Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Dev Neurosci ; 38(4): 251-263, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27614933

RESUMO

Term asphyxiated newborns remain at risk of developing brain injury despite available neuropreventive therapies such as hypothermia. Neurorestorative treatments may be an alternative. This study investigated the effect of sildenafil on brain injury induced by neonatal hypoxia-ischemia (HI) at term-equivalent age. Neonatal HI was induced in male Long-Evans rat pups at postnatal day 10 (P10) by left common carotid ligation followed by a 2-hour exposure to 8% oxygen; sham-operated rat pups served as the control. Both groups were randomized to oral sildenafil or vehicle twice daily for 7 consecutive days. Gait analysis was performed on P27. At P30, the rats were sacrificed, and their brains were extracted. The surfaces of both hemispheres were measured on hematoxylin and eosin-stained brain sections. Mature neurons and endothelial cells were quantified near the infarct boundary zone using immunohistochemistry. HI caused significant gait impairment and a reduction in the size of the left hemisphere. Treatment with sildenafil led to an improvement in the neurological deficits as measured by gait analysis, as well as an improvement in the size of the left hemisphere. Sildenafil, especially at higher doses, also caused a significant increase in the number of neurons near the infarct boundary zone. In conclusion, sildenafil administered after neonatal HI may improve brain injury recovery by promoting neuronal populations.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Encéfalo/efeitos dos fármacos , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Recuperação de Função Fisiológica/efeitos dos fármacos , Citrato de Sildenafila/farmacologia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Masculino , Neurônios/efeitos dos fármacos
2.
Sci Rep ; 11(1): 22046, 2021 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-34764335

RESUMO

The hippocampus is a fundamental structure of the brain that plays an important role in neurodevelopment and is very sensitive to hypoxia-ischemia (HI). The purpose of this study was to investigate the effects of sildenafil on neonatal hippocampal brain injuries resulting from HI, and on neuronal development in this context. HI was induced in male Long-Evans rat pups at postnatal day 10 (P10) by a left common carotid ligation followed by a 2-h exposure to 8% oxygen. Rat pups were randomized to vehicle or sildenafil given orally twice daily for 7 days starting 12 h after HI. Hematoxylin and eosin staining was performed at P30 to measure the surface of the hippocampus; immunohistochemistry was performed to stain neurons, oligodendrocytes, and glial cells in the hippocampus. Western blots of the hippocampus were performed at P12, P17, and P30 to study the expression of neuronal markers and mTOR pathway. HI caused significant hippocampal atrophy and a significant reduction of the number of mature neurons, and induced reactive astrocytosis and microgliosis in the hippocampus. HI increased apoptosis and caused significant dysregulation of the normal neuronal development program. Treatment with sildenafil preserved the gross morphology of the hippocampus, reverted the number of mature neurons to levels comparable to sham rats, significantly increased both the immature and mature oligodendrocytes, and significantly reduced the number of microglia and astrocytes. Sildenafil also decreased apoptosis and reestablished the normal progression of post-natal neuronal development. The PI3K/Akt/mTOR pathway, whose activity was decreased after HI in the hippocampus, and restored after sildenafil treatment, may be involved. Sildenafil may have both neuroprotective and neurorestorative properties in the neonatal hippocampus following HI.


Assuntos
Hipocampo/efeitos dos fármacos , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Citrato de Sildenafila/uso terapêutico , Animais , Animais Recém-Nascidos , Hipocampo/patologia , Hipóxia-Isquemia Encefálica/patologia , Masculino , Neurogênese/efeitos dos fármacos , Ratos , Ratos Long-Evans , Vasodilatadores/uso terapêutico
3.
Neurosci Lett ; 440(1): 54-8, 2008 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-18541376

RESUMO

Feeding schedules that restrict food access to a predictable daytime meal induce in rodents food-anticipatory behaviors, changes in physiological rhythms and shifts in the rhythm of clock gene expression in the brain and periphery. However, little is known about the effects of nighttime restricted feeding. Previously, we showed that daytime restricted access to a highly palatable complete meal replacement, Ensure Plus (Ensure), shifts the rhythm of expression of the clock protein PER2 in limbic forebrain areas including the oval nucleus of the bed nucleus of the stria terminalis (BNSTov), central nucleus of the amygdala (CEA), basolateral amygdala (BLA) and dentate gyrus (DG), and induces a rhythm in the dorsomedial hypothalamic nucleus (DMH) in food deprived (restricted feeding), but not free-fed rats (restricted treat). In the present study we investigated the effects of nighttime restricted feeding (Ensure only, 2 h/night) and nighttime restricted treats (Ensure 2 h/night+free access to chow) in order to determine whether these effects were dependent on the time of day the meal was provided. We found that nighttime restricted feeding, like daytime restricted feeding, shifted the rhythm of PER2 expression in the BNSTov and CEA and peak expression was observed approximately 12 h after the mealtime. Also consistent with previous work, nighttime restricted feeding induced a rhythm of PER2 expression in the DMH and these effects occurred without affecting the rhythm in the suprachiasmatic nucleus (SCN). In contrast to previous work with daytime restricted feeding, nighttime restricted feeding had no effect on PER2 rhythms in the BLA and DG. Finally, nighttime restricted treats, as was the case for daytime restricted treats, had no effect on PER2 expression in any of the brain areas studied. The present results together with our previous findings show that the effect of restricted feeding on PER2 rhythms in the limbic forebrain and hypothalamus depend on a negative energy balance and vary as a function of time of day in a brain region-specific manner.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Ritmo Circadiano/fisiologia , Privação de Alimentos/fisiologia , Regulação da Expressão Gênica/fisiologia , Hipotálamo/metabolismo , Proteínas Nucleares/metabolismo , Prosencéfalo/metabolismo , Análise de Variância , Animais , Comportamento Animal , Proteínas de Ciclo Celular/genética , Masculino , Atividade Motora/fisiologia , Proteínas Nucleares/genética , Proteínas Circadianas Period , Ratos , Ratos Wistar , Fatores de Tempo
4.
Int J Dev Neurosci ; 49: 31-6, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26773167

RESUMO

Mesial temporal lobe epilepsy (MTLE) is a focal epileptic disorder that is frequently associated with hippocampal sclerosis. This study investigated whether blocking angiogenesis prevents the development of seizures and hippocampal atrophy in the pilocarpine rat model of MTLE. To block angiogenesis, a subset of animals were given sunitinib orally. Continuous video recordings were performed to identify seizures. Brains were then extracted and sectioned, and hippocampal surfaces and angiogenesis were assessed. After a latent period of 6.6 ± 2.6 days, the sham-treated pilocarpine rats presented convulsive seizures, while the pilocarpine rats treated with sunitinib did not develop seizures. Sham-treated pilocarpine rats but not sunitinib-treated pilocarpine rats had significantly smaller hippocampi. Endothelial cell counts in sham-treated pilocarpine rats were significantly greater than in controls and sunitinib-treated pilocarpine rats. Blocking angiogenesis immediately following the initial insult in this animal model prevented thus angiogenesis and hippocampal atrophy and averted the development of clinical seizures.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Epilepsia do Lobo Temporal/complicações , Hipocampo/irrigação sanguínea , Indóis/uso terapêutico , Neovascularização Patológica , Pirróis/uso terapêutico , Inibidores da Angiogênese/farmacologia , Animais , Contagem de Células , Modelos Animais de Doenças , Epilepsia do Lobo Temporal/induzido quimicamente , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Indóis/farmacologia , Lectinas/metabolismo , Masculino , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/etiologia , Neovascularização Patológica/patologia , Pilocarpina/toxicidade , Pirróis/farmacologia , Ratos , Ratos Sprague-Dawley , Estatísticas não Paramétricas , Sunitinibe , Fatores de Tempo
5.
Invest Ophthalmol Vis Sci ; 57(10): 4306-14, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27552408

RESUMO

PURPOSE: The purpose of this study was to investigate the effects of sildenafil on retinal injury following neonatal hypoxia-ischemia (HI) at term-equivalent age in rat pups. METHODS: Hypoxia-ischemia was induced in male Long-Evans rat pups at postnatal day 10 (P10) by a left common carotid ligation followed by a 2-hour exposure to 8% oxygen. Sham-operated rats served as the control group. Both groups were administered vehicle or 2, 10, or 50 mg/kg sildenafil, twice daily for 7 consecutive days. Retinal function was assessed by flash electroretinograms (ERGs) at P29, and retinal structure was assessed by retinal histology at P30. RESULTS: Hypoxia-ischemia caused significant functional (i.e., attenuation of the ERG a-wave and b-wave amplitudes and photopic negative response) and structural (i.e., thinning of the total retina, especially the inner retinal layers) retinal damage in the left eyes (i.e., ipsilateral to the carotid ligation). Treatment with the different doses of sildenafil led to a dose-dependent increase in the amplitudes of the ERG a- and b-waves and of the photopic negative response in HI animals, with higher doses associated with greater effect sizes. Similarly, a dose response was observed in terms of improvements in the retinal layer thicknesses. CONCLUSIONS: Hypoxia-ischemia at term-equivalent age induced functional and structural damage mainly to the inner retina. Treatment with sildenafil provided a dose-dependent recovery of retinal function and structure.


Assuntos
Hipóxia/tratamento farmacológico , Traumatismo por Reperfusão/tratamento farmacológico , Retina/patologia , Retina/fisiopatologia , Doenças Retinianas/tratamento farmacológico , Citrato de Sildenafila/administração & dosagem , Administração Oral , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Eletrorretinografia/efeitos dos fármacos , Feminino , Seguimentos , Hipóxia/complicações , Hipóxia/patologia , Masculino , Inibidores da Fosfodiesterase 5/administração & dosagem , Ratos , Ratos Long-Evans , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/patologia , Retina/efeitos dos fármacos , Doenças Retinianas/etiologia , Doenças Retinianas/fisiopatologia
6.
Biol Open ; 5(4): 475-83, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26988760

RESUMO

Many premature newborns develop bronchopulmonary dysplasia (BPD), a chronic lung disease resulting from prolonged mechanical ventilation and hyperoxia. BPD survivors typically suffer long-term injuries not only to the lungs, but also to the brain and retina. However, currently it is not clear whether the brain and retinal injuries in these newborns are related only to their prematurity, or also to BPD. We investigated whether the hyperoxia known to cause histologic changes in the lungs similar to BPD in an animal model also causes brain and retinal injuries. Sprague Dawley rat pups were exposed to hyperoxia (95% O2, 'BPD' group) or room air (21% O2, 'control' group) from postnatal day 4-14 (P4-14); the rat pups were housed in room air between P14 and P28. At P28, they were sacrificed, and their lungs, brain, and eyes were extracted. Hematoxylin and eosin staining was performed on lung and brain sections; retinas were stained with Toluidine Blue. Hyperoxia exposure resulted in an increased mean linear intercept in the lungs (P<0.0001). This increase was associated with a decrease in some brain structures [especially the whole-brain surface (P=0.02)], as well as a decrease in the thickness of the retinal layers [especially the total retina (P=0.0008)], compared to the room air control group. In addition, a significant negative relationship was observed between the lung structures and the brain (r=-0.49,P=0.02) and retina (r=-0.70,P=0.0008) structures. In conclusion, hyperoxia exposure impaired lung, brain, and retina structures. More severe lung injuries correlated with more severe brain and retinal injuries. This result suggests that the same animal model of chronic neonatal hyperoxia can be used to simultaneously study lung, brain and retinal injuries related to hyperoxia.

7.
PLoS One ; 10(5): e0128028, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25996847

RESUMO

BACKGROUND: Neonatal encephalopathy following birth asphyxia is a major predictor of long-term neurological impairment. Therapeutic hypothermia is currently the standard of care to prevent brain injury in asphyxiated newborns but is not protective in all cases. More robust and versatile treatment options are needed. Angiogenesis is a demonstrated therapeutic target in adult stroke. However, no systematic study examines the expression of angiogenesis-related markers following birth asphyxia in human newborns. OBJECTIVE: This study aimed to evaluate the expression of angiogenesis-related protein markers in asphyxiated newborns developing and not developing brain injury compared to healthy control newborns. DESIGN/METHODS: Twelve asphyxiated newborns treated with hypothermia were prospectively enrolled; six developed eventual brain injury and six did not. Four healthy control newborns were also included. We used Rules-Based Medicine multi-analyte profiling and protein array technologies to study the plasma concentration of 49 angiogenesis-related proteins. Mean protein concentrations were compared between each group of newborns. RESULTS: Compared to healthy newborns, asphyxiated newborns not developing brain injury showed up-regulation of pro-angiogenic proteins, including fatty acid binding protein-4, glucose-6-phosphate isomerase, neuropilin-1, and receptor tyrosine-protein kinase erbB-3; this up-regulation was not evident in asphyxiated newborns eventually developing brain injury. Also, asphyxiated newborns developing brain injury showed a decreased expression of anti-angiogenic proteins, including insulin-growth factor binding proteins -1, -4, and -6, compared to healthy newborns. CONCLUSIONS: These findings suggest that angiogenesis pathways are dysregulated following birth asphyxia and are putatively involved in brain injury pathology and recovery.


Assuntos
Asfixia Neonatal/sangue , Asfixia Neonatal/terapia , Hipotermia Induzida , Neovascularização Fisiológica/fisiologia , Biomarcadores/sangue , Feminino , Humanos , Recém-Nascido , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA