Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 319
Filtrar
1.
Br J Clin Pharmacol ; 90(6): 1418-1427, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38450797

RESUMEN

AIMS: Little is known about the population pharmacokinetics (PPK) of vancomycin in neonates with perinatal asphyxia treated with therapeutic hypothermia (TH). We aimed to describe the PPK of vancomycin and propose an initial dosing regimen for the first 48 h of treatment with pharmacokinetic/pharmacodynamic target attainment. METHODS: Neonates with perinatal asphyxia treated with TH were included from birth until Day 6 in a multicentre prospective cohort study. A vancomycin PPK model was constructed using nonlinear mixed-effects modelling. The model was used to evaluate published dosing guidelines with regard to pharmacokinetic/pharmacodynamic target attainment. The area under the curve/minimal inhibitory concentration ratio of 400-600 mg*h/L was used as target range. RESULTS: Sixteen patients received vancomycin (median gestational age: 41 [range: 38-42] weeks, postnatal age: 4.4 [2.5-5.5] days, birth weight: 3.5 [2.3-4.7] kg), and 112 vancomycin plasma concentrations were available. Most samples (79%) were collected during the rewarming and normothermic phase, as vancomycin was rarely initiated during the hypothermic phase due to its nonempirical use. An allometrically scaled 1-compartment model showed the best fit. Vancomycin clearance was 0.17 L/h, lower than literature values for term neonates of 3.5 kg without perinatal asphyxia (range: 0.20-0.32 L/h). Volume of distribution was similar. Published dosing regimens led to overexposure within 24 h of treatment. A loading dose of 10 mg/kg followed by 24 mg/kg/day in 4 doses resulted in target attainment. CONCLUSION: Results of this study suggest that vancomycin clearance is reduced in term neonates with perinatal asphyxia treated with TH. Lower dosing regimens should be considered followed by model-informed precision dosing.


Asunto(s)
Antibacterianos , Asfixia Neonatal , Hipotermia Inducida , Modelos Biológicos , Vancomicina , Humanos , Recién Nacido , Vancomicina/farmacocinética , Vancomicina/administración & dosificación , Hipotermia Inducida/métodos , Asfixia Neonatal/terapia , Asfixia Neonatal/tratamiento farmacológico , Estudios Prospectivos , Masculino , Femenino , Antibacterianos/farmacocinética , Antibacterianos/administración & dosificación , Área Bajo la Curva , Edad Gestacional , Relación Dosis-Respuesta a Droga
2.
Sci Rep ; 13(1): 9467, 2023 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-37301929

RESUMEN

Intrapartum hypoxia-ischemia leading to neonatal encephalopathy (NE) results in significant neonatal mortality and morbidity worldwide, with > 85% of cases occurring in low- and middle-income countries (LMIC). Therapeutic hypothermia (HT) is currently the only available safe and effective treatment of HIE in high-income countries (HIC); however, it has shown limited safety or efficacy in LMIC. Therefore, other therapies are urgently required. We aimed to compare the treatment effects of putative neuroprotective drug candidates following neonatal hypoxic-ischemic (HI) brain injury in an established P7 rat Vannucci model. We conducted the first multi-drug randomized controlled preclinical screening trial, investigating 25 potential therapeutic agents using a standardized experimental setting in which P7 rat pups were exposed to unilateral HI brain injury. The brains were analysed for unilateral hemispheric brain area loss after 7 days survival. Twenty animal experiments were performed. Eight of the 25 therapeutic agents significantly reduced brain area loss with the strongest treatment effect for Caffeine, Sonic Hedgehog Agonist (SAG) and Allopurinol, followed by Melatonin, Clemastine, ß-Hydroxybutyrate, Omegaven, and Iodide. The probability of efficacy was superior to that of HT for Caffeine, SAG, Allopurinol, Melatonin, Clemastine, ß-hydroxybutyrate, and Omegaven. We provide the results of the first systematic preclinical screening of potential neuroprotective treatments and present alternative single therapies that may be promising treatment options for HT in LMIC.


Asunto(s)
Asfixia Neonatal , Lesiones Encefálicas , Hipotermia Inducida , Hipoxia-Isquemia Encefálica , Melatonina , Fármacos Neuroprotectores , Animales , Humanos , Recién Nacido , Ratas , Alopurinol/farmacología , Animales Recién Nacidos , Asfixia Neonatal/tratamiento farmacológico , Encéfalo , Lesiones Encefálicas/tratamiento farmacológico , Cafeína/farmacología , Clemastina/farmacología , Modelos Animales de Enfermedad , Proteínas Hedgehog , Hidroxibutiratos/farmacología , Hipotermia Inducida/métodos , Hipoxia/tratamiento farmacológico , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Isquemia/terapia , Melatonina/farmacología , Melatonina/uso terapéutico , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico
3.
Epilepsy Behav ; 142: 109189, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37037061

RESUMEN

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.


Asunto(s)
Asfixia Neonatal , Epilepsia , Humanos , Recién Nacido , Ratas , Animales , Bumetanida/uso terapéutico , Bumetanida/farmacología , Midazolam/uso terapéutico , Anticonvulsivantes/uso terapéutico , Anticonvulsivantes/farmacología , Asfixia/complicaciones , Asfixia/tratamiento farmacológico , Nacimiento a Término , Miembro 2 de la Familia de Transportadores de Soluto 12 , Fenobarbital/uso terapéutico , Fenobarbital/farmacología , Epilepsia/tratamiento farmacológico , Asfixia Neonatal/complicaciones , Asfixia Neonatal/tratamiento farmacológico , Convulsiones/tratamiento farmacológico , Convulsiones/etiología
4.
Molecules ; 28(3)2023 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-36770769

RESUMEN

Perinatal asphyxia is considered to be one of the major causes of brain neurodegeneration in full-term newborns. The worst consequence of perinatal asphyxia is neurodegenerative brain damage, also known as hypoxic-ischemic encephalopathy. Hypoxic-ischemic encephalopathy is the leading cause of mortality in term newborns. To date, due to the complex mechanisms of brain damage, no effective or causal treatment has been developed that would ensure complete neuroprotection. Although hypothermia is the standard of care for hypoxic-ischemic encephalopathy, it does not affect all changes associated with encephalopathy. Therefore, there is a need to develop effective treatment strategies, namely research into new agents and therapies. In recent years, it has been pointed out that natural compounds with neuroprotective properties, such as melatonin, can be used in the treatment of hypoxic-ischemic encephalopathy. This natural substance with anti-inflammatory, antioxidant, anti-apoptotic and neurofunctional properties has been shown to have pleiotropic prophylactic or therapeutic effects, mainly against experimental brain neurodegeneration in hypoxic-ischemic neonates. Melatonin is a natural neuroprotective hormone, which makes it promising for the treatment of neurodegeneration after asphyxia. It is supposed that melatonin alone or in combination with hypothermia may improve neurological outcomes in infants with hypoxic-ischemic encephalopathy. Melatonin has been shown to be effective in the last 20 years of research, mainly in animals with perinatal asphyxia but, so far, no clinical trials have been performed on a sufficient number of newborns. In this review, we summarize the advantages and limitations of melatonin research in the treatment of experimental and clinical perinatal asphyxia.


Asunto(s)
Asfixia Neonatal , Lesiones Encefálicas , Hipotermia , Hipoxia-Isquemia Encefálica , Melatonina , Humanos , Animales , Embarazo , Femenino , Recién Nacido , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Asfixia/complicaciones , Melatonina/farmacología , Melatonina/uso terapéutico , Asfixia Neonatal/tratamiento farmacológico , Asfixia Neonatal/complicaciones
5.
Ann Neurol ; 93(2): 226-243, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36054632

RESUMEN

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.


Asunto(s)
Asfixia Neonatal , Epilepsia , Humanos , Recién Nacido , Ratas , Animales , Midazolam/farmacología , Midazolam/uso terapéutico , Asfixia/complicaciones , Asfixia/tratamiento farmacológico , Enfermedades Neuroinflamatorias , Benzodiazepinas/uso terapéutico , Epilepsia/complicaciones , Asfixia Neonatal/complicaciones , Asfixia Neonatal/tratamiento farmacológico
6.
Ann Neurol ; 92(6): 1066-1079, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36054160

RESUMEN

OBJECTIVE: Seizures are more common in the neonatal period than at any other stage of life. Phenobarbital is the first-line treatment for neonatal seizures and is at best effective in approximately 50% of babies, but may contribute to neuronal injury. Here, we assessed the efficacy of phenobarbital versus the synthetic neurosteroid, ganaxolone, to moderate seizure activity and neuropathology in neonatal lambs exposed to perinatal asphyxia. METHODS: Asphyxia was induced via umbilical cord occlusion in term lambs at birth. Lambs were treated with ganaxolone (5mg/kg/bolus then 5mg/kg/day for 2 days) or phenobarbital (20mg/kg/bolus then 5mg/kg/day for 2 days) at 6 hours. Abnormal brain activity was classified as stereotypic evolving (SE) seizures, epileptiform discharges (EDs), and epileptiform transients (ETs) using continuous amplitude-integrated electroencephalographic recordings. At 48 hours, lambs were euthanized for brain pathology. RESULTS: Asphyxia caused abnormal brain activity, including SE seizures that peaked at 18 to 20 hours, EDs, and ETs, and induced neuronal degeneration and neuroinflammation. Ganaxolone treatment was associated with an 86.4% reduction in the number of seizures compared to the asphyxia group. The total seizure duration in the asphyxia+ganaxolone group was less than the untreated asphyxia group. There was no difference in the number of SE seizures between the asphyxia and asphyxia+phenobarbital groups or duration of SE seizures. Ganaxolone treatment, but not phenobarbital, reduced neuronal degeneration within hippocampal CA1 and CA3 regions, and cortical neurons, and ganaxolone reduced neuroinflammation within the thalamus. INTERPRETATION: Ganaxolone provided better seizure control than phenobarbital in this perinatal asphyxia model and was neuroprotective for the newborn brain, affording a new therapeutic opportunity for treatment of neonatal seizures. ANN NEUROL 2022;92:1066-1079.


Asunto(s)
Asfixia Neonatal , Epilepsia , Pregnanolona , Animales , Humanos , Recién Nacido , Anticonvulsivantes/uso terapéutico , Asfixia Neonatal/complicaciones , Asfixia Neonatal/tratamiento farmacológico , Epilepsia/tratamiento farmacológico , Fenobarbital/uso terapéutico , Convulsiones/tratamiento farmacológico , Ovinos , Animales Recién Nacidos , Modelos Animales de Enfermedad
7.
Minerva Pediatr (Torino) ; 74(2): 107-115, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-33107271

RESUMEN

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.


Asunto(s)
Asfixia Neonatal , Hipotermia Inducida , Hipoxia-Isquemia Encefálica , Adulto , Asfixia/complicaciones , Asfixia/tratamiento farmacológico , Asfixia Neonatal/complicaciones , Asfixia Neonatal/tratamiento farmacológico , Humanos , Hipotermia Inducida/métodos , Hipoxia-Isquemia Encefálica/terapia , Recién Nacido , Fenobarbital/farmacocinética , Fenobarbital/uso terapéutico
8.
Am J Perinatol ; 39(8): 824-829, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-33129208

RESUMEN

Hypoxic-ischemic encephalopathy (HIE) is the main cause of long-term neurodevelopmental morbidity in term born infants worldwide. Melatonin is a hormone with antioxidant and anti-inflammatory effects that make it a promising molecule for the treatment of perinatal asphyxia. Probably, the synergistic use of hypothermia associated with melatonin treatment may improve the neurological outcome in infants with HIE. In the past 20 years, the efficacy of melatonin in reducing oxidative stress has been demonstrated in animals; however, clinical trials with sufficient sample size of newborns are lacking to date. Since in 2000 we were among the first to study the neuroprotective properties of melatonin on infants, in this review, we want to summarize the advantages and limitations of the investigations conducted to date. KEY POINTS: · HIE is the main cause of morbidity in term born infants worldwide.. · Melatonin is a promising molecule for the treatment of perinatal asphyxia.. · This review summarizes advantages and limitations of the investigations conducted on melatonin..


Asunto(s)
Asfixia Neonatal , Hipotermia Inducida , Hipoxia-Isquemia Encefálica , Melatonina , Asfixia , Asfixia Neonatal/complicaciones , Asfixia Neonatal/tratamiento farmacológico , Femenino , Humanos , Hipoxia-Isquemia Encefálica/complicaciones , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Recién Nacido , Melatonina/uso terapéutico , Embarazo
10.
J Coll Physicians Surg Pak ; 31(12): 1511-1512, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34794300

RESUMEN

An observational cohort study was performed at Neonatal Intensive Care Units of two military hospitals to see the effects of citicoline in neonates diagnosed to have moderate to severe hypoxic ischemic encephalopathy. Twenty newborns fulfilling the inclusion criteria were selected for the study and were given injection citicoline via the IV route. The outcomes in the immediate newborn period, including neurological features and mortality, were studied. Only one baby, who was given injection citicoline, died during the study period and 19 babies were discharged and sent home after establishment of oral feeds. Citicoline was found promising in treatment of newborns with moderate to severe birth asphyxia. Key Words: Hypoxia, Ischemia, Birth asphyxia, HIE.


Asunto(s)
Asfixia Neonatal , Citidina Difosfato Colina , Puntaje de Apgar , Asfixia , Asfixia Neonatal/tratamiento farmacológico , Citidina Difosfato Colina/uso terapéutico , Humanos , Recién Nacido , Proyectos Piloto
11.
Acta Neurol Belg ; 121(6): 1401-1406, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34494216

RESUMEN

BACKGROUND: Perinatal asphyxia (PA) is a devastating neonatal condition characterized by a lack of oxygen supporting the organ systems. PA can lead to hypoxic-ischemic encephalopathy (HIE), a brain dysfunction due to oxygen deprivation with a complex neurological sequela. The pathophysiology of HIE and PA is not entirely understood, with therapeutic hypothermia being the standard treatment with only limited value. However, alternative neuroprotective therapies can be a potential treatment modality. METHODS: In this review, we will characterize the biochemical mechanisms of PA and HIE, while also giving insight into cerebrolysin, a neuroprotective treatment used for HIE and PA. RESULTS: We found that cerebrolysin has up to 6-month treatment window post-ischemic insult. Cerebrolysin injections of 0.1 ml/kg of body weight twice per week were found to provide gross motor and speech deficit improvement. CONCLUSION: Our literature search emphasizes the positive effects of cerebrolysin for general improvement outcomes. Nevertheless, biomarker establishment is warranted to improve patient outcomes.


Asunto(s)
Aminoácidos/uso terapéutico , Asfixia Neonatal/tratamiento farmacológico , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Aminoácidos/farmacología , Asfixia Neonatal/complicaciones , Asfixia Neonatal/fisiopatología , Circulación Cerebrovascular/efectos de los fármacos , Circulación Cerebrovascular/fisiología , Humanos , Hipoxia-Isquemia Encefálica/etiología , Hipoxia-Isquemia Encefálica/fisiopatología , Recién Nacido , Fármacos Neuroprotectores/farmacología , Ensayos Clínicos Controlados Aleatorios como Asunto/métodos , Resultado del Tratamiento
12.
Epilepsia ; 62(11): 2826-2844, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34458992

RESUMEN

OBJECTIVE: Birth asphyxia is a major cause of hypoxic-ischemic encephalopathy (HIE) in neonates and often associated with mortality, neonatal seizures, brain damage, and later life motor, cognitive, and behavioral impairments and epilepsy. Preclinical studies on rodent models are needed to develop more effective therapies for preventing HIE and its consequences. Thus far, the most popular rodent models have used either exposure of intact animals to hypoxia-only, or a combination of hypoxia and carotid occlusion, for the induction of neonatal seizures and adverse outcomes. However, such models lack systemic hypercapnia, which is a fundamental constituent of birth asphyxia with major effects on neuronal excitability. Here, we use a recently developed noninvasive rat model of birth asphyxia with subsequent neonatal seizures to study later life adverse outcome. METHODS: Intermittent asphyxia was induced for 30 min by exposing male and female postnatal day 11 rat pups to three 7 + 3-min cycles of 9% and 5% O2 at constant 20% CO2 . All pups exhibited convulsive seizures after asphyxia. A set of behavioral tests were performed systematically over 14 months following asphyxia, that is, a large part of the rat's life span. Video-electroencephalographic (EEG) monitoring was used to determine whether asphyxia led to the development of epilepsy. Finally, structural brain alterations were examined. RESULTS: The animals showed impaired spatial learning and memory and increased anxiety when tested at an age of 3-14 months. Video-EEG at ~10 months showed an abundance of spontaneous seizures, which was paralleled by neurodegeneration in the hippocampus and thalamus, and by aberrant mossy fiber sprouting. SIGNIFICANCE: The present model of birth asphyxia recapitulates several of the later life consequences associated with human HIE. This model thus allows evaluation of the efficacy of novel therapies designed to prevent HIE and seizures following asphyxia, and of how such therapies might alleviate long-term adverse consequences.


Asunto(s)
Asfixia Neonatal , Disfunción Cognitiva , Epilepsia , Hipoxia-Isquemia Encefálica , Animales , Animales Recién Nacidos , Ansiedad , Asfixia/complicaciones , Asfixia Neonatal/complicaciones , Asfixia Neonatal/tratamiento farmacológico , Encéfalo , Disfunción Cognitiva/complicaciones , Epilepsia/complicaciones , Femenino , Humanos , Hipoxia/complicaciones , Hipoxia-Isquemia Encefálica/complicaciones , Recién Nacido , Masculino , Ratas , Convulsiones/tratamiento farmacológico
13.
Int J Mol Sci ; 22(9)2021 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-34062911

RESUMEN

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


Asunto(s)
Asfixia Neonatal/tratamiento farmacológico , Isquemia Encefálica/tratamiento farmacológico , Ácido Quinurénico/análogos & derivados , Neuronas/metabolismo , Animales , Asfixia Neonatal/metabolismo , Asfixia Neonatal/patología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Región CA1 Hipocampal/diagnóstico por imagen , Región CA1 Hipocampal/efectos de los fármacos , Región CA3 Hipocampal/diagnóstico por imagen , Región CA3 Hipocampal/efectos de los fármacos , Modelos Animales de Enfermedad , Electroencefalografía , Potenciales Evocados Visuales/efectos de los fármacos , Humanos , Ácido Quinurénico/farmacología , Neuronas/efectos de los fármacos , Neuronas/patología , Ratas , Investigación Biomédica Traslacional
14.
Epilepsia ; 62(6): 1460-1471, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33955541

RESUMEN

OBJECTIVES: Bumetanide was suggested as an adjunct to phenobarbital for suppression of neonatal seizures. This suggestion was based on the idea that bumetanide, by reducing intraneuronal chloride accumulation through inhibition of the Na-K-2Cl cotransporter NKCC1, may attenuate or abolish depolarizing γ-aminobutyric acid (GABA) responses caused by birth asphyxia. However, a first proof-of-concept clinical trial failed. This could have had several reasons, including bumetanide's poor brain penetration, the wide cellular NKCC1 expression pattern in the brain, and problems with the general concept of NKCC1's role in neonatal seizures. We recently replicated the clinical failure of bumetanide to potentiate phenobarbital's effect in a novel rat model of birth asphyxia. In this study, a clinically relevant dose (0.3 mg/kg) of bumetanide was used that does not lead to NKCC1-inhibitory brain levels. The aim of the present experiments was to examine whether a much higher dose (10 mg/kg) of bumetanide is capable of potentiating phenobarbital in this rat model. Furthermore, the effects of the two lipophilic bumetanide derivatives, the ester prodrug N,N-dimethylaminoethylester of bumetanide (DIMAEB) and the benzylamine derivative bumepamine, were examined at equimolar doses. METHODS: Intermittent asphyxia was induced for 30 min by exposing male and female P11 rat pups to three 7 + 3 min cycles of 9% and 5% O2 at constant 20% CO2 . All control pups exhibited neonatal seizures after the asphyxia. RESULTS: Even at 10 mg/kg, bumetanide did not potentiate the effect of a submaximal dose (15 mg/kg) of phenobarbital on seizure incidence, whereas a significant suppression of neonatal seizures was determined for combinations of phenobarbital with DIMAEB or, more effectively, bumepamine, which, however, does not inhibit NKCC1. Of interest, the bumepamine/phenobarbital combination prevented the neurodegenerative consequences of asphyxia and seizures in the hippocampus. SIGNIFICANCE: Both bumepamine and DIMAEB are promising tools that may help to develop more effective lead compounds for clinical trials.


Asunto(s)
Anticonvulsivantes/farmacología , Asfixia Neonatal/complicaciones , Asfixia Neonatal/tratamiento farmacológico , Bencilaminas/uso terapéutico , Bumetanida/uso terapéutico , Hipocampo/patología , Degeneración Nerviosa/patología , Fenobarbital/farmacología , Convulsiones/tratamiento farmacológico , Convulsiones/etiología , Animales , Animales Recién Nacidos , Anticonvulsivantes/farmacocinética , Bencilaminas/farmacocinética , Encéfalo/metabolismo , Bumetanida/análogos & derivados , Bumetanida/farmacocinética , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Femenino , Masculino , Fenobarbital/farmacocinética , Embarazo , Ratas , Miembro 2 de la Familia de Transportadores de Soluto 12/biosíntesis
15.
J Matern Fetal Neonatal Med ; 34(18): 3080-3088, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31558088

RESUMEN

AIM: To evaluate the effects of prophylactic theophylline in renal function and survival rates of asphyxiated newborns. METHODS: Medline, Scopus, Cochrane Central Register of Controlled Trials, Clinicaltrials.gov and Google Scholar databases were systematically searched. All randomized controlled trials evaluating the efficacy of theophylline in the prevention of perinatal asphyxia were selected. RESULTS: A total of seven studies were included with a total of 458 asphyxiated neonates. Incidence of acute kidney injury was significantly lower in neonates receiving theophylline (OR: 0.24, 95% CI: [0.16, 0.36]), while mortality rates were similar between the two groups (OR: 0.86, 95% CI: [0.46, 1.62]). Theophylline administration was associated with significantly decreased serum creatinine levels (MD: -0.57 mg/dl, 95% CI: [-0.68, -0.46]) and elevated glomerular filtration rate (MD: 13.79 ml/min/1.73 m2, 95% CI: [11.91, 15.68]) in the third day of life. Theophylline also lead to lower ß2-microglobulin levels, higher urine output and negative fluid balance. CONCLUSIONS: The present findings suggest the effectiveness of theophylline in ameliorating renal function of asphyxiated neonates. Future large-scale trials should assess potential long-term adverse outcomes in clinical practice.KeynotesAsphyxia is a major cause of acute kidney injury in neonatesAcute kidney injury is associated with adverse clinical outcomes in asphyxiated neonates.Theophylline administration leads to significantly lower incidence of acute kidney injury in asphyxiated neonates.


Asunto(s)
Lesión Renal Aguda , Asfixia Neonatal , Lesión Renal Aguda/etiología , Lesión Renal Aguda/prevención & control , Asfixia , Asfixia Neonatal/complicaciones , Asfixia Neonatal/tratamiento farmacológico , Tasa de Filtración Glomerular , Humanos , Recién Nacido , Teofilina/uso terapéutico
16.
Epilepsia ; 62(4): 920-934, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33258158

RESUMEN

OBJECTIVE: Neonatal seizures are the most frequent type of neurological emergency in newborn infants, often being a consequence of prolonged perinatal asphyxia. Phenobarbital is currently the most widely used antiseizure drug for treatment of neonatal seizures, but fails to stop them in ~50% of cases. In a neonatal hypoxia-only model based on 11-day-old (P11) rats, the NKCC1 inhibitor bumetanide was reported to potentiate the antiseizure activity of phenobarbital, whereas it was ineffective in a human trial in neonates. The aim of this study was to evaluate the effect of clinically relevant doses of bumetanide as add-on to phenobarbital on neonatal seizures in a noninvasive model of birth asphyxia in P11 rats, designed for better translation to the human term neonate. METHODS: Intermittent asphyxia was induced for 30 minutes by exposing the rat pups to three 7 + 3-minute cycles of 9% and 5% O2 at constant 20% CO2 . Drug treatments were administered intraperitoneally either before or immediately after asphyxia. RESULTS: All untreated rat pups had seizures within 10 minutes after termination of asphyxia. Phenobarbital significantly blocked seizures when applied before asphyxia at 30 mg/kg but not 15 mg/kg. Administration of phenobarbital after asphyxia was ineffective, whereas midazolam (0.3 or 1 mg/kg) exerted significant antiseizure effects when administered before or after asphyxia. In general, focal seizures were more resistant to treatment than generalized convulsive seizures. Bumetanide (0.3 mg/kg) alone or in combination with phenobarbital (15 or 30 mg/kg) exerted no significant effect on seizure occurrence. SIGNIFICANCE: The data demonstrate that bumetanide does not increase the efficacy of phenobarbital in a model of birth asphyxia, which is consistent with the negative data of the recent human trial. The translational data obtained with the novel rat model of birth asphyxia indicate that it is a useful tool to evaluate novel treatments for neonatal seizures.


Asunto(s)
Asfixia Neonatal/tratamiento farmacológico , Bumetanida/uso terapéutico , Modelos Animales de Enfermedad , Midazolam/uso terapéutico , Fenobarbital/uso terapéutico , Convulsiones/tratamiento farmacológico , Animales , Animales Recién Nacidos , Anticonvulsivantes/uso terapéutico , Asfixia Neonatal/complicaciones , Asfixia Neonatal/fisiopatología , Femenino , Hipnóticos y Sedantes/uso terapéutico , Masculino , Ratas , Ratas Wistar , Convulsiones/etiología , Convulsiones/fisiopatología , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/uso terapéutico , Resultado del Tratamiento
17.
Am J Physiol Regul Integr Comp Physiol ; 319(6): R653-R665, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33074015

RESUMEN

Antenatal glucocorticoids improve outcomes among premature infants but are associated with hyperglycemia, which can exacerbate hypoxic-ischemic injury. It is still unclear how antenatal glucocorticoids or hyperglycemia modulate fetal cardiovascular adaptations to severe asphyxia. In this study, preterm fetal sheep received either saline or 12 mg im maternal dexamethasone, followed 4 h later by complete umbilical cord occlusion (UCO) for 25 min. An additional cohort of fetuses received titrated glucose infusions followed 4 h later by UCO to control for the possibility that hyperglycemia contributed to the cardiovascular effects of dexamethasone. Fetuses were studied for 7 days after UCO. Maternal dexamethasone was associated with fetal hyperglycemia (P < 0.001), increased arterial pressure (P < 0.001), and reduced femoral (P < 0.005) and carotid (P < 0.05) vascular conductance before UCO. UCO was associated with bradycardia, femoral vasoconstriction, and transient hypertension. For the first 5 min of UCO, fetal blood pressure in the dexamethasone-asphyxia group was greater than saline-asphyxia (P < 0.001). However, the relative increase in arterial pressure was not different from saline-asphyxia. Fetal heart rate and femoral vascular conductance fell to similar nadirs in both saline and dexamethasone-asphyxia groups. Dexamethasone did not affect the progressive decline in femoral vascular tone or arterial pressure during continuing UCO. By contrast, there were no effects of glucose infusions on the response to UCO. In summary, maternal dexamethasone but not fetal hyperglycemia increased fetal arterial pressure before and for the first 5 min of prolonged UCO but did not augment the cardiovascular adaptations to acute asphyxia.


Asunto(s)
Asfixia Neonatal/tratamiento farmacológico , Glucemia/efectos de los fármacos , Dexametasona/toxicidad , Corazón Fetal/efectos de los fármacos , Glucocorticoides/toxicidad , Hemodinámica/efectos de los fármacos , Hiperglucemia/inducido químicamente , Nacimiento Prematuro/tratamiento farmacológico , Animales , Animales Recién Nacidos , Presión Arterial/efectos de los fármacos , Asfixia Neonatal/sangre , Asfixia Neonatal/fisiopatología , Biomarcadores/sangre , Glucemia/metabolismo , Dexametasona/administración & dosificación , Modelos Animales de Enfermedad , Corazón Fetal/fisiopatología , Edad Gestacional , Glucocorticoides/administración & dosificación , Frecuencia Cardíaca/efectos de los fármacos , Hiperglucemia/sangre , Hiperglucemia/fisiopatología , Nacimiento Prematuro/sangre , Nacimiento Prematuro/fisiopatología , Oveja Doméstica , Factores de Tiempo
18.
Int J Mol Sci ; 21(18)2020 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-32948011

RESUMEN

Hypoxic-ischemic encephalopathy (HIE) is still a major cause of neonatal death and disability as therapeutic hypothermia (TH) alone cannot afford sufficient neuroprotection. The present study investigated whether ventilation with molecular hydrogen (2.1% H2) or graded restoration of normocapnia with CO2 for 4 h after asphyxia would augment the neuroprotective effect of TH in a subacute (48 h) HIE piglet model. Piglets were randomized to untreated naïve, control-normothermia, asphyxia-normothermia (20-min 4%O2-20%CO2 ventilation; Tcore = 38.5 °C), asphyxia-hypothermia (A-HT, Tcore = 33.5 °C, 2-36 h post-asphyxia), A-HT + H2, or A-HT + CO2 treatment groups. Asphyxia elicited severe hypoxia (pO2 = 19 ± 5 mmHg) and mixed acidosis (pH = 6.79 ± 0.10). HIE development was confirmed by altered cerebral electrical activity and neuropathology. TH was significantly neuroprotective in the caudate nucleus but demonstrated virtually no such effect in the hippocampus. The mRNA levels of apoptosis-inducing factor and caspase-3 showed a ~10-fold increase in the A-HT group compared to naïve animals in the hippocampus but not in the caudate nucleus coinciding with the region-specific neuroprotective effect of TH. H2 or CO2 did not augment TH-induced neuroprotection in any brain areas; rather, CO2 even abolished the neuroprotective effect of TH in the caudate nucleus. In conclusion, the present findings do not support the use of these medical gases to supplement TH in HIE management.


Asunto(s)
Asfixia Neonatal/terapia , Daño Encefálico Crónico/prevención & control , Dióxido de Carbono/uso terapéutico , Hidrógeno/uso terapéutico , Hipotermia Inducida , Hipoxia-Isquemia Encefálica/terapia , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Acidosis/sangre , Acidosis/etiología , Acidosis/prevención & control , Administración por Inhalación , Animales , Animales Recién Nacidos , Factor Inductor de la Apoptosis/biosíntesis , Factor Inductor de la Apoptosis/genética , Asfixia Neonatal/complicaciones , Asfixia Neonatal/tratamiento farmacológico , Daño Encefálico Crónico/etiología , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Factor Neurotrófico Derivado del Encéfalo/genética , Dióxido de Carbono/administración & dosificación , Dióxido de Carbono/toxicidad , Caspasa 3/biosíntesis , Caspasa 3/genética , Núcleo Caudado/patología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Electroencefalografía , Potenciales Evocados Visuales/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/patología , Hidrógeno/administración & dosificación , Hidrógeno/análisis , Hipoxia-Isquemia Encefálica/complicaciones , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Hipoxia-Isquemia Encefálica/patología , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Fármacos Neuroprotectores/administración & dosificación , Especificidad de Órganos , Distribución Aleatoria , Porcinos
19.
Arch Iran Med ; 23(5): 312-318, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32383615

RESUMEN

BACKGROUND: As there are different views on the effects of aminophylline on neonatal renal function, we intended to observe the effects of aminophylline on renal dysfunction in neonates with prenatal asphyxia. METHODS: This randomized trial was conducted in the Obstetrics and Gynecology Hospital, Tehran, Iran, from June 2016 to May 2017, in neonates with moderate to severe asphyxia during birth. Fifty-six neonates were divided randomly into two groups. The intervention group received one dose of 5mg/kg slow intravenous aminophylline injection and the placebo group received 2 mL/kg of intravenous 10% solution of dextrose saline during the first hour of life. They were monitored and compared for renal functional indices, electrolytes, and complications of asphyxia during the three days of life. RESULTS: The mean of Cr (37.9 ± 8.8 vs 38.5 ± 9.4 and 20.8 ± 4.8 vs 30.1 ± 5.2 µmol/L), GFR (21.55 ± 4.7 vs 20.25 ± 4.4 and 30.8 ± 7.1 vs 20.1 ± 6.5 mL/minute/1.73 m2), Na (135.1 ± 12.4 vs134.5 ± 11.2 and 128.9 ± 11.5 vs 134.2 ± 10.9 mEq/L), and urine output (98.2 ± 25 vs 96.8 ± 23 and 148.7 ± 35 vs 108.8 ± 20 cc) were in the aminophylline treated and placebo group on the 1st and 3rd days, respectively. The mean difference of Cr (-9.3 (-8.9; -9.7) µmol/L); (P = 0.02), GFR (10.7 (10.1; 11.3) mL/minute/1.73 m2) (P = 0.009), Na (-5.3 (-5.9; -4.7) mEq/L) (P = 0.002), and urine volume (39.9 (24.9; 54.9) cc) (P = 0.001) presented statistically significant differences on the third day between the intervention and placebo group. CONCLUSION: Aminophylline was effective in preventing renal dysfunction in neonates with asphyxia. Neonates who received aminophylline indicated a significant improvement in GFR and urine output on the first day of life.


Asunto(s)
Lesión Renal Aguda/prevención & control , Aminofilina/uso terapéutico , Asfixia Neonatal/tratamiento farmacológico , Antagonistas de Receptores Purinérgicos P1/uso terapéutico , Lesión Renal Aguda/fisiopatología , Método Doble Ciego , Femenino , Tasa de Filtración Glomerular , Humanos , Recién Nacido , Irán , Masculino , Orina
20.
BMC Neurol ; 20(1): 171, 2020 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-32366288

RESUMEN

BACKGROUND: Perinatal asphyxia, more appropriately known as hypoxic-ischemic encephalopathy (HIE), is a condition characterized by clinical and laboratory evidence of acute or sub-acute brain injury resulting from systemic hypoxemia and/or reduced cerebral blood flow. HIE is a common and devastating clinical condition in resource-poor countries with poor treatment outcome. This paper describes the protocol for an ongoing study that aims to evaluate the neuroprotective effects of Erythropoietin (EPO) as compared to routine care in the management of moderate to severe HIE among term infants. METHODS: This study is a double-blind randomized controlled trial that will be conducted in the neonatal wards of the Lagos University Teaching Hospital (LUTH), Lagos, Nigeria, over a two-year period after ethical approvals and consents. One hundred and twenty-eight term newborns (≥ 37 weeks gestation) diagnosed with moderate/ severe HIE at admission will be allocated by randomization to receive either EPO or normal saline. All the participants will be offered standard care according to the unit protocol for HIE. Baseline investigations and close monitoring of the babies are done until discharge. Participants are followed up for 2 years to monitor their outcome (death or neurological development) using standard instruments. DISCUSSION: Previous trials had shown that EPO confers neuroprotective benefits and improve neurological and behavioral outcome in infants with HIE both singly or as an adjuvant to therapeutic hypothermia. This study hypothesized that administering EPO to newborns with moderate /severe HIE can positively influence their clinical and neurological outcomes and will provide evidence to either support or disprove the usefulness of Erythropoietin as a sole agent in the treatment of HIE, especially in resource-limited environment with the highest burden of the disease. TRIAL REGISTRATION: The study has been registered with the Pan African Clinical trials registry on the 2nd of December 2018, with registration number PACTR201812814507775.


Asunto(s)
Asfixia Neonatal/tratamiento farmacológico , Eritropoyetina/uso terapéutico , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Lesiones Encefálicas/terapia , Circulación Cerebrovascular , Método Doble Ciego , Humanos , Recién Nacido , Neuroprotección , Fármacos Neuroprotectores/uso terapéutico , Nigeria , Evaluación de Resultado en la Atención de Salud , Ensayos Clínicos Controlados Aleatorios como Asunto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...