Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Am J Physiol Renal Physiol ; 327(2): F314-F326, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38932694

RESUMEN

Perinatal asphyxia (PA) poses a significant threat to multiple organs, particularly the kidneys. Diagnosing PA-associated kidney injury remains challenging, and treatment options are inadequate. Furthermore, there is a lack of long-term follow-up data regarding the renal implications of PA. In this study, 7-day-old male Wistar rats were exposed to PA using a gas mixture (4% O2; 20% CO2 in N2 for 15 min) to investigate molecular pathways linked to renal tubular damage, hypoxia, angiogenesis, heat shock response, inflammation, and fibrosis in the kidney. In a second experiment, adult rats with a history of PA were subjected to moderate renal ischemia-reperfusion (IR) injury to test the hypothesis that PA exacerbates renal susceptibility. Our results revealed an increased gene expression of renal injury markers (kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin), hypoxic and heat shock factors (hypoxia-inducible factor-1α, heat shock factor-1, and heat shock protein-27), proinflammatory cytokines (interleukin-1ß, interleukin-6, tumor necrosis factor-α, and monocyte chemoattractant protein-1), and fibrotic markers (transforming growth factor-ß, connective tissue growth factor, and fibronectin) promptly after PA. Moreover, a machine learning model was identified through random forest analysis, demonstrating an impressive classification accuracy (95.5%) for PA. Post-PA rats showed exacerbated functional decline and tubular injury and more intense hypoxic, heat shock, proinflammatory, and profibrotic response after renal IR injury compared with controls. In conclusion, PA leads to subclinical kidney injury, which may increase the susceptibility to subsequent renal damage later in life. In addition, the parameters identified through random forest analysis provide a robust foundation for future biomarker research in the context of PA.NEW & NOTEWORTHY This article demonstrates that perinatal asphyxia leads to subclinical kidney injury that permanently increases renal susceptibility to subsequent ischemic injury. We identified major molecular pathways involved in perinatal asphyxia-induced renal complications, highlighting potential targets of therapeutic approaches. In addition, random forest analysis revealed a model that classifies perinatal asphyxia with 95.5% accuracy that may provide a strong foundation for further biomarker research. These findings underscore the importance of multiorgan follow-up for perinatal asphyxia-affected patients.


Asunto(s)
Lesión Renal Aguda , Modelos Animales de Enfermedad , Riñón , Ratas Wistar , Daño por Reperfusión , Animales , Masculino , Lesión Renal Aguda/patología , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/etiología , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Riñón/patología , Riñón/metabolismo , Fibrosis , Asfixia Neonatal/metabolismo , Asfixia Neonatal/complicaciones , Asfixia Neonatal/patología , Animales Recién Nacidos , Ratas , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Citocinas/metabolismo , Factores de Edad , Mediadores de Inflamación/metabolismo
2.
J Neurosci ; 41(9): 1982-1995, 2021 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-33468566

RESUMEN

Excessive fear learning and generalized, extinction-resistant fear memories are core symptoms of anxiety and trauma-related disorders. Despite significant evidence from clinical studies reporting hyperactivity of the bed nucleus of stria terminalis (BNST) under these conditions, the role of BNST in fear learning and expression is still not clarified. Here, we tested how BNST modulates fear learning in male mice using a chemogenetic approach. Activation of GABAergic neurons of BNST during fear conditioning or memory consolidation resulted in enhanced cue-related fear recall. Importantly, BNST activation had no acute impact on fear expression during conditioning or recalls, but it enhanced cue-related fear recall subsequently, potentially via altered activity of downstream regions. Enhanced fear memory consolidation could be replicated by selectively activating somatostatin (SOM), but not corticotropin-releasing factor (CRF), neurons of the BNST, which was accompanied by increased fear generalization. Our findings suggest the significant modulation of fear memory strength by specific circuits of the BNST.SIGNIFICANCE STATEMENT The bed nucleus of stria terminalis (BNST) mediates different defensive behaviors, and its connections implicate its integrative modulatory role in fear memory formation; however, the involvement of BNST in fear learning has yet to be elucidated in detail. Our data highlight that BNST stimulation enhances fear memory formation without direct effects on fear expression. Our study identified somatostatin (SOM) cells within the extended amygdala as specific neurons promoting fear memory formation. These data underline the importance of anxiety circuits in maladaptive fear memory formation, indicating elevated BNST activity as a potential vulnerability factor to anxiety and trauma-related disorders.


Asunto(s)
Aprendizaje/fisiología , Consolidación de la Memoria/fisiología , Neuronas/fisiología , Núcleos Septales/fisiología , Animales , Miedo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Somatostatina/metabolismo
3.
J Neurosci ; 40(23): 4551-4564, 2020 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-32350040

RESUMEN

Forming effective responses to threatening stimuli requires the adequate and coordinated emergence of stress-related internal states. Such ability depends on early-life experiences and, in connection, the adequate formation of neuromodulatory systems, particularly serotonergic signaling. Here, we assess the serotonergic background of experience-dependent behavioral responsiveness using male and female zebrafish (Danio rerio). For the first time, we have characterized a period during behavioral metamorphosis in which zebrafish are highly reactive to their environment. Absence of social stimuli during this phase established by isolated rearing fundamentally altered the behavioral phenotype of postmetamorphic zebrafish in a challenge-specific manner, partially due to reduced responsiveness and an inability to develop stress-associated arousal state. In line with this, isolation differentially affected whole-brain serotonergic signaling in resting and stress-induced conditions, an effect that was localized in the dorsal pallium and was negatively associated with responsiveness. Administration of the serotonin receptor 1A partial agonist buspirone prevented the isolation-induced serotonin response to novelty in the level of the whole brain and the forebrain as well, without affecting catecholamine levels, and rescued stress-induced arousal along with challenge-induced behaviors, which together indicates functional connection between these changes. In summary, there is a consistent negative association between behavioral responsiveness and serotonergic signaling in zebrafish, which is well recognizable through the modifying effects of developmental perturbation and pharmacological manipulations as well. Our results imply a conserved serotonergic mechanism that context-dependently modulates environmental reactivity and is highly sensitive to experiences acquired during a specific early-life time window, a phenomenon that was previously only suggested in mammals.SIGNIFICANCE STATEMENT The ability to respond to challenges is a fundamental factor in survival. We show that zebrafish that lack appropriate social stimuli in a sensitive developmental period show exacerbated alertness in nonstressful conditions while failing to react adequately to stressors. This shift is reflected inversely by central serotonergic signaling, a system that is implicated in numerous mental disorders in humans. Serotonergic changes in brain regions modulating responsivity and behavioral impairment were both prevented by the pharmacological blockade of serotonergic function. These results imply a serotonergic mechanism in zebrafish that transmits early-life experiences to the later phenotype by shaping stress-dependent behavioral reactivity, a phenomenon that was previously only suggested in mammals. Zebrafish provide new insights into early-life-dependent neuromodulation of behavioral stress-responses.


Asunto(s)
Nivel de Alerta/fisiología , Reacción de Prevención/fisiología , Conducta Animal/fisiología , Receptor de Serotonina 5-HT1A/fisiología , Agonistas de Receptores de Serotonina/farmacología , Serotonina/fisiología , Animales , Nivel de Alerta/efectos de los fármacos , Reacción de Prevención/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Femenino , Masculino , Aislamiento Social/psicología , Pez Cebra
4.
Psychiatr Hung ; 35(1): 30-36, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31854320

RESUMEN

BACKGROUND: Attention-deficit/hyperactivity disorder (ADHD) is the most common neurodevelopmental disorder. According to literature data, perinatal adversities might be associated with the occurrence of ADHD, but the results are inconclusive at the moment. The aim of the present study is to describe perinatal adversities in children with ADHD. METHODS: The data of children in County Fejér Pedagogical Service in three consecutive school years (2012-2015) was analyzed. In the present sample, 219 children with special education needs were diagnosed with ADHD (age: 9.0 years, SD: 3.1 years; 36 girls). The diagnosis was based on ICD-10 criteria. The present analysis is restricted to the presence of preterm birth, complicated/prolonged labor, intrauterine hypoxia and birth asphyxia in children with ADHD from different areas (central, urban or rural) of the county. Logistic regression analysis was performed to assess the effects of gender, age was used as a covariate. RESULTS: Within this sample, the presence of complicated/prolonged labor was significantly lower (OR: 0.378, p<0.039) in girls compared to boys. The other studied variables did not show significant differences regarding gender. Intrauterine hypoxia was registered in 28 cases, while birth asphyxia was registered in 15 cases. The presence of preterm birth was 8.7%, and showed distinctive differences according to living area (central: 3.6% vs rural: 11.3%). CONCLUSIONS: Our results indicate the importance of registering perinatal complications and long term follow up of these children in the direction of neurodevelopmental disorders, however a limitation of the present study is the lack of a control group.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/etiología , Complicaciones del Embarazo , Efectos Tardíos de la Exposición Prenatal , Trastorno por Déficit de Atención con Hiperactividad/diagnóstico , Niño , Femenino , Humanos , Recién Nacido , Masculino , Perinatología , Embarazo
5.
Pediatr Res ; 85(2): 225-233, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30341412

RESUMEN

Social adversities experienced in childhood can have a profound impact on the developing brain, leading to the emergence of psychopathologies in adulthood. Despite the burden this places on both the individual and society, the neurobiological aspects mediating this transition remain unclear. Recent advances in preclinical and clinical research have begun examining neuroplasticity-the nervous system's ability to form adaptive changes in response to new experience-in the context of early-life vulnerability to social adversities and plasticity-related alterations following such traumatic events. A key mediator of plasticity-related molecular processes is the brain-derived neurotrophic factor (BDNF), which has also been implicated in various psychiatric disorders related to childhood social adversities. Preclinical and clinical data suggest early-life social adversities (ELSA) might be associated with accelerated maturation of social network circuitry, a possible ontogenic adaptation to the adverse environment. Neural plasticity decreases by adulthood, lessening the efficacy of treatment in ELSA-related psychiatric disorders. However, literature data suggest that by increasing BDNF/TrkB signalling through antidepressant treatment a juvenile-like plasticity state can be induced, which allows for reorganization of the social circuitry when guided by psychotherapy and surrounded by a safe and positive environment.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/fisiología , Conducta Social , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Preescolar , Humanos , Lactante , Acontecimientos que Cambian la Vida , Plasticidad Neuronal , Polimorfismo de Nucleótido Simple , Transducción de Señal
6.
Dev Psychobiol ; 56(1): 49-57, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23168609

RESUMEN

As previously shown, rats isolated from weaning develop abnormal social and aggressive behavior characterized by biting attacks targeting vulnerable body parts of opponents, reduced attack signaling, and increased defensive behavior despite increased attack counts. Here we studied whether this form of violent aggression could be reversed by resocialization in adulthood. During the first weak of resocialization, isolation-reared rats showed multiple social deficits including increased defensiveness and decreased huddling during sleep. Deficits were markedly attenuated in the second and third weeks. Despite improved social functioning in groups, isolated rats readily showed abnormal features of aggression in a resident-intruder test performed after the 3-week-long resocialization. Thus, post-weaning social isolation-induced deficits in prosocial behavior were eliminated by resocialization during adulthood, but abnormal aggression was resilient to this treatment. Findings are compared to those obtained in humans who suffered early social maltreatment, and who also show social deficits and dysfunctional aggression in adulthood.


Asunto(s)
Agresión/fisiología , Conducta Animal/fisiología , Conducta Social , Aislamiento Social , Animales , Conducta Exploratoria/fisiología , Masculino , Ratas , Ratas Wistar , Destete
7.
Neurobiol Stress ; 32: 100662, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39183773

RESUMEN

Traumatic experiences result in the development of posttraumatic stress disorder (PTSD) in 10-25% of exposed individuals. While human clinical studies suggest that susceptibility is potentially linked to endocannabinoid (eCB) signaling, neurobiological PTSD susceptibility factors are poorly understood. Employing a rat model of contextual conditioned fear, we characterized distinct resilient and susceptible subpopulations based on lasting generalized fear, a core symptom of PTSD. In these groups, we assessed i.) eCB levels by mass spectrometry and ii.) expression variations of eCB system- and iii.) neuroplasticity-related genes by real-time quantitative PCR in the circuitry relevant in trauma-induced changes. Furthermore, employing unsupervised and semi-supervised machine learning based statistical analytical models, we assessed iv.) gene expression patterns with the most robust predictive power regarding PTSD susceptibility. According to our findings, in our model, generalized fear responses occurred with sufficient variability to characterize distinct resilient and susceptible subpopulations. Resilient subjects showed elevated prelimbic and lower ventral hippocampal levels of eCB 2-arachidonoyl-glycerol (2-AG) compared to resilient and non-shocked control subjects. Ventral hippocampal 2-AG content positively correlated with the strength of fear generalization. Furthermore, susceptibility was associated with i.) prefrontal, hippocampal and amygdalar neuronal hypoactivity, ii.) marked decrease in the expression of genes of transcription factors modulating neuroplasticity and iii.) an altered expression pattern of eCB-related genes, including enzymes involved in eCB metabolism. Unsupervised and semi-supervised statistical approaches highlighted that hippocampal gene expression patterns possess strong predictive power regarding susceptibility. Taken together, the marked eCB and neuroplasticity changes in susceptible individuals associated with abnormal activity patterns in the fear circuitry possibly contribute to context coding deficits, resulting in generalized fear.

8.
Horm Behav ; 63(5): 752-8, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23578952

RESUMEN

The hypothalamus-pituitary-adrenal-axis is strongly controlled by the endocannabinoid system. The specific impact of enhanced 2-arachidonoylglycerol signaling on corticosterone plasma levels, however, was not investigated so far. Here we studied the effects of the recently developed monoacylglycerol lipase inhibitor JZL184 on basal and stress-induced corticosterone levels in male CD1 mice, and found that this compound dramatically increased basal levels without affecting stress responses. Since acute changes in corticosterone levels can affect behavior, JZL184 was administered concurrently with the corticosterone synthesis inhibitor metyrapone, to investigate whether the previously shown behavioral effects of JZL184 are dependent on corticosterone. We found that in the elevated plus-maze, the effects of JZL184 on "classical" anxiety-related measures were abolished by corticosterone synthesis blockade. By contrast, effects on the "ethological" measures of anxiety (i.e. risk assessment) were not affected by metyrapone. In the open-field, the locomotion-enhancing effects of the compound were not changed either. These findings show that monoacylglycerol lipase inhibition dramatically increases basal levels of corticosterone. This endocrine effect partly affects the anxiolytic, but not the locomotion-enhancing effects of monoacylglycerol lipase blockade.


Asunto(s)
Ansiedad/enzimología , Benzodioxoles/farmacología , Corticosterona/sangre , Inhibidores Enzimáticos/farmacología , Monoacilglicerol Lipasas/antagonistas & inhibidores , Actividad Motora/efectos de los fármacos , Piperidinas/farmacología , Animales , Ansiedad/sangre , Masculino , Metirapona/farmacología , Ratones , Actividad Motora/fisiología
9.
Neurobiol Stress ; 25: 100546, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37323648

RESUMEN

Adverse social experiences during childhood increase the risk of developing aggression-related psychopathologies. The prefrontal cortex (PFC) is a key regulator of social behavior, where experience-dependent network development is tied to the maturation of parvalbumin-positive (PV+) interneurons. Maltreatment in childhood could impact PFC development and lead to disturbances in social behavior during later life. However, our knowledge regarding the impact of early-life social stress on PFC operation and PV+ cell function is still scarce. Here, we used post-weaning social isolation (PWSI) to model early-life social neglect in mice and to study the associated neuronal changes in the PFC, additionally distinguishing between the two main subpopulations of PV+ interneurons, i.e. those without or those enwrapped by perineuronal nets (PNN). For the first time to such detailed extent in mice, we show that PWSI induced disturbances in social behavior, including abnormal aggression, excessive vigilance and fragmented behavioral organization. PWSI mice showed altered resting-state and fighting-induced co-activation patterns between orbitofrontal and medial PFC (mPFC) subregions, with a particularly highly elevated activity in the mPFC. Surprisingly, aggressive interaction was associated with a higher recruitment of mPFC PV+ neurons that were surrounded by PNN in PWSI mice that seemed to mediate the emergence of social deficits. PWSI did not affect the number of PV+ neurons and PNN density, but enhanced PV and PNN intensity as well as cortical and subcortical glutamatergic drive onto mPFC PV+ neurons. Our results suggest that the increased excitatory input of PV+ cells could emerge as a compensatory mechanism for the PV+ neuron-mediated impaired inhibition of mPFC layer 5 pyramidal neurons, since we found lower numbers of GABAergic PV+ puncta on the perisomatic region of these cells. In conclusion, PWSI leads to altered PV-PNN activity and impaired excitatory/inhibitory balance in the mPFC, which possibly contributes to social behavioral disruptions seen in PWSI mice. Our data advances our understanding on how early-life social stress can impact the maturing PFC and lead to the development of social abnormalities in adulthood.

10.
Horm Behav ; 60(1): 28-36, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21316368

RESUMEN

We showed earlier that social isolation from weaning (a paradigm frequently used to model social neglect in children) induces abnormal forms of attack in rats, and assumed that these are associated with hyperarousal. To investigate this hypothesis, we deprived rats of social contacts from weaning and studied their behavior, glucocorticoid and autonomic stress responses in the resident-intruder paradigm at the age of 82 days. Social isolation resulted in abnormal attack patterns characterized by attacks on vulnerable targets, deficient social communication and increased defensive behaviors (defensive upright, flight, freezing). During aggressive encounters, socially deprived rats rapidly switched from one behavior to another, i.e. showed an increased number of behavioral transitions as compared to controls. We tentatively term this behavioral feature "behavioral fragmentation" and considered it a form of behavioral arousal. Basal levels of plasma corticosterone regularly assessed by radioimmunoassay between 27 and 78 days of age were not affected. In contrast, aggression-induced glucocorticoid responses were approximately doubled by socially isolation. Diurnal oscillations in heart rate assessed by in vivo biotelemetry were not affected by social isolation. In contrast, the aggression-induced increase in heart rate was higher in socially isolated than in socially housed rats. Thus, post-weaning social isolation induced abnormal forms of aggression that developed on the background of increased behavioral, endocrine and autonomic arousal. We suggest that this paradigm may be used to model aggression-related psychopathologies associated with hyperarousal, particularly those that are triggered by adverse rearing conditions.


Asunto(s)
Agresión/fisiología , Sistema Nervioso Autónomo/fisiología , Corticosterona/fisiología , Aislamiento Social/psicología , Estrés Psicológico/fisiopatología , Destete , Agresión/psicología , Animales , Conducta Animal/fisiología , Corticosterona/sangre , Frecuencia Cardíaca/fisiología , Masculino , Ratas , Ratas Wistar , Estrés Psicológico/sangre , Estrés Psicológico/psicología
11.
Behav Pharmacol ; 22(2): 113-21, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21285873

RESUMEN

N-methyl-D-asparate (NMDA)-mediated glutamatergic neurotransmission is strongly involved in the development of trauma-induced behavioral dysfunctions, and indirect evidence suggests that NR2B subunit-expressing NMDA receptors are primarily involved in this process. Earlier studies showed that NR2B blockers inhibit the acquisition of conditioned fear, a frequently used model of post-traumatic stress disorder, but their effects on the expression of conditioned fear was poorly studied. We investigated here the effects of the selective serotonin reuptake blocker, fluoxetine, the NMDA blocker, MK-801, and the NR2B subunit blocker, Ro25-6981 on the expression of conditioned fear. Rats received 10 foot shocks administered over 5 min and were tested 24 h later in the shocking context. Treatments were administered 1 h before testing. Shocks dramatically increased freezing and reduced exploration. MK-801 and Ro25-6981 significantly ameliorated both changes. The effects of fluoxetine were less pronounced. In the open field, MK-801 increased locomotion, ataxia, and stereotypy (effects typical of NMDA blockade). Neither fluoxetine nor Ro25-6981 affected locomotion in the open field. Thus, the NR2B-specific NMDA blockade preserved the beneficial effects of general NMDA antagonists on the expression of conditioned fear but did not produce the locomotor side-effects typical of the latter. These findings warrant further studies on the effects of NR2B antagonists in models of post-traumatic stress disorder.


Asunto(s)
Maleato de Dizocilpina/farmacología , Fluoxetina/farmacología , Fenoles/farmacología , Piperidinas/farmacología , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Animales , Ataxia/inducido químicamente , Modelos Animales de Enfermedad , Maleato de Dizocilpina/efectos adversos , Electrochoque , Antagonistas de Aminoácidos Excitadores/efectos adversos , Antagonistas de Aminoácidos Excitadores/farmacología , Conducta Exploratoria/efectos de los fármacos , Miedo/efectos de los fármacos , Fluoxetina/efectos adversos , Reacción Cataléptica de Congelación/efectos de los fármacos , Locomoción/efectos de los fármacos , Masculino , Fenoles/efectos adversos , Piperidinas/efectos adversos , Ratas , Ratas Wistar , Inhibidores Selectivos de la Recaptación de Serotonina/efectos adversos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Trastorno de Movimiento Estereotipado/inducido químicamente , Trastornos por Estrés Postraumático/tratamiento farmacológico , Trastornos por Estrés Postraumático/fisiopatología
12.
Endocr J ; 58(5): 395-407, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21505269

RESUMEN

Forced swim test (FST) is a widely used test for antidepressant development. Depression is a stress related disease, as hormones of the stress-axis can modify mood. However it is not clear, how the appearance of depressive-like behavior (floating) in FST is connected with changes in the stress-hormone levels. We hypothesized, that different manipulations would alter the behavior through changes in stress-hormone levels. First the effect of environmental alterations was studied. Increasing water-temperature enhanced floating time together with a decrease in adrenocorticotropin levels. During the dark phase of the day rats spent more time with floating independently from the actual lighting. Neither the phase nor the actual lighting had significant effect on adrenocorticotropin concentrations with higher corticosterone levels during the dark phase. At greater water depth rats float less but the size of animals had no effect. Water depth did not influence adrenocorticotropin and corticosterone responses, but the size of the rats significantly affected both factors. Secondly, administration of imipramine reduced floating and adrenocorticotropin level without affecting corticosterone. Despite the known connection between depression and stress we did not find a correlation between floating behavior and hormone levels. As an alternative mechanism imipramine-induced heart rate and core body temperature decrease was found by telemetric approach. This study is the first summary in rats examining the effect of wide range of environmental alterations during FST. It seems likely that both brain monoamines and stress-axis take part in the development of depression, but these pathways are regulated independently.


Asunto(s)
Hormona Adrenocorticotrópica/sangre , Conducta Animal/fisiología , Corticosterona/sangre , Depresión/fisiopatología , Estrés Psicológico/fisiopatología , Adaptación Psicológica/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Temperatura Corporal/efectos de los fármacos , Peso Corporal , Oscuridad , Frecuencia Cardíaca/efectos de los fármacos , Calor , Imipramina/farmacología , Masculino , Ratas , Natación
13.
Neurobiol Stress ; 15: 100415, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34765699

RESUMEN

Anxiety and trauma-related disorders are characterized by significant alterations in threat detection, resulting in inadequate fear responses evoked by weak threats or safety stimuli. Recent research pointed out the important role of the bed nucleus of stria terminalis (BNST) in threat anticipation and fear modulation under ambiguous threats, hence, exaggerated fear may be traced back to altered BNST function. To test this hypothesis, we chemogenetically inhibited specific BNST neuronal populations (corticotropin-releasing hormone - BNSTCRH and somatostatin - BNSTSST expressing neurons) in a predator odor-evoked innate fear paradigm. The rationale for this paradigm was threefold: (1) predatory cues are particularly strong danger signals for all vertebrate species evoking defensive responses on the flight-avoidance-freezing dimension (conservative mechanisms), (2) predator odor can be presented in a scalable manner (from weak to strong), and (3) higher-order processing of olfactory information including predatory odor stimuli is integrated by the BNST. Accordingly, we exposed adult male mice to low and high predatory threats presented by means of cat urine, or low- and high-dose of 2-methyl-2-thiazoline (2MT), a synthetic derivate of a fox anogenital product, which evoked low and high fear response, respectively. Then, we tested the impact of chemogenetic inhibition of BNSTCRH and BNSTSST neurons on innate fear responses using crh- and sst-ires-cre mouse lines. We observed that BNSTSST inhibition was effective only under low threat conditions, resulting in reduced avoidance and increased exploration of the odor source. In contrast, BNSTCRH inhibition had no impact on 2MT-evoked responses, but enhanced fear responses to cat odor, representing an even weaker threat stimulus. These findings support the notion that BNST is recruited by uncertain or remote, potential threats, and CRH and SST neurons orchestrate innate fear responses in complementary ways.

14.
Eur J Neurosci ; 32(10): 1744-53, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21039962

RESUMEN

Callous-unemotional violence associated with antisocial personality disorder is often called 'predatory' because it involves restricted intention signaling and low emotional/physiological arousal, including decreased glucocorticoid production. This epithet may be a mere metaphor, but may also cover a structural similarity at the level of the hypothalamus where the control of affective and predatory aggression diverges. We investigated this hypothesis in a laboratory model where glucocorticoid production is chronically limited by adrenalectomy with glucocorticoid replacement (ADXr). This procedure was proposed to model important aspects of antisocial violence. Sham and ADXr rats were submitted to resident/intruder conflicts, and the resulting neuronal activation patterns were investigated by c-Fos immunocytochemistry. In line with earlier findings, the share of attacks aimed at vulnerable targets (head, throat and belly) was dramatically increased by ADXr, while intention signaling by offensive threats was restricted. Aggressive encounters activated the mediobasal hypothalamus, a region involved in intra-specific aggression, but sham and ADXr rats did not differ in this respect. In contrast, the activation of the lateral hypothalamus that is tightly involved in predatory aggression was markedly larger in ADXr rats; moreover, c-Fos counts correlated positively with the share of vulnerable attacks and negatively with social signaling. Glucocorticoid deficiency increased c-Fos activation in the central amygdala, a region also involved in predatory aggression. In addition, activation patterns in the periaqueductal gray - involved in autonomic control - also resembled those seen in predatory aggression. These findings suggest that antisocial and predatory aggression are not only similar but are controlled by overlapping neural mechanisms.


Asunto(s)
Agresión/fisiología , Conducta Animal/fisiología , Glucocorticoides/deficiencia , Conducta Predatoria/fisiología , Adrenalectomía , Amígdala del Cerebelo/anatomía & histología , Amígdala del Cerebelo/metabolismo , Animales , Trastorno de Personalidad Antisocial/fisiopatología , Glucocorticoides/administración & dosificación , Humanos , Hipotálamo/anatomía & histología , Hipotálamo/metabolismo , Masculino , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Wistar , Conducta Social
15.
Front Neuroendocrinol ; 29(2): 273-91, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18054070

RESUMEN

Mounting evidence suggests that--beyond the well-known genomic effects--glucocorticoids affect cell function via non-genomic mechanisms. Such mechanisms operate in many major systems and organs including the cardiovascular, immune, endocrine and nervous systems, smooth and skeletal muscles, liver, and fat cells. Non-genomic effects are exerted by direct actions on membrane lipids (affecting membrane fluidity), membrane proteins (e.g. ion channels and neurotransmitter receptors), and cytoplasmic proteins (e.g. MAPKs, phospholipases, protein kinases, etc.). These actions are mediated by the glucocorticoids per se or by the proteins dissociated from the liganded glucocorticoid receptor complex. The MR and GR also activate non-genomic mechanisms in certain cases. Some effects of glucocorticoids are shared by a variety of steroids, whereas others are more selective. Moreover, "ultra-selective" effects-mediated by certain glucocorticoids only-were also shown. Disparate findings suggest that non-genomic mechanisms also show "demand-specificity", i.e. require the coincidence of two or more processes. Some of the non-genomic mechanisms activated by glucocorticoids are therapeutically relevant; moreover, the "non-genomic specificity" of certain glucocorticoids raises the possibility of therapeutic applications. Despite the large body of evidence, however, the non-genomic mechanisms of glucocorticoids are still poorly understood. Criteria for differentiating genomic and non-genomic mechanisms are often loosely applied; interactions between various mechanisms are unknown, and non-genomic mechanism-specific pharmacological (potentially therapeutic) agents are lacking. Nevertheless, the discovery of non-genomic mechanisms is a major breakthrough in stress research, and further insights into these mechanisms may open novel approaches for the therapy of various diseases.


Asunto(s)
Glucocorticoides/fisiología , Animales , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Sistema Nervioso Central/fisiología , Citoplasma/fisiología , Genoma , Glucocorticoides/farmacología , Humanos , Ligandos , Lípidos de la Membrana/fisiología , Proteínas de la Membrana/fisiología , Modelos Biológicos , Receptores de Superficie Celular/fisiología , Receptores de Glucocorticoides/fisiología , Receptores de Mineralocorticoides/fisiología , Sensibilidad y Especificidad , Conducta Sexual Animal/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Factores de Tiempo
16.
Behav Pharmacol ; 20(3): 265-72, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19421027

RESUMEN

Neuroanatomical findings revealed that CB1 cannabinoid and 5-HT3 receptors are coexpressed by a subtype of gamma-aminobutyric acid (GABA)ergic interneurons in the prefrontal cortex, hippocampus, and basolateral amygdala, three brain regions that are crucial for the control of anxiety. In these regions, serotonergic inputs increase GABA release through 5-HT3 receptors, the phenomenon being retrogradely controlled by cannabinoid neurotransmission. This suggests a functional interaction between 5-HT3 neurotransmission and CB1 signaling. In a first attempt to investigate the behavioral relevance of these interactions, we studied the effects of the selective 5-HT3 agonist 1-(m-chlorophenyl)-biguanide (mCPBG), on plus-maze behavior in NMRI, CD1 wild type, and CB1-KO mice. The genetic disruption of CB1 receptors consistently increased anxiety. This effect was significantly decreased by the 5-HT3 agonist, mCPBG. The dose-response curve was bell-shaped. Surprisingly, mCPBG did not affect the behavior of CD1 wild type and NMRI mice. We hypothesize that in the aforementioned regions, 5-HT3 activation decreases anxiety by promoting GABA release, but this effect is dampened by CB1 signaling. The disruption of CB1 receptors in CB1-KOs released GABA neurons from retrograde inhibition and made the effects of 5-HT3 stimulation conspicuous. Altogether, our findings reveal a functional interaction between 5-HT3 neurotransmission and CB1 signaling. Besides this interaction being an interesting aspect of anxiety control, it may also explain the notoriously inconsistent effects of 5-HT3 ligands on anxiety. If 5-HT3 neurotransmission and CB1 signaling interact, the anxiety-related effects of 5-HT3 ligands may depend on species, strain, and situation-related differences in both 5-HT3 and CB1 receptor expression and function.


Asunto(s)
Ansiedad/metabolismo , Receptor Cannabinoide CB1/genética , Receptores de Serotonina 5-HT3/metabolismo , Transmisión Sináptica/fisiología , Animales , Ansiedad/genética , Ansiedad/psicología , Conducta Animal/efectos de los fármacos , Biguanidas/farmacología , Moduladores de Receptores de Cannabinoides/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Noqueados , Receptor Cannabinoide CB1/fisiología , Agonistas del Receptor de Serotonina 5-HT3 , Especificidad de la Especie , Transmisión Sináptica/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo
17.
J Pharmacol Exp Ther ; 327(2): 482-90, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18725543

RESUMEN

Emerging evidence suggests that the rewarding, abuse-related effects of nicotine are modulated by the endocannabinoid system of the brain. For example, pharmacological blockade or genetic deletion of cannabinoid CB(1) receptors can reduce or eliminate many abuse-related behavioral and neurochemical effects of nicotine. Furthermore, doses of Delta(9)-tetrahydrocannabinol and nicotine that are ineffective when given alone can induce conditioned place preference when given together. These previous studies have used systemically administered CB(1) receptor agonists and antagonists and gene deletion techniques, which affect cannabinoid CB(1) receptors throughout the brain. A more functionally selective way to alter endocannabinoid activity is to inhibit fatty acid amide hydrolase (FAAH), thereby magnifying and prolonging the effects of the endocannabinoid anandamide only when and where it is synthesized and released on demand. Here, we combined behavioral and neurochemical approaches to evaluate whether the FAAH inhibitor URB597 (cyclohexyl carbamic acid 3'-carbamoyl-3-yl ester) could alter the abuse-related effects of nicotine in rats. We found that URB597, at a dose (0.3 mg/kg) that had no behavioral effects by itself, prevented development of nicotine-induced conditioned place preference (CPP) and acquisition of nicotine self-administration. URB597 also reduced nicotine-induced reinstatement in both CPP and self-administration models of relapse. Furthermore, in vivo microdialysis showed that URB597 reduced nicotine-induced dopamine elevations in the nucleus accumbens shell, the terminal area of the brain's mesolimbic reward system. These findings suggest that FAAH inhibition can counteract the addictive properties of nicotine and that FAAH may serve as a new target for development of medications for treatment of tobacco dependence.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Ácidos Araquidónicos/metabolismo , Benzamidas/farmacología , Carbamatos/farmacología , Condicionamiento Psicológico/efectos de los fármacos , Dopamina/análisis , Nicotina/farmacología , Núcleo Accumbens/efectos de los fármacos , Alcamidas Poliinsaturadas/metabolismo , Tabaquismo/tratamiento farmacológico , Amidohidrolasas/fisiología , Animales , Endocannabinoides , Hidrólisis , Masculino , Actividad Motora/efectos de los fármacos , Núcleo Accumbens/química , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Recompensa , Autoadministración , Tabaquismo/enzimología
18.
Neuropharmacology ; 54(1): 129-40, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17904589

RESUMEN

Converging evidence suggests that the endocannabinoid system is an important constituent of neuronal substrates involved in brain reward processes and emotional responses to stress. Here, we evaluated motivational effects of intravenously administered anandamide, an endogenous ligand for cannabinoid CB1-receptors, in Sprague-Dawley rats, using a place-conditioning procedure in which drugs abused by humans generally produce conditioned place preferences (reward). Anandamide (0.03-3 mg/kg intravenous) produced neither conditioned place preferences nor aversions. However, when rats were pre-treated with the fatty acid amide hydrolase (FAAH) inhibitor URB597 (cyclohexyl carbamic acid 3'-carbamoyl-3-yl ester; 0.3 mg/kg intraperitoneal), which blocks anandamide's metabolic degradation, anandamide produced dose-related conditioned place aversions. In contrast, URB597 alone showed no motivational effects. Like URB597 plus anandamide, the synthetic CB1-receptor ligand WIN 55,212-2 (50-300 microg/kg, intravenous) produced dose-related conditioned place aversions. When anxiety-related effects of anandamide and URB597 were evaluated in a light/dark box, both a low anandamide dose (0.3 mg/kg) and URB597 (0.1 and 0.3 mg/kg) produced anxiolytic effects when given alone, but produced anxiogenic effects when combined. A higher dose of anandamide (3 mg/kg) produced anxiogenic effects and depressed locomotor activity when given alone and these effects were potentiated after URB597 treatment. Finally, anxiogenic effects of anandamide plus URB597 and development of place aversions with URB597 plus anandamide were prevented by the CB1-receptor antagonist AM251 (3 mg/kg intraperitoneal). Thus, additive interactions between the effects of anandamide on brain reward processes and on anxiety may account for its aversive effects when intravenously administered during FAAH inhibition with URB597.


Asunto(s)
Amidohidrolasas/metabolismo , Ansiedad/enzimología , Ácidos Araquidónicos/metabolismo , Motivación , Alcamidas Poliinsaturadas/metabolismo , Analgésicos/farmacología , Análisis de Varianza , Animales , Ansiedad/tratamiento farmacológico , Ácidos Araquidónicos/farmacología , Reacción de Prevención/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Benzamidas/farmacología , Benzoxazinas/farmacología , Carbamatos/farmacología , Condicionamiento Operante/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Endocannabinoides , Inhibidores Enzimáticos/farmacología , Masculino , Morfolinas/farmacología , Actividad Motora/efectos de los fármacos , Naftalenos/farmacología , Alcamidas Poliinsaturadas/farmacología , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
19.
Psychoneuroendocrinology ; 33(9): 1198-210, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18656313

RESUMEN

Neuronal plasticity within the amygdala mediates many behavioral effects of traumatic experience, and this brain region also controls various aspects of social behavior. However, the specific involvement of the amygdala in trauma-induced social deficits has never been systematically investigated. We exposed rats to a single series of electric foot-shocks--a frequently used model of trauma--and studied their behavior in the social avoidance and psychosocial stimulation tests (non-contact versions of the social interaction test) at different time intervals. Social interaction-induced neuronal activation patterns were studied in the prefrontal cortex (orbitofrontal and medial), amygdala (central, medial, and basolateral), dorsal raphe and locus coeruleus. Shock exposure markedly inhibited social behavior in both tests. The effect lasted at least 4 weeks, and amplified over time. As shown by c-Fos immunocytochemistry, social interactions activated all the investigated brain areas. Traumatic experience exacerbated this activation in the central and basolateral amygdala, but not in other regions. The tight correlation between the social deficit and amygdala activation patterns suggest that the two phenomena were associated. A real-time PCR study showed that CRF mRNA expression in the amygdala was temporarily reduced 14, but not 1 and 28 days after shock exposure. In contrast, amygdalar NK1 receptor mRNA expression increased throughout. Thus, the trauma-induced social deficits appear to be associated with, and possibly caused by, plastic changes in fear-related amygdala subdivisions.


Asunto(s)
Amígdala del Cerebelo/fisiología , Reacción de Prevención/fisiología , Corteza Prefrontal/metabolismo , Conducta Social , Estrés Psicológico/fisiopatología , Análisis de Varianza , Animales , Hormona Liberadora de Corticotropina/genética , Hormona Liberadora de Corticotropina/metabolismo , Regulación de la Expresión Génica/fisiología , Masculino , Norepinefrina/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/análisis , Ratas , Ratas Wistar , Receptores de Neuroquinina-1/genética , Receptores de Neuroquinina-1/metabolismo , Serotonina/metabolismo , Estadísticas no Paramétricas
20.
Behav Neurosci ; 122(4): 849-54, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18729638

RESUMEN

Disturbed social relations during childhood (e.g., social neglect) often lead to aggression-related psychopathologies in adulthood. Social isolation also increased aggressiveness in laboratory animals. Here the authors show in rats, that social isolation from weaning not only increases the level of aggressiveness, but results in abnormal attack patterns and deficits in social communication. In socially deprived rats, the share of attacks aimed at vulnerable body parts of opponents (head, throat, and belly) dramatically increased and the attack/threat ratio was shifted toward attacks, suggesting a decrease in intention signaling. Moreover, a Multiple Regression Analysis showed that the nonassociation of attacks with offensive threats predicted the occurrence of vulnerable attacks with 81.1% accuracy. The authors suggest that the social deprivation-induced abnormal aggression models the aggression-related problems resulting from early social neglect in humans, and studies on its brain mechanisms may increase our understanding of the mechanisms underlying psychopathologies resulting from early social problems.


Asunto(s)
Conducta Agonística/fisiología , Relaciones Interpersonales , Aislamiento Social , Análisis de Varianza , Animales , Animales Recién Nacidos , Conducta Animal , Conducta Exploratoria/fisiología , Masculino , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda