Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Glia ; 68(3): 528-542, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31670865

RESUMEN

Mild-traumatic brain injury (mTBI) represents ~80% of all emergency room visits and increases the probability of developing long-term cognitive disorders in children. To date, molecular and cellular mechanisms underlying post-mTBI cognitive dysfunction are unknown. Astrogliosis has been shown to significantly alter astrocytes' properties following brain injury, potentially leading to significant brain dysfunction. However, such alterations have never been investigated in the context of juvenile mTBI (jmTBI). A closed-head injury model was used to study jmTBI on postnatal-day 17 mice. Astrogliosis was evaluated using glial fibrillary acidic protein (GFAP), vimentin, and nestin immunolabeling in somatosensory cortex (SSC), dentate gyrus (DG), amygdala (AMY), and infralimbic area (ILA) of prefrontal cortex in both hemispheres from 1 to 30 days postinjury (dpi). In vivo T2-weighted-imaging (T2WI) and diffusion tensor imaging (DTI) were performed at 7 and 30 dpi to examine tissue level structural alterations. Increased GFAP-labeling was observed up to 30 dpi in the ipsilateral SSC, the initial site of the impact. However, vimentin and nestin expression was not perturbed by jmTBI. The morphology of GFAP positive cells was significantly altered in the SSC, DG, AMY, and ILA up to 7 dpi that some correlated with magnetic resonance imaging changes. T2WI and DTI values were significantly altered at 30 dpi within these brain regions most prominently in regions distant from the impact site. Our data show that jmTBI triggers changes in astrocytic phenotype with a distinct spatiotemporal pattern. We speculate that the presence and time course of astrogliosis may contribute to pathophysiological processes and long-term structural alterations following jmTBI.


Asunto(s)
Astrocitos/metabolismo , Conmoción Encefálica/patología , Lesiones Encefálicas/patología , Traumatismos Cerrados de la Cabeza/patología , Animales , Encéfalo/patología , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis/patología , Imagen por Resonancia Magnética/métodos , Ratones
2.
J Neurosci ; 32(40): 13860-72, 2012 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-23035095

RESUMEN

Excitatory-inhibitory imbalance has been identified within specific brain microcircuits in models of Rett syndrome (RTT) and other autism spectrum disorders (ASDs). However, macrocircuit dysfunction across the RTT brain as a whole has not been defined. To approach this issue, we mapped expression of the activity-dependent, immediate-early gene product Fos in the brains of wild-type (Wt) and methyl-CpG-binding protein 2 (Mecp2)-null (Null) mice, a model of RTT, before and after the appearance of overt symptoms (3 and 6 weeks of age, respectively). At 6 weeks, Null mice exhibit significantly less Fos labeling than Wt in limbic cortices and subcortical structures, including key nodes in the default mode network. In contrast, Null mice exhibit significantly more Fos labeling than Wt in the hindbrain, most notably in cardiorespiratory regions of the nucleus tractus solitarius (nTS). Using nTS as a model, whole-cell recordings demonstrated that increased Fos expression in Nulls at 6 weeks of age is associated with synaptic hyperexcitability, including increased frequency of spontaneous and miniature EPSCs and increased amplitude of evoked EPSCs in Nulls. No such effect of genotype on Fos or synaptic function was seen at 3 weeks. In the mutant forebrain, reduced Fos expression, as well as abnormal sensorimotor function, were reversed by the NMDA receptor antagonist ketamine. In light of recent findings that the default mode network is hypoactive in autism, our data raise the possibility that hypofunction within this meta-circuit is a shared feature of RTT and other ASDs and is reversible.


Asunto(s)
Sistema Nervioso Autónomo/fisiopatología , Antagonistas de Aminoácidos Excitadores/farmacología , Ketamina/farmacología , Proteína 2 de Unión a Metil-CpG/fisiología , Red Nerviosa/fisiopatología , Prosencéfalo/fisiopatología , Núcleo Solitario/fisiopatología , Animales , Cerebelo/metabolismo , Cerebelo/fisiopatología , Modelos Animales de Enfermedad , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes fos , Humanos , Masculino , Proteína 2 de Unión a Metil-CpG/deficiencia , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Noqueados , Potenciales Postsinápticos Miniatura/efectos de los fármacos , Potenciales Postsinápticos Miniatura/fisiología , Red Nerviosa/efectos de los fármacos , Red Nerviosa/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Especificidad de Órganos , Técnicas de Placa-Clamp , Prosencéfalo/metabolismo , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Síndrome de Rett/genética , Síndrome de Rett/fisiopatología , Filtrado Sensorial/efectos de los fármacos , Filtrado Sensorial/fisiología , Núcleo Solitario/química , Núcleo Solitario/metabolismo , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología
3.
J Neurosci ; 32(5): 1803-10, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22302819

RESUMEN

Rett syndrome (RTT) results from loss-of-function mutations in the gene encoding the methyl-CpG-binding protein 2 (MeCP2) and is characterized by abnormal motor, respiratory and autonomic control, cognitive impairment, autistic-like behaviors and increased risk of seizures. RTT patients and Mecp2-null mice exhibit reduced expression of brain-derived neurotrophic factor (BDNF), which has been linked in mice to increased respiratory frequency, a hallmark of RTT. The present study was undertaken to test the hypotheses that BDNF deficits in Mecp2 mutants are associated with reduced activation of the BDNF receptor, TrkB, and that pharmacologic activation of TrkB would improve respiratory function. We characterized BDNF protein expression, TrkB activation and respiration in heterozygous female Mecp2 mutant mice (Het), a model that recapitulates the somatic mosaicism for mutant MECP2 found in typical RTT patients, and evaluated the ability of a small molecule TrkB agonist, LM22A-4, to ameliorate biochemical and functional abnormalities in these animals. We found that Het mice exhibit (1) reduced BDNF expression and TrkB activation in the medulla and pons and (2) breathing dysfunction, characterized by increased frequency due to periods of tachypnea, and increased apneas, as in RTT patients. Treatment of Het mice with LM22A-4 for 4 weeks rescued wild-type levels of TrkB phosphorylation in the medulla and pons and restored wild-type breathing frequency. These data provide new insight into the role of BDNF signaling deficits in the pathophysiology of RTT and highlight TrkB as a possible therapeutic target in this disease.


Asunto(s)
Benzamidas/metabolismo , Modelos Animales de Enfermedad , Agonismo Parcial de Drogas , Receptor trkB/agonistas , Receptor trkB/metabolismo , Mecánica Respiratoria/efectos de los fármacos , Síndrome de Rett/metabolismo , Animales , Femenino , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fosforilación/efectos de los fármacos , Fosforilación/genética , Mecánica Respiratoria/genética , Síndrome de Rett/tratamiento farmacológico , Síndrome de Rett/genética
4.
Psychiatry Res ; 316: 114755, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35963061

RESUMEN

Face masks have proven to be key to slowing down the SARS-Cov2 virus spread in the COVID-19 pandemic context. However, wearing face masks is not devoid of "side-effects", at both the physical and psychosocial levels. In particular, masks hinder emotion reading from facial expressions as they hide a significant part of the face. This disturbs both holistic and featural processing of facial expressions and, therefore, impairs emotion recognition, and influences many aspects of human social behavior. Communication in general is disrupted by face masks, as they modify the wearer's voice and prevent the audience from using lip reading or other non-verbal cues for speech comprehension. Individuals suffering from psychiatric conditions with impairment of communication, are at higher risk of distress because masks increase their difficulties to read emotions from faces. The identification and acknowledgement of these "side-effects" on communication are necessary because they warrant further work on adaptive solutions that will help foster the use of face masks by the greatest number.


Asunto(s)
COVID-19 , Pandemias , Emociones , Humanos , Pandemias/prevención & control , ARN Viral , SARS-CoV-2
5.
Sci Rep ; 12(1): 10248, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35715469

RESUMEN

Patients with drug-resistant epilepsy can experience respiratory alterations, notably during seizures. The mechanisms underlying long-term alterations in respiratory function remain unclear. As the brainstem 5-HT system is a prominent modulator of respiratory function, this study aimed at determining whether epilepsy is associated with alterations in both the respiratory function and brainstem serotonin (5-HT) system in rats. Epilepsy was triggered by pilocarpine-induced status epilepticus in rats. Our results showed that 30-50% of epileptic (EPI) rats exhibited a sharp decrease in oxygen consumption (SDOC), low metabolic rate of oxygen, and slow regular ventilation (EPI/SDOC + rats). These alterations were detected only in rats with chronic epilepsy, independent of behavioral seizures, were persistent over time, and not associated with death. In these rats, 5-HT fiber density in the nucleus tractus solitarius was lower than that in the control and EPI/SDOC- rats. Both EPI/SDOC + rats and DBA/2 mice that present with audiogenic-induced seizure followed by fatal respiratory arrest-a model of sudden and expected death in epilepsy-had increased transcript levels of tryptophan hydroxylase 2 and 5-HT presynaptic transporter. Thus, our data support that 5-HT alterations are associated with chronic and acute epilepsy-related respiratory dysfunction.


Asunto(s)
Epilepsia Refleja , Trastornos Respiratorios , Animales , Tronco Encefálico/metabolismo , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos DBA , Ratas , Roedores/metabolismo , Convulsiones , Serotonina/metabolismo
6.
J Neurosci ; 30(15): 5303-10, 2010 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-20392952

RESUMEN

Postnatal deficits in brain-derived neurotrophic factor (BDNF) are thought to contribute to pathogenesis of Rett syndrome (RTT), a progressive neurodevelopmental disorder caused by mutations in the gene encoding methyl-CpG-binding protein 2 (MeCP2). In Mecp2-null mice, a model of RTT, BDNF deficits are most pronounced in structures important for autonomic and respiratory control, functions that are severely affected in RTT patients. However, relatively little is known about how these deficits affect neuronal function or how they may be linked to specific RTT endophenotypes. To approach these issues, we analyzed synaptic function in the brainstem nucleus tractus solitarius (nTS), the principal site for integration of primary visceral afferent inputs to central autonomic pathways and a region in which we found markedly reduced levels of BDNF in Mecp2 mutants. Our results demonstrate that the amplitude of spontaneous miniature and evoked EPSCs in nTS neurons is significantly increased in Mecp2-null mice and, accordingly, that mutant cells are more likely than wild- type cells to fire action potentials in response to primary afferent stimulation. These changes occur without any increase in intrinsic neuronal excitability and are unaffected by blockade of inhibitory GABA currents. However, this synaptopathy is associated with decreased BDNF availability in the primary afferent pathway and can be rescued by application of exogenous BDNF. On the basis of these findings, we hypothesize that altered sensory gating in nTS contributes to cardiorespiratory instability in RTT and that nTS is a site at which restoration of normal BDNF signaling could help reestablish normal homeostatic controls.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteína 2 de Unión a Metil-CpG/metabolismo , Neuronas/fisiología , Núcleo Solitario/fisiología , Sinapsis/fisiología , Transmisión Sináptica/fisiología , Potenciales de Acción/fisiología , Animales , Modelos Animales de Enfermedad , Potenciales Evocados/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Técnicas In Vitro , Bulbo Raquídeo/fisiología , Proteína 2 de Unión a Metil-CpG/deficiencia , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Noqueados , Inhibición Neural/fisiología , Neuronas Aferentes/fisiología , Síndrome de Rett , Vías Visuales/fisiología , Ácido gamma-Aminobutírico/metabolismo
7.
Front Bioeng Biotechnol ; 9: 786881, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35155403

RESUMEN

Exposure to blast is one of the major causes of death and disability in recent military conflicts. Therefore, it is crucial to evaluate the protective capability of the ballistic-proof equipment worn by soldiers against the effects of blast overpressure (i.e., primary blast injuries). A focus will be made on thoracic protective equipment (TPE). An anthropomorphic mannequin, called BOPMAN, and anesthetized swine both wearing soft, hard or no ballistic protection, were subjected to an open-field high-intensity blast. For swine, thoracic wall motion (acceleration and velocity) was recorded during blast exposure and severity of lung injury was evaluated postmortem. Different data were collected from BOPMAN thoracic responses, including reflected and internal pressure, as well as the force at the rear face of the instrumented part. The severity of blast-induced lung injuries (contusion extent, Axelsson Severity Scale) and the thoracic wall motion were decreased in animals protected with thoracic ceramic hard plates as compared to those wearing soft or no protection. There was a clear trend towards greater lung injury in animals protected with the soft body armor used, even when compared to unprotected animals. In line with these experimental data, the measured force as well as the force impulse measured using BOPMAN were also decreased with a ceramic hard plate protection and increased when a soft ballistic pack was used compared to no protection. Comparison of data collected on BOPMAN and swine equipped with the same protection level revealed that those two force parameters were well correlated with the level of blast-induced lung injury (force, R2 = 0.74 and force impulse, R2 = 0.77, p < 0.05). Taken together, our results suggest that the force and the force impulse data from BOPMAN may help estimate the efficiency of existing TPE regarding lung protection under blast exposure and may represent an important tool for development of future TPE.

8.
Neuroscience ; 466: 205-221, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33895341

RESUMEN

Insulin-like growth factors (IGF) are potent neurotrophic and neurorepair factors that were recently proposed as biomarkers of traumatic brain injury (TBI) and associated psychiatric comorbidities, in particular post-traumatic stress disorder (PSTD). We tested the hypothesis that the IGF system is differentially deregulated in the acute and early chronic stages of TBI, and under acute stress. Plasma and brain IGF1 and IGF2 levels were evaluated in mice 3 weeks and 3 days after a controlled cortical impact (CCI)-induced mild-to-moderate TBI. The effects of conditioned fear on IGF levels and its interaction with TBI (TBI followed, 3 weeks later, by fear-inducing procedures) were also evaluated. In the plasma, IGF1 decreased 3 weeks post-TBI only (-9%), whereas IGF2 remained unaffected. In the brain, IGF1 increased only in the cortex and hippocampus at 3 weeks post-TBI (up to +650%). At 3 days, surpringly, this increase was more diffuse and more important in sham (craniotomized) animals. Additionally, IGF2 immunostaining in brain ventricles was reorganized in TBI animals at both post-TBI stages. Conditioned fear exposure did not influence the effects of early chronic TBI on plasma IGF1 levels, but reduced plasma IGF2 (-6%) levels. It also dampened the effects of TBI on brain IGF systems, but brain IGF1 level and IGF2 tissue distribution remained statistically different from controls under these conditions. In co-exposed animals, DNA methylation increased at the hippocampal Igf1 gene promoter. These results show that blood IGF1 and IGF2 are most reduced in the early chronic phase of TBI and after exposure to a stressful event, and that the brain IGF system is up-regulated after TBI, and more so in the acute phase.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Animales , Biomarcadores , Encéfalo/metabolismo , Miedo , Hipocampo/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ratones
9.
J Neurosci ; 29(39): 12187-95, 2009 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-19793977

RESUMEN

Rett syndrome (RTT) is a neurodevelopmental disorder caused by loss-of-function mutations in the Methyl-CpG-binding protein-2 (MECP2) gene and is characterized by derangements in cognition, behavior, motor control, respiration and autonomic homeostasis, as well as seizures. Deficits in norepinephrine (NE) are thought to contribute to RTT pathogenesis, but little is known about how MeCP2 regulates function of noradrenergic neurons. We therefore characterized morphological, electrical, and neurochemical properties of neurons in the locus ceruleus (LC), the major source of noradrenergic innervation to the central neuraxis, in Mecp2 mutant mice. We found that MeCP2 null LC neurons are electrically hyperexcitable, smaller in size, and express less of the NE-synthesizing enzyme tyrosine hydroxylase (TH) compared with wild-type neurons. Increased excitability of mutant neurons is associated with reductions in passive membrane conductance and the amplitude of the slow afterhyperpolarization. Studies in Mecp2 heterozygotes, which are mosaic for the null allele, demonstrated that electrical hyperexcitability and reduced neuronal size are cell-autonomous consequences of MeCP2 loss, whereas reduced TH expression appears to reflect both cell-autonomous and non-autonomous influences. Finally, we found reduced levels of TH and norepinephrine in cingulate cortex, a forebrain target of the LC. Thus, genetic loss of MeCP2 results in a somewhat paradoxical LC neuron phenotype, characterized by both electrical hyperexcitability and reduced indices of noradrenergic function. Given the importance of the LC in modulating activity in brainstem and forebrain networks, we hypothesize that dysregulation of LC function in the absence of MeCP2 plays a key role in the pathophysiology of RTT.


Asunto(s)
Modelos Animales de Enfermedad , Locus Coeruleus/fisiopatología , Síndrome de Rett/fisiopatología , Animales , Femenino , Masculino , Potenciales de la Membrana/genética , Proteína 2 de Unión a Metil-CpG/deficiencia , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Síndrome de Rett/genética
10.
Brain Behav Immun Health ; 5: 100081, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32427134

RESUMEN

This review intends to provide an overview of the current knowledge on neurologic sequelae of COVID-19 and their possible etiology, and, based on available data, proposes possible improvements in current medical care procedures. We conducted a thorough review of the scientific literature on neurologic manifestations of COVID-19, the neuroinvasive propensity of known coronaviruses (CoV) and their possible effects on brain structural and functional integrity. It appears that around one third of COVID-19 patients admitted to intensive care units (ICU) for respiratory difficulties exhibit neurologic symptoms. This may be due to progressive brain damage and dysfunction triggered by severe hypoxia and hypoxemia, heightened inflammation and SARS-CoV-2 dissemination into brain parenchyma, as suggested by current reports and analyses of previous CoV outbreaks. Viral invasion of the brain may particularly target and alter brainstem and thalamic functions and, consequently, result in sensorimotor dysfunctions and psychiatric disorders. Moreover, data collected from other structurally homologous CoV suggest that SARS-CoV-2 infection may lead to brain cell degeneration and demyelination similar to multiple sclerosis (MS). Hence, current evidence warrants further evaluation and long-term follow-up of possible neurologic sequelae in COVID-19 patients. It may be particularly relevant to evaluate brainstem integrity in recovered patients, as it is suspected that this cerebral area may particularly be dysfunctional following SARS-CoV-2 infection. Because CoV infection can potentially lead to chronic neuroinflammation and progressive demyelination, neuroimaging features and signs of MS may also be evaluated in the long term in recovered COVID-19 patients.

11.
Soins ; 65(849): 59-62, 2020 Oct.
Artículo en Francés | MEDLINE | ID: mdl-33357621

RESUMEN

Our social environment shapes our eating habits, notably our consumption of fruit and vegetables rich in micronutrients (vitamins and trace elements), essential for regulating the immune system. Ensuring a balanced intake of micronutrients could prove to be particularly beneficial for patients with severe forms of COVID-19 suffering from critical immune dysregulation.


Asunto(s)
COVID-19/inmunología , COVID-19/terapia , Sistema Inmunológico/fisiología , Micronutrientes/administración & dosificación , Vitaminas/administración & dosificación , Humanos
12.
Eur J Neurosci ; 30(4): 602-10, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19674087

RESUMEN

Rett syndrome (RTT) is a progressive developmental disorder resulting from loss-of-function mutations in the gene encoding methyl-CpG-binding protein 2 (MeCP2), a transcription regulatory protein. The RTT phenotype is complex and includes severe cardiorespiratory abnormalities, dysautonomia and behavioral symptoms of elevated stress. These findings have been attributed to an apparent hyperactivity of the sympathetic nervous system due to defects in brainstem development; however, the possibility that the peripheral sympathoadrenal axis itself is abnormal has not been explored. The present study demonstrates that the adrenal medulla and sympathetic ganglia of Mecp2 null mice exhibit markedly reduced catecholamine content compared with wild-type controls. Despite this, null animals exhibit significantly higher plasma epinephrine levels, suggesting enhanced secretory granule function in adrenal chromaffin cells. Indeed, we find that Mecp2 null chromaffin cells exhibit a cell autonomous hypersecretory phenotype characterized by significant increases in the speed and size of individual secretory granule fusion events in response to electrical stimulation. These findings appear to indicate accelerated formation and enhanced dilation of the secretory granule fusion pore, resulting in elevated catecholamine release. Our data therefore highlight abnormal catecholamine function in the sympathoadrenal axis as a potential source of autonomic dysfunction in RTT. These findings may help to explain the apparent 'overactivity' of the sympathetic nervous system reported in patients with RTT.


Asunto(s)
Glándulas Suprarrenales/fisiopatología , Gránulos Cromafines/metabolismo , Modelos Animales de Enfermedad , Síndrome de Rett/fisiopatología , Sistema Nervioso Simpático/fisiopatología , Glándulas Suprarrenales/química , Glándulas Suprarrenales/patología , Animales , Cromatografía Líquida de Alta Presión , Dopamina/análisis , Electrofisiología , Epinefrina/análisis , Femenino , Inmunohistoquímica , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Ratones , Ratones Noqueados , Mutación , Norepinefrina/análisis , Ganglio Cervical Superior/química , Sistema Nervioso Simpático/patología
13.
J Neurosci ; 27(40): 10912-7, 2007 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-17913925

RESUMEN

Rett syndrome (RTT) is caused by loss-of-function mutations in the gene encoding methyl-CpG-binding protein 2 (MeCP2). Although MeCP2 is thought to act as a transcriptional repressor of brain-derived neurotrophic factor (BDNF), Mecp2 null mice, which develop an RTT-like phenotype, exhibit progressive deficits in BDNF expression. These deficits are particularly significant in the brainstem and nodose cranial sensory ganglia (NGs), structures critical for cardiorespiratory homeostasis, and may be linked to the severe respiratory abnormalities characteristic of RTT. Therefore, the present study used Mecp2 null mice to further define the role of MeCP2 in regulation of BDNF expression and neural function, focusing on NG neurons and respiratory control. We find that mutant neurons express significantly lower levels of BDNF than wild-type cells in vitro, as in vivo, under both depolarizing and nondepolarizing conditions. However, BDNF levels in mutant NG cells can be increased by chronic depolarization in vitro or by treatment of Mecp2 null mice with CX546, an ampakine drug that facilitates activation of glutamatergic AMPA receptors. Ampakine-treated Mecp2 null mice also exhibit marked functional improvement, characterized by restoration of normal breathing frequency and minute volume. These data demonstrate that BDNF expression remains plastic in Mecp2 null mice and raise the possibility that ampakine compounds could be of therapeutic value in the treatment of RTT.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Depsipéptidos/efectos de los fármacos , Dioxoles/uso terapéutico , Regulación de la Expresión Génica/efectos de los fármacos , Piperidinas/uso terapéutico , Síndrome de Rett/tratamiento farmacológico , Síndrome de Rett/fisiopatología , Análisis de Varianza , Anestésicos Locales/farmacología , Animales , Animales Recién Nacidos , Factor Neurotrófico Derivado del Encéfalo/genética , Células Cultivadas , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ganglio Nudoso/citología , Pletismografía/métodos , ARN Mensajero/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Tetrodotoxina/farmacología
14.
Respir Physiol Neurobiol ; 164(1-2): 55-63, 2008 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-18534925

RESUMEN

Severely arrhythmic breathing is a hallmark of Rett syndrome (RTT) and profoundly affects quality of life for patients and their families. The last decade has seen the identification of the disease-causing gene, methyl-CpG-binding protein 2 (Mecp2) and the development of mouse models that phenocopy many aspects of the human syndrome, including breathing dysfunction. Recent studies have begun to characterize the breathing phenotype of Mecp2 mutant mice and to define underlying electrophysiological and neurochemical deficits. The picture that is emerging is one of defects in synaptic transmission throughout the brainstem respiratory network associated with abnormal expression in several neurochemical signaling systems, including brain-derived neurotrophic factor (BDNF), biogenic amines and gamma-amino-butyric acid (GABA). Based on such findings, potential therapeutic strategies aimed at improving breathing by targeting deficits in neurochemical signaling are being explored. This review details our current understanding of respiratory dysfunction and underlying mechanisms in RTT with a particular focus on insights gained from mouse models.


Asunto(s)
Epigénesis Genética , Trastornos Respiratorios , Síndrome de Rett/complicaciones , Síndrome de Rett/genética , Animales , Dioxoles/uso terapéutico , Modelos Animales de Enfermedad , Humanos , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Mutación/genética , Piperidinas/uso terapéutico , Trastornos Respiratorios/tratamiento farmacológico , Trastornos Respiratorios/etiología , Trastornos Respiratorios/genética , Trastornos Respiratorios/patología , Centro Respiratorio/metabolismo , Centro Respiratorio/fisiopatología , Sistema Respiratorio/metabolismo , Sistema Respiratorio/fisiopatología
15.
J Neurosci ; 26(42): 10911-5, 2006 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-17050729

RESUMEN

Disruptions in brain-derived neurotrophic factor (BDNF) expression are proposed to contribute to the molecular pathogenesis of Rett syndrome (RTT), a severe neurological disorder caused by loss-of-function mutations in methyl-CpG-binding protein-2 (MeCP2). Although MeCP2 is a transcriptional regulator of BDNF, it is unknown how MeCP2 mutations affect transsynaptic BDNF signaling. Our findings demonstrate an early, abnormal neurosecretory phenotype in MeCP2-deficient neurons characterized by significant increases in the percentage of cellular BDNF content available for release. However, loss of MeCP2 also results in deficits in total cell BDNF content that are developmentally regulated in a cell-type-specific manner. Thus, the net effect of MeCP2 loss on absolute BDNF secretion changes with age and is determined by both the amount of BDNF available for release and progressive declines in total cellular BDNF. We propose, therefore, that loss of MeCP2 function disrupts transsynaptic BDNF signaling by perturbing the normal balance between BDNF protein levels and secretion. However, mutant neurons are capable of secreting wild-type levels of BDNF in response to high-frequency electrical stimulation. In addition, we found elevated exocytic function in Mecp2(-/y) adrenal chromaffin cells, indicating that the Mecp2 null mutation is associated with alterations of neurosecretion that are not restricted to BDNF. These findings are the first examples of abnormal neuropeptide and catecholamine secretion in a mouse model of RTT.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Factor Neurotrófico Derivado del Encéfalo/genética , Regulación de la Expresión Génica/genética , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Sinapsis/metabolismo , Glándulas Suprarrenales/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/fisiología , Células Cultivadas , Proteína 2 de Unión a Metil-CpG/deficiencia , Ratones , Ratones Noqueados , Transducción de Señal/genética , Sinapsis/genética
16.
J Neurotrauma ; 34(8): 1645-1657, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-27901414

RESUMEN

Traumatic brain injury (TBI) is common in both military and civilian populations, and often results in neurobehavioral sequelae that impair quality of life in both patients and their families. Although individuals who are chronically exposed to stress are more likely to experience TBI, it is still unknown whether pre-injury stress influences the outcome after TBI. The present study tested whether behavioral and cognitive long-term outcome after TBI in rats is affected by prior exposure to an innate stress stimulus. Young adult male Sprague-Dawley rats were exposed to the predator odor 2,5-dihydro-2,4,5-trimethylthiazoline (TMT) or to water (WAT); exposure was repeated eight times at irregular intervals over a 2-week period. Rats were subsequently subjected to either mild-to-moderate bilateral brain injury (lateral fluid percussion [LFP]) or sham surgery (Sham). Four experimental groups were studied: Sham-WAT, Sham-TMT, LFP-WAT and LFP-TMT. Compared with Sham-WAT rats, LFP-WAT rats exhibited transient locomotor hyperactivity without signs of anxiety, minor spatial learning acquisition and hippocampal long-term potentiation deficits, and lower baseline activity of the hypothalamic-pituitary-adrenal axis with slightly stronger reactivity to restraint stress. Exposure to TMT had only negligible effects on Sham rats, whereas it exacerbated all deficits in LFP rats except for locomotor hyperactivity. Early brain inflammatory response (8 h post-trauma) was aggravated in rats pre-exposed to TMT, suggesting that increased brain inflammation may sustain functional deficits in these rats. Hence, these data suggest that pre-exposure to stressful conditions can aggravate long-term deficits induced by TBI, leading to severe stress response deficits, possibly due to dysregulated inflammatory response.


Asunto(s)
Conducta Animal , Lesiones Traumáticas del Encéfalo/complicaciones , Disfunción Cognitiva/etiología , Inflamación/etiología , Estrés Psicológico/complicaciones , Animales , Conducta Animal/fisiología , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/fisiopatología , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/fisiopatología , Modelos Animales de Enfermedad , Inflamación/metabolismo , Inflamación/fisiopatología , Masculino , Aprendizaje por Laberinto , Ratas , Ratas Sprague-Dawley , Estrés Psicológico/metabolismo , Estrés Psicológico/fisiopatología
17.
CNS Neurosci Ther ; 22(3): 230-7, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26775713

RESUMEN

AIMS: Transgenic TGR(ASrAOGEN)680 (TGR) rats with specific downregulation of glial angiotensinogen (AOGEN) synthesis develop cardiovascular deficits, anxiety, altered response to stress, and depression. Here, we evaluated whether these deficits are associated with alteration of the integrity of the noradrenergic system originating from locus coeruleus (LC) neurons. METHODS: Adult TGR rats were compared to control Sprague Dawley rats in terms of the following: tissue levels of transcripts encoding noradrenergic markers, tissue tyrosine hydroxylase (TH) protein level, in vivo TH activity, density of TH-containing fibers, behavioral response to novelty, locomotor activity, and polysomnography. RESULTS: TH expression was increased in the LC of TGR rats compared to controls. In LC terminal fields, there was an increase in density of TH-containing fibers in TGR rats that was associated with an elevation of in vivo TH activity. TGR rats also displayed locomotor hyperactivity in response to novelty. Moreover, polysomnographic studies indicated that daily paradoxical sleep duration was increased in TGR rats and that the paradoxical sleep rebound triggered by total sleep deprivation was blunted in these rats. CONCLUSIONS: Altogether, these results suggest that disruption of astroglial AOGEN synthesis leads to cardiovascular, cognitive, behavioral, and sleep disorders that might be partly due to LC dysfunction.


Asunto(s)
Angiotensinógeno/genética , Angiotensinógeno/metabolismo , Locus Coeruleus/metabolismo , Locus Coeruleus/patología , Animales , Temperatura Corporal , Estudios de Casos y Controles , Cerebelo/patología , Dopamina beta-Hidroxilasa/genética , Dopamina beta-Hidroxilasa/metabolismo , Regulación de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Levodopa/metabolismo , Masculino , Actividad Motora/genética , Neuronas/metabolismo , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/genética , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Sueño/genética , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo , Vigilia/genética
18.
Brain Res ; 1030(1): 1-10, 2004 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-15567332

RESUMEN

Our study was devoted to determine in freely moving rats whether the increase in tissue concentration of tyrosine hydroxylase (TH) elicited by a single administration of RU 24722 could modify the catecholaminergic reactivity of neuronal processes present in the rostrolateral part of the pericerulean area (r-lPCA) in response to tail pinch. Catecholaminergic activity was monitored by measuring in vivo the concentration of dopamine metabolite 3,4-dihydroxyphenylacetic acid (DOPAC) using microdialysis coupled to HPLC detection. In this study, the microdialysis probe was implanted at a sufficient distance from the lateral border of rostral nucleus locus ceruleus (LC) to avoid a large contribution of the noradrenergic cell bodies in the measurements performed. We first evidenced that DOPAC measured in the r-lPCA indicated the functional state of catecholaminergic metabolism in neuronal processes (dendrites and fibers) laying in this region. We also showed that the enhancement of TH protein concentration in the r-lPCA following RU 24722 treatment supported an increased in vivo catecholaminergic metabolism in this region. Furthermore, catecholaminergic metabolism response to tail pinch was potentiated in animals with greater TH tissue concentration. Thus, our study reveals that the modulation of both TH concentration and catecholaminergic metabolism in the r-lPCA may be critical in the functioning of cells and neuronal elements present in this region, notably in adaptive responses to noxious stimuli.


Asunto(s)
Ácido 3,4-Dihidroxifenilacético/metabolismo , Locus Coeruleus/efectos de los fármacos , Locus Coeruleus/metabolismo , Nociceptores/metabolismo , Vincamina/análogos & derivados , Vincamina/farmacología , Animales , Ácido Hidroxiindolacético/metabolismo , Masculino , Microdiálisis , Estimulación Física , Ratas , Ratas Sprague-Dawley , Cola (estructura animal) , Tirosina 3-Monooxigenasa/metabolismo
19.
Compr Physiol ; 3(3): 1125-34, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23897682

RESUMEN

Neurotrophic factors (NTFs) are a heterogeneous group of extracellular signaling molecules that play critical roles in the development, maintenance, modulation and plasticity of the central and peripheral nervous systems. A subset of these factors, including members of three multigene families-the neurotrophins, neuropoetic cytokines and the glial cell line-derived neurotrophic factor ligands-are particularly important for development and regulation of neurons involved in respiratory control. Here, we review the functional biology of these NTFs and their receptors, as well as their roles in regulating survival, maturation, synaptic strength and plasticity in respiratory control pathways. In addition, we highlight recent progress in identifying the role of abnormal NTF signaling in the molecular pathogenesis of respiratory dysfunction in Rett syndrome and in the development of potential new NTF-targeted therapeutic strategies.


Asunto(s)
Factores de Crecimiento Nervioso/fisiología , Mecánica Respiratoria/fisiología , Adaptación Fisiológica/fisiología , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/fisiología , Humanos , Ratones , Neuronas Aferentes/fisiología , Receptores de Factor de Crecimiento Nervioso/fisiología , Trastornos Respiratorios/genética , Trastornos Respiratorios/fisiopatología , Sistema Respiratorio/inervación , Sistema Respiratorio/metabolismo , Síndrome de Rett/fisiopatología , Transducción de Señal/fisiología , Sinapsis/fisiología
20.
Front Neurol ; 4: 146, 2013 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-24130548

RESUMEN

Ischemia and metabolic crisis are frequent post-traumatic secondary brain insults that negatively influence outcome. Clinicians commonly mix up these two types of insults, mainly because high lactate/pyruvate ratio (LPR) is the common marker for both ischemia and metabolic crisis. However, LPR elevations during ischemia and metabolic crisis reflect two different energetic imbalances: ischemia (Type 1 LPR elevations with low oxygenation) is characterized by a drastic deprivation of energetic substrates, whereas metabolic crisis (Type 2 LPR elevations with normal or high oxygenation) is associated with profound mitochondrial dysfunction but normal supply of energetic substrates. The discrimination between ischemia and metabolic crisis is crucial because conventional recommendations against ischemia may be detrimental for patients with metabolic crisis. Multimodal monitoring, including microdialysis and brain tissue oxygen monitoring, allows such discrimination, but these techniques are not easily accessible to all head-injured patients. Thus, a new "gold standard" and adapted medical education are required to optimize the management of patients with metabolic crisis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA