Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Neurooncol Adv ; 5(1): vdad108, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37781088

RESUMEN

Background: Hypersomnolence is a common and disruptive side effect of cranial radiotherapy and is associated with fatigue and disturbances in mood and cognition in primary brain tumor (PBT) patients. The biological underpinnings of this effect are not understood. Our laboratory has previously found that the presence of a single nucleotide polymorphism (rs934945, G-E mutation) in the PERIOD2 (PER2) clock gene was associated with a decreased likelihood of fatigue in PBT patients. Here, we aim to understand the effects of PER2 polymorphism on radiation susceptibility within a murine model of cranial-irradiation-induced hypersomnolence (C-RIH). Methods: Male and female transgenic mice were generated using CRISPR-Cas9, replacing the endogenous mouse PER2:CRY1 binding domain with its human isoform with (hE1244 KI) or without the SNP rs934945 (hG1244 KI). Activity and sleep were monitored continuously 10 days before and after cranial irradiation (whole brain, 15Gy, single fraction). Behavioral assessments measuring anxiety, depression, and working memory were used to assess mood and cognitive changes 2 months postradiation. Results: During their active phase, hE1244 knock-ins (KIs) had less radiation-induced suppression of activity relative to hG1244 KIs and female hE1244 KIs saw a reduction of hypersomnolence over 10 days. hE1244 KIs displayed less anxiety behavior and were more ambulatory within all behavioral tests. Conclusions: The PER2 rs934945 polymorphism had long-lasting behavioral effects associated with radiation toxicity, particularly in sleep in females and the activity of all animals. Our findings shed light on biological mechanisms underlying C-RIH.

2.
J Pathol ; 260(3): 276-288, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37185821

RESUMEN

The effect of cytokines on non-traditional immunological targets under conditions of chronic inflammation is an ongoing subject of study. Fatigue is a symptom often associated with autoimmune diseases. Chronic inflammatory response and activated cell-mediated immunity are associated with cardiovascular myopathies which can be driven by muscle weakness and fatigue. Thus, we hypothesize that immune dysfunction-driven changes in myocyte mitochondria may play a critical role in fatigue-related pathogenesis. We show that persistent low-level expression of IFN-γ in designated IFN-γ AU-Rich Element deletion mice (ARE mice) under androgen exposure resulted in mitochondrial and metabolic deficiencies in myocytes from male or castrated ARE mice. Most notably, echocardiography unveiled that low ejection fraction in the left ventricle post-stress correlated with mitochondrial deficiencies, explaining how heart function decreases under stress. We report that inefficiencies and structural changes in mitochondria, with changes to expression of mitochondrial genes, are linked to male-biased fatigue and acute cardiomyopathy under stress. Our work highlights how male androgen hormone backgrounds and active autoimmunity reduce mitochondrial function and the ability to cope with stress and how pharmacological blockade of stress signal protects heart function. These studies provide new insight into the diverse actions of IFN-γ in fatigue, energy metabolism, and autoimmunity. © 2023 The Pathological Society of Great Britain and Ireland. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.


Asunto(s)
Andrógenos , Interferón gamma , Animales , Masculino , Ratones , Andrógenos/metabolismo , Citocinas/metabolismo , Inflamación/metabolismo , Mitocondrias/metabolismo , Células Musculares/metabolismo
3.
Front Neurosci ; 16: 847572, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35321093

RESUMEN

Brain-derived neurotrophic factor (BDNF) belongs to the neurotrophin family of secreted growth factors and binds with high affinity to the TrkB tyrosine kinase receptors. BDNF is a critical player in the development of the central (CNS) and peripheral (PNS) nervous system of vertebrates and its strong pro-survival function on neurons has attracted great interest as a potential therapeutic target for the management of neurodegenerative disorders such as Amyotrophic Lateral Sclerosis (ALS), Huntington, Parkinson's and Alzheimer's disease. The TrkB gene, in addition to the full-length receptor, encodes a number of isoforms, including some lacking the catalytic tyrosine kinase domain. Importantly, one of these truncated isoforms, namely TrkB.T1, is the most widely expressed TrkB receptor in the adult suggesting an important role in the regulation of BDNF signaling. Although some progress has been made, the mechanism of TrkB.T1 function is still largely unknown. Here we critically review the current knowledge on TrkB.T1 distribution and functions that may be helpful to our understanding of how it regulates and participates in BDNF signaling in normal physiological and pathological conditions.

4.
Transl Psychiatry ; 12(1): 111, 2022 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-35301275

RESUMEN

Recent studies have suggested that the use of cognitive enhancers as adjuncts to exposure-based therapy in individuals suffering from post-traumatic stress disorder (PTSD) may be beneficial. Brain cholinergic signaling through basal forebrain projections to the hippocampus is an established pathway mediating fear response and cognitive flexibility. Here we employed a genetic strategy to enhance cholinergic activity through increased signaling of the NGF receptor TrkA. This strategy leads to increased levels of the marker of cholinergic activation, acetylcholine synthesizing enzyme choline acetyltransferase, in forebrain cholinergic regions and their projection areas such as the hippocampus. Mice with increased cholinergic activity do not display any neurobehavioral abnormalities except a selective attenuation of fear response and lower fear expression in extinction trials. Reduction in fear response is rescued by the GABA antagonist picrotoxin in mutant mice, and, in wild-type mice, is mimicked by the GABA agonist midazolam suggesting that GABA can modulate cholinergic functions on fear circuitries. Importantly, mutant mice also show a reduction in fear processing under stress conditions in a single prolonged stress (SPS) model of PTSD-like behavior, and augmentation of cholinergic signaling by the drug donepezil in wild-type mice promotes extinction learning in a similar SPS model of PTSD-like behavior. Donepezil is already in clinical use for the treatment of dementia suggesting a new translational application of this drug for improving exposure-based psychotherapy in PTSD patients.


Asunto(s)
Prosencéfalo Basal , Trastornos por Estrés Postraumático , Animales , Prosencéfalo Basal/metabolismo , Colinérgicos/uso terapéutico , Extinción Psicológica/fisiología , Miedo/fisiología , Humanos , Ratones , Trastornos por Estrés Postraumático/psicología
5.
Exp Neurol ; 337: 113576, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33359475

RESUMEN

The pathophysiology of Amyotrophic Lateral Sclerosis (ALS), a disease caused by the gradual degeneration of motoneurons, is still largely unknown. Insufficient neurotrophic support has been cited as one of the causes of motoneuron cell death. Neurotrophic factors such as BDNF have been evaluated in ALS human clinical trials, but yielded disappointing results attributed to the poor pharmacokinetics and pharmacodynamics of BDNF. In the inherited ALS G93A SOD1 animal model, deletion of the BDNF receptor TrkB.T1 delays spinal cord motoneuron cell death and muscle weakness through an unknown cellular mechanism. Here we show that TrkB.T1 is expressed ubiquitously in the spinal cord and its deletion does not change the SOD1 mutant spinal cord inflammatory state suggesting that TrkB.T1 does not influence microglia or astrocyte activation. Although TrkB.T1 knockout in astrocytes preserves muscle strength and co-ordination at early stages of disease, its specific conditional deletion in motoneurons or astrocytes does not delay motoneuron cell death during the early stage of the disease. These data suggest that TrkB.T1 may limit the neuroprotective BDNF signaling to motoneurons via a non-cell autonomous mechanism providing new understanding into the reasons for past clinical failures and insights into the design of future clinical trials employing TrkB agonists in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Glicoproteínas de Membrana/genética , Proteínas Tirosina Quinasas/genética , Receptor trkB/genética , Esclerosis Amiotrófica Lateral/patología , Esclerosis Amiotrófica Lateral/psicología , Animales , Señalización del Calcio , Eliminación de Gen , Interleucina-1beta/metabolismo , Activación de Macrófagos , Glicoproteínas de Membrana/agonistas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/patología , Neuronas Motoras/patología , Desempeño Psicomotor , Médula Espinal/metabolismo , Médula Espinal/patología , Superóxido Dismutasa-1/genética , Factor de Necrosis Tumoral alfa/metabolismo
6.
Nat Commun ; 11(1): 1950, 2020 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-32327658

RESUMEN

BDNF signaling in hypothalamic circuitries regulates mammalian food intake. However, whether BDNF exerts metabolic effects on peripheral organs is currently unknown. Here, we show that the BDNF receptor TrkB.T1 is expressed by pancreatic ß-cells where it regulates insulin release. Mice lacking TrkB.T1 show impaired glucose tolerance and insulin secretion. ß-cell BDNF-TrkB.T1 signaling triggers calcium release from intracellular stores, increasing glucose-induced insulin secretion. Additionally, BDNF is secreted by skeletal muscle and muscle-specific BDNF knockout phenocopies the ß-cell TrkB.T1 deletion metabolic impairments. The finding that BDNF is also secreted by differentiated human muscle cells and induces insulin secretion in human islets via TrkB.T1 identifies a new regulatory function of BDNF on metabolism that is independent of CNS activity. Our data suggest that muscle-derived BDNF may be a key factor mediating increased glucose metabolism in response to exercise, with implications for the treatment of diabetes and related metabolic diseases.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Calcio/metabolismo , Células Cultivadas , Glucosa/metabolismo , Intolerancia a la Glucosa , Humanos , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Noqueados , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptor trkB/química , Receptor trkB/genética , Receptor trkB/metabolismo , Transducción de Señal
7.
Int J Mol Sci ; 21(4)2020 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-32079227

RESUMEN

Vav1 is a Rho/Rac (Ras-related C3 botulinum toxin substrate) guanine nucleotide exchange factor expressed in hematopoietic and endothelial cells that are involved in a wide range of cellular functions. It is also stabilized under hypoxic conditions when it regulates the accumulation of the transcription factor HIF (Hypoxia Inducible Factor)-1α, which activates the transcription of target genes to orchestrate a cellular response to low oxygen. One of the genes induced by HIF-1α is GLUT (Glucose Transporter)-1, which is the major glucose transporter expressed in vessels that supply energy to the brain. Here, we identify a role for Vav1 in providing glucose to the brain. We found that Vav1 deficiency downregulates HIF-1α and GLUT-1 levels in endothelial cells, including blood-brain barrier cells. This downregulation of GLUT-1, in turn, reduced glucose uptake to endothelial cells both in vitro and in vivo, and reduced glucose levels in the brain. Furthermore, endothelial cell-specific Vav1 knock-out in mice, which caused glucose uptake deficiency, also led to a learning delay in fear conditioning experiments. Our results suggest that Vav1 promotes learning by activating HIF-1α and GLUT-1 and thereby distributing glucose to the brain. We further demonstrate the importance of glucose transport by endothelial cells in brain functioning and reveal a potential new axis for targeting GLUT-1 deficiency syndromes and other related brain diseases.


Asunto(s)
Glucemia/metabolismo , Encéfalo/metabolismo , Regulación de la Expresión Génica , Transportador de Glucosa de Tipo 1/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Proteínas Proto-Oncogénicas c-vav/genética , Animales , Femenino , Transportador de Glucosa de Tipo 1/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Proteínas Proto-Oncogénicas c-vav/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transfección
8.
Elife ; 82019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31429825

RESUMEN

Brain-derived neurotrophic factor (BDNF) is a potent modulator of brain synaptic plasticity. Signaling defects caused by dysregulation of its Ntrk2 (TrkB) kinase (TrkB.FL) and truncated receptors (TrkB.T1) have been linked to the pathophysiology of several neurological and neurodegenerative disorders. We found that upregulation of Rbfox1, an RNA binding protein associated with intellectual disability, epilepsy and autism, increases selectively hippocampal TrkB.T1 isoform expression. Physiologically, increased Rbfox1 impairs BDNF-dependent LTP which can be rescued by genetically restoring TrkB.T1 levels. RNA-seq analysis of hippocampi with upregulation of Rbfox1 in conjunction with the specific increase of TrkB.T1 isoform expression also shows that the genes affected by Rbfox1 gain of function are surprisingly different from those influenced by Rbfox1 deletion. These findings not only identify TrkB as a major target of Rbfox1 pathophysiology but also suggest that gain or loss of function of Rbfox1 regulate different genetic landscapes.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Hipocampo/fisiología , Potenciación a Largo Plazo , Glicoproteínas de Membrana/biosíntesis , Proteínas Tirosina Quinasas/biosíntesis , Factores de Empalme de ARN/biosíntesis , Regulación hacia Arriba , Animales , Perfilación de la Expresión Génica , Ratones , Isoformas de Proteínas/biosíntesis , Análisis de Secuencia de ARN
9.
Mol Cell Neurosci ; 89: 33-41, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29580900

RESUMEN

Amyotrophic lateral sclerosis (ALS) causes rapidly progressive paralysis and death within 5 years from diagnosis due to degeneration of the motor circuits. However, a significant population of ALS patients also shows cognitive impairments and progressive hippocampal pathology. Likewise, the mutant SOD1(G93A) mouse model of ALS (mSOD1), in addition to loss of spinal motor neurons, displays altered spatial behavior and hippocampal abnormalities including loss of parvalbumin-positive interneurons (PVi) and enhanced long-term potentiation (LTP). However, the cellular and molecular mechanisms underlying these morpho-functional features are not well understood. Since removal of TrkB.T1, a receptor isoform of the brain-derived neurotrophic factor, can partially rescue the phenotype of the mSOD1 mice, here we tested whether removal of TrkB.T1 can normalize the number of PVi and the LTP in this model. Stereological analysis of hippocampal PVi in control, TrkB.T1-/-, mSOD1, and mSOD1 mice deficient for TrkB.T1 (mSOD1/T1-/-) showed that deletion of TrkB.T1 restored the number of PVi to physiological level in the mSOD1 hippocampus. The rescue of PVi neuron number is paralleled by a normalization of high-frequency stimulation-induced LTP in the pre-symptomatic mSOD1/T1-/- mice. Our experiments identified TrkB.T1 as a cellular player involved in the homeostasis of parvalbumin expressing interneurons and, in the context of murine ALS, show that TrkB.T1 is involved in the mechanism underlying structural and functional hippocampal degeneration. These findings have potential implications for hippocampal degeneration and cognitive impairments reported in ALS patients at early stages of the disease.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Región CA1 Hipocampal/metabolismo , Interneuronas/metabolismo , Potenciación a Largo Plazo , Receptor trkB/genética , Esclerosis Amiotrófica Lateral/genética , Animales , Región CA1 Hipocampal/fisiología , Eliminación de Gen , Interneuronas/fisiología , Ratones , Ratones Endogámicos C57BL , Parvalbúminas/genética , Parvalbúminas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptor trkB/metabolismo , Superóxido Dismutasa-1/genética
10.
Neurosci Lett ; 644: 76-82, 2017 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-28237805

RESUMEN

The adult rodent piriform cortex has been reported to harbor immature neurons that express markers associated with neurodevelopment and plasticity, namely polysialylated neural cell adhesion molecule (PSA-NCAM) and doublecortin (DCX). We characterized the expression of PSA-NCAM and DCX across the rostrocaudal axis of the rat piriform cortex and observed higher numbers of PSA-NCAM and DCX positive cells in the posterior subdivision. As observed in the rat piriform cortex, Nestin-GFP reporter mice also revealed a similar gradient of GFP-positive cells with an increasing rostro-caudal gradient of expression. Given the extensive noradrenergic innervation of the piriform cortex and its role in regulating piriform cortex function and synaptic plasticity, we addressed the influence of norepinephrine (NE) on piriform cortex plasticity marker expression. Depletion of NE by treatment with the noradrenergic neurotoxin DSP-4 significantly increased the number of DCX and PSA-NCAM immunopositive cells in the piriform cortex of adult rats. Similarly, DSP-4 treated Nestin-GFP reporter mice revealed a robust induction of GFP-positive cells within the piriform cortex following NE depletion. Genetic loss of NE in dopamine ß-hydroxylase knockout (Dbh -/-) mice phenocopied the effects of DSP-4, with an increase noted in PSA-NCAM and DCX positive cells in the piriform cortex. Further, chronic α2-adrenergic receptor stimulation with the agonist guanabenz increased PSA-NCAM and DCX positive cells in the piriform cortex of adult rats and GFP-positive cells in the piriform cortex of Nestin-GFP mice. By contrast, chronic α2-adrenergic receptor blockade with the antagonist yohimbine reduced PSA-NCAM and DCX positive cells in the piriform cortex of adult rats. Our results provide novel evidence for a role of NE in regulating the expression of plasticity markers, including PSA-NCAM, DCX, and nestin, within the adult mouse and rat piriform cortex.


Asunto(s)
Plasticidad Neuronal/fisiología , Norepinefrina/metabolismo , Corteza Piriforme/metabolismo , Animales , Biomarcadores/análisis , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/biosíntesis , Nestina/biosíntesis , Moléculas de Adhesión de Célula Nerviosa/biosíntesis , Neuropéptidos/biosíntesis , Ratas , Ratas Wistar
11.
Sci Rep ; 6: 32957, 2016 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-27604420

RESUMEN

Nedd4-2 (NEDD4L in humans) is a ubiquitin protein ligase best known for its role in regulating ion channel internalization and turnover. Nedd4-2 deletion in mice causes perinatal lethality associated with increased epithelial sodium channel (ENaC) expression in lung and kidney. Abundant data suggest that Nedd4-2 plays a role in neuronal functions and may be linked to epilepsy and dyslexia in humans. We used a mouse model of Nedd4-2 haploinsufficiency to investigate whether an alteration in Nedd4-2 levels of expression affects general nervous system functions. We found that Nedd4-2 heterozygous mice are hyperactive, have increased basal synaptic transmission and have enhanced sensitivity to inflammatory pain. Thus, Nedd4-2 heterozygous mice provide a new genetic model to study inflammatory pain. These data also suggest that in human, SNPs affecting NEDD4L levels may be involved in the development of neuropsychological deficits and peripheral neuropathies and may help unveil the genetic basis of comorbidities.


Asunto(s)
Hipercinesia/enzimología , Hipercinesia/genética , Ubiquitina-Proteína Ligasas Nedd4/deficiencia , Ubiquitina-Proteína Ligasas Nedd4/genética , Animales , Región CA1 Hipocampal/enzimología , Región CA1 Hipocampal/fisiopatología , Modelos Animales de Enfermedad , Epilepsia/enzimología , Epilepsia/genética , Epilepsia/fisiopatología , Potenciales Postsinápticos Excitadores , Haploinsuficiencia , Heterocigoto , Humanos , Hipercinesia/fisiopatología , Potenciación a Largo Plazo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Dolor/enzimología , Dolor/genética , Dolor/fisiopatología , Transmisión Sináptica
12.
J Neurosci ; 34(11): 4090-8, 2014 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-24623787

RESUMEN

TrkA is a tyrosine kinase receptor required for development and survival of the peripheral nervous system. In the adult, TrkA and its ligand NGF are peripheral pain mediators, particularly in inflammatory pain states. However, how TrkA regulates the function of nociceptive neurons and whether its activity levels may lead to sensory abnormalities is still unclear. Here we report the characterization of a 3 aa (KFG) domain that negatively regulates TrkA level and function in response to NGF. Deletion of this domain in mouse causes a reduction of TrkA ubiquitination leading to an increase in TrkA protein levels and activity. The number of dorsal root ganglia neurons is not affected by the mutation. However, mutant mice have enhanced thermal sensitivity and inflammatory pain. Together, these data suggest that ubiquitination is a mechanism used in nociceptive neurons to regulate TrkA level and function. Our results may enhance our understanding of how ubiquitination affects TrkA activation following noxious thermal stimulation and inflammatory pain.


Asunto(s)
Factor de Crecimiento Nervioso/metabolismo , Nociceptores/metabolismo , Receptor trkA/genética , Receptor trkA/metabolismo , Ubiquitinación/fisiología , Animales , Línea Celular , Regulación hacia Abajo/fisiología , Células Madre Embrionarias/citología , Femenino , Ganglios Espinales/citología , Células HEK293 , Humanos , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Inflamación/metabolismo , Inflamación/fisiopatología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Mutantes , Factor de Crecimiento Nervioso/farmacología , Estructura Terciaria de Proteína , Receptor trkA/química , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo
13.
PLoS One ; 7(6): e39946, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22761934

RESUMEN

Brain Derived Neurotrophic Factor (BDNF) exerts strong pro-survival effects on developing and injured motoneurons. However, in clinical trials, BDNF has failed to benefit patients with amyotrophic lateral sclerosis (ALS). To date, the cause of this failure remains unclear. Motoneurons express the TrkB kinase receptor but also high levels of the truncated TrkB.T1 receptor isoform. Thus, we investigated whether the presence of this receptor may affect the response of diseased motoneurons to endogenous BDNF. We deleted TrkB.T1 in the hSOD1(G93A) ALS mouse model and evaluated the impact of this mutation on motoneuron death, muscle weakness and disease progression. We found that TrkB.T1 deletion significantly slowed the onset of motor neuron degeneration. Moreover, it delayed the development of muscle weakness by 33 days. Although the life span of the animals was not affected we observed an overall improvement in the neurological score at the late stage of the disease. To investigate the effectiveness of strategies aimed at bypassing the TrkB.T1 limit to BDNF signaling we treated SOD1 mutant mice with the adenosine A2A receptor agonist CGS21680, which can activate motoneuron TrkB receptor signaling independent of neurotrophins. We found that CGS21680 treatment slowed the onset of motor neuron degeneration and muscle weakness similarly to TrkB.T1 removal. Together, our data provide evidence that endogenous TrkB.T1 limits motoneuron responsiveness to BDNF in vivo and suggest that new strategies such as Trk receptor transactivation may be used for therapeutic intervention in ALS or other neurodegenerative disorders.


Asunto(s)
Esclerosis Amiotrófica Lateral/fisiopatología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Receptor trkB/fisiología , Adenosina/análogos & derivados , Adenosina/farmacología , Agonistas del Receptor de Adenosina A2/farmacología , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ratones , Fenetilaminas/farmacología , Receptor trkB/genética , Superóxido Dismutasa/genética
14.
J Neurosci ; 32(7): 2252-62, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22396401

RESUMEN

The molecular mechanisms underlying the effects of electroconvulsive shock (ECS) therapy, a fast-acting and very effective antidepressant therapy, are poorly understood. Changes related to neuroplasticity, including enhanced adult hippocampal neurogenesis and neuronal arborization, are believed to play an important role in mediating the effects of ECS. Here we show a dynamic upregulation of the scaffold protein tamalin, selectively in the hippocampus of animals subjected to ECS. Interestingly, this gene upregulation is functionally significant because tamalin deletion in mice abrogated ECS-induced neurogenesis in the adult mouse hippocampus. Furthermore, loss of tamalin blunts mossy fiber sprouting and dendritic arborization caused by ECS. These data suggest an essential role for tamalin in ECS-induced adult neuroplasticity and provide new insight into the pathways that are involved in mediating ECS effects.


Asunto(s)
Proteínas Portadoras/fisiología , Electrochoque , Hipocampo/crecimiento & desarrollo , Proteínas de la Membrana/fisiología , Plasticidad Neuronal/fisiología , Animales , Electrochoque/métodos , Células Madre Embrionarias/fisiología , Femenino , Hipocampo/citología , Hipocampo/fisiología , Péptidos y Proteínas de Señalización Intracelular , Masculino , Proteínas de la Membrana/deficiencia , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes Neurológicos , Neurogénesis/fisiología , Distribución Aleatoria
15.
PLoS One ; 7(3): e32869, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22396798

RESUMEN

BACKGROUND: Previous studies suggest that the responsiveness of TrkB receptor to BDNF is developmentally regulated in rats. Antidepressant drugs (AD) have been shown to increase TrkB signalling in the adult rodent brain, and recent findings implicate a BDNF-independent mechanism behind this phenomenon. When administered during early postnatal life, ADs produce long-lasting biochemical and behavioural alterations that are observed in adult animals. METHODOLOGY: We have here examined the responsiveness of brain TrkB receptors to BDNF and ADs during early postnatal life of mouse, measured as autophosphorylation of TrkB (pTrkB). PRINCIPAL FINDINGS: We found that ADs fail to induce TrkB signalling before postnatal day 12 (P12) after which an adult response of TrkB to ADs was observed. Interestingly, there was a temporally inverse correlation between the appearance of the responsiveness of TrkB to systemic ADs and the marked developmental reduction of BDNF-induced TrkB in brain microslices ex vivo. Basal p-TrkB status in the brain of BDNF deficient mice was significantly reduced only during early postnatal period. Enhancing cAMP (cyclic adenosine monophosphate) signalling failed to facilitate TrkB responsiveness to BDNF. Reduced responsiveness of TrkB to BDNF was not produced by the developmental increase in the expression of dominant-negative truncated TrkB.T1 because this reduction was similarly observed in the brain microslices of trkB.T1(-/-) mice. Moreover, postnatal AD administration produced long-lasting behavioural alterations observable in adult mice, but the responses were different when mice were treated during the time when ADs did not (P4-9) or did (P16-21) activate TrkB. CONCLUSIONS: We have found that ADs induce the activation of TrkB only in mice older than 2 weeks and that responsiveness of brain microslices to BDNF is reduced during the same time period. Exposure to ADs before and after the age when ADs activate TrkB produces differential long-term behavioural responses in adult mice.


Asunto(s)
Antidepresivos/farmacología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Encéfalo/embriología , Regulación del Desarrollo de la Expresión Génica , Receptor trkB/metabolismo , Factores de Edad , Animales , Antidepresivos/uso terapéutico , Conducta Animal , Sistema Libre de Células , Clomipramina/farmacología , AMP Cíclico/metabolismo , Femenino , Imipramina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Fosforilación , Transducción de Señal , Factores de Tiempo
16.
J Neurosci ; 30(3): 1096-109, 2010 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-20089918

RESUMEN

Slow-onset adaptive changes that arise from sustained antidepressant treatment, such as enhanced adult hippocampal neurogenesis and increased trophic factor expression, play a key role in the behavioral effects of antidepressants. alpha(2)-Adrenoceptors contribute to the modulation of mood and are potential targets for the development of faster acting antidepressants. We investigated the influence of alpha(2)-adrenoceptors on adult hippocampal neurogenesis. Our results indicate that alpha(2)-adrenoceptor agonists, clonidine and guanabenz, decrease adult hippocampal neurogenesis through a selective effect on the proliferation, but not the survival or differentiation, of progenitors. These effects persist in dopamine beta-hydroxylase knock-out (Dbh(-/-)) mice lacking norepinephrine, supporting a role for alpha(2)-heteroceptors on progenitor cells, rather than alpha(2)-autoreceptors on noradrenergic neurons that inhibit norepinephrine release. Adult hippocampal progenitors in vitro express all the alpha(2)-adrenoceptor subtypes, and decreased neurosphere frequency and BrdU incorporation indicate direct effects of alpha(2)-adrenoceptor stimulation on progenitors. Furthermore, coadministration of the alpha(2)-adrenoceptor antagonist yohimbine with the antidepressant imipramine significantly accelerates effects on hippocampal progenitor proliferation, the morphological maturation of newborn neurons, and the increase in expression of brain derived neurotrophic factor and vascular endothelial growth factor implicated in the neurogenic and behavioral effects of antidepressants. Finally, short-duration (7 d) yohimbine and imipramine treatment results in robust behavioral responses in the novelty suppressed feeding test, which normally requires 3 weeks of treatment with classical antidepressants. Our results demonstrate that alpha(2)-adrenoceptors, expressed by progenitor cells, decrease adult hippocampal neurogenesis, while their blockade speeds up antidepressant action, highlighting their importance as targets for faster acting antidepressants.


Asunto(s)
Antagonistas de Receptores Adrenérgicos alfa 2 , Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Hipocampo/efectos de los fármacos , Imipramina/farmacología , Neurogénesis/efectos de los fármacos , Agonistas alfa-Adrenérgicos/farmacología , Antagonistas Adrenérgicos alfa/farmacología , Agonistas Adrenérgicos beta/farmacología , Análisis de Varianza , Animales , Animales Recién Nacidos , Antiparkinsonianos/farmacología , Ácido Ascórbico/farmacología , Factor Neurotrófico Derivado del Encéfalo , Bromodesoxiuridina/metabolismo , Células Cultivadas , Dopamina beta-Hidroxilasa/genética , Proteínas de Dominio Doblecortina , Droxidopa/farmacología , Esquema de Medicación , Interacciones Farmacológicas , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Técnicas In Vitro , Isoproterenol/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuropéptidos/metabolismo , Fenilefrina/farmacología , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ratas , Ratas Wistar , Tiempo de Reacción/efectos de los fármacos , Receptores Adrenérgicos alfa 2/fisiología , Células Madre/efectos de los fármacos , Yohimbina/farmacología
17.
J Neurosci ; 29(3): 678-85, 2009 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-19158294

RESUMEN

Pathological or in vitro overexpression of the truncated TrkB (TrkB.T1) receptor inhibits signaling through the full-length TrkB (TrkB.FL) tyrosine kinase receptor. However, to date, the role of endogenous TrkB.T1 is still unknown. By studying mice lacking the truncated TrkB.T1 isoform but retaining normal spatiotemporal expression of TrkB.FL, we have analyzed TrkB.T1-specific physiological functions and its effect on endogenous TrkB kinase signaling in vivo. We found that TrkB.T1-deficient mice develop normally but show increased anxiety in association with morphological abnormalities in the length and complexity of neurites of neurons in the basolateral amygdala. However, no behavioral abnormalities were detected in hippocampal-dependent memory tasks, which correlated with lack of any obvious hippocampal morphological deficits or alterations in basal synaptic transmission and long-term potentiation. In vivo reduction of TrkB signaling by removal of one BDNF allele could be partially rescued by TrkB.T1 deletion, which was revealed by an amelioration of the enhanced aggression and weight gain associated with BDNF haploinsufficiency. Our results suggest that, at the physiological level, TrkB.T1 receptors are important regulators of TrkB.FL signaling in vivo. Moreover, TrkB.T1 selectively affects dendrite complexity of certain neuronal populations.


Asunto(s)
Encéfalo/anomalías , Encéfalo/anatomía & histología , Mutación , Neuronas/fisiología , Receptor trkB/genética , Animales , Peso Corporal/genética , Encéfalo/ultraestructura , Factor Neurotrófico Derivado del Encéfalo/genética , Condicionamiento Psicológico/fisiología , Conducta Exploratoria/fisiología , Miedo , Hipocampo/citología , Técnicas In Vitro , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor trkB/deficiencia , Tinción con Nitrato de Plata/métodos
18.
Life Sci ; 76(12): 1325-38, 2005 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-15670613

RESUMEN

We assessed the effect of Azadirachta indica (A. indica), a plant that has been reported to possess antioxidant, anti-inflammatory and anxiolytic properties, on cerebral reperfusion injury and long term cerebral hypoperfusion. When blood flow to brain region that has undergone critical period of ischemia is re-established, additional injury is to be expected from the reperfusion. In the present study, bilateral common carotid artery (BCCA) occlusion for 30 min followed by 45 min reperfusion resulted in increase in lipid peroxidation, superoxide dismutase (SOD) activity and fall in total tissue sulfhydryl (T-SH) groups. A. indica pretreatment (500 mg/kg/day x 7 days) attenuated the reperfusion induced enhanced lipid peroxidation, SOD activity and prevented fall in T-SH groups. Moreover, A.indica per se increased brain ascorbic acid level, which was unchanged during reperfusion insult. Long-term cerebral hypoperfusion induced by permanent BCCA occlusion has been reported to cause behavioral and histopathological abnormalities. In the present study, as tested by open field paradigm and Morris' water maze, a propensity towards anxiety and disturbances of learning/memory were observed in animals subjected to hypoperfusion for 2 weeks. A. indica (500 mg/kg/day x 15 days) significantly reduced these hypoperfusion induced functional disturbances. Reactive changes in brain histology like gliosis, perivascular lymphocytic infiltration, recruitment of macrophages and cellular edema following long term hypoperfusion were also attenuated effectively by A. indica. We conclude that our study provides an experimental evidence for possible neuroprotective potentiality of A. indica.


Asunto(s)
Azadirachta/química , Ataque Isquémico Transitorio/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Extractos Vegetales/uso terapéutico , Daño por Reperfusión/tratamiento farmacológico , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Conducta Exploratoria/efectos de los fármacos , Ataque Isquémico Transitorio/metabolismo , Ataque Isquémico Transitorio/patología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Fitoterapia , Ratas , Ratas Endogámicas , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA