Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Mol Psychiatry ; 28(11): 4719-4728, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37674017

RESUMEN

In the wild, animals face a highly variable world full of predators. Most predator attacks are unsuccessful, and the prey survives. According to the conventional perspective, the fear responses elicited by predators are acute and transient in nature. However, the long-term, non-lethal effects of predator exposure on prey behavioral stress sequelae, such as anxiety and post-traumatic symptoms, remain poorly understood. Most experiments on animal models of anxiety-related behavior or post-traumatic stress disorder have been carried out using commercial strains of rats and mice. A fundamental question is whether laboratory rodents appropriately express the behavioral responses of wild species in their natural environment; in other words, whether behavioral responses to stress observed in the laboratory can be generalized to natural behavior. To further elucidate the relative contributions of the natural selection pressures influences, this study investigated the bio-behavioral and morphological effects of auditory predator cues (owl territorial calls) in males and females of three wild rodent species in a laboratory set-up: Acomys cahirinus; Gerbillus henleyi; and Gerbillus gerbillus. Our results indicate that owl territorial calls elicited not only "fight or flight" behavioral responses but caused PTSD-like behavioral responses in wild rodents that have never encountered owls in nature and could cause, in some individuals, enduring physiological and morphological responses that parallel those seen in laboratory rodents or traumatized people. In all rodent species, the PTSD phenotype was characterized by a blunting of fecal cortisol metabolite response early after exposure and by a lower hypothalamic orexin-A level and lower total dendritic length and number in the dentate gyrus granule cells eight days after predator exposure. Phenotypically, this refers to a significant functional impairment that could affect reproduction and survival and thus fitness and population dynamics.


Asunto(s)
Trastornos por Estrés Postraumático , Humanos , Masculino , Femenino , Ratas , Animales , Trastornos por Estrés Postraumático/metabolismo , Roedores , Ansiedad/etiología , Señales (Psicología) , Neuronas/metabolismo , Modelos Animales de Enfermedad
2.
Neuropsychopharmacol Rep ; 43(1): 2-11, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36622038

RESUMEN

Sleep figures in numerous ancient texts, for example, Epic of Gilgamesh, and has been a focus for countless mystical and philosophical texts. Even in the present century, sleep remains one of the most complex behaviors whose function still remains to be further explored. Current hypotheses suggest that among other functions, sleep contributes to memory processes. Memory is a core topic of study in post-traumatic stress disorder (PTSD) and other stress-related phenomena. It is widely accepted that sleep plays a major role in the consolidation of newly encoded hippocampus-dependent memories to pre-existing knowledge networks. Conversely, sleep deprivation disrupts consolidation and impairs memory retrieval. Along this line, sleep deprivation following a potentially traumatic event may interfere with the consolidation of event-related memories and, thereby, may reduce long-term post-traumatic stress-related symptoms. This review consolidates clinical and animal studies on the relationships between sleep, sleep deprivation, memory processes, and trauma exposure while introducing new contemporary insights into an ancient African tribal ritual (Àìsùn Oku) and Japanese ceremony ritual (Tsuya). We propose that these findings, focusing specifically on the effects of sleep deprivation in the immediate aftermath of traumatic events, may be explored as a possible therapeutic measure. Along with a summary of the field questions on whether sleep is performed "to remember" or "to forget" we lay the rationale for using sleep deprivation as a clinical tool. A tool that may partially prevent the long-term persistence of these traumatic events' memory and thereby, at least partly, attenuating the development of PTSD.


Asunto(s)
Trastornos por Estrés Postraumático , Humanos , Animales , Trastornos por Estrés Postraumático/prevención & control , Privación de Sueño , Conducta Ceremonial , Pueblos del Este de Asia , Sueño
3.
Transl Psychiatry ; 12(1): 181, 2022 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-35504866

RESUMEN

MDMA (3,4-methylenedioxymethamphetamine), a synthetic ring-substituted amphetamine, combined with psychotherapy has demonstrated efficacy for the treatment of chronic posttraumatic stress disorder (PTSD) patients. This controlled prospective study aimed to assess the bio-behavioral underpinnings of MDMA in a translational model of PTSD. Rats exposed to predator-scent stress (PSS) were subjected to a trauma-cue at day 7 shortly after single-dose MDMA injection (5 mg/kg). The elevated plus maze and acoustic startle response tests were assessed on day 14 and served for classification into behavioral response groups. Freezing response to a further trauma-reminder was assessed on Day 15. The morphological characteristics of the dentate gyrus (DG) and basolateral amygdala (BLA) were subsequently examined. Hypothalamic-pituitary-adrenal axis and 5-hydroxytryptamine involvement were evaluated using: (1) corticosterone measurements at 2 h and 4 h after MDMA treatment, (2) Lewis strain rats with blunted HPA-response and (3) pharmacological receptor-blockade. MDMA treatment was effective in attenuating stress behavioral responses only when paired with memory reactivation by a trauma-cue. The effects of the treatment on behavior were associated with a commensurate normalization of the dendritic cytoarchitecture of DG and BLA neurons. Pretreatment with RU486, Ketanserin, or Pindolol prevented the above improvement in anxiety-like behavioral responses. MDMA treatment paired with memory reactivation reduced the prevalence rate of PTSD-phenotype 14 days later and normalized the cytoarchitecture changes induced by PSS (in dendritic complexities) compared to saline control. MDMA treatment paired with a trauma-cue may modify or update the original traumatic memory trace through reconsolidation processes. These anxiolytic-like effects seem to involve the HPA axis and 5-HT systems.


Asunto(s)
Ansiolíticos , N-Metil-3,4-metilenodioxianfetamina , Trastornos por Estrés Postraumático , Animales , Ansiolíticos/uso terapéutico , Señales (Psicología) , Modelos Animales de Enfermedad , Sistema Hipotálamo-Hipofisario , N-Metil-3,4-metilenodioxianfetamina/uso terapéutico , Sistema Hipófiso-Suprarrenal , Estudios Prospectivos , Ratas , Ratas Endogámicas Lew , Reflejo de Sobresalto , Trastornos por Estrés Postraumático/tratamiento farmacológico , Trastornos por Estrés Postraumático/prevención & control
4.
Transl Psychiatry ; 10(1): 10, 2020 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-32066707

RESUMEN

Converging evidence indicates that orexins (ORXs), the regulatory neuropeptides, are implicated in anxiety- and depression-related behaviors via the modulation of neuroendocrine, serotonergic, and noradrenergic systems. This study evaluated the role of the orexinergic system in stress-associated physiological responses in a controlled prospective animal model. The pattern and time course of activation of hypothalamic ORX neurons in response to predator-scent stress (PSS) were examined using c-Fos as a marker for neuronal activity. The relationship between the behavioral response pattern 7 days post-exposure and expressions of ORXs was evaluated. We also investigated the effects of intracerebroventricular microinfusion of ORX-A or almorexant (ORX-A/B receptor antagonist) on behavioral responses 7 days following PSS exposure. Hypothalamic levels of ORX-A, neuropeptide Y (NPY), and brain-derived neurotrophic factor (BDNF) were assessed. Compared with rats whose behaviors were extremely disrupted (post-traumatic stress disorder [PTSD]-phenotype), those whose behaviors were minimally selectively disrupted displayed significantly upregulated ORX-A and ORX-B levels in the hypothalamic nuclei. Intracerebroventricular microinfusion of ORX-A before PSS reduced the prevalence of the PTSD phenotype compared with that of artificial cerebrospinal fluid or almorexant, and rats treated with almorexant displayed a higher prevalence of the PTSD phenotype than did untreated rats. Activated ORX neurons led to upregulated expressions of BDNF and NPY, which might provide an additional regulatory mechanism for the modulation of adaptive stress responses. The study indicates that the activated ORX system might promote adaptive responses to PSS probably via stimulation of BDNF and NPY secretion, and early intervention with ORX-A reduces the prevalence of the PTSD phenotype and increases the prevalence of adaptive phenotypes. The findings provide some insights into the mechanisms underlying the involvement of the ORX system in stress-related disorders.


Asunto(s)
Trastornos por Estrés Postraumático , Animales , Modelos Animales de Enfermedad , Neuropéptido Y , Estudios Prospectivos , Ratas , Ratas Sprague-Dawley
5.
J Neurotrauma ; 36(2): 380-394, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29947272

RESUMEN

The complex interactions and overlapping symptoms of comorbid post-traumatic stress disorder (PTSD) and mild traumatic brain injury (mTBI) induced by an explosive blast wave have become a focus of attention in recent years, making clinical distinction and effective intervention difficult. Because dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis is thought to underlie trauma-related (psycho)pathology, we evaluated both the endogenous corticosterone response and the efficacy of exogenous hydrocortisone treatment provided shortly after blast exposure. We employed a controlled experimental blast-wave paradigm in which unanesthetized animals were exposed to visual, auditory, olfactory, and tactile effects of an explosive blast wave produced by exploding a thin copper wire. Endogenous corticosterone concentrations were evaluated at different time points (before, and 3 h, 5 h and 17 days) after blast exposure. Subsequently, the efficacy of exogenous hydrocortisone (25 mg/kg-1 or 125 mg/kg-1) injected intraperitoneally 1 h after exposure was compared with that of a similarly timed saline injection. Validated cognitive and behavioral tests were used to assess both PTSD and mTBI phenotypes on days 7-14 following the blast. Retrospective analysis revealed that animals demonstrating the PTSD phenotype exhibited a significantly blunted endogenous corticosterone response to the blast compared with all other groups. Moreover, a single 125 mg/kg-1 dose of hydrocortisone administered 1 h after exposure significantly reduced the occurrence of the PTSD phenotype. Hydrocortisone treatment did not have a similar effect on the mTBI phenotype. Results of this study indicate that an inadequate corticosteroid response following blast exposure increases risk for PTSD phenotype, and corticosteroid treatment is a potential clinical intervention for attenuating PTSD. The differences in patterns of physiological and therapeutic response between PTSD and mTBI phenotypes lend credence to the retrospective behavioral and cognitive classification criteria we designed, and is in keeping with the assumption that mTBI and PTSD phenotypes may reflect distinct underlying biological and clinical profiles.


Asunto(s)
Antiinflamatorios , Traumatismos por Explosión , Conmoción Encefálica , Corticosterona , Trastornos por Estrés Postraumático , Animales , Antiinflamatorios/sangre , Antiinflamatorios/farmacología , Traumatismos por Explosión/sangre , Traumatismos por Explosión/psicología , Conmoción Encefálica/sangre , Conmoción Encefálica/etiología , Conmoción Encefálica/psicología , Corticosterona/sangre , Corticosterona/farmacología , Masculino , Ratas , Ratas Sprague-Dawley , Trastornos por Estrés Postraumático/sangre , Trastornos por Estrés Postraumático/etiología
6.
Psychoneuroendocrinology ; 87: 35-42, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29035710

RESUMEN

The basal activity of the hypothalamic-pituitary-adrenal axis is highly dynamic and is characterized by both circadian and ultradian (pulsatile) patterns of hormone secretion. Pulsatility of glucocorticoids has been determined to be critical for optimal transcriptional, neuroendocrine, and behavioral responses. We used an animal model of post-traumatic stress disorder (PTSD) to assess whether stress-induced impairment of behavioral responses is correlated with aberrant secretion of corticosterone. Serial blood samples were collected manually via the jugular vein cannula during the light-(inactive)-phase in conscious male rats at 20-min intervals for a period of 5h before and 6.5h after exposure to predator scent stress. The outcome measures included behavior in an elevated plus-maze and acoustic startle response 7days after exposure. Individual animals were retrospectively classified as having "extreme", "partial", or "minimal" behavioral responses according to pre-set cut-off criteria for behavioral response patterns. Corticosterone secretion patterns were analyzed retrospectively. Under basal conditions, the amplitude of ultradian oscillations of corticosterone levels, rather than the mean corticosterone level or the frequency of corticosterone pulsatility, was significantly reduced in individuals who displayed PTSD-phenotype 8days later. In addition, extreme disruption of behavior on day 8 post-exposure was also characterized by a blunting of corticosterone response to the stressor. Animals with behavior that was only partially affected or unaffected displayed none of the above changes. Blunted basal corticosterone pulse amplitude is a pre-existing susceptibility or risk factor for PTSD, which originates from prior (life) experiences and may therefore predict post-exposure PTSD-phenotype in rats.


Asunto(s)
Corticosterona/fisiología , Trastornos por Estrés Postraumático/fisiopatología , Estimulación Acústica , Animales , Biomarcadores/sangre , Corticosterona/sangre , Corticosterona/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/fisiopatología , Glucocorticoides/análisis , Glucocorticoides/sangre , Sistema Hipotálamo-Hipofisario/fisiopatología , Masculino , Aprendizaje por Laberinto , Fenotipo , Sistema Hipófiso-Suprarrenal/fisiopatología , Ratas , Ratas Sprague-Dawley , Reflejo de Sobresalto/fisiología , Estrés Psicológico/fisiopatología , Ritmo Ultradiano/fisiología
7.
J Neurotrauma ; 34(1): 145-164, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-26885687

RESUMEN

The intense focus in the clinical literature on the mental and neurocognitive sequelae of explosive blast-wave exposure, especially when comorbid with post-traumatic stress-related disorders (PTSD) is justified, and warrants the design of translationally valid animal studies to provide valid complementary basic data. We employed a controlled experimental blast-wave paradigm in which unanesthetized animals were exposed to visual, auditory, olfactory, and tactile effects of an explosive blast-wave produced by exploding a thin copper wire. By combining cognitive-behavioral paradigms and ex vivo brain MRI to assess mild traumatic brain injury (mTBI) phenotype with a validated behavioral model for PTSD, complemented by morphological assessments, this study sought to examine our ability to evaluate the biobehavioral effects of low-intensity blast overpressure on rats, in a translationally valid manner. There were no significant differences between blast- and sham-exposed rats on motor coordination and strength, or sensory function. Whereas most male rats exposed to the blast-wave displayed normal behavioral and cognitive responses, 23.6% of the rats displayed a significant retardation of spatial learning acquisition, fulfilling criteria for mTBI-like responses. In addition, 5.4% of the blast-exposed animals displayed an extreme response in the behavioral tasks used to define PTSD-like criteria, whereas 10.9% of the rats developed both long-lasting and progressively worsening behavioral and cognitive "symptoms," suggesting comorbid PTSD-mTBI-like behavioral and cognitive response patterns. Neither group displayed changes on MRI. Exposure to experimental blast-wave elicited distinct behavioral and morphological responses modelling mTBI-like, PTSD-like, and comorbid mTBI-PTSD-like responses. This experimental animal model can be a useful tool for elucidating neurobiological mechanisms underlying the effects of blast-wave-induced mTBI and PTSD and comorbid mTBI-PTSD.


Asunto(s)
Traumatismos por Explosión/diagnóstico por imagen , Conmoción Encefálica/diagnóstico por imagen , Modelos Animales de Enfermedad , Trastornos por Estrés Postraumático/diagnóstico por imagen , Animales , Ansiedad/diagnóstico por imagen , Ansiedad/etiología , Ansiedad/psicología , Traumatismos por Explosión/complicaciones , Traumatismos por Explosión/psicología , Conmoción Encefálica/complicaciones , Conmoción Encefálica/psicología , Comorbilidad , Masculino , Aprendizaje por Laberinto/fisiología , Presión/efectos adversos , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Trastornos por Estrés Postraumático/etiología , Trastornos por Estrés Postraumático/psicología
8.
Behav Brain Res ; 308: 128-42, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27105958

RESUMEN

It is unclear whether the poor autonomic flexibility or dysregulation observed in patients with posttraumatic stress disorder (PTSD) represents a pre-trauma vulnerability factor or results from exposure to trauma. We used an animal model of PTSD to assess the association between the behavioral response to predator scent stress (PSS) and the cardiac autonomic modulation in male and female rats. The rats were surgically implanted with radiotelemetry devices to measure their electrocardiograms and locomotor activity (LMA). Following baseline telemetric monitoring, the animals were exposed to PSS or sham-PSS. Continuous telemetric monitoring (24h/day sampling) was performed over the course of 7days. The electrocardiographic recordings were analyzed using the time- and frequency-domain indexes of heart rate variability (HRV). The behavioral response patterns were assessed using the elevated plus maze and acoustic startle response paradigms for the retrospective classification of individuals according to the PTSD-related cut-off behavioral criteria. During resting conditions, the male rats had significantly higher heart rates (HR) and lower HRV parameters than the female rats during both the active and inactive phases of the daily cycle. Immediately after PSS exposure, both the female and male rats demonstrated a robust increase in HR and a marked drop in HRV parameters, with a shift of sympathovagal balance towards sympathetic predominance. In both sexes, autonomic system habituation and recovery were selectively inhibited in the rats whose behavior was extremely disrupted after exposure to PSS. However, in the female rats, exposure to the PSS produced fewer EBR rats, with a more rapid recovery curve than that of the male rats. PSS did not induce changes to the circadian rhythm of the LMA. According to our results, PTSD can be conceptualized as a disorder that is related to failure-of-recovery mechanisms that impede the restitution of physiological homeostasis.


Asunto(s)
Enfermedades del Sistema Nervioso Autónomo/etiología , Caracteres Sexuales , Trastornos por Estrés Postraumático/complicaciones , Estrés Psicológico/fisiopatología , Estimulación Acústica , Análisis de Varianza , Animales , Modelos Animales de Enfermedad , Electrocardiografía , Femenino , Frecuencia Cardíaca/fisiología , Masculino , Aprendizaje por Laberinto , Ratas , Ratas Sprague-Dawley , Reflejo de Sobresalto/fisiología , Telemetría
9.
Neuropsychopharmacology ; 40(3): 774-90, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25241802

RESUMEN

The hypothalamic-pituitary-adrenal (HPA) axis displays a characteristic circadian pattern of corticosterone release, with higher levels at the onset of the active phase and lower levels at the onset of the inactive phase. As corticosterone levels modify the response to stress and influence the susceptibility to and/or severity of stress-related sequelae, we examined the effects of an acute psychological trauma applied at different zeitgeber times (ZTs) on behavioral stress responses. Rats were exposed to stress either at the onset of the inactive-(light) phase (ZT=0) or at the onset of the active-(dark) phase (ZT=12). Their behavior in the elevated plus-maze and acoustic startle response paradigms were assessed 7 days post exposure for retrospective classification into behavioral response groups. Serum corticosterone levels and the dexamethasone suppression test were used to assess the stress response and feedback inhibition of the HPA axis. Immunoreactivity for neuropeptide Y (NPY) and NPY-Y1 receptor (Y1R) in the paraventricular (PVN) and arcuate (ARC) hypothalamic nuclei, hippocampus, and basolateral amygdala were measured. The behavioral effects of NPY/Y1R antagonist microinfused into the PVN 30 min before stress exposure during the inactive or active phase, respectively, were evaluated. PVN immunoreactivity for NPY and Y1R was measured 1 day after the behavioral tests. The time of day of the traumatic exposure markedly affected the pattern of the behavioral stress response and the prevalence of rats showing an extreme behavioral response. Rats exposed to the stressor at the onset of their inactive phase displayed a more traumatic behavioral response, faster post-exposure corticosterone decay, and a more pronounced stress-induced decline in NPY and Y1R expression in the PVN and arcuate hypothalamic nuclei. Blocking PVN Y1R before stress applied in the active phase, or administering NPY to the PVN before stress applied in the inactive phase, had a resounding behavioral effect. The time at which stress occurred significantly affected the behavioral stress response. Diurnal variations in HPA and NPY/Y1R significantly affect the behavioral response, conferring more resilience at the onset of the active phase and more vulnerability at the onset of the inactive phase, implying that NPY has a significant role in conferring resilience to stress-related psychopathology.


Asunto(s)
Ritmo Circadiano/fisiología , Sistema Hipotálamo-Hipofisario/fisiología , Neuropéptido Y/metabolismo , Sistema Hipófiso-Suprarrenal/fisiología , Trauma Psicológico/fisiopatología , Receptores de Neuropéptido Y/fisiología , Estrés Psicológico/fisiopatología , Administración Intranasal , Amígdala del Cerebelo/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Arginina/administración & dosificación , Arginina/análogos & derivados , Arginina/farmacología , Corticosterona/sangre , Hipocampo/metabolismo , Masculino , Aprendizaje por Laberinto/fisiología , Microinyecciones , Neuropéptido Y/administración & dosificación , Neuropéptido Y/farmacología , Núcleo Hipotalámico Paraventricular/metabolismo , Pruebas de Función Adreno-Hipofisaria , Trauma Psicológico/sangre , Trauma Psicológico/metabolismo , Trauma Psicológico/psicología , Ratas , Receptores de Neuropéptido Y/antagonistas & inhibidores , Receptores de Neuropéptido Y/metabolismo , Reflejo de Sobresalto/fisiología , Estrés Psicológico/sangre , Estrés Psicológico/metabolismo , Estrés Psicológico/psicología , Factores de Tiempo
10.
Eur Neuropsychopharmacol ; 24(12): 1925-44, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25451698

RESUMEN

Alterations in cytoarchitecture and molecular signaling have been observed in adaptive and maladaptive responses to stress and presumably underlie the physiological and behavioral changes observed. The relationship between behavioral responses to stress exposure and changes in cytoarchitecture of subregions of the hippocampus and amygdala was investigated in an animal model of PTSD. Behaviors in elevated plus-maze and acoustic startle response tests were assessed in rats 7 days after exposure to predator scent stress. Brains were harvested 24h later. Neurons from CA1, CA3, and dentate gyrus subregions and basolateral amygdala were reconstructed and subjected to Sholl analysis and spine density estimation. Glucocorticoid receptor, brain-derived neurotrophic factor, phospho-NR1-Ser-889, phospho-GluR1-Ser-845, phospho-calcium/calmodulin dependent protein kinase II-Thy-286, post-synaptic density protein 95 and phospho-CREB-Ser-133 were evaluated in the hippocampus. Data were analyzed by retrospective classification of individual rats into three behavioral response groups. The extent and distribution of changes in the morphology of hippocampal and amygdalar dendrites was significantly associated with stress-induced behavioral response classification. Extreme (PTSD-like) behavioral disruption was associated with extensive neuronal retraction in the hippocampus and proliferation in the amygdala. Neither structure displayed such changes in minimal behavioral responders. Partial behavioral response was associated with identical changes in the hippocampus only. Patterns of change in requisite molecular signaling genes and endophenotypic markers corresponded to the structural and behavioral responses. The extent and distribution of changes in the cytoarchitecture of hippocampal and amygdalar subregions is directly related to the pattern of behavioral response of the individual to stress exposure.


Asunto(s)
Amígdala del Cerebelo/patología , Modelos Animales de Enfermedad , Hipocampo/patología , Trastornos por Estrés Postraumático/patología , Amígdala del Cerebelo/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteína de Unión a CREB/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Dendritas/patología , Dendritas/ultraestructura , Homólogo 4 de la Proteína Discs Large , Hipocampo/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Aprendizaje por Laberinto , Proteínas de la Membrana/metabolismo , Neuronas/patología , Ratas , Receptores AMPA/metabolismo , Receptores de Glucocorticoides/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Reflejo de Sobresalto , Trastornos por Estrés Postraumático/metabolismo
11.
ILAR J ; 55(2): 233-45, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25225303

RESUMEN

The diagnosis of Post-Traumatic Stress Disorder (PTSD) is conditional on directly experiencing or witnessing a significantly threatening event and the presence of a certain minimal number of symptoms from each of four symptom clusters (re-experiencing, avoidance, negative cognition and mood, and hyperarousal) at least one month after the event (DSM 5) (American Psychiatric Association 2013). Only a proportion of the population exposed develops symptoms fulfilling the criteria. The individual heterogeneity in responses of stress-exposed animals suggested that adapting clearly defined and reliably reproducible "diagnostic", i.e. behavioral, criteria for animal responses would augment the clinical validity of the analysis of study data. We designed cut-off (inclusion/exclusion) behavioral criteria (CBC) which classify study subjects as being severely, minimally or partially affected by the stress paradigm, to be applied retrospectively in the analysis of behavioral data. Behavioral response classification enables the researcher to correlate (retrospectively) specific anatomic, bio-molecular and physiological parameters with the degree and pattern of the individual behavioral response, and also introduces "prevalence rates" as a valid study-parameter. The cumulative results of our studies indicate that, by classifying the data from individual subjects according to their response patterns, the animal study can more readily be translated into clinical "follow-up" studies and back again. This article will discuss the concept of the model and its background, and present a selection of studies employing and examining the model, alongside the underlying translational rationale of each.


Asunto(s)
Conducta Animal/fisiología , Síntomas Conductuales/fisiopatología , Biomarcadores , Modelos Animales de Enfermedad , Individualidad , Trastornos por Estrés Postraumático/diagnóstico , Trastornos por Estrés Postraumático/fisiopatología , Animales , Síntomas Conductuales/diagnóstico , Clasificación/métodos , Humanos , Modelos Teóricos
13.
Cell Tissue Res ; 354(1): 127-39, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23912242

RESUMEN

Post-traumatic stress disorder (PTSD) is clinically defined in DSM-4 by exposure to a significantly threatening and/or horrifying event and the presence of a certain number of symptoms from each of three symptom clusters at least one month after the event. Since humans clearly do not respond homogeneously to a potentially traumatic experience, the heterogeneity in animal responses might be regarded as confirming the validity of animal studies, rather than as representing a problem. A model of diagnostic criteria for psychiatric disorders could therefore be applied to animal responses to augment the validity of study data, providing that the criteria for classification are clearly defined, reliably reproducible and yield results that conform to findings in human subjects. The method described herein was developed in an attempt to model diagnostic criteria in terms of individual patterns of response by using behavioral measures and determining cut-off scores to distinguish between extremes of response or non-response, leaving a sizeable proportion of subjects in a middle group, outside each set of cut-off criteria. The cumulative results of our studies indicate that the contribution of animal models can be further enhanced by classifying individual animal study subjects according to their response patterns. The animal model also enables the researcher to go one step further and correlate specific anatomic, bio-molecular and physiological parameters with the degree and pattern of the individual behavioral response and introduces "prevalence rates" as a parameter. The translational value of the classification method and future directions are discussed.


Asunto(s)
Modelos Animales de Enfermedad , Trastornos por Estrés Postraumático/diagnóstico , Animales , Humanos , Ratones , Ratas , Roedores , Investigación Biomédica Traslacional
14.
Curr Protoc Neurosci ; Chapter 9: Unit 9.45, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23853112

RESUMEN

Animal behavioral studies have commonly regarded the entire group of animals subjected to the study conditions as homogeneous, disregarding individual differences in response patterns. The following discussion will focus on a method of analyzing data that aims to model clinical diagnostic criteria applied to individual patterns of response using data from behavioral measures, and employing cut-off scores to distinguish between extremes of response versus non-response and the sizeable proportion of study subjects in-between them. This protocol unit will present the concept of the model and its background, provide detailed protocols for each of its components, and present a selection of studies employing and examining the model, alongside the underlying translational rationale of each.


Asunto(s)
Modelos Animales de Enfermedad , Trastornos por Estrés Postraumático , Estimulación Acústica , Animales , Investigación Biomédica , Señales (Psicología) , Conducta Exploratoria , Femenino , Masculino , Aprendizaje por Laberinto , Ratones , Ratas , Reflejo de Sobresalto/fisiología , Trastornos por Estrés Postraumático/fisiopatología
15.
Neuropsychopharmacology ; 37(11): 2388-404, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22713910

RESUMEN

Reliable evidence supports the role of sleep in learning and memory processes. In rodents, sleep deprivation (SD) negatively affects consolidation of hippocampus-dependent memories. As memory is integral to post-traumatic stress symptoms, the effects of post-exposure SD on various aspect of the response to stress in a controlled, prospective animal model of post-traumatic stress disorder (PTSD) were evaluated. Rats were deprived of sleep for 6 h throughout the first resting phase after predator scent stress exposure. Behaviors in the elevated plus-maze and acoustic startle response tests were assessed 7 days later, and served for classification into behavioral response groups. Freezing response to a trauma reminder was assessed on day 8. Urine samples were collected daily for corticosterone levels, and heart rate (HR) was also measured. Finally, the impact of manipulating the hypothalamus-pituitary-adrenal axis and adrenergic activity before SD was assessed. Mifepristone (MIFE) and epinephrine (EPI) were administered systemically 10-min post-stress exposure and behavioral responses and response to trauma reminder were measured on days 7-8. Hippocampal expression of glucocorticoid receptors (GRs) and morphological assessment of arborization and dendritic spines were subsequently evaluated. Post-exposure SD effectively ameliorated long-term, stress-induced, PTSD-like behavioral disruptions, reduced trauma reminder freezing responses, and decreased hippocampal expression of GR compared with exposed-untreated controls. Although urine corticosterone levels were significantly elevated 1 h after SD and the HR was attenuated, antagonizing GRs with MIFE or stimulation of adrenergic activity with EPI effectively abolished the effect of SD. MIFE- and EPI-treated animals clearly demonstrated significantly lower total dendritic length, fewer branches and lower spine density along dentate gyrus dendrites with increased levels of GR expression 8 days after exposure, as compared with exposed-SD animals. Intentional prevention of sleep in the early aftermath of stress exposure may well be beneficial in attenuating traumatic stress-related sequelae. Post-exposure SD may disrupt the consolidation of aversive or fearful memories by facilitating correctly timed interactions between glucocorticoid and adrenergic systems.


Asunto(s)
Agonistas Adrenérgicos/uso terapéutico , Epinefrina/uso terapéutico , Antagonistas de Hormonas/uso terapéutico , Mifepristona/uso terapéutico , Privación de Sueño/fisiopatología , Trastornos por Estrés Postraumático/prevención & control , Estimulación Acústica/efectos adversos , Agonistas Adrenérgicos/farmacología , Análisis de Varianza , Animales , Corticosterona/orina , Dendritas/efectos de los fármacos , Dendritas/metabolismo , Dendritas/ultraestructura , Modelos Animales de Enfermedad , Electrocardiografía , Reacción Cataléptica de Congelación/efectos de los fármacos , Reacción Cataléptica de Congelación/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/ultraestructura , Antagonistas de Hormonas/farmacología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Mifepristona/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/ultraestructura , Ratas , Ratas Sprague-Dawley , Receptores de Glucocorticoides/metabolismo , Reflejo de Sobresalto/efectos de los fármacos , Reflejo de Sobresalto/fisiología , Tinción con Nitrato de Plata , Trastornos por Estrés Postraumático/fisiopatología , Telemetría , Factores de Tiempo
16.
Neuropharmacology ; 62(2): 715-24, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21565209

RESUMEN

In humans, the diagnosis of PTSD is made only if an individual exhibits a certain number of symptoms from each of three quite well defined symptom clusters over a certain period of time. Animal behavioral studies, however, have generally tended to overlook this aspect and have commonly regarded the entire group of animals subjected to certain study conditions as homogeneous. Thus, in an attempt to develop animal models of long-term chronic behavioral responses to stress (i.e. PTSD) in a comparable manner to human diagnosis, we applied cut-off inclusion/exclusion criteria to behavioral data for a cohort of animals exposed to a stress paradigm. This grouped them as behaviorally affected or unaffected by the stress. This model takes into account the variability in degree of the individual's response to the stress paradigm, thereby modeling the fact that not all humans exposed to traumatic stress respond with affective disorder. This article will present and discuss findings from a series of studies employing a model of individual behavioral response classification. This article will discuss the concept of the model and its background and present a selection of studies employing and examining the model, alongside the underlying translational rationale of each. This article is part of a Special Issue entitled 'Post-Traumatic Stress Disorder'.


Asunto(s)
Conducta Animal/fisiología , Trastornos por Estrés Postraumático/psicología , Animales , Modelos Animales de Enfermedad , Humanos , Investigación Biomédica Traslacional
17.
Eur Neuropsychopharmacol ; 22(3): 205-21, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21925847

RESUMEN

In mammals, the circadian and stress systems are involved in adaptation to predictable and unpredictable stimuli, respectively. A series of experiments examined the relationship between stress-induced posttraumatic stress (PTSD)-like behavioral response patterns in rats and brain levels of genes related to circadian rhythms. The effects of agomelatine, administered immediately after exposure, on stress-related behavior and on local expression of Per1 and Per2 were assessed. Animals were exposed to predator scent stress. The outcome measures included behavior in an elevated plus-maze (EPM) and acoustic startle response (ASR) 7days after the exposure. Pre-set cut-off behavioral criteria classified exposed animals according to behavioral responses in EPM and ASR paradigms as those with 'extreme behavioral response' (EBR), 'minimal behavioral response (MBR),' or 'partial behavioral response' (PBR). Per1 and Per2 expression in hippocampal subregions, frontal cortex and suprachiasmatic nucleus (SCN) 8days after exposure were evaluated using immunohistochemical and RT-PCR techniques at zeitgeber-times 19 and 13. The effects of agomelatine, on behavioral tests were evaluated on Day 8. Local brain expression of Per1 and Per2 mRNA was subsequently assessed. Data were analyzed in relation to individual behavior patterns. Animals with extreme behavioral response (EBR) displayed a distinct pattern of Per1 and Per2 expression in the SCN, which was the opposite of that observed in the control and MBR animals. In the DG, no variation in Per2 expression was observed in the EBR and PBR animals. Immediate post-exposure treatment with agomelatine significantly reduced percentage of extreme-responders and normalized the expression of Per1 and Per2 as compared to controls. Stress-induced alterations in Per genes in the EBR animals may represent an imbalance between normally precisely orchestrated physiological and behavioral processes and psychopathological processes. These findings indicate that these circadian-related genes play a role in the neurobiological response to predator scent stress and provide supportive evidence that the use of agomelatine immediately after traumatic experience may be protective against the subsequent development of PTSD.


Asunto(s)
Acetamidas/farmacología , Encéfalo/efectos de los fármacos , Ritmo Circadiano/genética , Expresión Génica/efectos de los fármacos , Proteínas Circadianas Period/genética , Estrés Psicológico/genética , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Encéfalo/metabolismo , Proteínas Circadianas Period/metabolismo , Ratas , Ratas Sprague-Dawley , Reflejo de Sobresalto/efectos de los fármacos , Reflejo de Sobresalto/fisiología , Estrés Psicológico/metabolismo
18.
Neuropsychopharmacology ; 36(11): 2286-302, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21734649

RESUMEN

Nuclear factor-κB (NF-κB) is a ubiquitously expressed transcription factor for genes involved in cell survival, differentiation, inflammation, and growth. This study examined the role of NF-κB pathway in stress-induced PTSD-like behavioral response patterns in rats. Immunohistochemical technique was used to detect the expression of the NF-κB p50 and p65 subunits, I-κBα, p38, and phospho-p38 in the hippocampal subregions at 7 days after exposure to predator scent stress. Expression of p65 nuclear translocation was quantified by western blot as the level of NF-κB activation. The effects of intraperitoneally administered corticosterone or a selective NF-κB inhibitor (pyrrolidine dithiocarbamate (PDTC)) at 1 h post exposure on behavioral tests (elevated plus-maze and acoustic startle response) were evaluated 7 days later. Hippocampal expressions of those genes were subsequently evaluated. All data were analyzed in relation to individual behavior patterns. Extreme behavioral responder animals displayed significant upregulation of p50 and p65 with concomitant downregulation of I-κBα, p38, and phospho-p38 levels in hippocampal structures compared with minimal behavioral responders and controls. Immediate post-exposure treatment with high-dose corticosterone and PDTC significantly reduced prevalence rates of extreme responders and normalized the expression of those genes. Stress-induced upregulation of NF-κB complex in the hippocampus may contribute to the imbalance between what are normally precisely orchestrated and highly coordinated physiological and behavioral processes, thus associating it with stress-related disorders.


Asunto(s)
Corticosterona/administración & dosificación , FN-kappa B/biosíntesis , Pirrolidinas/administración & dosificación , Trastornos por Estrés Postraumático/tratamiento farmacológico , Estrés Psicológico/tratamiento farmacológico , Tiocarbamatos/administración & dosificación , Regulación hacia Arriba/efectos de los fármacos , Animales , Gatos , Esquema de Medicación , Masculino , FN-kappa B/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Trastornos por Estrés Postraumático/metabolismo , Estrés Psicológico/metabolismo , Factores de Tiempo , Regulación hacia Arriba/fisiología
19.
Eur Neuropsychopharmacol ; 21(11): 796-809, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21741804

RESUMEN

High-dose corticosteroids have been reported to reduce symptoms of acute stress and post-traumatic stress in polytrauma patients and in animal studies. The underlying mechanism of action remains largely unclear. These issues were addressed in parallel in the clinical and preclinical studies below. In this preliminary study, 25 patients with acute stress symptoms were administered a single intravenous bolus of high-dose hydrocortisone (100-140 mg) or placebo within 6 h of a traumatic event in a prospective, randomized, double-blind, placebo-controlled pilot study. Early single high-dose hydrocortisone intervention attenuated the core symptoms of both the acute stress and of subsequent PTSD in patients. High-dose hydrocortisone treatment given in the first few hours after a traumatic experience was associated with significant favorable changes in the trajectory of exposure to trauma, as expressed by the reduced risk of the development of PTSD post-trauma. In parallel, a comparative study of morphological arborization in dentate gyrus and its modulating molecules was performed in stress-exposed animals treated with high-dose hydrocortisone. Steroid-treated stressed animals displayed significantly increased dendritic growth and spine density, with increased levels of brain-derived neurotrophic factor (BDNF) and obtunded postsynaptic density-95 (PSD-95) levels. The animal study provided insights into the potential mechanism of this intervention, as it identified relevant morphological and biochemical associations to the clinical observations. Thus, evidence from clinical and animal studies suggests that there is a "window of opportunity" in the early aftermath of trauma to help those who are vulnerable to the development of chronic PTSD.


Asunto(s)
Antiinflamatorios/uso terapéutico , Hidrocortisona/uso terapéutico , Trastornos por Estrés Postraumático/prevención & control , Heridas y Lesiones/tratamiento farmacológico , Estimulación Acústica , Adulto , Análisis de Varianza , Animales , Ansiedad/tratamiento farmacológico , Ansiedad/etiología , Encéfalo/patología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Espinas Dendríticas/patología , Espinas Dendríticas/ultraestructura , Modelos Animales de Enfermedad , Homólogo 4 de la Proteína Discs Large , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Femenino , Estudios de Seguimiento , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Neuronas/patología , Neuronas/ultraestructura , Dimensión del Dolor , Ratas , Ratas Sprague-Dawley , Reflejo de Sobresalto/efectos de los fármacos , Tinción con Nitrato de Plata , Trastornos por Estrés Postraumático/etiología , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/patología , Factores de Tiempo , Índices de Gravedad del Trauma , Resultado del Tratamiento , Heridas y Lesiones/complicaciones , Heridas y Lesiones/patología , Adulto Joven
20.
Eur Neuropsychopharmacol ; 21(3): 230-40, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21194896

RESUMEN

The therapeutic value of ß-adrenoceptor blockage, using propranolol, in the aftermath of traumatic experience is uncertain. A prospective, controlled animal model of posttraumatic stress disorder (PTSD) was employed to assess the effects of propranolol on long-term behavioral responses to stress. Animals exposed to predator scent stress received a single bolus of propranolol (10 or 15mg/kg) or vehicle 1h post-exposure. Outcomes were assessed using the elevated plus-maze (EPM) and acoustic startle response (ASR) at 30days and freezing response to a trauma reminder (unsoiled litter) on Day 31. Individual animals were classified as having "extreme", "partial" and "minimal" behavioral responses, according to pre-set cut-off criteria for EPM and ASR response patterns. The physiological efficacy of the doses of propranolol was verified by collecting cardiovascular data telemetrically (from exposed or unexposed individuals given propranolol or vehicle). The effect of propranolol on long-term memory was verified using a non-spatial memory task. Both doses of propranolol effectively reduced mean heart rate and impaired the object-recognition task, as expected. No significant effect on prevalence rates of PTSD-like behavioral responses or on trauma reminder response was observed for either dose of propranolol as compared to vehicle. Despite adequate efficacy in terms of heart rate and disruption of memory, single-dose, post-stress ß-blockage with propranolol was ineffective in reducing onset of PTSD-like behavioral disruption and trauma cue responses in the long term. Traumatic stress-related processes appear to be affected differently than the others.


Asunto(s)
Antagonistas Adrenérgicos beta/farmacología , Nadolol/farmacología , Propranolol/farmacología , Trastornos por Estrés Postraumático/prevención & control , Estrés Psicológico/tratamiento farmacológico , Animales , Ansiedad/tratamiento farmacológico , Conducta Animal/efectos de los fármacos , Señales (Psicología) , Modelos Animales de Enfermedad , Frecuencia Cardíaca , Inyecciones Subcutáneas , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Memoria a Largo Plazo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Reflejo de Sobresalto/efectos de los fármacos , Trastornos por Estrés Postraumático/fisiopatología , Trastornos por Estrés Postraumático/psicología , Estrés Psicológico/fisiopatología , Estrés Psicológico/psicología , Factores de Tiempo , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...