Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Neurobiol Dis ; 145: 105074, 2020 11.
Article in English | MEDLINE | ID: mdl-32890773

ABSTRACT

In utero alcohol exposure can induce severe neurodevelopmental disabilities leading to long-term behavioral deficits. Because alcohol induces brain defects, many studies have focused on nervous cells. However, recent reports have shown that alcohol markedly affects cortical angiogenesis in both animal models and infants with fetal alcohol spectrum disorder (FASD). In addition, the vascular system is known to contribute to controlling gamma-aminobutyric acid (GABA)ergic interneuron migration in the developing neocortex. Thus, alcohol-induced vascular dysfunction may contribute to the neurodevelopmental defects in FASD. The present study aimed at investigating the effects of alcohol on endothelial activity of pial microvessels. Ex vivo experiments on cortical slices from mouse neonates revealed that in endothelial cells from pial microvessels acute alcohol exposure inhibits both glutamate-induced calcium mobilization and activities of matrix metalloproteinase-9 (MMP-9) and tissue plasminogen activator (tPA). The inhibitory effect of alcohol on glutamate-induced MMP-9 activity was abrogated in tPA-knockout and Grin1flox/VeCadcre mice suggesting that alcohol interacts through the endothelial NMDAR/tPA/MMP-9 vascular pathway. Contrasting with the effects from acute alcohol exposure, in mouse neonates exposed to alcohol in utero during the last gestational week, glutamate exacerbated both calcium mobilization and endothelial protease activities from pial microvessels. This alcohol-induced vascular dysfunction was associated with strong overexpression of the N-methyl-d-aspartate receptor subunit GluN1 and mispositioning of the Gad67-GFP interneurons that normally populate the superficial cortical layers. By comparing several human control fetuses with a fetus chronically exposed to alcohol revealed that alcohol exposure led to mispositioning of the calretinin-positive interneurons, whose density was decreased in the superficial cortical layers II-III and increased in deepest layers. This study provides the first mechanistic and functional evidence that alcohol impairs glutamate-regulated activity of pial microvessels. Endothelial dysfunction is characterized by altered metalloproteinase activity and interneuron mispositioning, which was also observed in a fetus with fetal alcohol syndrome. These data suggest that alcohol-induced endothelial dysfunction may contribute in ectopic cortical GABAergic interneurons, that has previously been described in infants with FASD.


Subject(s)
Endothelial Cells/drug effects , Fetal Alcohol Spectrum Disorders/enzymology , Fetal Alcohol Spectrum Disorders/pathology , Interneurons/pathology , Neurogenesis/drug effects , Pia Mater/drug effects , Animals , Central Nervous System Depressants/toxicity , Endothelial Cells/enzymology , Ethanol/toxicity , Female , GABAergic Neurons/drug effects , GABAergic Neurons/pathology , Humans , Interneurons/drug effects , Metalloproteases/metabolism , Mice , Pia Mater/enzymology , Pregnancy , Prenatal Exposure Delayed Effects
2.
Med Sci (Paris) ; 35(11): 859-865, 2019 Nov.
Article in French | MEDLINE | ID: mdl-31845877

ABSTRACT

Alcohol consumption during pregnancy constitutes a major cause of neurodevelopmental and behavioral disabilities. Whereas it is possible for clinicians to establish a perinatal diagnosis of fetal alcohol syndrome, the more severe expression of fetal alcohol spectrum disorder (FASD), most FASD children are late or mis-diagnosed due to a lack of clear morphological and neurodevelopmental abnormalities. Several precious years of care are consequently lost. Recent data revealed a functional placenta-brain axis involved in the control of the fetal brain angiogenesis which is impaired by in utero alcohol exposure. Because in the developing fetal brain a correct angiogenesis is required for a correct neurodevelopment, these preclinical and clinical advances pave the way for a new generation of placental biomarkers for early diagnosis of FASD.


TITLE: Alcoolisation fœtale - Le placenta au secours du diagnostic précoce des troubles du développement cérébral de l'enfant. ABSTRACT: La consommation d'alcool au cours de la grossesse constitue une cause majeure de troubles du comportement et de handicap. Alors qu'il est possible pour un clinicien d'établir un diagnostic néonatal du syndrome d'alcoolisation fœtale, l'atteinte la plus sévère des troubles causés par l'alcoolisation fœtale (TCAF), une grande majorité des enfants échappe à un diagnostic précoce en raison de l'absence d'anomalies morphologiques évidentes. Plusieurs années de prise en charge sont alors perdues. Des avancées récentes ont permis d'établir l'existence d'un axe fonctionnel placenta-cerveau impliqué dans le contrôle de l'angiogenèse cérébrale, qui se trouve dérégulé chez les enfants exposés in utero à l'alcool. Une angiogenèse cérébrale normale étant un prérequis à l'établissement d'un neurodéveloppement correct, ces avancées ouvrent la voie à l'identification d'une nouvelle génération de biomarqueurs placentaires d'atteinte cérébrale pour le diagnostic précoce des enfants TCAF.


Subject(s)
Brain Diseases/diagnosis , Fetal Alcohol Spectrum Disorders/diagnosis , Placenta , Animals , Brain Diseases/etiology , Early Diagnosis , Female , Humans , Infant, Newborn , Neovascularization, Physiologic , Placenta/metabolism , Pregnancy
3.
Front Neurol ; 10: 407, 2019.
Article in English | MEDLINE | ID: mdl-31068895

ABSTRACT

Background: Remifentanil, a synthetic opioid used for analgesia during cesarean sections, has been shown in ex vivo experiments to exert anti-apoptotic activity on immature mice brains. The present study aimed to characterize the impact of remifentanil on brain lesions using an in vivo model of excitotoxic neonatal brain injury. Methods: Postnatal day 2 (P2) mice received three intraperitoneal injections of remifentanil (500 ng/g over a 10-min period) or saline just before an intracortical injection of ibotenate (10 µg). Cerebral reactive oxygen species (ROS) production, cell death, in situ labeling of cortical caspase activity, astrogliosis, inflammation mediators, and lesion size were determined at various time points after ibotenate injection. Finally, behavioral tests were performed until P18. Results: In the injured neonatal brain, remifentanil significantly decreased ROS production, cortical caspase activity, DNA fragmentation, interleukin-1ß levels, and reactive astrogliosis. At P7, the sizes of the ibotenate-induced lesions were significantly reduced by remifentanil treatment. Performance on negative geotaxis (P6-8) and grasping reflex (P10-12) tests was improved in the remifentanil group. At P18, a sex specificity was noticed; remifentanil-treated females spent more time in the open field center than did the controls, suggesting less anxiety in young female mice. Conclusions: In vivo exposure to remifentanil exerts a beneficial effect against excitotoxicity on the developing mouse brain, which is associated with a reduction in the size of ibotenate-induced brain lesion as well as prevention of some behavioral deficits in young mice. The long-term effect of neonatal exposure to remifentanil should be investigated.

4.
Pharmacol Res Perspect ; 5(4)2017 Aug.
Article in English | MEDLINE | ID: mdl-28805973

ABSTRACT

Clinical studies showed beneficial effects of magnesium sulfate regarding the risk of cerebral palsy. However, regimen protocols fluctuate worldwide and risks of adverse effects impacting the vascular system have been reported for human neonates, keeping open the question of the optimal dosing. Using clinically relevant concentrations and doses of magnesium sulfate, experiments consisted of characterizing, respectively, ex vivo and in vivo, the effects of magnesium sulfate on the nervous and vascular systems of mouse neonates by targeting neuroprotection, angiogenesis, and hemodynamic factors and in measuring, in human fetuses, the impact of a 4-g neuroprotective loading dose of magnesium sulfate on brain hemodynamic parameters. Preclinical experiments using cultured cortical slices from mouse neonates showed that the lowest and highest tested concentrations of magnesium sulfate were equally potent to prevent excitotoxic-induced cell death, cell edema, cell burst, and intracellular calcium increase, whereas no side effects were found regarding apoptosis. In contrast, in vivo data revealed that magnesium sulfate exerted dose-dependent vascular effects on the fetal brain. In particular, it induced brain hypoperfusion, stabilization of Hif-1α, long-term upregulation of VEGF-R2 expression, impaired endothelial viability, and altered cortical angiogenesis. Clinically, in contrast to 6-g loading doses used in some protocols, a 4-g bolus of magnesium sulfate did not altered fetal brain hemodynamic parameters. In conclusion, these data provide the first mechanistic evidence of double-sword and dose-dependent actions of magnesium sulfate on nervous and vascular systems. They strongly support the clinical use of neuroprotection protocols validated for the lowest (4-g) loading dose of magnesium sulfate.

5.
Acta Neuropathol Commun ; 5(1): 44, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28587682

ABSTRACT

Most children with in utero alcohol exposure do not exhibit all features of fetal alcohol syndrome (FAS), and a challenge for clinicians is to make an early diagnosis of fetal alcohol spectrum disorders (FASD) to avoid lost opportunities for care. In brain, correct neurodevelopment requires proper angiogenesis. Since alcohol alters brain angiogenesis and the placenta is a major source of angiogenic factors, we hypothesized that it is involved in alcohol-induced brain vascular defects. In mouse, using in vivo repression and overexpression of PLGF, we investigated the contribution of placenta on fetal brain angiogenesis. In human, we performed a comparative molecular and morphological analysis of brain/placenta angiogenesis in alcohol-exposed fetuses. Results showed that prenatal alcohol exposure impairs placental angiogenesis, reduces PLGF levels and consequently alters fetal brain vasculature. Placental repression of PLGF altered brain VEGF-R1 expression and mimicked alcohol-induced vascular defects in the cortex. Over-expression of placental PGF rescued alcohol effects on fetal brain vessels. In human, alcohol exposure disrupted both placental and brain angiogenesis. PLGF expression was strongly decreased and angiogenesis defects observed in the fetal brain markedly correlated with placental vascular impairments. Placental PGF disruption impairs brain angiogenesis and likely predicts brain disabilities after in utero alcohol exposure. PLGF assay at birth could contribute to the early diagnosis of FASD.


Subject(s)
Brain/drug effects , Fetal Alcohol Spectrum Disorders/metabolism , Placenta Growth Factor/metabolism , Placenta/drug effects , Animals , Brain/blood supply , Brain/embryology , Brain/pathology , Disease Models, Animal , Ethanol/toxicity , Female , Humans , Mice , Neovascularization, Pathologic/chemically induced , Neovascularization, Pathologic/embryology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Placenta/blood supply , Placenta/metabolism , Placenta/pathology , Placenta Growth Factor/genetics , Pregnancy , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism
6.
Cell Death Dis ; 8(2): e2610, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28182007

ABSTRACT

Brain developmental lesions are a devastating consequence of prenatal alcohol exposure (PAE). We recently showed that PAE affects cortical vascular development with major effects on angiogenesis and endothelial cell survival. The underlying molecular mechanisms of these effects remain poorly understood. This study aimed at characterizing the ethanol exposure impact on the autophagic process in brain microvessels in human fetuses with fetal alcohol syndrome (FAS) and in a PAE mouse model. Our results indicate that PAE induces an increase of autophagic vacuole number in human fetal and neonatal mouse brain cortical microvessels. Subsequently, ex vivo studies using green fluorescent protein (GFP)-LC3 mouse microvessel preparations revealed that ethanol treatment alters autophagy in endothelial cells. Primary cultures of mouse brain microvascular endothelial cells were used to characterize the underlying molecular mechanisms. LC3 and p62 protein levels were significantly increased in endothelial cells treated with 50 mM ethanol. The increase of autophagic vacuole number may be due to excessive autophagosome formation associated with the partial inhibition of the mammalian target of rapamycin pathway upon ethanol exposure. In addition, the progression from autophagosomes to autolysosomes, which was monitored using autophagic flux inhibitors and mRFP-EGFP vector, showed a decrease in the autolysosome number. Besides, a decrease in the Rab7 protein level was observed that may underlie the impairment of autophagosome-lysosome fusion. In addition, our results showed that ethanol-induced cell death is likely to be mediated by decreased mitochondrial integrity and release of apoptosis-inducing factor. Interestingly, incubation of cultured cells with rapamycin prevented ethanol effects on autophagic flux, ethanol-induced cell death and vascular plasticity. Taken together, these results are consistent with autophagy dysregulation in cortical microvessels upon ethanol exposure, which could contribute to the defects in angiogenesis observed in patients with FAS. Moreover, our results suggest that rapamycin represents a potential therapeutic strategy to reduce PAE-related brain developmental disorders.


Subject(s)
Autophagy/drug effects , Brain/drug effects , Ethanol/adverse effects , Microvessels/drug effects , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/pathology , Animals , Autophagosomes/drug effects , Autophagosomes/metabolism , Brain/metabolism , Brain/pathology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Fetal Alcohol Spectrum Disorders/metabolism , Fetal Alcohol Spectrum Disorders/pathology , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Microvessels/metabolism , Microvessels/pathology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Models, Animal , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , RNA-Binding Proteins/metabolism , Sirolimus/pharmacology
7.
Dev Neurobiol ; 75(3): 315-33, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25220981

ABSTRACT

Ketamine is a NMDA receptor (NMDAR) antagonist used in pediatric anesthesia. Given the role of glutamatergic signaling during brain maturation, we studied the effects of a single ketamine injection (40 mg/kg s.c) in mouse neonates depending on postnatal age at injection (P2, P5, or P10) on cortical NMDAR subunits expression and association with Membrane-Associated Guanylate Kinases PSD95 and SAP102. The effects of ketamine injection at P2, P5, or P10 on motor activity were compared in adulthood. Ketamine increased GluN2A and GluN2B mRNA levels in P2-treated mice without change in proteins, while it decreased GluN2B protein in P10-treated mice without change in mRNA. Ketamine reduced GluN2A mRNA and protein levels in P5-treated mice without change in GluN2B and GluN1. Ketamine affected the GluN2A/PSD95 association regardless of the age at injection, while GluN2B/PSD95 association was enhanced only in P5-treated mice. Microdissection of ketamine-treated mouse cortex showed a decrease in GluN2A mRNA level in superficial layers (I-IV) and an increase in all subunit expressions in deep layers (V-VI) in P5- and P10-treated mice, respectively. Our data suggest that ketamine impairs cortical NMDAR subunit developmental profile and delays the synaptic targeting of GluN2A-enriched NMDAR. Ketamine injection at P2 or P10 resulted in hyperlocomotion in adult male mice in an open field, without change in females. Voluntary running-wheel exercise showed age- and sex-dependent alterations of the mouse activity, especially during the dark phase. Overall, a single neonatal ketamine exposure led to short-term NMDAR cortical developmental profile impairments and long-term motor activity alterations persisting in adulthood.


Subject(s)
Aging/drug effects , Behavior, Animal/drug effects , Cerebral Cortex/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Receptors, N-Methyl-D-Aspartate/drug effects , Aging/metabolism , Aging/psychology , Animals , Cerebral Cortex/metabolism , Disks Large Homolog 4 Protein , Excitatory Amino Acid Antagonists/administration & dosage , Female , Guanylate Kinases/metabolism , Ketamine/administration & dosage , Locomotion/drug effects , Male , Membrane Proteins/metabolism , Mice , Motor Activity/drug effects , RNA, Messenger/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism
8.
Anesth Analg ; 118(5): 1041-51, 2014 May.
Article in English | MEDLINE | ID: mdl-24781573

ABSTRACT

BACKGROUND: The use of remifentanil in a context of potential prematurity led us to explore ex vivo the opioid effects on the immature mouse brain. Remifentanil enhances medullary glutamatergic N-methyl-D-aspartate (NMDA) receptor activity. Furthermore, in neonatal mouse cortex, NMDA was previously shown to exert either excitotoxic or antiapoptotic effects depending on the cortical layers. With the use of a model of acute cultured brain slices, we evaluated the potential necrotic and apoptotic effects of remifentanil, alone or associated with its glycine vehicle (commercial preparation of remifentanil, C.P. remifentanil), on the immature brain. METHODS: Cerebral slices from postnatal day 2 mice were treated up to 5 hours with the different compounds, incubated alone or in the presence of NMDA. The necrotic effect was studied by measuring lactate dehydrogenase activity and 7-Aminoactinomycin D labeling. Apoptotic death was evaluated by measurement of caspase-3 activity and cleaved caspase-3 protein levels, using Western blot and immunohistochemistry. Extrinsic and intrinsic apoptotic pathways were investigated by measuring caspase-8, caspase-9 activities, Bax protein levels, and mitochondrial integrity. RESULTS: C.P. remifentanil was ineffective on necrotic death, whereas it significantly reduced caspase-3 activity and cortical cleaved caspase-3 levels. C.P. remifentanil inhibited cortical Bax protein expression, caspase-9 activity, and preserved mitochondrial integrity, whereas it had no effect on caspase-8 activity. Its action targeted the neocortex superficial layers, and it was reversed by the opioid receptors antagonist naloxone and the NMDA antagonist MK801. Remifentanil and glycine acted synergistically to inhibit apoptotic death. In addition, C.P. remifentanil enhanced the antiapoptotic effect of NMDA, whereas it did not improve NMDA excitotoxicity in brain slices. CONCLUSION: The present data indicate that at a supraclinical concentration C.P. remifentanil had no pronecrotic effect but exerted ex vivo antiapoptotic action on the immature mouse brain, involving the opioid and NMDA receptors, and the mitochondrial-dependent apoptotic pathway. Assessment of the impact of the antiapoptotic effect of remifentanil in in vivo neonatal mouse models of brain injury will also be essential to measure its consequences on the developing brain.


Subject(s)
Analgesics, Opioid/pharmacology , Apoptosis/drug effects , Brain/cytology , Brain/drug effects , Piperidines/pharmacology , Analgesics, Opioid/pharmacokinetics , Animals , Animals, Newborn , Blotting, Western , Caspase 3/metabolism , Cell Death/drug effects , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Drug Synergism , Glycine/pharmacology , Half-Life , Immunohistochemistry , In Vitro Techniques , L-Lactate Dehydrogenase/metabolism , Mice , Microscopy, Electron , Mitochondria/drug effects , Mitochondria/ultrastructure , Piperidines/pharmacokinetics , Receptors, N-Methyl-D-Aspartate/drug effects , Remifentanil , bcl-2-Associated X Protein/metabolism
9.
J Cereb Blood Flow Metab ; 34(5): 764-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24517976

ABSTRACT

Glutamate transporters (excitatory amino-acid transporters (EAATs)) are essential for brain homeostasis. While previous studies indicate that the vascular endothelium contributes to glutamate efflux in the adult brain, little information is available regarding glutamate uptake in the immature brain. The present study shows a differential expression pattern of EAATs between cortical microvessels in adults and newborns. In addition, adult cortical endothelial cells take up glutamate more efficiently than neonatal cells. Our findings indicate age-specific changes in extracellular glutamate regulation by brain endothelial cells, suggesting differences in the efficiency of glutamate efflux during an excitotoxic process that, in turn, may contribute to age-specific brain vulnerability.


Subject(s)
Amino Acid Transport System X-AG/genetics , Cerebellar Cortex/blood supply , Cerebellar Cortex/growth & development , Endothelial Cells/metabolism , Glutamic Acid/metabolism , Amino Acid Transport System X-AG/analysis , Amino Acid Transport System X-AG/metabolism , Animals , Cells, Cultured , Cerebellar Cortex/cytology , Cerebellar Cortex/metabolism , Endothelial Cells/cytology , Gene Expression Regulation, Developmental , Mice
10.
Neurobiol Dis ; 45(3): 871-86, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22209711

ABSTRACT

In industrialized countries, cerebral palsy affects 2.5‰ of preterm and term infants. At a neurochemical level, the massive release of glutamate constitutes a major process leading to excitotoxicity and neonatal brain lesions. Previous studies, conducted in the laboratory, revealed that, in (δ/δ)VEGF(A) transgenic mice, glutamate-induced brain lesions are exacerbated suggesting that VEGF(A) could play a protective action against excitotoxicity. Using a model of cultured cortical brain slices, the aim of the study was to characterize the central effects of VEGF against glutamate-induced excitotoxicity in neonates. Exposure of brain slices to glutamate induced a strong increase of necrotic cell death in the deep cortical layer VI and a decrease of apoptotic death in superficial layers II-IV. When administered alone, a 6-h treatment with VEGF(A) had no effect on both apoptotic and necrotic deaths. In contrast, VEGF(A) abolished the glutamate-induced necrosis observed in layer VI. While MEK and PI3-K inhibitors had no effect on the protective action of VEGF(A), L-NAME, a pan inhibitor of NOS, abrogated the effect of VEGF(A) and exacerbated the excitotoxic action of glutamate. Calcimetry experiments performed on brain slices revealed that VEGF(A) reduced the massive calcium influx induced by glutamate in layer VI and this effect was blocked by L-NAME. Neuroprotective effect of VEGF(A) was also blocked by LNIO and NPLA, two inhibitors of constitutive NOS, while AGH, an iNOS inhibitor, had no effect. Nitrite measurements, electron paramagnetic resonance spectroscopy and immunohistochemistry indicated that glutamate was a potent inducer of NO production via activation of nNOS in the cortical layer VI. In vivo administration of nNOS siRNA promoted excitotoxicity and mimicked the effects of L-NAME, LNIO and NPLA. A short-term glutamate treatment increased nNOS Ser1412 phosphorylation, while a long-term exposure inhibited nNOS/NR2B protein-protein interactions. Altogether, these findings indicate that, in deep cortical layers of mice neonates, glutamate stimulates nNOS activity. Contrasting with mature brain, NO production induced by high concentrations of glutamate is neuroprotective and is required for the anti-necrotic effect of VEGF(A).


Subject(s)
Apoptosis/drug effects , Cerebral Cortex/growth & development , Neurons/metabolism , Neuroprotective Agents/pharmacology , Nitric Oxide/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Calcium/metabolism , Caspase 3/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Citrulline/metabolism , Dose-Response Relationship, Drug , Electron Spin Resonance Spectroscopy/methods , Enzyme Inhibitors/pharmacology , Enzyme-Linked Immunosorbent Assay/methods , Excitatory Amino Acid Agents/pharmacology , Gene Expression Regulation, Developmental/drug effects , Glutamate Decarboxylase/genetics , Glutamic Acid/toxicity , Green Fluorescent Proteins/genetics , In Vitro Techniques , L-Lactate Dehydrogenase/metabolism , Mice , Mice, Transgenic , NADPH Dehydrogenase/metabolism , Neurons/drug effects , Nitric Oxide Synthase/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , S-Nitroso-N-Acetylpenicillamine/pharmacology , Time Factors
11.
Ann Neurol ; 72(6): 952-60, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23280843

ABSTRACT

OBJECTIVE: In humans, antenatal alcohol exposure elicits various developmental disorders, in particular in the brain. Numerous studies focus on the deleterious effects of alcohol on neural cells. Although recent studies suggest that alcohol can affect angiogenesis in adults, the impact of prenatal alcohol exposure on brain microvasculature remains poorly understood. METHODS: We used a mouse model to investigate effects of prenatal alcohol exposure on the cortical microvascular network in vivo and ex vivo and the action of alcohol, glutamate, and vascular endothelial growth factor A (VEGF) on activity, plasticity, and survival of microvessels. We used quantitative reverse transcriptase polymerase chain reaction, Western blot, immunohistochemistry, calcimetry, and videomicroscopy. We characterized the effect of prenatal alcohol exposure on the cortical microvascular network in human controls and fetal alcohol syndrome (FAS)/partial FAS (pFAS) patients at different developmental stages. RESULTS: In mice, prenatal alcohol exposure induced a reduction of cortical vascular density, loss of the radial orientation of microvessels, and altered expression of VEGF receptors. Time-lapse experiments performed on brain slices revealed that ethanol inhibited glutamate-induced calcium mobilization in endothelial cells, affected plasticity, and promoted death of microvessels. These effects were prevented by VEGF. In humans, we evidenced a stage-dependent alteration of the vascular network in the cortices of fetuses with pFAS/FAS. Whereas no modification was observed from gestational week 20 (WG20) to WG22, the radial organization of cortical microvessels was clearly altered in pFAS/FAS patients from WG30 to WG38. INTERPRETATION: Prenatal alcohol exposure affects cortical angiogenesis both in mice and in pFAS/FAS patients, suggesting that vascular defects contribute to alcohol-induced brain abnormalities.


Subject(s)
Central Nervous System Depressants/adverse effects , Cerebral Cortex/pathology , Ethanol/adverse effects , Fetal Alcohol Spectrum Disorders/pathology , Microvessels/growth & development , Microvessels/pathology , Prenatal Exposure Delayed Effects/pathology , Age Factors , Animals , Animals, Newborn , CD13 Antigens/metabolism , Calcium/metabolism , Cell Death/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Fetal Alcohol Spectrum Disorders/metabolism , Fetus , Gene Expression Regulation, Developmental/drug effects , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glutamic Acid/pharmacology , Humans , In Vitro Techniques , Locomotion/drug effects , Male , Maze Learning/drug effects , Mice , Microscopy, Video , Microvessels/metabolism , Muscle Strength/physiology , Neovascularization, Pathologic/pathology , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Vascular Endothelial Growth Factor/genetics , Receptors, Vascular Endothelial Growth Factor/metabolism , Time Factors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
12.
Cereb Cortex ; 20(5): 1092-108, 2010 May.
Article in English | MEDLINE | ID: mdl-19759125

ABSTRACT

In term and preterm neonates, massive glutamate release can lead to excitotoxic white-matter and cortical lesions. Because of its high permeability toward calcium, the N-methyl-D-aspartic acid (NMDA) receptor is thought to play an important role in excitotoxic lesions and NMDA antagonists therefore hold promise for neuroprotection. We found that, in neonatal mouse cortex, a given NMDA concentration exerted either excitotoxic or antiapoptotic effects depending on the cortical layers. In layer VI, NMDA led to excitotoxicity, sustained calcium mobilization, and necrosis of Gad67GFP neurons. In the immature layers II-IV, NMDA decreased apoptosis and induced transient calcium mobilization. The NMDA antagonist MK801 acted as a potent caspase-3 activator in immature layers II-IV and affected gamma aminobutyric acid (GABA)ergic interneurons. The apoptotic effect of MK801-induced BAX expression, mitochondrial potential collapse and caspase-9 activation. In vivo Bax small interfering ribonucleic acid and a caspase-9 inhibitor abrogated MK801-induced apoptosis and pyknotic nucleus formation. Ketamine, an anesthetic with NMDA antagonist properties, mimicked the apoptotic effects of MK801. These data indicate a dual effect of glutamate on survival of immature and mature GABAergic neurons and suggest that ketamine may induce apoptosis of immature GABAergic neurons.


Subject(s)
Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Glutamic Acid/pharmacology , Interneurons/physiology , gamma-Aminobutyric Acid/metabolism , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Apoptosis/drug effects , Calcium/metabolism , Caspase 3/metabolism , Dizocilpine Maleate/pharmacology , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation, Developmental/drug effects , Glutamate Decarboxylase/genetics , Green Fluorescent Proteins/genetics , In Vitro Techniques , L-Lactate Dehydrogenase/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Transgenic , N-Methylaspartate/pharmacology , Necrosis/chemically induced , RNA, Small Interfering/pharmacology , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
13.
Endocrinology ; 150(5): 2342-50, 2009 May.
Article in English | MEDLINE | ID: mdl-19164468

ABSTRACT

26RFa is a hypothalamic RFamide neuropeptide that was identified as the endogenous ligand of the orphan G protein-coupled receptor, GPR103, and that stimulates appetite in mice. Up until now, the mechanism of action of 26RFa in the hypothalamic control of food intake remains unknown. The high density of GPR103 in the arcuate nucleus (Arc) prompted us to investigate, in the present study, the effects of 26RFa on the rat neuropeptide Y (NPY)/proopiomelanocortin (POMC) system. Intracerebroventricular injection of 26RFa stimulated NPY expression and release in the basal hypothalamus, whereas it decreased POMC expression and alpha-MSH release, and these effects were associated with an increase in food intake. A double in situ hybridization procedure indicated that the 26RFa receptor is present in NPY neurons of the Arc, but not in POMC neurons. Central administration of NPY Y1 and Y5 receptor antagonists abolished the inhibitory effects of 26RFa on POMC expression and alpha-MSH release, and reversed 26RFa-induced food consumption. Finally, 26RFa antagonized the effects of leptin on NPY expression and release, POMC expression and alpha-MSH release, and food intake. Altogether, the present data demonstrate for the first time that 26RFa exerts its orexigenic activity by stimulating the release of NPY in the Arc, which in turn inhibits POMC neurons by activating the Y1 and Y5 receptors. It is also suggested that the balance 26RFa/leptin is an important parameter in the maintenance of energy homeostasis.


Subject(s)
Appetite Regulation/drug effects , Arcuate Nucleus of Hypothalamus/physiology , Neuropeptide Y/metabolism , Neuropeptides/pharmacology , Pro-Opiomelanocortin/metabolism , Animals , Appetite Regulation/genetics , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Eating/drug effects , Eating/genetics , Energy Metabolism/drug effects , Energy Metabolism/genetics , Gene Expression Regulation/drug effects , Hypothalamic Hormones/administration & dosage , Hypothalamic Hormones/pharmacology , Hypothalamus/drug effects , Hypothalamus/metabolism , Injections, Intraventricular , Leptin/metabolism , Male , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Neuropeptide Y/genetics , Neuropeptide Y/physiology , Neuropeptides/administration & dosage , Pro-Opiomelanocortin/physiology , Rats , Rats, Wistar , alpha-MSH/metabolism
14.
Neuropsychopharmacology ; 34(2): 424-35, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18536705

ABSTRACT

Pituitary adenylate cyclase-activating polypeptide (PACAP) and the proopiomelanocortin (POMC)-derived peptide, alpha-melanocyte-stimulating hormone (alpha-MSH), exert anorexigenic activities. While alpha-MSH is known to inhibit food intake and stimulate catabolism via activation of the central melanocortin-receptor MC4-R, little is known regarding the mechanism by which PACAP inhibits food consumption. We have recently found that, in the arcuate nucleus of the hypothalamus, a high proportion of POMC neurons express PACAP receptors. This observation led us to investigate whether PACAP may inhibit food intake through a POMC-dependent mechanism. In mice deprived of food for 18 h, intracerebroventricular administration of PACAP significantly reduced food intake after 30 min, and this effect was reversed by the PACAP antagonist PACAP6-38. In contrast, vasoactive intestinal polypeptide did not affect feeding behavior. Pretreatment with the MC3-R/MC4-R antagonist SHU9119 significantly reduced the effect of PACAP on food consumption. Central administration of PACAP induced c-Fos mRNA expression and increased the proportion of POMC neuron-expressing c-Fos mRNA in the arcuate nucleus. Furthermore, PACAP provoked an increase in POMC and MC4-R mRNA expression in the hypothalamus, while MC3-R mRNA level was not affected. POMC mRNA level in the arcuate nucleus of PACAP-specific receptor (PAC1-R) knock-out mice was reduced as compared with wild-type animals. Finally, i.c.v. injection of PACAP provoked a significant increase in plasma glucose level. Altogether, these results indicate that PACAP, acting through PAC1-R, may inhibit food intake via a melanocortin-dependent pathway. These data also suggest a central action of PACAP in the control of glucose metabolism.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Eating/drug effects , Hypothalamus/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Pro-Opiomelanocortin/metabolism , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Blood Glucose/analysis , Corticosterone/blood , Dose-Response Relationship, Drug , Eating/physiology , Hypothalamus/drug effects , Male , Melanocyte-Stimulating Hormones/pharmacology , Mice , Mice, Knockout , Neurons/drug effects , Neurons/metabolism , Neuropeptide Y/metabolism , Peptide Fragments/pharmacology , Pituitary Adenylate Cyclase-Activating Polypeptide/antagonists & inhibitors , Pro-Opiomelanocortin/genetics , RNA, Messenger/metabolism , Receptor, Melanocortin, Type 3/antagonists & inhibitors , Receptor, Melanocortin, Type 3/metabolism , Receptor, Melanocortin, Type 4/antagonists & inhibitors , Receptor, Melanocortin, Type 4/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Vasoactive Intestinal Peptide/pharmacology
15.
J Neurochem ; 107(2): 361-74, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18710417

ABSTRACT

Urotensin II (UII) and UII-related peptide (URP) are paralog neuropeptides whose existence and distribution in mouse have not yet been investigated. In this study, we showed by HPLC/RIA analysis that the UII-immunoreactive molecule in the mouse brain corresponds to a new UII(17) isoform. Moreover, calcium mobilization assays indicated that UII(17) and URP were equally potent in stimulating UII receptor (UT receptor). Quantitative RT-PCR and in situ hybridization analysis revealed that in the CNS UII and URP mRNAs were predominantly expressed in brainstem and spinal motoneurons. Besides, they were differentially expressed in the medial vestibular nucleus, locus coeruleus and the ventral medulla. In periphery, both mRNAs were expressed in skeletal muscle, testis, vagina, stomach, and gall bladder, whereas only URP mRNA could be detected in the seminal vesicle, heart, colon, and thymus. By contrast, the UT receptor mRNA was widely expressed, and notably, very high amounts of transcript occurred in skeletal muscle and prostate. In the biceps femoris muscle, UII-like immunoreactivity was shown to coexist with synaptophysin in muscle motor end plate regions. Altogether these results suggest that (i) UII and URP may have many redundant biological effects, especially at the neuromuscular junction; (ii) URP may more specifically participate to autonomic, cardiovascular and reproductive functions.


Subject(s)
Brain/metabolism , Neuromuscular Junction/metabolism , Peptide Hormones/metabolism , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/genetics , Urotensins/metabolism , Animals , Brain/anatomy & histology , CHO Cells , Calcium/metabolism , Chromatography, High Pressure Liquid , Cricetinae , Cricetulus , Female , Male , Mice , Radioimmunoassay/methods , Receptors, G-Protein-Coupled/metabolism , Synaptophysin/metabolism , Tyrosine 3-Monooxygenase/metabolism , Urotensins/chemistry
16.
Endocrinology ; 149(6): 2840-52, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18292192

ABSTRACT

Chromaffin cells of the adrenal medulla elaborate and secrete catecholamines and neuropeptides for hormonal and paracrine signaling in stress and during inflammation. We have recently documented the action of the cytokine TNF-alpha on neuropeptide secretion and biosynthesis in isolated bovine chromaffin cells. Here, we demonstrate that the type 2 TNF-alpha receptor (TNF-R2) mediates TNF-alpha signaling in chromaffin cells via activation of nuclear factor (NF)-kappaB. Microarray and suppression subtractive hybridization have been used to identify TNF-alpha target genes in addition to those encoding the neuropeptides galanin, vasoactive intestinal polypeptide, and secretogranin II in chromaffin cells. TNF-alpha, acting through the TNF-R2, causes an early up-regulation of NF-kappaB, long-lasting induction of the NF-kappaB target gene inhibitor kappaB (IkappaB), and persistent stimulation of other NF-kappaB-associated genes including mitogen-inducible gene-6 (MIG-6), which acts as an IkappaB signaling antagonist, and butyrate-induced transcript 1. Consistent with long-term activation of the NF-kappaB signaling pathway, delayed induction of neuropeptide gene transcription by TNF-alpha in chromaffin cells is blocked by an antagonist of NF-kappaB signaling. TNF-alpha-dependent signaling in neuroendocrine cells thus leads to a unique, persistent mode of NF-kappaB activation that features long-lasting transcription of both IkappaB and MIG-6, which may play a role in the long-lasting effects of TNF-alpha in regulating neuropeptide output from the adrenal, a potentially important feedback station for modulating long-term cytokine effects in inflammation.


Subject(s)
Chromaffin Cells/physiology , Inflammation/physiopathology , NF-kappa B/physiology , Neuropeptides/genetics , Signal Transduction/physiology , TNF Receptor-Associated Factor 2/physiology , Tumor Necrosis Factor-alpha/pharmacology , Adrenal Glands/cytology , Adrenal Glands/physiology , Animals , Cattle , Chromaffin Cells/drug effects , Gene Expression Regulation , Humans , In Situ Hybridization , Oligonucleotide Array Sequence Analysis , RNA/genetics , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
17.
J Comp Neurol ; 503(4): 573-91, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17534937

ABSTRACT

The novel RFamide peptide 26RFa, the endogenous ligand of the orphan receptor GPR103, affects food intake, locomotion, and activity of the gonadotropic axis. However, little is known regarding the localization of 26RFa receptors. The present report provides the first detailed mapping of 26RFa binding sites and GPR103 mRNA in the rat central nervous system (CNS). 26RFa binding sites were widely distributed in the brain and spinal cord, whereas the expression of GPR103 mRNA was more discrete, notably in the midbrain, the pons, and the medulla oblongata, suggesting that 26RFa can bind to a receptor(s) other than GPR103. Competition experiments confirmed that 26RFa interacts with an RFamide peptide receptor distinct from GPR103 that may be NPFF2. High densities of 26RFa binding sites were observed in olfactory, hypothalamic, and brainstem nuclei involved in the control of feeding behavior, including the piriform cortex, the ventromedial and dorsomedial hypothalamic nuclei, the paraventricular nucleus, the arcuate nucleus, the lateral hypothalamic area, and the nucleus of the solitary tract. The preoptic and anterior hypothalamic areas were also enriched with 26RFa recognition sites, supporting a physiological role of the neuropeptide in the regulation of the gonadotropic axis. A high density of 26RFa binding sites was detected in regions of the CNS involved in the processing of pain, such as the dorsal horn of the spinal cord and the parafascicular thalamic nucleus. The wide distribution of 26RFa binding sites suggests that 26RFa has multiple functions in the CNS that are mediated by at least two distinct receptors.


Subject(s)
Central Nervous System/metabolism , Neuropeptides/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Binding Sites/drug effects , Binding Sites/physiology , Binding, Competitive/drug effects , Brain Mapping , Dose-Response Relationship, Drug , In Situ Hybridization/methods , Iodine Isotopes/pharmacokinetics , Male , Neuropeptides/pharmacokinetics , RNA, Messenger/metabolism , Radioligand Assay/methods , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/genetics
18.
Ann N Y Acad Sci ; 1070: 457-61, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16888209

ABSTRACT

Neuropeptide Y (NPY) and pituitary adenylate cyclase-activating polypeptide (PACAP) exert opposite actions in energy homeostasis: NPY is a potent orexigenic peptide whereas PACAP reduces food intake. PAC1-R and VPAC2-R mRNAs are actively expressed in the arcuate nucleus of the hypothalamus which contains a prominent population of NPY neurons. By using a double-labeling in situ hybridization technique, we now show that a significant proportion of NPY neurons express PAC1-R or VPAC2-R mRNA. This observation indicates that PACAP may regulate the activity of NPY neurons, suggesting that the inhibitory effect of PACAP on food intake may be mediated, at least in part, through modulation of NPY neurotransmission.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Neuropeptide Y/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Animals , RNA, Messenger/genetics , Rats
19.
Ann N Y Acad Sci ; 1070: 512-7, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16888217

ABSTRACT

Two VIP receptors, shared with a similar affinity by pituitary adenylate cyclase-activating polypeptide (PACAP), have been cloned: VPAC1 and VPAC2. PHI binds to these receptors with a lower affinity. We previously showed that VIP protects against excitotoxic white matter damage in newborn mice. This article aimed to determine the receptor involved in VIP-induced neuroprotection. VIP effects were mimicked with a similar potency by VPAC2 agonists and PHI but not by VPAC1 agonists, PACAP 27 or PACAP 38. VIP neuroprotective effects were lost in mice lacking VPAC2 receptor. In situ hybridization confirmed the presence of VPAC2 mRNA. These data suggest that, in this model, VIP-induced neuroprotection is mediated by VPAC2 receptors. The pharmacology of this VPAC2 receptor seems unconventional as PACAP does not mimic VIP effects and PHI acts with a comparable potency.


Subject(s)
Neurons/drug effects , Neuroprotective Agents/pharmacology , Neurotoxins/pharmacology , Vasoactive Intestinal Peptide/pharmacology , Animals , Animals, Newborn , Brain/drug effects , Brain-Derived Neurotrophic Factor/genetics , Mice , Neurons/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Receptors, Vasoactive Intestinal Peptide, Type II/metabolism
20.
J Comp Neurol ; 495(1): 21-36, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16432902

ABSTRACT

The vasoactive peptide urotensin II (UII) is primarily expressed in motoneurons of the brainstem and spinal cord. Intracerebroventricular injection of UII provokes various behavioral, cardiovascular, motor, and endocrine responses in the rat, but the distribution of the UII receptor in the central nervous system (CNS) has not yet been determined. In the present study, we have investigated the localization of UII receptor (GPR14) mRNA and UII binding sites in the rat CNS. RT-PCR analysis revealed that the highest density of GPR14 mRNA occurred in the pontine nuclei. In situ hybridization histochemistry showed that the GPR14 gene is widely expressed in the brain and spinal cord. In particular, a strong hybridization signal was observed in the olfactory system, hippocampus, olfactory and medial amygdala, hypothalamus, epithalamus, several tegmental nuclei, locus coeruleus, pontine nuclei, motor nuclei, nucleus of the solitary tract, dorsal motor nucleus of the vagus, inferior olive, cerebellum, and spinal cord. Autoradiographic labeling of brain slices with radioiodinated UII showed the presence of UII-binding sites in the lateral septum, bed nucleus of the stria terminalis, medial amygdaloid nucleus, anteroventral thalamus, anterior pretectal nucleus, pedunculopontine tegmental nucleus, pontine nuclei, geniculate nuclei, parabigeminal nucleus, dorsal endopiriform nucleus, and cerebellar cortex. Intense expression of the GPR14 gene in some hypothalamic nuclei (supraoptic, paraventricular, ventromedian, and arcuate nuclei), in limbic structures (amygdala and hippocampus), in medullary nuclei (solitary tract, dorsal motor nucleus of the vagus), and in motor control regions (cerebral and cerebellar cortex, substantia nigra, pontine nuclei) provides the anatomical substrate for the central effects of UII on behavioral, cardiovascular, neuroendocrine, and motor functions. The occurrence of GPR14 mRNA in cranial and spinal motoneurons is consistent with the reported autocrine/paracrine action of UII on motoneurons.


Subject(s)
Central Nervous System/metabolism , Neurons/metabolism , Receptors, G-Protein-Coupled/genetics , Urotensins/metabolism , Animals , Binding, Competitive/physiology , Brain/anatomy & histology , Brain/metabolism , Central Nervous System/anatomy & histology , Iodine Radioisotopes , Male , Motor Neurons/metabolism , Neurons/cytology , RNA, Messenger/metabolism , Radioligand Assay , Rats , Rats, Wistar , Spinal Cord/anatomy & histology , Spinal Cord/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL