Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
2.
J Neurosci Res ; 91(11): 1463-72, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24038156

RESUMO

Supraphysiological oxygen concentrations are toxic to the developing brain. Inflammatory processes increase the risk for brain injury. Sigma-1 receptor agonists are potent suppressors of inflammation-related events and are powerful immunomodulatory and antioxidative agents. Neuroprotective effects of sigma-1 receptor agonists have been described previously for neonatal and adult models of brain injury. The aim of this study was to assess the selective sigma-1 receptor agonist 2-(4-morpholinethyl)1-phenylcyclohexanecarboxylate (PRE-084) in models of inflammation-sensitized hyperoxia-induced developing brain injury. For in vivo studies, rat pups were randomly presensitized with 1) lipopolysaccharide or 2) vehicle on postnatal day 3. On day 6, pups received either 1) PRE-084 or 2) vehicle and were subsequently exposed to hyperoxic conditions for 6, 12, or 24 hr. At the end of exposure, animals were sacrificed and brains were processed for caspase-3 analysis using immunohistochemistry and Western blotting. For in vitro studies, oligodendroglial cells were subjected to hyperoxic conditions in the presence or absence of proinflammatory cytokines and PRE-084. Cell membrane integrity and cell viability were assessed by means of lactate dehydrogenase and 2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide (XTT) assays. Inflammatory presensitization significantly increased hyperoxia-induced injury both in vivo and in vitro. PRE-084 administration did not attenuate damage. Sigma-1 receptor agonists have been described as a promising therapeutic strategy for brain injury. We were not able to confirm this in the present model. The exact mechanisms of action of sigma-1 receptor agonists as well as the pathophysiologic pathways involved in hyperoxia-induced injury in the developing brain remain to be elucidated.


Assuntos
Lesões Encefálicas/metabolismo , Hiperóxia/metabolismo , Inflamação/metabolismo , Morfolinas/farmacologia , Fármacos Neuroprotetores/farmacologia , Animais , Western Blotting , Lesões Encefálicas/etiologia , Modelos Animais de Doenças , Imunofluorescência , Hiperóxia/complicações , Imuno-Histoquímica , Inflamação/complicações , Ratos , Ratos Wistar , Receptores sigma/agonistas , Receptor Sigma-1
3.
PLoS One ; 18(4): e0284096, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37023080

RESUMO

Neurodevelopmental impairment is a significant complication among survivors of preterm birth. To improve outcomes, reliable biomarkers for early detection of brain injury and prognostic assessment are required. Secretoneurin is a promising early biomarker of brain injury in adults and full-term neonates suffering from perinatal asphyxia. Data on preterm infants is currently lacking. The aim of this pilot study was to determine secretoneurin concentrations in preterm infants in the neonatal period, and to assess secretoneurin's potential as a biomarker of preterm brain injury. We included 38 very preterm infants (VPI) born at <32 weeks' gestation in the study. Secretoneurin concentrations were measured in serum samples obtained from the umbilical cord, at 48 hours and 3 weeks of life. Outcome measures included repeated cerebral ultrasonography, magnetic resonance imaging at term-equivalent age, general movements assessment, and neurodevelopmental assessment at a corrected age of 2 years by the Bayley Scales of Infant and Toddler Development, third edition (Bayley-III). In comparison to a term-born reference population, VPI had lower secretoneurin serum concentrations in umbilical cord blood and blood collected at 48 hours of life. When measured at 3 weeks of life, concentrations correlated with gestational age at birth. Secretoneurin concentrations did not differ between VPI with an imaging-based diagnosis of brain injury and those without, but when measured in umbilical cord blood and at 3 weeks of life correlated with and were predictive of Bayley-III motor and cognitive scale scores. Secretoneurin levels in VPI differ from term-born neonates. Secretoneurin seems unsuitable as a diagnostic biomarker of preterm brain injury, but bears some prognostic potential and is worthy of further investigation as a blood-based biomarker of preterm brain injury.


Assuntos
Lesões Encefálicas , Doenças do Prematuro , Nascimento Prematuro , Lactente , Gravidez , Feminino , Humanos , Recém-Nascido , Pré-Escolar , Recém-Nascido Prematuro , Projetos Piloto , Nascimento Prematuro/patologia , Lesões Encefálicas/diagnóstico , Lesões Encefálicas/patologia , Idade Gestacional , Biomarcadores , Doenças do Prematuro/patologia , Encéfalo/diagnóstico por imagem , Encéfalo/patologia
4.
Neonatology ; 119(2): 204-213, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35073542

RESUMO

INTRODUCTION: Perinatal asphyxia is a leading cause of neonatal death. Up to one-third of asphyxiated neonates suffer from hypoxic-ischaemic encephalopathy (HIE) with substantial long-term morbidity. Currently available diagnostic and prognostic tools bear limitations, and additional reliable biomarkers are needed for all stages of clinical management. A novel tool in neuroscientific research is micro-ribonucleic acid (miRNA) profiling. The aim of the present study was to determine miRNA expression profiles of healthy and asphyxiated neonates with and without HIE and to assess their potential as diagnostic and prognostic biomarkers. METHODS: We prospectively enrolled 49 neonates with a gestational age of ≥36 weeks, 15 of which fulfilled the diagnostic criteria of perinatal asphyxia and 34 served as healthy controls. Dried blood spots were collected from umbilical cord blood (UCB) and from venous blood upon admission to neonatal intensive care unit (NICU) and at 48 h of life. Samples were analysed by means of FirePlex™ technology (Abcam, Cambridge, MA, USA). RESULTS: In the UCB, miRNA expression levels of hsa-mir-124-3p, hsa-mir-1285-5p, and hsa-mir-331-5p were significantly lower in asphyxiated neonates compared to healthy controls. Asphyxiated neonates requiring therapeutic hypothermia had significantly increased expression of hsa-miR-30e-5p and significantly decreased expression of hsa-miR-142-3p, hsa-miR-338-3p, hsa-miR-34b-3p, hsa-miR-497-5p, and hsa-miR-98-5p at the time of admission to the NICU. At 48 h, infants suffering from moderate/severe HIE with a poor long-term neurodevelopmental outcome showed a significant increase in hsa-mir-145-5p. DISCUSSION/CONCLUSION: MiRNA profiling shows promise as a biomarker for perinatal asphyxia, hypothermia-requiring HIE, and poor neurodevelopmental outcome. Confirmatory studies are called for.


Assuntos
Asfixia Neonatal , Hipóxia-Isquemia Encefálica , MicroRNAs , Asfixia , Asfixia Neonatal/complicações , Asfixia Neonatal/diagnóstico , Asfixia Neonatal/genética , Biomarcadores , Feminino , Humanos , Hipóxia-Isquemia Encefálica/diagnóstico , Hipóxia-Isquemia Encefálica/genética , Lactente , Recém-Nascido , MicroRNAs/genética , Gravidez , Prognóstico
5.
IBRO Rep ; 9: 247-257, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33024879

RESUMO

Levomepromazine (LMP) is a phenothiazine neuroleptic drug with strong analgesic and sedative properties that is increasingly used off-label in pediatrics and is being discussed as an adjunct therapy in neonatal intensive care. Basic research points towards neuroprotective potential of phenothiazines, but LMP's effect on the developing brain is currently unknown. The aim of the present study was to assess LMP as a pharmacologic strategy in established neonatal in vitro and in vivo models of the healthy and injured developing mouse brain. In vitro, HT-22 cells kept exposure-naïve or injured by glutamate were pre-treated with vehicle or increasing doses of LMP and cell viability was determined. In vivo, LMP's effects were first assessed in 5-day-old healthy, uninjured CD-1 mouse pups receiving a single intraperitoneal injection of vehicle or different dosages of LMP. In a second step, mouse pups were subjected to excitotoxic brain injury and subsequently treated with vehicle or LMP. Endpoints included somatometric data as well as histological and immunohistochemical analyses. In vitro, cell viability in exposure-naïve cells was significantly reduced by high doses of LMP, but remained unaffected in glutamate-injured cells. In vivo, no specific toxic effects of LMP were observed neither in healthy mouse pups nor in experimental animals subjected to excitotoxic injury, but body weight gain was significantly lower following higher-dose LMP treatment. Also, LMP failed to produce a neuroprotective effect in the injured developing brain. Additional studies are required prior to a routine clinical use of LMP in neonatal intensive care units.

6.
Biomed Res Int ; 2019: 5935279, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31001556

RESUMO

Hematopoietic growth factors are considered to bear neuroprotective potential. We have previously shown that delayed treatment with granulocyte colony-stimulating factor (G-CSF)/stem cell factor (SCF) and Fms-related tyrosine kinase 3 ligand (FL) ameliorates excitotoxic neonatal brain injury. The effect of these substances in combined-stressor neonatal brain injury models more closely mimicking clinical conditions has not been investigated. The aim of this study was to assess the short-, mid-, and long-term neuroprotective potential of G-CSF/SCF and FL in a neonatal model of hypoxic-hyperoxic ischemic brain injury. Five-day-old (P5) CD-1 mice were subjected to unilateral common carotid artery ligation and subsequent alternating periods of hypoxia and hyperoxia for 65 minutes. Sixty hours after injury, pups were randomly assigned to intraperitoneal treatment with (i) G-CSF (200 µg/kg)/SCF (50 µg/kg), (ii) FL (100 µg/kg), or (iii) vehicle every 24 hours for three or five consecutive days. Histopathological and functional outcomes were evaluated on P10, P18, and P90. Baseline outcome parameters were established in sham-treated and healthy control animals. Gross brain injury did not significantly differ between treatment groups at any time point. On P10, caspase-3 activation and caspase-independent apoptosis were similar between treatment groups; cell proliferation and the number of BrdU-positive vessels did not differ on P18 or P90. Neurobehavioral assessment did not reveal significant differences between treatment groups in accelerod performance, open field behavior, or novel object recognition capacity on P90. Turning behavior was more frequently observed in G-CSF/SCF- and FL-treated animals. No sex-specific differences were detected in any outcome parameter evaluated. In hypoxic-hyperoxic ischemic neonatal brain injury, G-CSF/SCF and FL treatment does not convey neuroprotection. Prior to potential clinical use, meticulous assessment of these hematopoietic growth factors is mandated.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Fator Estimulador de Colônias de Granulócitos/farmacologia , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Proteínas de Membrana/farmacologia , Fármacos Neuroprotetores/farmacologia , Fator de Células-Tronco/farmacologia , Animais , Animais Recém-Nascidos , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Lesões Encefálicas/fisiopatologia , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/patologia , Hipóxia-Isquemia Encefálica/fisiopatologia , Camundongos , Camundongos Endogâmicos ICR , Fatores de Tempo
7.
Neuroscience ; 410: 239-253, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31121260

RESUMO

Excitotoxicity plays an important role in the pathogenesis of developing brain injury. The neuropeptide secretoneurin (SN) has neuroprotective potential. The aim of this study was to investigate SN plasma concentrations following excitotoxicity and to evaluate the effect of SN as therapeutic strategy in excitotoxic newborn brain injury. Baseline SN plasma concentrations were established in healthy animals. To evaluate the effect of an excitotoxic insult on SN levels, mice pups were subjected to an intracranial injection of ibotenic acid and SN plasma concentrations were measured thereafter. To assess SN's neuroprotective potential, a subgroup of animals was randomly assigned to the following groups: i) "single treatment": vehicle 1× phosphate-buffered saline (PBS), SN 0.25 µg/g body weight (bw), SN 2.5 µg/g bw or SN 12.5 µg/g bw in a single dose 1 h after insult; ii) "acute repetitive treatment": vehicle 1× PBS or SN 0.25 µg/g bw every 24 h starting 1 h after insult; iii) "delayed repetitive treatment": vehicle 1× PBS or SN 0.25 µg/g bw every 24 h starting 60 h after insult. Animals subjected to excitotoxic injury showed significantly lower SN plasma concentrations 6 and 120 h after insult in comparison to healthy controls. Administration of SN did not positively affect lesion size, apoptotic cell death, microglial cell activation or cell proliferation. To conclude, endogenous SN plasma levels are lower in newborn mice subjected to an excitotoxic insult than in healthy controls. Supplementation with SN in various treatment regimens is not neuroprotective in the experimental animal model of excitotoxic newborn brain injury.


Assuntos
Lesões Encefálicas/sangue , Lesões Encefálicas/prevenção & controle , Ácido Ibotênico/toxicidade , Neuropeptídeos/sangue , Neuropeptídeos/uso terapêutico , Neurotoxinas/toxicidade , Secretogranina II/sangue , Secretogranina II/uso terapêutico , Animais , Animais Recém-Nascidos , Biomarcadores/sangue , Lesões Encefálicas/induzido quimicamente , Camundongos , Neuroproteção/efeitos dos fármacos , Neuroproteção/fisiologia , Distribuição Aleatória
8.
Eur J Neurosci ; 27(4): 874-83, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18279363

RESUMO

Enhanced glutamate release and inflammation play an important role in the pathogenesis of developmental brain injury. Although N-methyl-d-aspartate receptor (NMDAR) antagonists potently attenuate neonatal brain damage in several animal models, they can also impact trophic functions in the developing brain. As a consequence, high-affinity NMDAR antagonists have been shown to trigger widespread apoptotic neurodegeneration in the newborn brain. Dextromethorphan (DM), a low-affinity NMDAR antagonist with anti-inflammatory properties, may be neuroprotective against excitotoxic and inflammation-enhanced excitotoxic brain injury, without the associated stimulation of apoptotic degeneration. Using an established newborn mouse model of excitotoxic brain damage, we determined whether systemic injection of DM significantly attenuates excitotoxic lesion size. We investigated several doses and time regimens; a dose of 5 microg/g DM given in a combination of both pre-injury and repetitive post-injury treatment proved most effective. DM treatment significantly reduced lesion size in gray and white matter by reducing cell death as shown by a decreased Fluoro-Jade B staining and caspase-3 activation. Pre-treatment with interleukin-1beta and lipopolysaccharide enhanced NMDAR-mediated excitotoxic brain injury and microglial cell activation. This sensitizing effect was abolished by DM treatment, as the effectiveness of DM in reducing lesion size and microglial cell activation was similar to phosphate-buffered saline-pre-treated controls. In all cases, no gender-specific differences were detected. DM treatment did not trigger any apoptotic neurodegeneration (caspase-3 cleavage, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling, Fluoro-Jade B staining). Although functional parameters were not measured, our data corroborate reports that DM is neuroprotective and that it may therefore improve functional outcome following perinatal brain injury.


Assuntos
Encéfalo/efeitos dos fármacos , Dextrometorfano/uso terapêutico , Encefalomalacia/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Caspase 3/metabolismo , Modelos Animais de Doenças , Encefalomalacia/induzido quimicamente , Encefalomalacia/patologia , Agonistas de Aminoácidos Excitatórios/toxicidade , Antagonistas de Aminoácidos Excitatórios/uso terapêutico , Feminino , Ácido Ibotênico/toxicidade , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Inflamação/induzido quimicamente , Inflamação/prevenção & controle , Masculino , Camundongos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Reação em Cadeia da Polimerase
9.
Eur J Paediatr Neurol ; 11(3): 129-35, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17222575

RESUMO

BACKGROUND: Periventricular leukomalacia (PVL) is a major cause of neurological handicap in pre-term infants. At present, there are no effective or causal therapies available. Thyroid hormones play an essential role in brain development and are reported to be decreased in pre-terms and following brain injury in adults. HYPOTHESIS: Excitotoxic brain damage of newborn mice decreases thyroid hormone concentrations. Exogenous T3 administration restores thyroid hormone levels and reduces perinatal brain damage in an animal model of PVL. DESIGN AND METHOD: To create white and gray matter (WM/GM) lesion mimicking several key aspects of PVL, we injected ibotenic acid (Ibo), a glutamate analog, into the right hemisphere (intracranially (i.c.)) of 5-day-old mice. T3 (10 microg/kg body weight (bw)) was injected intraperitoneally (i.p.) 1 h or repeatedly 1/24/48/72/96 h post-insult. We determined lesion size, number of apoptotic cells in WM/GM and serum T3/T4 concentration at 24 and 120 h after injury. Serum T3/T4 concentration was also determined before and 1 and 2h after T3 administration. RESULTS: Excitotoxic brain damage did not alter serum T3/T4 concentrations within 120 h of injury. Serum T3 levels were distinctly elevated within 1 h of T3 injection; however, this elevation was relatively short-lived (half-life estimated to be less than 12 h). Neither single nor repetitive T3 treatment regimen reduced excitotoxic lesion size, but it did reduce apoptosis. CONCLUSIONS: T3 replacement does not prevent excitotoxic cell death, but it does reduce developmental neuronal apoptosis, which could participate to the beneficial neuropsychological effects of hormone therapy. Further study is therefore warranted.


Assuntos
Apoptose/efeitos dos fármacos , Neurônios/patologia , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/patologia , Tri-Iodotironina/análogos & derivados , Análise de Variância , Animais , Animais Recém-Nascidos , Caspase 3/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Vias de Administração de Medicamentos , Agonistas de Aminoácidos Excitatórios/toxicidade , Lateralidade Funcional , Ácido Ibotênico/toxicidade , Camundongos , Neurônios/efeitos dos fármacos , Síndromes Neurotóxicas/etiologia , Fatores de Tempo , Tri-Iodotironina/administração & dosagem , Tri-Iodotironina/sangue
10.
Neuroscience ; 352: 88-96, 2017 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-28391015

RESUMO

Neonatal brain injury is a problem of global importance. To date, no causal therapies are available. A substance with considerable therapeutic potential is the endogenous neuropeptide secretoneurin (SN), which has proven to be beneficial in adult stroke. The aim of this study was to assess its effect in neonatal hypoxic-ischemic brain injury models. In vitro, primary hippocampal neurons were pre-treated with vehicle, 1µg/ml, 10µg/ml, or 50µg/ml SN and subjected to oxygen-glucose deprivation (OGD) for six hours. Cell death was assessed after a 24-h recovery period. In vivo, seven day-old CD-1 mice underwent unilateral common carotid artery ligation and were exposed to 8% oxygen/nitrogen for 20 min. SN plasma concentrations were serially determined by ELISA after insult. One hour after hypoxia, a subgroup of animals was treated with vehicle or SN. SN plasma concentrations significantly decreased 48h after insult. The number of caspase-3-positive cells was significantly lower in the hypoxic-ischemic hemisphere in the thalamus of SN-treated animals. In the hypoxic-only hemisphere administration of SN significantly reduced the number of caspase-3-positive cells (in cortex, white matter, hippocampus, thalamus and striatum) and inhibited microglial cell activation in the thalamus. SN has neuroprotective potential in neonatal brain injury. Its main action seems to be inhibition of apoptosis in the aftermath of the insult, predominantly in the hypoxic-only hemisphere. This might be explained by the less pronounced injury in this hemisphere, where blood flow and thus nutrient supply are maintained.


Assuntos
Lesões Encefálicas/etiologia , Lesões Encefálicas/prevenção & controle , Lateralidade Funcional/efeitos dos fármacos , Hipóxia-Isquemia Encefálica/complicações , Neuropeptídeos/uso terapêutico , Secretogranina II/uso terapêutico , Animais , Animais Recém-Nascidos , Caspase 3/metabolismo , Técnicas de Cultura de Células , Hipóxia Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Glucose/deficiência , Hipocampo/citologia , Hipóxia-Isquemia Encefálica/sangue , Camundongos , Microglia/efeitos dos fármacos , Microglia/patologia , Neurônios/efeitos dos fármacos , Neuropeptídeos/sangue , Neuropeptídeos/farmacologia , Fármacos Neuroprotetores/sangue , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Antígeno Nuclear de Célula em Proliferação/metabolismo , Secretogranina II/sangue , Secretogranina II/farmacologia , Estatísticas não Paramétricas , Fatores de Tempo
11.
Brain Res ; 1634: 94-103, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26772988

RESUMO

BACKGROUND: Developmental brain injury results in cognitive and motor deficits in the preterm infant. Enhanced glutamate release and subsequent receptor activation are major pathogenetic factors. The effect of haematopoietic growth factors, such as granulocyte colony-stimulating factor (G-CSF), stem cell factor (SCF) and flt-3 ligand (FL) on neonatal brain injury is controversially discussed. Timing of treatment is known to be a crucial factor. Based on the hypothesis that an exacerbation of injury is caused by administration of substances in the acute phase, the objective of this study was to evaluate the effect of delayed administration of G-CSF/SCF and FL to protect against excitotoxic brain injury in vivo. METHODS: In an established neonatal mouse model of excitotoxic brain injury, we evaluated the effect of daily intraperitoneal doses of G-CSF/SCF or FL, starting 60 h after the excitotoxic insult. RESULTS: Intraperitoneal injections of G-CSF/SCF and FL, given 60 h after the excitotoxic insult, significantly reduced lesion size at postnatal days 10, 18 and 90. G-CSF/SCF treatment resulted in a decrease in apoptotic cell death indicated by reduced caspase-3 activation. G-CSF/SCF and FL treatment did not affect apoptosis-inducing factor-dependent apoptosis or cell proliferation. CONCLUSION: We show that delayed systemic treatment with the haematopoietic growth factors G-CSF/SCF and FL protects against N-methyl-D-aspartate receptor-mediated developmental excitotoxic brain damage. Our results suggest that neuroprotective effects in this neonatal animal model of excitotoxic brain injury depend on the timing of drug administration after the insult.


Assuntos
Apoptose/efeitos dos fármacos , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Córtex Cerebral/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Proteínas de Membrana/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Fator de Células-Tronco/administração & dosagem , Animais , Animais Recém-Nascidos , Lesões Encefálicas/induzido quimicamente , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Agonistas de Aminoácidos Excitatórios , Ácido Ibotênico , Camundongos , Receptores de N-Metil-D-Aspartato/agonistas
12.
Brain Res ; 1556: 10-8, 2014 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-24530252

RESUMO

BACKGROUND: Hypoxic-ischemic encephalopathy (HIE) resulting from perinatal asphyxia often leads to severe neurologic impairment or even death. There is a need to advance therapy for infants with HIE, for example to combine hypothermia with pharmacological treatment strategies. Levetiracetam (LEV) is approved for clinical administration to infants older than 4 weeks of age and is also used off-label in neonates. Furthermore, LEV was shown to be neuroprotective in adult animal models of brain injury. AIM OF THE STUDY: The aim of this study was to evaluate the neuroprotective potential of LEV in vitro using primary hippocampal neurons, and in vivo using an established model of neonatal hypoxic-ischemic brain injury. RESULTS: LEV treatment per se did not induce neurotoxicity in the developing rodent brain. Following oxygen glucose deprivation, we observed some, although not a significant, increase in cell death after LEV treatment. In vivo, LEV was administered under normothermic and hypothermic conditions following hypoxic-ischemic brain damage. LEV administration significantly increased brain injury under normothermic conditions. Compared to the normothermia-treated group, in the hypothermia group LEV administration did not increase hypoxic-ischemic brain injury. DISCUSSION: This study demonstrates that LEV treatment increases neonatal hypoxic-ischemic brain injury. Administration of LEV in the acute phase of the injury might interfere with the balanced activation and inactivation of excitatory and inhibitory receptors in the developing brain. The neurotoxic effect of LEV in the injured newborn brain might further suggest an agonistic effect of LEV on the GABAergic system. Hypothermia treatment attenuates glutamate release following hypoxic-ischemic brain injury and might therefore limit the potentially deleterious effects of LEV. As a consequence, our findings do not necessarily rule out a potentially beneficial effect, but argue for cautious use of LEV in newborn infants with pre-existing brain injury.


Assuntos
Hipotermia Induzida/métodos , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Piracetam/análogos & derivados , Animais , Fator de Indução de Apoptose/metabolismo , Caspase 3/metabolismo , Contagem de Células , Morte Celular/efeitos dos fármacos , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/deficiência , Hipocampo/citologia , Hipóxia , Hipóxia-Isquemia Encefálica/induzido quimicamente , Levetiracetam , Camundongos , Neurônios/efeitos dos fármacos , Piracetam/uso terapêutico
13.
Exp Neurol ; 261: 501-9, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25111531

RESUMO

Premature birth represents a clinical situation of risk for brain injury. The diversity of pathophysiological processes complicates efforts to find effective therapeutic strategies. Excitotoxicity is one important factor in the pathogenesis of preterm brain injury. The observation that sigma-1 receptor agonists possess neuroprotective potential, at least partly mediated by a variety of anti-excitotoxic mechanisms, has generated great interest in targeting those receptors to counteract brain injury. The objective of this study was to evaluate the effect of the highly specific sigma-1 receptor agonist, 4-phenyl-1-(4-phenylbutyl) piperidine (PPBP) to protect against excitotoxic developmental brain injury in vivo and in vitro. Primary hippocampal neurons were pre-treated with PPBP before glutamate was applied and subsequently analyzed for cell death (PI/calcein AM), mitochondrial activity (TMRM) and morphology of the neuronal network (WGA) using confocal microscopy. Using an established neonatal mouse model we also determined whether systemic injection of PPBP significantly attenuates excitotoxic brain injury. PPBP significantly reduced neuronal cell death in primary hippocampal neurons exposed to glutamate. Neurons treated with PPBP showed a less pronounced loss of mitochondrial membrane potential and fewer morphological changes after glutamate exposure. A single intraperitoneal injection of PPBP given one hour after the excitotoxic insult significantly reduced microglial cell activation and lesion size in cortical gray and white matter. The present study provides strong support for the consideration of sigma-1 receptor agonists as a candidate therapy for the reduction of neonatal excitotoxic brain lesions and might offer a novel target to counteract developmental brain injury.


Assuntos
Lesões Encefálicas/prevenção & controle , Haloperidol/análogos & derivados , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Microglia/efeitos dos fármacos , Receptores sigma/agonistas , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Fator de Indução de Apoptose/metabolismo , Lesões Encefálicas/induzido quimicamente , Caspase 3/metabolismo , Modelos Animais de Doenças , Agonistas de Aminoácidos Excitatórios/toxicidade , Ácido Glutâmico/farmacologia , Glicoproteínas/metabolismo , Haloperidol/uso terapêutico , Hipocampo/citologia , Ácido Ibotênico/toxicidade , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Estatísticas não Paramétricas , Receptor Sigma-1
14.
Brain Res ; 1355: 31-40, 2010 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-20692240

RESUMO

BACKGROUND: Perinatal brain injury in preterm infants is a major cause of neurological handicap. The role of the neurotrophin receptor p75 (p75(NTR)) in the pathogenesis and repair of neonatal excitotoxic brain injury is unknown. Depending on a complex interplay of neurotrophin signalling, p75(NTR) can, in addition to its trophic function, also induce apoptosis. HYPOTHESIS: We hypothesised that excitotoxicity increases p75(NTR) expression and p75(NTR) knockout (KO) mice have a significantly smaller lesion size upon excitotoxicity as compared to wild-type (WT) mice. METHODS: We used an established animal model of neonatal excitotoxic brain injury mimicking several key aspects of human preterm brain damage. We subjected five-day-old WT and KO mice to excitotoxic injury by means of a single intracranial ibotenate injection (N-methyl-D-aspartate receptor agonist, NMDAR) into one brain hemisphere. Lesion size, number of activated caspase-3- and apoptosis-inducing factor (AIF)-positive cells were determined as outcome parameters. Gender analyses were taken into account retrospectively. RESULTS: NMDAR-mediated excitotoxicity induced an upregulation of p75(NTR) expression in the peri-lesion area. Lesion size was significantly increased in female KO as compared to male KO animals. Knockout of p75(NTR) reduced the number of activated caspase-3 but not AIF-positive cells after NMDAR-mediated excitotoxic injury independently of gender. CONCLUSION: Since NMDAR-mediated excitotoxic brain injury induced p75(NTR) expression and caspase-3-activated apoptosis in p75(NTR) KO animals was decreased, we conclude that activation of p75(NTR) contributes to NMDAR-mediated apoptosis in the neonatal brain. An increase in lesion size in female animals after excitotoxic brain injury suggests that in females p75(NTR) seems to play a dual role.


Assuntos
Lesão Encefálica Crônica/metabolismo , Neurotoxinas/toxicidade , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores de Fator de Crescimento Neural/fisiologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Lesão Encefálica Crônica/induzido quimicamente , Lesão Encefálica Crônica/patologia , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Knockout , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de Fator de Crescimento Neural/genética
15.
Neurobiol Dis ; 24(2): 357-66, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16959492

RESUMO

Using an established mouse model of human periventricular leukomalacia, we investigated whether EPO could reduce excitotoxic damage. When administered 1 h following intracerebral injection of 10 microg ibotenic acid at day 5 of life, both a single injection of EPO (5000 IU/kg bw) and repetitive administrations of EPO reduced white and gray matter lesion size. The therapeutic window for protection was small as the protective effect of EPO was lost when EPO administration was delayed to 4 h post-insult. EPO-mediated upregulation of EPO-R, but not EPO, mRNA was observed within 4 h of the excitotoxic insult. The EPO effect was gender independent. Minor hematopoetic effects were observed following EPO treatment. We conclude that a single dose of EPO is sufficient to reduce excitotoxic brain injury and may therefore possess therapeutic relevance in the clinical setting.


Assuntos
Eritropoetina/farmacologia , Leucomalácia Periventricular/tratamento farmacológico , Degeneração Neural/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Neurotoxinas/antagonistas & inibidores , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Citoproteção/efeitos dos fármacos , Citoproteção/fisiologia , Modelos Animais de Doenças , Esquema de Medicação , Eritropoetina/uso terapêutico , Feminino , Ácido Glutâmico/metabolismo , Humanos , Ácido Ibotênico/antagonistas & inibidores , Ácido Ibotênico/metabolismo , Recém-Nascido , Injeções Intraventriculares , Leucomalácia Periventricular/metabolismo , Leucomalácia Periventricular/fisiopatologia , Masculino , Camundongos , Degeneração Neural/fisiopatologia , Degeneração Neural/prevenção & controle , Fármacos Neuroprotetores/uso terapêutico , Neurotoxinas/metabolismo , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Receptores da Eritropoetina/genética , Receptores de N-Metil-D-Aspartato/agonistas , Fatores de Tempo
16.
Pediatr Res ; 59(4 Pt 1): 549-53, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16549527

RESUMO

Granulocyte-colony stimulating factor (G-CSF) has been shown to reduce brain lesion size and mortality in adult mice after hypoxic-ischemic injury. Another hematopoietic growth factor, stem cell factor (SCF), has been shown to be up-regulated in the brains of adult rodents following brain damage, where it stimulates postlesional neurogenesis. Injection of the excitotoxic agent ibotenate into the brain of newborn mice produces a brain lesion characterized by neuronal death and white matter cysts, which is similar to periventricular leucomalacia. The aim of the present study was to investigate whether administration of SCF and G-CSF is neuroprotective against ibotenate lesions in neonatal mice. Contrary to our expectations, cortical and white matter brain lesions induced by ibotenate were enhanced following the administration of 50 microg/kg SCF or 200 microg/kg G-CSF. Dose-response studies indicated that G-CSF could increase grey matter lesions even at lower dosages (22 and 66 microg/kg). Administration of SCF and G-CSF in combination also increased cortical and white matter lesions, to 133 +/- 8% and 187 +/- 12%. In the undamaged brain, G-CSF or G-CSF+SCF treatment had no effect on apoptosis in the grey or white matter; however, these treatments significantly increased apoptosis in the damaged brain. Our data clearly demonstrate that G-CSF and SCF are not neuroprotective and result in deleterious enhancement of excitotoxic brain damage in newborn mice. We conclude that G-CSF and SCF should be used cautiously in newborn infants with brain lesions; if they are used, long term neurologic and neurodevelopmental follow-up is warranted.


Assuntos
Lesões Encefálicas/induzido quimicamente , Agonistas de Aminoácidos Excitatórios/toxicidade , Fator Estimulador de Colônias de Granulócitos , Ácido Ibotênico/toxicidade , Neurotoxinas/toxicidade , Fator de Células-Tronco , Animais , Animais Recém-Nascidos , Apoptose , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Relação Dose-Resposta a Droga , Fator Estimulador de Colônias de Granulócitos/metabolismo , Fator Estimulador de Colônias de Granulócitos/farmacologia , Camundongos , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Fator de Células-Tronco/metabolismo , Fator de Células-Tronco/farmacologia
17.
Br J Haematol ; 117(4): 935-9, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12060133

RESUMO

Donor lymphocyte infusions (DLI) can induce a graft-versus-leukaemia (GvL) reaction in patients with relapsed disease. However, the mechanisms involved in remission induction are not completely known. A patient with chemotherapy-refractory relapse 1 year after human leucocyte antigen (HLA)-identical, unrelated stem cell transplantation (SCT) for bcr/abl-positive common acute lymphoblastic leukaemia (ALL) received a DLI from the original donor, and achieved complete cytogenetic and molecular remission concomitantly with extensive graft-versus-host disease (GvHD). Seven CD8+, donor-derived, alloreactive T-cell clones were generated by stimulating post-DLI remission cells with the patient's pretransplant mature dendritic cells. The minor histocompatibility antigen (mHag) recognized by these T-cell clones was identified as HA-1, a mHag associated with acute GvHD after SCT. Our finding provides evidence of HA-1-associated GvL effects after DLI that paralleled the eradication of full-blown, chemotherapy-refractory ALL relapse after allogeneic SCT.


Assuntos
Doença Enxerto-Hospedeiro/imunologia , Transfusão de Linfócitos , Antígenos de Histocompatibilidade Menor , Oligopeptídeos , Leucemia-Linfoma Linfoblástico de Células Precursoras/cirurgia , Linfócitos T Citotóxicos/imunologia , Adulto , Linfócitos T CD8-Positivos/imunologia , Células Clonais/imunologia , Células Dendríticas/imunologia , Efeito Enxerto vs Leucemia/imunologia , Humanos , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Indução de Remissão , Transplante Homólogo
18.
Lab Invest ; 82(9): 1147-53, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12218075

RESUMO

The detection of micrometastatic disease remains a challenge for the diagnosis and monitoring of malignant disease. RT-PCR for human mammaglobin (hMAM) was recently shown to provide a sensitive method for assessing circulating breast cancer cells in peripheral blood. This study was aimed at investigating hMAM expression in normal and malignant tissue from the female genital tract and the prostate as well as in malignant effusions derived from gynecologic malignancies. hMAM expression was analyzed with nested RT-PCR in 152 samples of normal (n = 73) and malignant epithelial tissues (n = 79) and in 33 specimens of various normal mesenchymal tissue types. We found hMAM expression was not restricted to the normal mammary gland and breast carcinoma but was also detectable in most specimens of benign and malignant epithelial tissue from the ovary (97% versus 95%), uterus (both 100%), and cervix (91% versus 90%). Notably, hMAM expression was also found in benign prostatic hyperplasia (45%) and in prostate cancer (55%). A much lower expression rate was found in various normal and benign mesenchymal tissues (12%). In keeping with our previous data, hMAM expression was absent in all control samples (n = 124) of peripheral blood and bone marrow from healthy volunteers and patients with hematologic malignancies. In pleural or peritoneal effusions (n = 42) from patients with carcinomas of the breast, endometrium, or ovary, hMAM positivity was noticed in the majority of cases (74%), whereas only 52% of the specimens were cytologically positive for tumor cells. In conclusion, hMAM expression assessed by nested RT-PCR is a sensitive molecular marker for detecting micrometastatic tumor spread into pleural effusions and ascites from patients with breast cancer and various other gynecologic neoplasms.


Assuntos
Neoplasias dos Genitais Femininos/metabolismo , Proteínas de Neoplasias/genética , Derrame Pleural Maligno/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Uteroglobina/genética , Feminino , Genitália Feminina/metabolismo , Humanos , Masculino , Mamoglobina A , Próstata/metabolismo , Neoplasias da Próstata/metabolismo , RNA Mensageiro/análise , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA