Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
1.
Brain Behav Immun Health ; 38: 100791, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38818370

RESUMEN

Background: Immune system activation in the neonatal period is associated with white matter injury in preterm infants. In animal studies, neonatal priming of the immune system leads to chronic activation of i.e. microglia cells and altered neuroinflammatory responses potentially years after preterm birth. This may contribute further to brain injury and neurodevelopmental impairment. It is unknown to what extend this also occurs in human. Aim: To identify neuro-inflammatory markers at school age that relate to motor, cognitive and behavioral impairments in preterm born children in a pilot case-control study. Methods: We included n = 20 preterm born children (GA < 28 weeks) in this study, of which n = 10 with motor, cognitive and behavorial impairments and n = 10 preterm born controls next to n = 30 healthy adult controls. In the preterm children, at 8-9 years, 39 inflammatory markers were assessed by Luminex assay in blood serum samples. Firstly, the preterm concentrations of these markers were compared to n = 30 adult controls. Then a univariate analysis was performed to determine differences in values between preterm children with and without impairment at school age. Finally, a principal component analysis and hierarchical clustering was performed to identify protein profiles in preterm born children that relate to impairment at school age. Results: Inflammatory proteins in preterm children at school age differed from values of adult controls. Within the group of preterm children, we found significantly higher levels of GM-CSF in preterms with impairment (p < 0.01) and a trend towards significance for Gal1 and TRAIL (p = 0.06 and p = 0.06 respectively) when compared to preterms without impairment. In addition, differences in clustering of proteins between preterm children was observed, however this variance was not explained by presence of neurodevelopmental impairments. Conclusion: The inflammatory profile at school age in preterm children is different from that of adult controls. The immune modulating cytokines GM-CSF, Gal1 and TRAIL were higher in preterm children with impairment than control preterm children, suggesting that immune responses are altered in these children. No specific cluster of inflammatory markers could be identified. Results indicate that even at school age, neuroinflammatory pathways are activated in preterm born children with neurodevelopmental impairments.

2.
Sci Rep ; 13(1): 2237, 2023 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-36754974

RESUMEN

SARS-CoV-2 interacts with cellular cholesterol during many stages of its replication cycle. Pantethine was reported to reduce total cholesterol levels and fatty acid synthesis and potentially alter different processes that might be involved in the SARS-CoV-2 replication cycle. Here, we explored the potential antiviral effects of pantethine in two in vitro experimental models of SARS-CoV-2 infection, in Vero E6 cells and in Calu-3a cells. Pantethine reduced the infection of cells by SARS-CoV-2 in both preinfection and postinfection treatment regimens. Accordingly, cellular expression of the viral spike and nucleocapsid proteins was substantially reduced, and we observed a significant reduction in viral copy numbers in the supernatant of cells treated with pantethine. In addition, pantethine inhibited the infection-induced increase in TMPRSS2 and HECT E3 ligase expression in infected cells as well as the increase in antiviral interferon-beta response and inflammatory gene expression in Calu-3a cells. Our results demonstrate that pantethine, which is well tolerated in humans, was very effective in controlling SARS-CoV-2 infection and might represent a new therapeutic drug that can be repurposed for the prevention or treatment of COVID-19 and long COVID syndrome.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Chlorocebus aethiops , Humanos , Antivirales/farmacología , Síndrome Post Agudo de COVID-19 , Replicación Viral , Células Vero
3.
Neuropsychopharmacology ; 46(3): 579-602, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32781459

RESUMEN

Maternal immune activation (MIA) and poor maternal nutritional habits are risk factors for the occurrence of neurodevelopmental disorders (NDD). Human studies show the deleterious impact of prenatal inflammation and low n-3 polyunsaturated fatty acid (PUFA) intake on neurodevelopment with long-lasting consequences on behavior. However, the mechanisms linking maternal nutritional status to MIA are still unclear, despite their relevance to the etiology of NDD. We demonstrate here that low maternal n-3 PUFA intake worsens MIA-induced early gut dysfunction, including modification of gut microbiota composition and higher local inflammatory reactivity. These deficits correlate with alterations of microglia-neuron crosstalk pathways and have long-lasting effects, both at transcriptional and behavioral levels. This work highlights the perinatal period as a critical time window, especially regarding the role of the gut-brain axis in neurodevelopment, elucidating the link between MIA, poor nutritional habits, and NDD.


Asunto(s)
Ácidos Grasos Omega-3 , Efectos Tardíos de la Exposición Prenatal , Animales , Conducta Animal , Encéfalo , Femenino , Humanos , Inflamación , Microglía , Embarazo
4.
Neuropathol Appl Neurobiol ; 47(2): 297-315, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32898926

RESUMEN

AIMS: Impairment of blood-brain barrier (BBB) is involved in numerous neurological diseases from developmental to aging stages. Reliable imaging of increased BBB permeability is therefore crucial for basic research and preclinical studies. Today, the analysis of extravasation of exogenous dyes is the principal method to study BBB leakage. However, these procedures are challenging to apply in pups and embryos and may appear difficult to interpret. Here we introduce a novel approach based on agonist-induced internalization of a neuronal G protein-coupled receptor widely distributed in the mammalian brain, the somatostatin receptor type 2 (SST2). METHODS: The clinically approved SST2 agonist octreotide (1 kDa), when injected intraperitoneally does not cross an intact BBB. At sites of BBB permeability, however, OCT extravasates and induces SST2 internalization from the neuronal membrane into perinuclear compartments. This allows an unambiguous localization of increased BBB permeability by classical immunohistochemical procedures using specific antibodies against the receptor. RESULTS: We first validated our approach in sensory circumventricular organs which display permissive vascular permeability. Through SST2 internalization, we next monitored BBB opening induced by magnetic resonance imaging-guided focused ultrasound in murine cerebral cortex. Finally, we proved that after intraperitoneal agonist injection in pregnant mice, SST2 receptor internalization permits analysis of BBB integrity in embryos during brain development. CONCLUSIONS: This approach provides an alternative and simple manner to assess BBB dysfunction and development in different physiological and pathological conditions.


Asunto(s)
Barrera Hematoencefálica/patología , Permeabilidad Capilar , Inmunohistoquímica/métodos , Receptores de Somatostatina/análisis , Receptores de Somatostatina/metabolismo , Animales , Anticuerpos Monoclonales , Ratones , Ratones Endogámicos C57BL , Octreótido/metabolismo , Ratas , Ratas Wistar
5.
Nat Commun ; 11(1): 6133, 2020 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-33257673

RESUMEN

Omega-3 fatty acids (n-3 PUFAs) are essential for the functional maturation of the brain. Westernization of dietary habits in both developed and developing countries is accompanied by a progressive reduction in dietary intake of n-3 PUFAs. Low maternal intake of n-3 PUFAs has been linked to neurodevelopmental diseases in Humans. However, the n-3 PUFAs deficiency-mediated mechanisms affecting the development of the central nervous system are poorly understood. Active microglial engulfment of synapses regulates brain development. Impaired synaptic pruning is associated with several neurodevelopmental disorders. Here, we identify a molecular mechanism for detrimental effects of low maternal n-3 PUFA intake on hippocampal development in mice. Our results show that maternal dietary n-3 PUFA deficiency increases microglia-mediated phagocytosis of synaptic elements in the rodent developing hippocampus, partly through the activation of 12/15-lipoxygenase (LOX)/12-HETE signaling, altering neuronal morphology and affecting cognitive performance of the offspring. These findings provide a mechanistic insight into neurodevelopmental defects caused by maternal n-3 PUFAs dietary deficiency.


Asunto(s)
Encéfalo/efectos de los fármacos , Ácidos Grasos Omega-3/farmacología , Microglía/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Fagocitosis/efectos de los fármacos , Animales , Encéfalo/crecimiento & desarrollo , Suplementos Dietéticos , Ácidos Grasos Omega-3/deficiencia , Ácidos Grasos Omega-3/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hipocampo/citología , Hipocampo/crecimiento & desarrollo , Homeostasis , Humanos , Lipooxigenasa , Masculino , Ratones , Trastornos del Neurodesarrollo
6.
Brain Behav Immun ; 73: 427-440, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29879442

RESUMEN

Maternal immune activation (MIA) is a common environmental insult on the developing brain and represents a risk factor for neurodevelopmental disorders. Animal models of in utero inflammation further revealed a causal link between maternal inflammatory activation during pregnancy and behavioural impairment relevant to neurodevelopmental disorders in the offspring. Accumulating evidence point out that proinflammatory cytokines produced both in the maternal and fetal compartments are responsible for social, cognitive and emotional behavioral deficits in the offspring. Polyunsaturated fatty acids (PUFAs) are essential fatty acids with potent immunomodulatory activities. PUFAs and their bioactive derivatives can promote or inhibit many aspects of the immune and inflammatory response. PUFAs of the n-3 series ('n-3 PUFAs', also known as omega-3) exhibit anti-inflammatory/pro-resolution properties and promote immune functions, while PUFAs of the n-6 series ('n-6 PUFAs' or omega-6) favor pro-inflammatory responses. The present study aimed at providing insight into the effects of n-3 PUFAs on the consequences of MIA on brain development. We hypothesized that a reduction in n-3 PUFAs exacerbates both maternal and fetal inflammatory responses to MIA and later-life defects in memory in the offspring. Based on a lipopolysaccharide (LPS) model of MIA (LPS injection at embryonic day 17), we showed that n-3 PUFA deficiency 1) alters fatty acid composition of the fetal and adult offspring brain; 2) exacerbates maternal and fetal inflammatory processes with no significant alteration of microglia phenotype, and 3) induces spatial memory deficits in the adult offspring. We also showed a strong negative correlation between brain content in n-3 PUFA and cytokine production in MIA-exposed fetuses. Overall, our study is the first to address the deleterious effects of n-3 PUFA deficiency on brain lipid composition, inflammation and memory performances in MIA-exposed animals and indicates that it should be considered as a potent environmental risk factor for the apparition of neurodevelopmental disorders.


Asunto(s)
Ácidos Grasos Omega-3/deficiencia , Ácidos Grasos Omega-3/metabolismo , Memoria Espacial/efectos de los fármacos , Animales , Animales Recién Nacidos , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Citocinas/efectos de los fármacos , Suplementos Dietéticos , Modelos Animales de Enfermedad , Ácidos Grasos Omega-3/farmacología , Ácidos Grasos Omega-6/metabolismo , Ácidos Grasos Omega-6/fisiología , Femenino , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Trastornos del Neurodesarrollo/etiología , Trastornos del Neurodesarrollo/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/inmunología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Conducta Social
7.
Clin Genet ; 93(6): 1141-1147, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29508392

RESUMEN

Ephrin B2, one of the ligand of the EphB receptors, is involved in a complex signaling pathway regulating the development of the nervous system, neuronal migration, erythropoiesis and vasculogenesis. We report a patient with a de novo variant in EFNB2 and a family in which segregates a 610-kb deletion at chromosome 13q33 encompassing only ARGLU1 and EFNB2 genes. The de novo variant was observed in a patient with anal stenosis, hypoplastic left ventricle and mild developmental delay. The deletion was identified in 2 sibs with congenital heart defect and mild developmental delay. One of the affected sibs further had myoclonic epilepsy and bilateral sensorineural hearing loss. The carrier mother was apparently asymptomatic. Because EFNB2 is located in the subtelomeric region of 13q chromosome, we reviewed the previous reports of terminal 13q deletion. We suggest that haploinsufficiency of the EFNB2 could be at the origin of several clinical features reported in 13qter deletions, including intellectual disability, seizures, congenital heart defects, anorectal malformation and hearing loss.


Asunto(s)
Trastornos de los Cromosomas/genética , Efrina-B2/genética , Haploinsuficiencia/genética , Trastornos del Neurodesarrollo/genética , Preescolar , Deleción Cromosómica , Cromosomas Humanos Par 13/genética , Femenino , Estudios de Asociación Genética , Humanos , Lactante , Recién Nacido , Masculino , Linaje
8.
Neuroscience ; 311: 382-97, 2015 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-26542996

RESUMEN

The blood-brain barrier (BBB) is a complex structure that protects the central nervous system from peripheral insults. Understanding the molecular basis of BBB function and dysfunction holds significant potential for future strategies to prevent and treat neurological damage. The aim of our study was (1) to investigate BBB alterations following excitotoxicity and (2) to test the protective properties of melatonin. Ibotenate, a glutamate analog, was injected intracerebrally in postnatal day 5 (P5) rat pups to mimic excitotoxic injury. Animals were than randomly divided into two groups, one receiving intraperitoneal (i.p.) melatonin injections (5mg/kg), and the other phosphate buffer saline (PBS) injections. Pups were sacrificed 2, 4 and 18 h after ibotenate injection. We determined lesion size at 5 days by histology, the location and organization of tight junction (TJ) proteins by immunohistochemical studies, and BBB leakage by dextran extravasation. Expression levels of BBB genes (TJs, efflux transporters and detoxification enzymes) were determined in the cortex and choroid plexus by quantitative PCR. Dextran extravasation was seen 2h after the insult, suggesting a rapid BBB breakdown that was resolved by 4h. Extravasation was significantly reduced in melatonin-treated pups. Gene expression and immunohistochemical assays showed dynamic BBB modifications during the first 4h, partially prevented by melatonin. Lesion-size measurements confirmed white matter neuroprotection by melatonin. Our study is the first to evaluate BBB structure and function at a very early time point following excitotoxicity in neonates. Melatonin neuroprotects by preventing TJ modifications and BBB disruption at this early phase, before its previously demonstrated anti-inflammatory, antioxidant and axonal regrowth-promoting effects.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Melatonina/farmacología , Fármacos Neuroprotectores/farmacología , Animales , Animales Recién Nacidos , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/fisiología , Modelos Animales de Enfermedad , Fármacos actuantes sobre Aminoácidos Excitadores/toxicidad , Expresión Génica/efectos de los fármacos , Ácido Glutámico/análogos & derivados , Ácido Glutámico/toxicidad , Inmunohistoquímica , Distribución Aleatoria , Ratas Sprague-Dawley
9.
Acta Anaesthesiol Scand ; 58(6): 733-42, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24724965

RESUMEN

BACKGROUND: The highly selective α2 -adrenoreceptor agonist, dexmedetomidine, exerts neuroprotective, analgesic, anti-inflammatory and sympatholytic properties that may be beneficial for perinatal asphyxia. The optimal safe dose for pre-clinical newborn neuroprotection studies is unknown. METHODS: Following cerebral hypoxia-ischaemia, dexmedetomidine was administered to nine newborn piglets in a de-escalation dose study in combination with hypothermia (whole body cooling to 33.5°C). Dexmedetomidine was administered with a loading dose of 1 µg/kg and maintenance infusion at doses from 10 to 0.6 µg/kg/h. One additional piglet was not subjected to hypoxia-ischaemia. Blood for pharmacokinetic analysis was sampled pre-insult and frequently post-insult. A one-compartment linear disposition model was used to fit data. Population parameter estimates were obtained using non-linear mixed effects modelling. RESULTS: All dexmedetomidine infusion regimens led to plasma concentrations above those associated with sedation in neonates and children (0.4-0.8 µg/l). Seven out of the nine piglets with hypoxia-ischaemia experienced periods of bradycardia, hypotension, hypertension and cardiac arrest; all haemodynamic adverse events occurred in piglets with plasma concentrations greater than 1 µg/l. Dexmedetomidine clearance was 0.126 l/kg/h [coefficient of variation (CV) 46.6.%] and volume of distribution was 3.37 l/kg (CV 191%). Dexmedetomidine clearance was reduced by 32.7% at a temperature of 33.5°C. Dexmedetomidine clearance was reduced by 55.8% following hypoxia-ischaemia. CONCLUSIONS: Dexmedetomidine clearance was reduced almost tenfold compared with adult values in the newborn piglet following hypoxic-ischaemic brain injury and subsequent therapeutic hypothermia. Reduced clearance was related to cumulative effects of both hypothermia and exposure to hypoxia. High plasma levels of dexmedetomidine were associated with major cardiovascular complications.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacocinética , Asfixia Neonatal/complicaciones , Dexmedetomidina/farmacocinética , Hipotermia Inducida , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Fármacos Neuroprotectores/farmacocinética , Agonistas de Receptores Adrenérgicos alfa 2/sangre , Agonistas de Receptores Adrenérgicos alfa 2/uso terapéutico , Animales , Dexmedetomidina/sangre , Dexmedetomidina/uso terapéutico , Modelos Animales de Enfermedad , Hipoxia-Isquemia Encefálica/etiología , Masculino , Tasa de Depuración Metabólica , Fármacos Neuroprotectores/sangre , Fármacos Neuroprotectores/uso terapéutico , Dinámicas no Lineales , Sus scrofa , Porcinos
10.
Artículo en Inglés | MEDLINE | ID: mdl-25598870

RESUMEN

Systems biology is an interdisciplinary effort to integrate molecular, cellular, tissue, organ, and organism levels of function into computational models that facilitate the identification of general principles. Systems medicine adds a disease focus. Systems epidemiology adds yet another level consisting of antecedents that might contribute to the disease process in populations. In etiologic and prevention research, systems-type thinking about multiple levels of causation will allow epidemiologists to identify contributors to disease at multiple levels as well as their interactions. In public health, systems epidemiology will contribute to the improvement of syndromic surveillance methods. We encourage the creation of computational simulation models that integrate information about disease etiology, pathogenetic data, and the expertise of investigators from different disciplines.

11.
Ann Fr Anesth Reanim ; 32(12): 863-71, 2013 Dec.
Artículo en Francés | MEDLINE | ID: mdl-24169200

RESUMEN

Cerebral ischemia is a common thread of acute cerebral lesions, whether vascular or traumatic origin. Hyperbaric oxygen (HBO) improves tissue oxygenation and may prevent impairment of reversible lesions. In experimental models of cerebral ischemia or traumatic brain injury, HBO has neuroprotective effects which are related to various mechanisms such as modulation of oxidative stress, neuro-inflammation or cerebral and mitochondrial metabolism. However, results of clinical trials failed to prove any neuroprotective effects for cerebral ischemia and remained to be confirmed for traumatic brain injury despite preliminary encouraging results. The addition of inert gases to HBO sessions, especially argon or xenon which show neuroprotective experimental effects, may provide an additional improvement of cerebral lesions. Further multicentric studies with a strict methodology and a better targeted definition are required before drawing definitive conclusions about the efficiency of combined therapy with HBO and inert gases in acute cerebral lesions.


Asunto(s)
Lesiones Encefálicas/terapia , Isquemia Encefálica/terapia , Oxigenoterapia Hiperbárica , Gases Nobles/uso terapéutico , Animales , Lesiones Encefálicas/fisiopatología , Isquemia Encefálica/fisiopatología , Humanos , Estrés Oxidativo
12.
Dev Neurosci ; 35(2-3): 130-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23548575

RESUMEN

Toll-like receptors (TLRs) are members of the pattern recognition receptor family that detect components of foreign pathogens or endogenous molecules released in response to injury. Recent studies demonstrate that TLRs also have a functional role in regulating neuronal proliferation in the developing brain. This study investigated cellular expression of TLR3 using immunohistochemistry on human brain tissue. The tissue sections analysed contained anterior and lateral periventricular white matter from the frontal and parietal lobes in post-mortem neonatal cases with a postmenstrual age range of 23.6-31.4 weeks. In addition to preterm brains without overt pathology (control), preterm pathology cases with evidence of white matter injuries (WMI) were also examined. In order to identify TLR-positive cells, we utilized standard double-labelling immunofluorescence co-labelling techniques and confocal microscopy to compare co-expression of TLR3 with a neuronal marker (NeuN) or with glial markers (GFAP for astrocytes, Iba-1 for microglia and Olig2 for oligodendrocytes). We observed an increase in the neuronal (28 vs. 17%) and astroglial (38 vs. 21%) populations in the WMI group compared to controls in the anterior regions of the periventricular white matter in the frontal lobe. The increase in neurons and astrocytes in the WMI cases was associated with an increase in TLR3 immunoreactivity. This expression was significantly increased in the astroglia. The morphology of the TLR3 signal in the control cases was globular and restricted to the perinuclear region of the neurons and astrocytes, whilst in the cases of WMI, both neuronal, axonal and astroglial TLR3 expression was more diffuse (i.e., a different intracellular distribution) and could be detected along the extensions of the processes. This study demonstrates for the first time that neurons and glial cells in human neonatal periventricular white matter express TLR3 during development. The patterns of TLR3 expression were altered in the presence of WMI, which might influence normal developmental processes within the immature brain. Identifying changes in TLR3 expression during fetal development may be key to understanding the reduced volumes of grey matter and impaired cortical development seen in preterm infants.


Asunto(s)
Encéfalo/metabolismo , Recien Nacido Extremadamente Prematuro/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Receptor Toll-Like 3/biosíntesis , Encéfalo/crecimiento & desarrollo , Encéfalo/patología , Lesiones Encefálicas/metabolismo , Humanos , Recien Nacido Extremadamente Prematuro/crecimiento & desarrollo , Recién Nacido , Fibras Nerviosas Mielínicas/metabolismo , Fibras Nerviosas Mielínicas/patología , Receptor Toll-Like 3/análisis
13.
Arch Pediatr ; 19(9): 946-52, 2012 Sep.
Artículo en Francés | MEDLINE | ID: mdl-22885003

RESUMEN

Perinatal inflammation can lead to fetal/neonatal inflammatory syndrome, a risk factor for brain lesions, especially in the white matter. Perinatal inflammation is associated with increased incidence of cerebral palsy in humans and animal models and there is a strong relationship with increased incidence of autism and schizophrenia in humans. Perinatal inflammation causes acute microglial and astroglial activation, blood-brain barrier dysfunction, and disrupts oligodendrocyte maturation leading to hypomyelination. Inflammation also sensitizes the brain to additional perinatal insults, including hypoxia-ischemia. Furthermore, long after the primary cause of inflammation has resolved, gliosis may also persist and predispose to neurodegenerative diseases including Alzheimer's and Parkinson's disease, but this relation is still hypothetical. Finding of acute and chronic changes in brain structure and function due to perinatal inflammation highlights the need for treatments. As gliosis appears to be involved in the acute and chronic effects of perinatal inflammation, modulating the glial phenotype may be an effective strategy to prevent damage to the brain.


Asunto(s)
Encefalopatías/etiología , Inflamación/complicaciones , Encéfalo/crecimiento & desarrollo , Humanos , Recién Nacido , Trastornos Mentales/etiología , Microglía/fisiología , Enfermedades del Sistema Nervioso/etiología , Factores de Tiempo
14.
Neuroscience ; 199: 103-15, 2011 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-22001490

RESUMEN

Identification of novel molecules that can induce neuronal differentiation of embryonic stem (ES) cells is essential for deciphering the molecular mechanisms of early development and for exploring cell therapy approaches. Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal polypeptide (VIP) are known to be implicated early during ontogenesis in cell proliferation and neuronal differentiation. The aim of the present study was to determine the effects of VIP and PACAP on functional differentiation of ES cells. Quantitative-reverse transcription-polymerase chain reaction analysis showed an inversion of the expression pattern of PAC1 and VPAC1 receptors with time. ES cells expressed genes encoding extracellular signal-regulated kinase 1 and 2 and c-jun amino terminal kinase1. ES cells also expressed T-type α1I and α1G, L-type α1C and α1D, and N-type α1B calcium channel subunit mRNAs. Both peptides modified the shape of undifferentiated ES cells into bipolar cells expressing the neuronal marker neuron-specific enolase (NSE). Immunostaining indicated that PACAP intensified T-type α1I subunit immunoreactivity, whereas VIP increased L-types α1C and α1D, as well as N-type α1B subunit. Electrophysiological recording showed that VIP and PACAP enhanced transient calcium current. Moreover, VIP generated sustained calcium current. These findings demonstrate that PACAP and VIP induce morphological and functional differentiation of ES cells into a neuronal phenotype. Both peptides promote functional maturation of calcium channel subunits, suggesting that they can facilitate the genesis of cellular excitability.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Péptido Intestinal Vasoactivo/metabolismo , Animales , Canales de Calcio/genética , Canales de Calcio/metabolismo , Células Madre Embrionarias/citología , Perfilación de la Expresión Génica , Inmunohistoquímica , Ratones , Células-Madre Neurales/citología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Péptido Intestinal Vasoactivo/genética
15.
Cell Death Dis ; 2: e203, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21881605

RESUMEN

Brain protection of the newborn remains a challenging priority and represents a totally unmet medical need. Pharmacological inhibition of caspases appears as a promising strategy for neuroprotection. In a translational perspective, we have developed a pentapeptide-based group II caspase inhibitor, TRP601/ORPHA133563, which reaches the brain, and inhibits caspases activation, mitochondrial release of cytochrome c, and apoptosis in vivo. Single administration of TRP601 protects newborn rodent brain against excitotoxicity, hypoxia-ischemia, and perinatal arterial stroke with a 6-h therapeutic time window, and has no adverse effects on physiological parameters. Safety pharmacology investigations, and toxicology studies in rodent and canine neonates, suggest that TRP601 is a lead compound for further drug development to treat ischemic brain damage in human newborns.


Asunto(s)
Inhibidores de Caspasas , Inhibidores de Cisteína Proteinasa/uso terapéutico , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Isquemia/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Oligopéptidos/uso terapéutico , Quinolinas/farmacología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Sitios de Unión , Caspasas/metabolismo , Inhibidores de Cisteína Proteinasa/química , Citocromos c/metabolismo , Modelos Animales de Enfermedad , Hipoxia-Isquemia Encefálica/patología , Isquemia/patología , Ratones , Fármacos Neuroprotectores/química , Oligopéptidos/química , Oligopéptidos/farmacología , Quinolinas/química , Ratas
16.
Neuroscience ; 173: 156-68, 2011 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-21073926

RESUMEN

Activity-dependent neuroprotective protein (ADNP) was shown to be essential for embryogenesis and brain development while NAP, an active motif of ADNP, is neuroprotective in a broad range of neurodegenerative disorders. In the present study, we examined the protective potential of ADNP/NAP in a mouse model of excitotoxic brain lesion mimicking brain damage associated with cerebral palsy. We demonstrated that NAP had a potent neuroprotective effect against ibotenate-induced excitotoxic damage in the cortical plate and the white matter of P5 mice, and moderate against brain lesions of P0 mice. In contrast, endogenous ADNP appears not to be involved in the response to excitotoxic challenge in the studied model. Our findings further show that NAP reduced the number of apoptotic neurons through activation of PI-3K/Akt pathway in the cortical plate or both PI-3K/Akt and MAPK/MEK1 kinases in the white matter. In addition, NAP prevented ibotenate-induced loss of pre-oligodendrocytes without affecting the number of astrocytes or activated microglia around the site of injection. These findings indicate that protective actions of NAP are mediated by triggering transduction pathways that are crucial for neuronal and oligodendroglial survival, thus, NAP might be a promising therapeutic agent for treating developing brain damage.


Asunto(s)
Lesiones Encefálicas/prevención & control , Proteínas de Homeodominio/farmacología , Proteínas del Tejido Nervioso/farmacología , Fármacos Neuroprotectores/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Western Blotting , Lesiones Encefálicas/inducido químicamente , Lesiones Encefálicas/metabolismo , Parálisis Cerebral/metabolismo , Parálisis Cerebral/patología , Modelos Animales de Enfermedad , Agonistas de Aminoácidos Excitadores/toxicidad , Femenino , Proteínas de Homeodominio/metabolismo , Ácido Iboténico/toxicidad , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Brain Behav Immun ; 24(5): 776-83, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19903519

RESUMEN

Although the role of microglial activation in neural injury remains controversial, there is increasing evidence for a detrimental effect in the immature brain, which may occur in response to release of neurotoxic substances including pro-inflammatory cytokines. However, the signaling mechanisms involved in microglial-induced neuronal cell death are unclear. Microglia isolated from the brains of wild-type (WT) or MyD88 knockout (KO) mice were exposed to PBS or the TLR4-ligand LPS (100 ng/mL) for 2, 6, 14, or 24 h, and the microglia-conditioned medium (MCM) collected. Detection of multiple inflammatory molecules in MCM was performed using a mouse 22-plex cytokine microbead array kit. Primary neuronal cultures were supplemented with the 14 or 24 h MCM, and the degree of neuronal apoptosis examined after exposure for 24 h. Results showed a rapid and sustained elevation in multiple inflammatory mediators in the MCM of WT microglia exposed to LPS, which was largely inhibited in MyD88 KO microglia. There was a significant increase in apoptotic death measured at 24 h in cultured neurons exposed to CM from either 14 or 24 h LPS-stimulated WT microglia (p<.05 vs. WT control). By contrast, there was no increase in apoptotic death in cultured neurons exposed to CM from 14 or 24 h LPS-stimulated MyD88 KO microglia (p=.15 vs. MyD88 KO control). These data suggest that MyD88-dependent activation of microglia by LPS causes release of factors directly toxic to neurons.


Asunto(s)
Citocinas/metabolismo , Microglía/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Neuronas/metabolismo , Análisis de Varianza , Animales , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Encéfalo/inmunología , Encéfalo/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Citocinas/inmunología , Lipopolisacáridos/farmacología , Ratones , Ratones Noqueados , Microglía/efectos de los fármacos , Microglía/inmunología , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Neuronas/efectos de los fármacos , Neuronas/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
20.
Arch Pediatr ; 16 Suppl 1: S56-63, 2009 Sep.
Artículo en Francés | MEDLINE | ID: mdl-19836669

RESUMEN

Review of recent publications about perinatal consequences of cocaine use during pregnancy points out that: - dramatic obstetrical, neonatal and developmental abnormalities, reported during 1980-90', are less frequent in recent cohort studies; - pregnant women who use cocaine or crack, also consume other psychoactive drugs (alcohol, tobacco, benzodiazepines, cannabis, opiates, ...) and have a very chaotic life-style; so, it is difficult to distinguish abnormalities caused by cocaine per se, even with numerous cohorts, control groups and multivariate analysis.


Asunto(s)
Trastornos Relacionados con Cocaína/epidemiología , Complicaciones del Embarazo/epidemiología , Efectos Tardíos de la Exposición Prenatal/epidemiología , Estudios de Cohortes , Cocaína Crack , Femenino , Humanos , Análisis Multivariante , Embarazo , Trastornos Relacionados con Sustancias/epidemiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...