Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
1.
Neurotox Res ; 41(6): 526-545, 2023 Dec.
Article En | MEDLINE | ID: mdl-37378827

Neonatal hypoxia-ischemia (HI) is one of the main causes of tissue damage, cell death, and imbalance between neuronal excitation and inhibition and synaptic loss in newborns. GABA, the major inhibitory neurotransmitter of the central nervous system (CNS) in adults, is excitatory at the onset of neurodevelopment and its action depends on the chloride (Cl-) cotransporters NKCC1 (imports Cl-) and KCC2 (exports Cl-) expression. Under basal conditions, the NKCC1/KCC2 ratio decreases over neurodevelopment. Thus, changes in this ratio caused by HI may be related to neurological disorders. The present study evaluated the effects of bumetanide (NKCC cotransporters inhibitor) on HI impairments in two neurodevelopmental periods. Male Wistar rat pups, 3 (PND3) and 11 (PND11) days old, were submitted to the Rice-Vannucci model. Animals were divided into 3 groups: SHAM, HI-SAL, and HI-BUM, considering each age. Bumetanide was administered intraperitoneally at 1, 24, 48, and 72 h after HI. NKCC1, KCC2, PSD-95, and synaptophysin proteins were analyzed after the last injection by western blot. Negative geotaxis, righting reflex, open field, object recognition test, and Morris water maze task were performed to assess neurological reflexes, locomotion, and memory function. Tissue atrophy and cell death were evaluated by histology. Bumetanide prevented neurodevelopmental delay, hyperactivity, and declarative and spatial memory deficits. Furthermore, bumetanide reversed HI-induced brain tissue damage, reduced neuronal death and controlled GABAergic tone, maintained the NKCC1/KCC2 ratio, and synaptogenesis close to normality. Thereby, bumetanide appears to play an important therapeutic role in the CNS, protecting the animals against HI damage and improving functional performance.


Bumetanide , Hypoxia-Ischemia, Brain , Rats , Animals , Male , Bumetanide/pharmacology , Bumetanide/therapeutic use , Rats, Wistar , Solute Carrier Family 12, Member 2/metabolism , Ischemia/drug therapy , Hypoxia/drug therapy , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/drug therapy , Brain/metabolism , Cognition , Animals, Newborn
2.
Neurotox Res ; 41(2): 119-140, 2023 Apr.
Article En | MEDLINE | ID: mdl-36580261

L-2-Hydroxyglutaric aciduria (L-2-HGA) is an inherited neurometabolic disorder caused by deficient activity of L-2-hydroxyglutarate dehydrogenase. L-2-Hydroxyglutaric acid (L-2-HG) accumulation in the brain and biological fluids is the biochemical hallmark of this disease. Patients present exclusively neurological symptoms and brain abnormalities, particularly in the cerebral cortex, basal ganglia, and cerebellum. Since the pathogenesis of this disorder is still poorly established, we investigated the short-lived effects of an intracerebroventricular injection of L-2-HG to neonatal rats on redox homeostasis in the cerebellum, which is mostly affected in this disorder. We also determined immunohistochemical landmarks of neuronal viability (NeuN), astrogliosis (S100B and GFAP), microglia activation (Iba1), and myelination (MBP and CNPase) in the cerebral cortex and striatum following L-2-HG administration. Finally, the neuromotor development and cognitive abilities were examined. L-2-HG elicited oxidative stress in the cerebellum 6 h after its injection, which was verified by increased reactive oxygen species production, lipid oxidative damage, and altered antioxidant defenses (decreased concentrations of reduced glutathione and increased glutathione peroxidase and superoxide dismutase activities). L-2-HG also decreased the content of NeuN, MBP, and CNPase, and increased S100B, GFAP, and Iba1 in the cerebral cortex and striatum at postnatal days 15 and 75, implying long-standing neuronal loss, demyelination, astrocyte reactivity, and increased inflammatory response, respectively. Finally, L-2-HG administration caused a delay in neuromotor development and a deficit of cognition in adult animals. Importantly, the antioxidant melatonin prevented L-2-HG-induced deleterious neurochemical, immunohistochemical, and behavioral effects, indicating that oxidative stress may be central to the pathogenesis of brain damage in L-2-HGA.


Antioxidants , Oxidative Stress , Rats , Animals , Antioxidants/pharmacology , Animals, Newborn
3.
Int J Dev Neurosci ; 83(2): 165-177, 2023 Apr.
Article En | MEDLINE | ID: mdl-36453268

Nowadays, the only treatment for human babies suffering from hypoxia-ischemia (HI) is therapeutic hypothermia (TH). However, a better understanding of the specific effects of TH in males and females is important to improve its clinical application. The present study evaluated the short-term effects of TH on the brain injury and behavioral outcomes in male and female neonatal rats submitted to neonatal HI. Seven-day-old Wistar rats underwent a surgery for unilateral occlusion of the right common carotid artery and were exposed to a hypoxic atmosphere (8% oxygen) for 75 min. Then, the animals in the TH group were submitted to TH (scalp temperature of 32°C) for 5 h. In the behavioral tests, no remarkable differences triggered by HI or TH were observed relative to SHAM animals. Only females of the HI group presented lower latency to complete the righting reflex test. TH reduced the volume of brain injury in males, but not in females. The animals of the HI group showed a reduction in the number of neurons in the CA1 and dentate gyrus (DG) regions of the hippocampus and TH partially prevented neuronal death. In the CA1 region of the hippocampus, animals from the HI group showed more degenerating cells relative to the SHAM, which was reversed by TH. In the DG, animals from the HI group showed an increase in the number of degenerating neurons, which was partially reversed by TH only in males. Our data show that HI leads to a brain injury, which was attenuated by TH in a sex-dependent way and clarify the importance of the assessment of males and females in order to outline specific strategies for the treatment of each sex in newborns suffering from HI.


Brain Injuries , Hypothermia, Induced , Hypoxia-Ischemia, Brain , Humans , Rats , Animals , Male , Female , Rats, Wistar , Animals, Newborn , Hypoxia-Ischemia, Brain/therapy , Ischemia/therapy , Hypoxia , Brain
4.
Nutrients ; 14(2)2022 Jan 17.
Article En | MEDLINE | ID: mdl-35057576

The disruption of redox homeostasis and neuroinflammation are key mechanisms in the pathogenesis of brain hypoxia-ischemia (HI); medicinal plants have been studied as a therapeutic strategy, generally associated with the prevention of oxidative stress and inflammatory response. This study evaluates the neuroprotective role of the Plinia trunciflora fruit extract (PTE) in neonatal rats submitted to experimental HI. The HI insult provoked a marked increase in the lipoperoxidation levels and glutathione peroxidase (GPx) activity, accompanied by a decrease in the brain concentration of glutathione (GSH). Interestingly, PTE was able to prevent most of the HI-induced pro-oxidant effects. It was also observed that HI increased the levels of interleukin-1ß in the hippocampus, and that PTE-treatment prevented this effect. Furthermore, PTE was able to prevent neuronal loss and astrocyte reactivity induced by HI, as demonstrated by NeuN and GFAP staining, respectively. PTE also attenuated the anxiety-like behavior and prevented the spatial memory impairment caused by HI. Finally, PTE prevented neural tissue loss in the brain hemisphere, the hippocampus, cerebral cortex, and the striatum ipsilateral to the HI. Taken together our results provide good evidence that the PTE extract has the potential to be investigated as an adjunctive therapy in the treatment of brain insult caused by neonatal hypoxia-ischemia.


Hypoxia-Ischemia, Brain/drug therapy , Myrtaceae/chemistry , Neuroinflammatory Diseases/prevention & control , Neuroprotective Agents , Plant Extracts/administration & dosage , Animals , Animals, Newborn , Behavior, Animal/drug effects , Brain/drug effects , Brain/pathology , Brain/physiopathology , Fruit/chemistry , Glutathione Peroxidase/metabolism , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/physiopathology , Lipid Peroxidation/drug effects , Male , Neurons/pathology , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Rats , Rats, Wistar
5.
JIMD Rep ; 60(1): 23-31, 2021 Jul.
Article En | MEDLINE | ID: mdl-34258138

BACKGROUND: Morquio B disease (MBD) is a distinct GLB1-related dysostosis multiplex presenting a mild phenocopy of GALNS-related Morquio A disease. Previously reported cases from European countries carry the W273L variant on at least one GLB1 allele and exhibit a pure skeletal phenotype (pure MBD). Only a minority of MBD cases have been described with additional neuronopathic findings (MBD plus). OBJECTIVES AND METHODS: With the aim to further describe patterns of MBD-related dysostosis multiplex, we analyzed clinical, biochemical, and genetic features in 17 cases with GLB1-related dysostosis multiplex living and diagnosed in Brazil. RESULTS: About 14 of the 17 individuals had three or more skeletal findings characteristic of Morquio syndrome. Two had no additional neuronopathic features (pure MBD) and 12 exhibited additional neuronopathic features (MBD plus). Three of the 17 cases had mild dysostosis without distinct features of MBD. Seven of the 12 MBD plus patients had signs of spinal cord compression (SCC), as a result of progressive spinal vertebral dysostosis. There was an age-dependent increase in the number of skeletal findings and in the severity of growth impairment. GLB1 mutation analysis was completed in 10 of the 14 MBD patients. T500A occurred in compound heterozygosity in 8 of the 19 alleles. CONCLUSION: Our study extends the phenotypic spectrum of GLB1-related conditions by describing a cohort of patients with MBD and GM1-gangliosidosis (MBD plus). Targeting the progressive nature of the skeletal manifestations in the development of new therapies for GLB1-related conditions is warranted.

6.
J Neurochem ; 157(6): 1911-1929, 2021 06.
Article En | MEDLINE | ID: mdl-33098090

Prenatal and early postnatal periods are important for brain development and neural function. Neonatal insults such as hypoxia-ischemia (HI) causes prolonged neural and metabolic dysregulation, affecting central nervous system maturation. There is evidence that brain hypometabolism could increase the risk of adult-onset neurodegenerative diseases. However, the impact of non-pharmacologic strategies to attenuate HI-induced brain glucose dysfunction is still underexplored. This study investigated the long-term effects of early environmental enrichment in metabolic, cell, and functional responses after neonatal HI. Thereby, male Wistar rats were divided according to surgical procedure, sham, and HI (performed at postnatal day 3), and the allocation to standard (SC) or enriched condition (EC) during gestation and lactation periods. In-vivo cerebral metabolism was assessed by means of [18 F]-FDG micro-positron emission tomography, and cognitive, biochemical, and histological analyses were performed in adulthood. Our findings reveal that HI causes a reduction in glucose metabolism and glucose transporter levels as well as hyposynchronicity in metabolic brain networks. However, EC during prenatal or early postnatal period attenuated these metabolic disturbances. A positive correlation was observed between [18 F]-FDG values and volume ratios in adulthood, indicating that preserved tissue by EC is metabolically active. EC promotes better cognitive scores, as well as down-regulation of amyloid precursor protein in the parietal cortex and hippocampus of HI animals. Furthermore, growth-associated protein 43 was up-regulated in the cortex of EC animals. Altogether, results presented support that EC during gestation and lactation period can reduce HI-induced impairments that may contribute to functional decline and progressive late neurodegeneration.


Brain/metabolism , Environment , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/prevention & control , Neuronal Plasticity/physiology , Prenatal Exposure Delayed Effects/metabolism , Animals , Animals, Newborn , Female , Hypoxia-Ischemia, Brain/psychology , Lactation/metabolism , Lactation/psychology , Male , Maze Learning/physiology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/prevention & control , Neurodegenerative Diseases/psychology , Positron-Emission Tomography/methods , Pregnancy , Prenatal Exposure Delayed Effects/psychology , Rats , Rats, Wistar
7.
Res Dev Disabil ; 108: 103807, 2021 Jan.
Article En | MEDLINE | ID: mdl-33161308

INTRODUCTION: Cerebral palsy (CP) is one of the main causes of physical disabilities in childhood. There is evidence that CP children display high levels of stress, which could interfere with learning processes and interpretation of relevant sensory information during motor skills acquisition and socialization. OBJECTIVE: This study aims to compare basal levels of stress biomarkers (cortisol and alpha-amylase) of healthy children (HC) and children with CP, and to investigate whether a physical therapy session using the neurodevelopmental technique (NDT) interferes with these levels. METHODS: A cross-sectional design was used. A total of 86 children (HC: n = 45 and CP: n = 41) with matching age, sex, socioeconomic status, and sampling time. Salivary cortisol and alpha-amylase levels were measured by means of electrochemiluminescence and spectrophotometry methods. A single saliva sample was collected in the HC group to determine basal levels. For CP group three samples were collected: a first sample was taken 20-30 min prior to the intervention, while two post-intervention samples were collected (5 and 20 min) to evaluate individual changes in salivary stress biomarkers. RESULTS: Higher basal cortisol concentration was found in CP children when compared to HC group. Moreover, CP children showed a significant reduction in cortisol levels 20 min after NDT intervention. No significant differences were observed in alpha-amylase values. CONCLUSION: Present results show that CP causes alteration in basal cortisol values at childhood and suggest that CP children respond to environmental regulatory factors such as NDT, in attempt to reduce stress.


Cerebral Palsy , Salivary alpha-Amylases , Biomarkers , Child , Cross-Sectional Studies , Humans , Hydrocortisone , Physical Therapy Modalities , Saliva , Stress, Psychological
8.
Neurobiol Learn Mem ; 171: 107207, 2020 05.
Article En | MEDLINE | ID: mdl-32147586

BACKGROUND AND PURPOSE: Hypoxia and cerebral ischemia (HI) events are capable of triggering important changes in brain metabolism, including glucose metabolism abnormalities, which may be related to the severity of the insult. Using positron emission microtomography (microPET) with [18F]fluorodeoxyglucose (18F-FDG), this study proposes to assess abnormalities of brain glucose metabolism in adult rats previously submitted to the neonatal HI model. We hypothesize that cerebral metabolic outcomes will be associated with cognitive deficits and magnitude of brain injury. METHODS: Seven-day-old rats were subjected to an HI model, induced by permanent occlusion of the right common carotid artery and systemic hypoxia. 18F-FDG-microPET was used to assess regional and whole brain glucose metabolism in rats at 60 postnatal days (PND 60). An interregional cross-correlation matrix was utilized to construct metabolic brain networks (MBN). Rats were also subjected to the Morris Water Maze (MWM) to evaluate spatial memory and their brains were processed for volumetric evaluation. RESULTS: Brain glucose metabolism changes were observed in adult rats after neonatal HI insult, limited to the right brain hemisphere. However, not all HI animals exhibited significant cerebral hypometabolism. Hippocampal glucose metabolism was used to stratify HI animals into HI hypometabolic (HI-h) and HI non-hypometabolic (HI non-h) groups. The HI-h group had drastic MBN disturbance, cognitive deficit, and brain tissue loss, concomitantly. Conversely, HI non-h rats had normal brain glucose metabolism and brain tissue preserved, but also presented MBN changes and spatial memory impairment. Furthermore, data showed that brain glucose metabolism correlated with cognitive deficits and brain volume outcomes. CONCLUSIONS: Our findings demonstrated that long-term changes in MBN drive memory impairments in adult rats subjected to neonatal hypoxic ischemia, using in vivo imaging microPET-FDG. The MBN analyses identified glucose metabolism abnormalities in HI non-h animals, which were not detected by conventional 18F-FDG standardized uptake value (SUVr) measurements. These animals exhibited a metabolic brain signature that may explain the cognitive deficit even with no identifiable brain damage.


Brain/metabolism , Hypoxia-Ischemia, Brain/metabolism , Memory Disorders/metabolism , Nerve Net/metabolism , Animals , Brain/diagnostic imaging , Disease Models, Animal , Glucose/metabolism , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/diagnostic imaging , Male , Memory Disorders/diagnostic imaging , Memory Disorders/etiology , Nerve Net/diagnostic imaging , Positron-Emission Tomography , Rats , Rats, Wistar
9.
Pediatr Res ; 82(3): 544-553, 2017 Sep.
Article En | MEDLINE | ID: mdl-28426648

BackgroundHypoxia-ischemia (HI) is a major cause of neurological damage in preterm newborn. Swimming during pregnancy alters the offspring's brain development. We tested the effects of swimming during pregnancy in the very immature rat brain.MethodsFemale Wistar rats (n=12) were assigned to the sedentary (SE, n=6) or the swimming (SW, n=6) group. From gestational day 0 (GD0) to GD21 the rats in the SW group were made to swim for 20 min/day. HI on postnatal day (PND) 3 rats caused sensorimotor and cognitive impairments. Animals were distributed into SE sham (SESH), sedentary HIP3 (SEHI), swimming sham (SWSH), and swimming HIP3 (SWHI) groups. At PND4 and PND5, Na+/K+-ATPase activity and brain-derived neurotrophic factor (BDNF) levels were assessed. During lactation and adulthood, neurological reflexes, sensorimotor, anxiety-related, and cognitive evaluations were made, followed by histological assessment at PND60.ResultsAt early stages, swimming caused an increase in hippocampal BDNF levels and in the maintenance of Na+/K+-ATPase function in the SWHI group. The SWHI group showed smaller lesions and the preservation of white matter tracts. SEHI animals showed a delay in reflex maturation, which was reverted in the SWHI group. HIP3 induced spatial memory deficits and hypomyelination in SEHI rats, which was reverted in the SWHI group.ConclusionSwimming during pregnancy neuroprotected the brains against HI in very immature neonatal rats.


Hypoxia-Ischemia, Brain/prevention & control , Neuroprotection , Swimming , Animals , Behavior, Animal , Brain-Derived Neurotrophic Factor/metabolism , Female , Hippocampus/metabolism , Parietal Lobe/enzymology , Pregnancy , Rats , Rats, Wistar , Reflex , Sodium-Potassium-Exchanging ATPase/metabolism
10.
J Neurosci Res ; 95(1-2): 409-421, 2017 01 02.
Article En | MEDLINE | ID: mdl-27870406

Neonatal hypoxia-ischemia (HI) is an important cause of neurological deficits in humans, and the Levine-Rice model of experimental HI in the rat mimics the human brain lesion and the following sensory motor deficits and cognitive disabilities. With the growing evidence that sex influences all levels of brain functions, this Mini-Review highlights studies in which sex was a controlled variable and that provided evidence of sexual dimorphism in behavioral outcome, extension of brain damage, mechanisms of lesion, and treatment efficacy in the rat neonatal HI model. It was shown that 1) females have greater memory deficits; 2) cell death is dependent mainly on caspase activation in females; 3) males are more susceptible to oxidative stress; and 4) treatments acting on distinct cell death pathways afford sex-dependent neuroprotection. These tentative conclusions, along with growing evidence from other fields of neurobiology, support the need for scientists to design their experiments considering sex as an important variable; otherwise, important knowledge will continue to be missed. It is conceivable that sex can influence the development of efficacious therapeutic tools to treat neonates suffering from brain HI. © 2016 Wiley Periodicals, Inc.


Brain/pathology , Developmental Disabilities/etiology , Hypoxia-Ischemia, Brain/complications , Sex Characteristics , Animals , Animals, Newborn , Disease Models, Animal , Female , Hypoxia-Ischemia, Brain/pathology , Hypoxia-Ischemia, Brain/physiopathology , Male , Rats
...