Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Mol Cell Neurosci ; 107: 103525, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32629109

RESUMEN

The brain is exceptionally demanding in terms of energy metabolism. Approximately 20% of the calories consumed are devoted to our cerebral faculties, with the lion's share provided in the form of glucose. The brain's stringent energy dependency requires a high degree of harmonization between the elements responsible for supplying- and metabolizing energetic substrates. However, chronic stress may jeopardize this homeostatic energy balance by disruption of critical metabolic processes. In agreement, stress-related mental disorders have been linked with perturbations in energy metabolism. Prominent stress-induced metabolic alterations include the actions of hormones, glucose uptake and mitochondrial adjustments. Importantly, fundamental stress-responsive metabolic adjustments in humans and animal models bear a striking resemblance. Here, an overview is provided of key findings, demonstrating the pervasive impact of chronic stress on energy metabolism. Furthermore, I argue that medications, aimed primarily at restoring metabolic homeostasis, may constitute a novel approach to treat mental disorders.


Asunto(s)
Encéfalo/metabolismo , Metabolismo Energético/fisiología , Glucosa/metabolismo , Estrés Fisiológico/fisiología , Animales , Homeostasis/fisiología , Humanos , Mitocondrias/metabolismo
2.
Proc Natl Acad Sci U S A ; 115(43): E10187-E10196, 2018 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-30301805

RESUMEN

Stringent glucose demands render the brain susceptible to disturbances in the supply of this main source of energy, and chronic stress may constitute such a disruption. However, whether stress-associated cognitive impairments may arise from disturbed glucose regulation remains unclear. Here we show that chronic social defeat (CSD) stress in adult male mice induces hyperglycemia and directly affects spatial memory performance. Stressed mice developed hyperglycemia and impaired glucose metabolism peripherally as well as in the brain (demonstrated by PET and induced metabolic bioluminescence imaging), which was accompanied by hippocampus-related spatial memory impairments. Importantly, the cognitive and metabolic phenotype pertained to a subset of stressed mice and could be linked to early hyperglycemia 2 days post-CSD. Based on this criterion, ∼40% of the stressed mice had a high-glucose (glucose >150 mg/dL), stress-susceptible phenotype. The relevance of this biomarker emerges from the effects of the glucose-lowering sodium glucose cotransporter 2 inhibitor empagliflozin, because upon dietary treatment, mice identified as having high glucose demonstrated restored spatial memory and normalized glucose metabolism. Conversely, reducing glucose levels by empagliflozin in mice that did not display stress-induced hyperglycemia (resilient mice) impaired their default-intact spatial memory performance. We conclude that hyperglycemia developing early after chronic stress threatens long-term glucose homeostasis and causes spatial memory dysfunction. Our findings may explain the comorbidity between stress-related and metabolic disorders, such as depression and diabetes, and suggest that cognitive impairments in both types of disorders could originate from excessive cerebral glucose accumulation.


Asunto(s)
Conducta Animal/fisiología , Enfermedad Crónica/psicología , Hiperglucemia/fisiopatología , Trastornos de la Memoria/fisiopatología , Memoria Espacial/fisiología , Estrés Psicológico/fisiopatología , Animales , Conducta Animal/efectos de los fármacos , Compuestos de Bencidrilo/farmacología , Encéfalo/metabolismo , Encéfalo/fisiopatología , Enfermedad Crónica/tratamiento farmacológico , Glucosa/metabolismo , Glucósidos/farmacología , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/metabolismo , Hiperglucemia/psicología , Masculino , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/psicología , Ratones , Ratones Endogámicos C57BL , Deseabilidad Social , Memoria Espacial/efectos de los fármacos , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/metabolismo , Estrés Psicológico/psicología
3.
Proc Natl Acad Sci U S A ; 112(50): 15486-91, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26621716

RESUMEN

Dominance hierarchies are integral aspects of social groups, yet whether personality traits may predispose individuals to a particular rank remains unclear. Here we show that trait anxiety directly influences social dominance in male outbred rats and identify an important mediating role for mitochondrial function in the nucleus accumbens. High-anxious animals that are prone to become subordinate during a social encounter with a low-anxious rat exhibit reduced mitochondrial complex I and II proteins and respiratory capacity as well as decreased ATP and increased ROS production in the nucleus accumbens. A causal link for these findings is indicated by pharmacological approaches. In a dyadic contest between anxiety-matched animals, microinfusion of specific mitochondrial complex I or II inhibitors into the nucleus accumbens reduced social rank, mimicking the low probability to become dominant observed in high-anxious animals. Conversely, intraaccumbal infusion of nicotinamide, an amide form of vitamin B3 known to enhance brain energy metabolism, prevented the development of a subordinate status in high-anxious individuals. We conclude that mitochondrial function in the nucleus accumbens is crucial for social hierarchy establishment and is critically involved in the low social competitiveness associated with high anxiety. Our findings highlight a key role for brain energy metabolism in social behavior and point to mitochondrial function in the nucleus accumbens as a potential marker and avenue of treatment for anxiety-related social disorders.


Asunto(s)
Encéfalo/fisiopatología , Dominación-Subordinación , Mitocondrias/metabolismo , Conducta Social , Animales , Ansiedad , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Masculino , Niacinamida/metabolismo , Núcleo Accumbens/fisiopatología , Ratas Wistar
4.
Pediatr Res ; 73(1): 18-23, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23085819

RESUMEN

BACKGROUND: Both hypothermia and erythropoietin (EPO) are reported to have neuroprotective effects after perinatal hypoxia-ischemia (HI). We investigated a possible additive effect of the use of a combination of hypothermia-EPO in a rat model of neonatal HI. METHODS: At postnatal day 7, rats were subjected to HI and then randomized to 3 h of hypothermia, EPO, or both. Sensorimotor function was assessed by the cylinder-rearing test (CRT) at 2 and 5 wk after HI. Brain lesion volume and white matter loss were determined by hematoxylin-eosin and luxol fast blue staining, respectively. RESULTS: Multivariable analysis using general linear modeling showed that hypothermia, EPO, and the interaction hypothermia × gender were determinants of sensorimotor function, both at 2 and 5 wk after HI. Neuroprotective effects of hypothermia at 5 wk were more pronounced in females, showing 52% improvement in the CRT. Maximal improvement in males was 26% after combined treatment with hypothermia and EPO. Histological outcome was improved by hypothermia only with no additional effect of EPO or gender. CONCLUSION: Hypothermia after HI improved sensorimotor function in females more than in males. There was a borderline additive effect of EPO when combined with hypothermia. Histology of brain lesion volume and white matter damage was improved only by hypothermia.


Asunto(s)
Eritropoyetina/uso terapéutico , Hipotermia Inducida/métodos , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Hipoxia-Isquemia Encefálica/terapia , Fármacos Neuroprotectores/uso terapéutico , Animales , Animales Recién Nacidos , Glucemia , Temperatura Corporal , Femenino , Técnicas Histológicas , Hipoxia-Isquemia Encefálica/patología , Modelos Lineales , Masculino , Ratas , Factores Sexuales
5.
Ann Neurol ; 70(2): 255-64, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21674585

RESUMEN

OBJECTIVE: To investigate whether inhibition of mitochondrial p53 association using pifithrin-µ (PFT-µ) represents a potential novel neuroprotective strategy to combat perinatal hypoxic-ischemic (HI) brain damage. METHODS: Seven-day-old rats were subjected to unilateral carotid artery occlusion and hypoxia followed by intraperitoneal treatment with PFT-µ, an inhibitor of p53 mitochondrial association or PFT-α an inhibitor of p53 transcriptional activity. Cerebral damage, sensorimotor and cognitive function, apoptotic pathways (cytosolic cytochrome c, Smac/DIABLO, active caspase 3), and oxidative stress (lipid peroxidation and PARP-1 cleavage) were investigated. RESULTS: PFT-µ treatment completely prevented the HI-induced increase in mitochondrial p53 association at 3 hours and reduced neuronal damage at 48 hours post-HI. PFT-µ had long-term (6-10 weeks post-HI) beneficial effects as sensorimotor and cognitive outcome improved and infarct size was reduced by ~79%. Neuroprotection by PFT-µ treatment was associated with strong inhibition of apoptotic pathways and reduced oxidative stress. Unexpectedly, PFT-µ also inhibited HI-induced upregulation of p53 target genes. However, the neuroprotective effect of inhibiting only p53 transcriptional activity by PFT-α was significantly smaller and did not involve reduced oxidative stress. INTERPRETATION: We are the first to show that prevention of mitochondrial p53 association by PFT-µ strongly improves functional outcome and decreases lesion size after neonatal HI. PFT-µ not only inhibits mitochondrial release of cytochrome c, but also inhibits oxidative stress. We propose that as a consequence nuclear accumulation of p53 and transcription of proapoptotic target genes are prevented. In conclusion, targeting p53 mitochondrial association by PFT-µ may develop into a novel and powerful neuroprotective strategy.


Asunto(s)
Benzotiazoles/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Hipoxia-Isquemia Encefálica/prevención & control , Fármacos Neuroprotectores/farmacología , Tolueno/análogos & derivados , Proteína p53 Supresora de Tumor/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/fisiología , Benzotiazoles/uso terapéutico , Biomarcadores/metabolismo , Encéfalo/patología , Hipoxia-Isquemia Encefálica/patología , Mitocondrias/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Estrés Oxidativo , Ratas , Ratas Wistar , Tolueno/farmacología , Tolueno/uso terapéutico , Resultado del Tratamiento , Proteína p53 Supresora de Tumor/antagonistas & inhibidores
6.
J Mol Med (Berl) ; 100(10): 1441-1453, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35943566

RESUMEN

Chronic stress has the potential to impair health and may increase the vulnerability for psychiatric disorders. Emerging evidence suggests that specific neurometabolic dysfunctions play a role herein. In mice, chronic social defeat (CSD) stress reduces cerebral glucose uptake despite hyperglycemia. We hypothesized that this metabolic decoupling would be reflected by changes in contact sites between mitochondria and the endoplasmic reticulum, important intracellular nutrient sensors, and signaling hubs. We thus analyzed the proteome of their biochemical counterparts, mitochondria-associated membranes (MAMs) from whole brain tissue obtained from CSD and control mice. This revealed a lack of the glucose-metabolizing enzyme hexokinase 3 (HK3) in MAMs from CSD mice. In controls, HK3 protein abundance in MAMs and also in striatal synaptosomes correlated positively with peripheral blood glucose levels, but this connection was lost in CSD. We conclude that the ability of HK3 to traffic to sites of need, such as MAMs or synapses, is abolished upon CSD and surmise that this contributes to a cellular dysfunction instigated by chronic stress. KEY MESSAGES : Chronic social defeat (CSD) alters brain glucose metabolism CSD depletes hexokinase 3 (HK3) from mitochondria-associated membranes (MAMs) CSD results in loss of positive correlation between blood glucose and HK3 in MAMs and synaptosomes.


Asunto(s)
Glucemia , Hexoquinasa , Animales , Glucemia/metabolismo , Encéfalo/metabolismo , Glucosa/metabolismo , Hexoquinasa/metabolismo , Humanos , Ratones , Membranas Mitocondriales/metabolismo
7.
Pediatr Res ; 69(1): 56-61, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20856165

RESUMEN

There are mixed reports on the neuroprotective properties of erythropoietin (EPO) in animal models of birth asphyxia. We investigated the effect of EPO on short- and long-term outcome after neonatal hypoxic-ischemic (HI) brain injury in mice and compared the effect of two different dose regimens of EPO. Nine-day-old mice were subjected to HI, and EPO was injected i.p. at 0, 24, and 48 h after HI in a dose of either 5 or 20 kU/kg. Paw preference in the cylinder rearing test (CRT) was used as a measure of sensorimotor function. Only in female mice, administration of EPO at 5 kU/kg but not 20 kU/kg improved sensorimotor function, reduced striatum atrophy and hippocampal lesion volume, and enhanced myelin basic protein (MBP) staining as determined at 4 and 9 wk after HI. In addition, at 72 h after HI, more Ki 67 cells were found in the subventricular zone and dentate gyrus after EPO 5 kU/kg treatment, indicating an increase in progenitor cell proliferation. In conclusion, EPO improves sensorimotor function after neonatal HI and protects against striatum atrophy, hippocampus injury, and white matter loss. The protective effect of EPO is dose-dependent and only present in females.


Asunto(s)
Eritropoyetina/farmacología , Eritropoyetina/uso terapéutico , Hipocampo/citología , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Hipoxia-Isquemia Encefálica/patología , Vaina de Mielina/efectos de los fármacos , Desempeño Psicomotor/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Eritropoyetina/administración & dosificación , Femenino , Hipocampo/efectos de los fármacos , Humanos , Recién Nacido , Masculino , Ratones , Proteína Básica de Mielina/metabolismo , Neurogénesis/efectos de los fármacos , Factores Sexuales , Factores de Tiempo
8.
Transl Psychiatry ; 11(1): 4, 2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33414410

RESUMEN

Major depressive disorder is the most prevalent mental illness worldwide, still its pharmacological treatment is limited by various challenges, such as the large heterogeneity in treatment response and the lack of insight into the neurobiological pathways underlying this phenomenon. To decode the molecular mechanisms shaping antidepressant response and to distinguish those from general paroxetine effects, we used a previously established approach targeting extremes (i.e., good vs poor responder mice). We focused on the dentate gyrus (DG), a subregion of major interest in the context of antidepressant mechanisms. Transcriptome profiling on micro-dissected DG granule cells was performed to (i) reveal cell-type-specific changes in paroxetine-induced gene expression (paroxetine vs vehicle) and (ii) to identify molecular signatures of treatment response within a cohort of paroxetine-treated animals. We identified 112 differentially expressed genes associated with paroxetine treatment. The extreme group comparison (good vs poor responder) yielded 211 differentially expressed genes. General paroxetine effects could be distinguished from treatment response-associated molecular signatures, with a differential gene expression overlap of only 4.6% (15 genes). Biological pathway enrichment and cluster analyses identified candidate mechanisms associated with good treatment response, e.g., neuropeptide signaling, synaptic transmission, calcium signaling, and regulation of glucocorticoid secretion. Finally, we examined glucocorticoid receptor (GR)-dependent regulation of selected response-associated genes to analyze a hypothesized interplay between GR signaling and good antidepressant treatment response. Among the most promising candidates, we suggest potential targets such as the developmental gene Otx2 or Htr2c for further investigations into antidepressant treatment response in the future.


Asunto(s)
Trastorno Depresivo Mayor , Animales , Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Giro Dentado , Trastorno Depresivo Mayor/tratamiento farmacológico , Trastorno Depresivo Mayor/genética , Hipocampo , Ratones , Paroxetina/farmacología , Paroxetina/uso terapéutico
9.
J Endocrinol ; 249(1): 19-30, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33608492

RESUMEN

Stress has a major impact on the modulation of metabolism, as previously evidenced by hyperglycemia following chronic social defeat (CSD) stress in mice. Although CSD-triggered metabolic dysregulation might predispose to pre-diabetic conditions, insulin sensitivity remained intact, and obesity did not develop, when animals were fed with a standard diet (SD). Here, we investigated whether a nutritional challenge, a high-fat diet (HFD), aggravates the metabolic phenotype and whether there are particularly sensitive time windows for the negative consequences of HFD exposure. Chronically stressed male mice and controls (CTRL) were kept under (i) SD-conditions, (ii) with HFD commencing post-CSD, or (iii) provided with HFD lasting throughout and after CSD. Under SD conditions, stress increased glucose levels early post-CSD. Both HFD regimens increased glucose levels in non-stressed mice but not in stressed mice. Nonetheless, when HFD was provided after CSD, stressed mice did not differ from controls in long-term body weight gain, fat tissue mass and plasma insulin, and leptin levels. In contrast, when HFD was continuously available, stressed mice displayed reduced body weight gain, lowered plasma levels of insulin and leptin, and reduced white adipose tissue weights as compared to their HFD-treated non-stressed controls. Interestingly, stress-induced adrenal hyperplasia and hypercortisolemia were observed in mice treated with SD and with HFD after CSD but not in stressed mice exposed to a continuous HFD treatment. The present work demonstrates that CSD can reduce HFD-induced metabolic dysregulation. Hence, HFD during stress may act beneficially, as comfort food, by decreasing stress-induced metabolic demands.


Asunto(s)
Antígenos de Grupos Sanguíneos/análisis , Dieta Alta en Grasa , Estrés Psicológico/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Dieta Alta en Grasa/psicología , Ingestión de Energía , Prueba de Tolerancia a la Glucosa , Insulina/sangre , Leptina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Obesidad/psicología , Derrota Social , Estrés Psicológico/sangre , Aumento de Peso
10.
Neurobiol Stress ; 15: 100404, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34632008

RESUMEN

Delayed onset of antidepressant action is a shortcoming in depression treatment. Ketamine and its metabolite (2R,6R)-hydroxynorketamine (HNK) have emerged as promising rapid-acting antidepressants. However, their mechanism of action remains unknown. In this study, we first described the anxious and depression-prone inbred mouse strain, DBA/2J, as an animal model to assess the antidepressant-like effects of ketamine and HNK in vivo. To decode the molecular mechanisms mediating HNK's rapid antidepressant effects, a longitudinal cerebrospinal fluid (CSF) proteome profiling of its acute and sustained effects was conducted using an unbiased, hypothesis-free mass spectrometry-based proteomics approach. A total of 387 proteins were identified, with a major implication of significantly differentially expressed proteins in the glucocorticoid receptor (GR) signaling pathway, providing evidence for a link between HNK and regulation of the stress hormone system. Mechanistically, we identified HNK to repress GR-mediated transcription and reduce hormonal sensitivity of GR in vitro. In addition, mammalian target of rapamycin (mTOR) and brain-derived neurotrophic factor (BDNF) were predicted to be important upstream regulators of HNK treatment. Our results contribute to precise understanding of the temporal dynamics and molecular targets underlying HNK's rapid antidepressant-like effects, which can be used as a benchmark for improved treatment strategies for depression in future.

11.
Neurobiol Stress ; 15: 100338, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34095364

RESUMEN

The precise mechanisms underlying the detrimental effects of early life adversity (ELA) on adult mental health remain still elusive. To date, most studies have exclusively targeted neuronal populations and not considered neuron-glia crosstalk as a crucially important element for the integrity of stress-related brain function. Here, we have investigated the impact of ELA, in the form of a limited bedding and nesting material (LBN) paradigm, on a glial subpopulation with unique properties in brain homeostasis, the NG2+ cells. First, we have established a link between maternal behavior, activation of the offspring's stress response and heterogeneity in the outcome to LBN manipulation. We further showed that LBN targets the hippocampal NG2+ transcriptome with glucocorticoids being an important mediator of the LBN-induced molecular changes. LBN altered the NG2+ transcriptome and these transcriptional effects were correlated with glucocorticoids levels. The functional relevance of one LBN-induced candidate gene, Scn7a, could be confirmed by an increase in the density of voltage-gated sodium (Nav) channel activated currents in hippocampal NG2+ cells. Scn7a remained upregulated until adulthood in LBN animals, which displayed impaired cognitive performance. Considering that Nav channels are important for NG2+ cell-to-neuron communication, our findings provide novel insights into the disruption of this process in LBN mice.

12.
Neurobiol Dis ; 38(2): 266-72, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20132887

RESUMEN

We recently demonstrated that inhibition of the NF-kappaB-pathway by the specific peptide inhibitor TAT-NBD markedly reduced cerebral injury in a rat model of perinatal hypoxic-ischemic (HI) brain damage. The aim of the current study was to assess whether neuroprotection by TAT-NBD is associated with long-term functional improvements after neonatal HI. Postnatal-day 7 rats subjected to HI showed motor deficits in the cylinder rearing test and adhesive removal task. HI-treated animals also showed cognitive impairments in a visuo-spatial learning task (modified hole board) as defined by an increased latency to complete this task and increased numbers of short- and long-term memory errors. HI animals treated with TAT-NBD [20mg/kg i.p.] at 0 and 3h post-HI did not show impairments in the cylinder rearing test, adhesive removal task and modified hole board. In conclusion, the almost complete reduction in lesion size observed after TAT-NBD treatment was associated with long-lasting normalization of sensorimotor and cognitive functions.


Asunto(s)
Encéfalo/fisiopatología , Cognición/efectos de los fármacos , Hipoxia-Isquemia Encefálica/fisiopatología , Destreza Motora/efectos de los fármacos , FN-kappa B/metabolismo , Análisis de Varianza , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Señales (Psicología) , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/patología , FN-kappa B/antagonistas & inhibidores , Neuronas/metabolismo , Neuronas/patología , Ratas , Transducción de Señal
13.
Brain Behav Immun ; 24(5): 850-6, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19748566

RESUMEN

To understand and potentially treat the lifelong cognitive and motor deficits in humans resulting from perinatal mild cerebral hypoxic-ischemic (HI) events, valid animal models are of high importance. Nowadays the murine model of neonatal cerebral HI-injury (unilateral carotid artery occlusion followed by hypoxia) is applied more frequently. In the present study we investigated motor, behavioral and cognitive functioning in mice with mild cerebral HI-injury (45 min of hypoxia; HI-45) in comparison to mice exposed to severe HI (HI-75) and sham-control mice. Lateralizing motor disturbances as measured using the cylinder rearing test developed in both HI-45 and HI-75 mice and was significantly more severe in HI-75 animals. To assess behavior and cognitive functions, we used the modified hole board (mHB) test in two stages. First, the ability of the animals to find the three food rewards in cued holes over time was determined. The results revealed an overall learning impairment in HI-75 mice, while HI-45 mice were not different from sham controls. In the second stage, a reversal test was performed with rewarded cylinders being non-cued and non-rewarded cylinders being cued. This reversal-task revealed impairments in cognitive flexibility in HI-45 mice as compared to sham-control animals. Our data indicate that both the cylinder rearing task and the two stages of the mHB are suitable behavioral approaches to differentiate consequences of neonatal mild and severe brain damage on executive functioning.


Asunto(s)
Trastornos del Conocimiento/fisiopatología , Cognición/fisiología , Hipoxia-Isquemia Encefálica/fisiopatología , Actividad Motora/fisiología , Análisis de Varianza , Animales , Animales Recién Nacidos , Conducta Animal/fisiología , Ratones , Recompensa
14.
Brain Behav Immun ; 24(5): 812-21, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19766183

RESUMEN

Perinatal hypoxic-ischemic (HI) brain damage continues to be a major clinical problem. We investigated the contribution of the MAP kinase c-Jun N-terminal kinase (JNK), to neonatal HI brain damage. JNK regulates several transcriptional (via AP-1 activation) and non-transcriptional processes involved in brain damage such as inflammation and cell death/survival. P7 rats were subjected to HI by unilateral carotid artery occlusion and hypoxia. HI-induced activation of cerebral AP-1 peaked at 3-6h post-HI. Intraperitoneal administration of the JNK-inhibitor TAT-JBD immediately after HI prevented AP-1 activation. TAT-JBD treatment within 3h after HI reduced early neuronal damage by approximately 30%. JNK/AP-1 inhibition did not reduce HI-induced cytokine/chemokine expression. Analysis of indicators of apoptotic cell death revealed that TAT-JBD markedly reduced the HI-induced increase in active caspase 3. However, the upstream mediators of apoptosis: active caspase 8, cleaved Bid, mitochondrial cytochrome c release and caspase 9 cleavage were not reduced after TAT-JBD. TAT-JBD inhibited the HI-induced increase in Smac/DIABLO, an inhibitor of IAPs that prevent activation of caspase 3. TAT-JBD treatment also reduced cleavage of alpha-fodrin, indicating that calpain-mediated brain damage was reduced. Neuroprotection by TAT-JBD treatment was long-lasting as gray- and white matter damage was diminished by approximately 50% at 14 weeks post-HI concomitantly with marked improvement of sensorimotor behavior and cognitive functioning. In conclusion, JNK inhibition by TAT-JBD treatment reduced neonatal HI brain damage with a therapeutic window of 3h and long-lasting anatomical and behavioral improvements. We propose that inhibition of mitochondrial Smac/DIABLO release and calpain activation contribute to neuroprotection by TAT-JBD.


Asunto(s)
Apoptosis/efectos de los fármacos , Encéfalo/metabolismo , Hipoxia-Isquemia Encefálica/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Neuronas/metabolismo , Análisis de Varianza , Animales , Conducta Animal , Western Blotting , Encéfalo/patología , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Hipoxia-Isquemia Encefálica/patología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neuronas/efectos de los fármacos , Neuronas/patología , Péptidos/farmacología , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Prueba de Desempeño de Rotación con Aceleración Constante , Transducción de Señal/efectos de los fármacos , Factor de Transcripción AP-1/metabolismo
15.
Curr Alzheimer Res ; 17(1): 80-92, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32065104

RESUMEN

BACKGROUND: Function of the Amyloid Precursor Protein (AßPP) and its various cleavage products still is not unraveled down to the last detail. While its role as a source of the neurotoxic Amyloid beta (Aß) peptides in Alzheimer's Disease (AD) is undisputed and its property as a cell attachment protein is intriguing, while functions outside the neuronal context are scarcely investigated. This is particularly noteworthy because AßPP has a ubiquitous expression profile and its longer isoforms, AßPP750 and 770, are found in various tissues outside the brain and in non-neuronal cells. OBJECTIVE: Here, we aimed at analyzing the 5xFAD Alzheimer's disease mouse model in regard to male sexual function. The transgenes of this mouse model are regulated by Thy1 promoter activity and Thy1 is expressed in testes, e.g. by Sertoli cells. This allows speculation about an influence on sexual behavior. METHODS: We analyzed morphological as well as biochemical properties of testicular tissue from 5xFAD mice and wild type littermates and testosterone levels in serum, testes and the brain. Sexual behavior was assessed by a urine scent marking test at different ages for both groups. RESULTS: While sperm number, testes weight and morphological phenotypes of sperms were nearly indistinguishable from those of wild type littermates, testicular testosterone levels were significantly increased in the AD model mice. This was accompanied by elevated and prolonged sexual interest as displayed within the urine scent marking test. CONCLUSION: We suggest that overexpression of AßPP, which mostly is used to mimic AD in model mice, also affects male sexual behavior as assessed additional by the Urine Scent Marking (USM) test. The elevated testosterone levels might have an additional impact on central nervous system androgen receptors and also have to be considered when assessing learning and memory capabilities.


Asunto(s)
Enfermedad de Alzheimer/sangre , Encéfalo/metabolismo , Conducta Sexual Animal/fisiología , Testículo/patología , Testosterona/sangre , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Transgénicos , Tamaño de los Órganos/fisiología , Presenilina-1/genética , Recuento de Espermatozoides
16.
Neurosci Lett ; 451(2): 109-13, 2009 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-19103262

RESUMEN

Deferoxamine (DFO) and erythropoietin (EPO) have each been shown to provide neuroprotection in neonatal rodent models of brain injury. In view of the described anti-oxidative actions of DFO and the anti-apoptotic and anti-inflammatory effects of EPO, we hypothesized that the combination of DFO and EPO would increase neuroprotection after neonatal hypoxic-ischemic brain injury as compared to single DFO or EPO treatment. At postnatal day 7 rats underwent right common carotid artery occlusion followed by a 90-min exposure to 8% oxygen. Rats were treated intraperitoneally with DFO (200mg/kg), recombinant human EPO (1 kU/kg), a combination of DFO-EPO or vehicle at 0, 24 and 48 h after hypoxia-ischemia (HI) and were sacrificed at 72 h. DFO-EPO administration reduced the number of cleaved caspase 3-positive cells in the ipsilateral cerebral cortex. Early neuronal damage was assessed by staining for microtubuli-associated protein (MAP)-2. In our model 63+/-9% loss of ipsilateral MAP-2 was observed after HI, indicating extensive brain injury. DFO, EPO or DFO-EPO treatment did not improve neuronal integrity as defined by MAP-2. Cerebral white matter tracts were stained for myelin basic protein (MBP), a constituent of myelin. Hypoxia-ischemia strongly reduced MBP staining which suggests white matter damage. However, DFO, EPO and DFO-EPO treatment had no effect on the loss of MBP staining. Finally, HI-induced loss of striatal tyrosine hydroxylase staining was not attenuated by DFO, EPO or DFO-EPO. Although DFO-EPO treatment reduced the number of cleaved caspase 3(+) cells, treatment with DFO, EPO, or with the combination of DFO and EPO did not protect against gray or white matter damage in the experimental setting applied.


Asunto(s)
Citoprotección/efectos de los fármacos , Deferoxamina/farmacología , Eritropoyetina/farmacología , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Animales , Animales Recién Nacidos , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Antioxidantes/farmacología , Infarto Encefálico/tratamiento farmacológico , Infarto Encefálico/metabolismo , Infarto Encefálico/fisiopatología , Caspasa 3/efectos de los fármacos , Caspasa 3/metabolismo , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/patología , Corteza Cerebral/fisiopatología , Citoprotección/fisiología , Deferoxamina/uso terapéutico , Modelos Animales de Enfermedad , Combinación de Medicamentos , Sinergismo Farmacológico , Eritropoyetina/uso terapéutico , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/fisiopatología , Proteínas Asociadas a Microtúbulos/metabolismo , Proteína Básica de Mielina/metabolismo , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Fibras Nerviosas Mielínicas/efectos de los fármacos , Fibras Nerviosas Mielínicas/metabolismo , Fibras Nerviosas Mielínicas/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Ratas , Ratas Wistar
17.
Brain Res Rev ; 59(1): 22-33, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18514916

RESUMEN

Besides its established function in erythropoiesis, erythropoietin (EPO) is currently also appreciated for its neuroprotective effects. The detrimental sequelae of prolonged cerebral hypoxia and ischemia have been shown to attenuate by EPO treatment. After binding to the EPO receptor, EPO is capable of initiating a cascade of events which--via different pathways--may lead to neuroprotection. The circumstances that determine which specific signalling route(s) are activated by EPO are largely unknown. We aim to provide the reader with a timely overview on the use of EPO in models of stroke and hypoxia-ischemia and to discuss the molecular events that underlie its neuroprotection.


Asunto(s)
Eritropoyetina/uso terapéutico , Hipoxia-Isquemia Encefálica/prevención & control , Fármacos Neuroprotectores/uso terapéutico , Animales , Modelos Animales de Enfermedad , Eritropoyetina/metabolismo , Humanos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factores de Tiempo
18.
Neurosci Biobehav Rev ; 105: 136-177, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30970272

RESUMEN

Fear is an emotion that serves as a driving factor in how organisms move through the world. In this review, we discuss the current understandings of the subjective experience of fear and the related biological processes involved in fear learning and memory. We first provide an overview of fear learning and memory in humans and animal models, encompassing the neurocircuitry and molecular mechanisms, the influence of genetic and environmental factors, and how fear learning paradigms have contributed to treatments for fear-related disorders, such as posttraumatic stress disorder. Current treatments as well as novel strategies, such as targeting the perisynaptic environment and use of virtual reality, are addressed. We review research on the subjective experience of fear and the role of autobiographical memory in fear-related disorders. We also discuss the gaps in our understanding of fear learning and memory, and the degree of consensus in the field. Lastly, the development of linguistic tools for assessments and treatment of fear learning and memory disorders is discussed.


Asunto(s)
Miedo/fisiología , Aprendizaje/fisiología , Memoria Episódica , Trastornos Fóbicos , Psicolingüística , Trastornos por Estrés Postraumático , Animales , Humanos , Trastornos Fóbicos/fisiopatología , Trastornos Fóbicos/terapia , Trastornos por Estrés Postraumático/fisiopatología , Trastornos por Estrés Postraumático/terapia
19.
Neuropharmacology ; 138: 275-281, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29933007

RESUMEN

Social dominance can be attained through social competitions. Recent work in both humans and rodents has identified trait anxiety as a crucial predictor of social competitiveness. In addition, the anxiolytic GABAA positive modulator, diazepam, injected either systemically or into the ventral tegmental area (VTA) was shown to increase social dominance. Here, we investigated the impact of pharmacologically targeting GABAA receptors in the VTA for the outcome of a social competition between two unfamiliar male rats, one of them infused with vehicle and the other one with the drug under study. We show that infusion of the GABAA receptor agonist, muscimol, reduced anxiety-like behaviors and enhanced social competition, the GABAA receptor antagonist, bicuculline had the opposite effects. Importantly, intra-VTA muscimol administration also counteracted the disadvantage of high anxious rats to win a social competition against low anxious rats. Furthermore, we assessed the effectiveness of targeting specific GABAA receptor subunits by infusing zolpidem (α1-subunit agonist) or TCS1105 (a benzodiazepine ligand with α2-subunit agonistic and α1-subunit antagonistic effects) into the VTA. While zolpidem infusion did not affect the outcome of the social competition, TCS1105 enhanced social dominance. Our data highlight GABAergic mechanisms involving the engagement of α2-subunit containing GABAA receptors in the VTA in the attainment of dominance rank. The involvement of α2-subunit containing GABAA receptors in the VTA in the regulation of social competitiveness supports the potential therapeutic relevance of targeting these receptors to ameliorate anxiety-related social dysfunctions.


Asunto(s)
Conducta Competitiva/fisiología , Receptores de GABA-A/metabolismo , Área Tegmental Ventral/metabolismo , Animales , Ansiedad/tratamiento farmacológico , Ansiedad/metabolismo , Conducta Competitiva/efectos de los fármacos , Dominación-Subordinación , GABAérgicos/farmacología , Masculino , Distribución Aleatoria , Ratas Wistar , Área Tegmental Ventral/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo
20.
Psychoneuroendocrinology ; 91: 149-158, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29555365

RESUMEN

Understanding the neurobiological mechanisms underlying the response to an acute stressor may provide novel insights into successful stress-coping strategies. Acute behavioral stress-effects may be restricted to a specific time window early after stress-induction. However, existing behavioral test batteries typically span multiple days or even weeks, limiting the feasibility for a broad behavioral analysis following acute stress. Here, we designed a novel comprehensive behavioral test battery in male mice that assesses multiple behavioral dimensions within a sufficiently brief time window to capture acute stress-effects and its temporal profile. Using this battery, we investigated the behavioral impact of acute social defeat stress (ASD) early thereafter (ASD-early, ∼4 h), when circulating corticosterone levels were elevated, and late after stress-induction (ASD-late, ∼8 h), when corticosterone were returned to timed control levels. ASD-early, but not ASD-late, displayed hippocampal-dependent cognitive impairments in the Y-maze and in the spatial object recognition test. The actin-binding protein (ABP) Tumor suppressor down-regulated in renal cell carcinoma 1 (DRR1) has been described as resilience-promoting factor but the potential of DRR1 to curb stress-effects has not been investigated. Hippocampal DRR1 mRNA-expression was increased in ASD-early and ASD-late whereas DRR1-protein levels were increased only in ASD-late. We hypothesized that the absence of hippocampal DRR1 protein-upregulation in ASD-early caused the associated cognitive impairments. Hence, virus-mediated hippocampal DRR1-overexpression was induced as putative treatment, but cognitive deficits in ASD-early were not improved. We conclude that hippocampal DRR1-overexpression is insufficient to protect from the detrimental cognitive effects following acute social stress where perhaps a more global response in local actin dynamics, involving multiple stress-responsive ABPs that act synergistically, was warranted.


Asunto(s)
Estrés Psicológico/patología , Proteínas Supresoras de Tumor/fisiología , Animales , Conducta Animal/fisiología , Encéfalo/metabolismo , Trastornos del Conocimiento/metabolismo , Corticosterona/metabolismo , Hipocampo/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Fenotipo , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA