUSIONS: The absorption of ETT epinephrine is low and delayed at birth. RA and low UVC epinephrine rapidly achieve high plasma concentrations resulting in ROSC.


Assuntos
Asfixia/complicações , Reanimação Cardiopulmonar/métodos , Epinefrina/administração & dosagem , Parada Cardíaca/tratamento farmacológico , Animais , Animais Recém-Nascidos , Asfixia/tratamento farmacológico , Asfixia/metabolismo , Modelos Animais de Doenças , Epinefrina/farmacocinética , Parada Cardíaca/etiologia , Parada Cardíaca/metabolismo , Infusões Intravenosas , Instilação de Medicamentos , Ovinos , Traqueia , Veias Umbilicais , Vasoconstritores/administração & dosagem , Vasoconstritores/farmacocinética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA