Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
J Exp Biol ; 223(Pt 1)2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31896721

RESUMEN

Serotonin (5-HT) has largely been accepted to be inhibitory to vertebrate aggression, whereas an opposing stimulatory role has been proposed for invertebrates. Herein, we argue that critical gaps in our understanding of the nuanced role of 5-HT in invertebrate systems drove this conclusion prematurely, and that emerging data suggest a previously unrecognized level of phylogenetic conservation with respect to neurochemical mechanisms regulating the expression of aggressive behaviors. This is especially apparent when considering the interplay among factors governing 5-HT activity, many of which share functional homology across taxa. We discuss recent findings using insect models, with an emphasis on the stalk-eyed fly, to demonstrate how particular 5-HT receptor subtypes mediate the intensity of aggression with respect to discrete stages of the interaction (initiation, escalation and termination), which mirrors the complex behavioral regulation currently recognized in vertebrates. Further similarities emerge when considering the contribution of neuropeptides, which interact with 5-HT to ultimately determine contest progression and outcome. Relative to knowledge in vertebrates, much less is known about the function of 5-HT receptors and neuropeptides in invertebrate aggression, particularly with respect to sex, species and context, prompting the need for further studies. Our Commentary highlights the need to consider multiple factors when determining potential taxonomic differences, and raises the possibility of more similarities than differences between vertebrates and invertebrates with regard to the modulatory effect of 5-HT on aggression.


Asunto(s)
Agresión/fisiología , Dípteros/fisiología , Modelos Animales , Receptores de Serotonina/metabolismo , Serotonina/metabolismo , Animales , Femenino , Masculino
2.
Eur J Neurosci ; 48(2): 1833-1850, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29904960

RESUMEN

Adult psychiatric disorders characterized by cognitive deficits reliant on prefrontal cortex (PFC) dopamine are promoted by teenage bullying. Similarly, male Sprague-Dawley rats exposed to social defeat in mid-adolescence (P35-39) show impaired working memory in adulthood (P56-70), along with decreased medial PFC (mPFC) dopamine activity that results in part from increased dopamine transporter-mediated clearance. Here, we determined if dopamine synthesis and D2 autoreceptor-mediated inhibition of dopamine release in the adult mPFC are also enhanced by adolescent defeat to contribute to later dopamine hypofunction. Control and previously defeated rats did not differ in either DOPA accumulation following amino acid decarboxylase inhibition (NSD-1015 100 mg/kg ip.) or total/phosphorylated tyrosine hydroxylase protein expression, suggesting dopamine synthesis in the adult mPFC is not altered by adolescent defeat. However, exposure to adolescent defeat caused greater decreases in extracellular dopamine release (measured using in vivo chronoamperometry) in the adult mPFC upon local infusion of the D2 receptor agonist quinpirole (3 nM), implying greater D2 autoreceptor function. Equally enhanced D2 autoreceptor-mediated inhibition of dopamine release is seen in the adolescent (P40 or P49) mPFC, which declines in control rats by adulthood. However, this developmental decrease in autoreceptor function is absent following adolescent defeat, suggesting retention of an adolescent-like phenotype into adulthood. Current and previous findings indicate adolescent defeat decreases extracellular dopamine availability in the adult mPFC via both enhanced inhibition of dopamine release and increased dopamine clearance, which may be viable targets for improving treatment of cognitive deficits seen in neuropsychiatric disorders promoted by adolescent stress.


Asunto(s)
Autorreceptores/metabolismo , Dominación-Subordinación , Dopamina/metabolismo , Corteza Prefrontal/metabolismo , Receptores de Dopamina D2/metabolismo , Estrés Psicológico/metabolismo , Factores de Edad , Animales , Conducta Animal/fisiología , Masculino , Ratas , Ratas Sprague-Dawley
3.
Proc Biol Sci ; 285(1878)2018 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-29720416

RESUMEN

Multiple cave populations of the teleost Astyanax mexicanus have repeatedly reduced or lost eye and body pigmentation during adaptation to dark caves. Albinism, the complete absence of melanin pigmentation, is controlled by loss-of-function mutations in the oca2 gene. The mutation is accompanied by an increase in the melanin synthesis precursor l-tyrosine, which is also a precursor for catecholamine synthesis. In this study, we show a relationship between pigmentation loss, enhanced catecholamine synthesis and responsiveness to anaesthesia, determined as a proxy for catecholamine-related behaviours. We demonstrate that anaesthesia resistance (AR) is enhanced in multiple depigmented and albino cavefish (CF), inversely proportional to the degree of pigmentation loss, controlled by the oca2 gene, and can be modulated by experimental manipulations of l-tyrosine or the catecholamine norepinephrine (NE). Moreover, NE is increased in the brains of multiple albino and depigmented CF relative to surface fish. The results provide new insights into the evolution of pigment modification because NE controls a suite of adaptive behaviours similar to AR that may represent a target of natural selection. Thus, understanding the relationship between loss of pigmentation and AR may provide insight into the role of natural selection in the evolution of albinism via a melanin-catecholamine trade-off.


Asunto(s)
Ciclos de Actividad , Albinismo Oculocutáneo/genética , Anestésicos/farmacología , Catecolaminas/metabolismo , Characidae/fisiología , Proteínas de Peces/genética , Pigmentación , Albinismo Oculocutáneo/metabolismo , Anestesia , Animales , Evolución Biológica , Characidae/genética , Proteínas de Peces/metabolismo , Norepinefrina/metabolismo , Tirosina/metabolismo
4.
Stress ; 20(2): 223-230, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28345385

RESUMEN

Acute activation of the hypothalamic-pituitary-adrenal (HPA) axis, leading to the release of corticosteroid hormones into the circulation, is an adaptive response to perceived threats. Persistent activation of the HPA axis can lead to impaired physiological or behavioral function with maladaptive consequences. Thus, efficient control and termination of stress responses is essential for well-being. However, inhibitory control mechanisms governing the HPA axis are poorly understood. Previous studies suggest that serotonergic systems, acting within the medial hypothalamus, play an important role in inhibitory control of stress-induced HPA axis activity. To test this hypothesis, we surgically implanted chronic jugular cannulae in adult male rats and conducted bilateral microinjection of vehicle or the 5-HT1A receptor agonist, 8-hydroxy-2-(di-n-propylamino) tetralin hydrobromide (8-OH-DPAT; 8 nmol, 0.2 µL, 0.1 µL/min, per side) into the dorsomedial hypothalamus (DMH) immediately prior to a 40 min period of restraint stress. Repeated blood sampling was conducted using an automated blood sampling system and plasma corticosterone concentrations were determined using enzyme-linked immunosorbent assay. Bilateral intra-DMH microinjections of 8-OH-DPAT suppressed stress-induced increases in plasma corticosterone within 10 min of the onset of handling prior to restraint and, as measured by area-under-the-curve analysis of plasma corticosterone concentrations, during the 40 min period of restraint. These data support an inhibitory role for serotonergic systems, acting within the DMH, on stress-induced activation of the HPA axis. Lay summary: Inhibitory control of the hypothalamic-pituitary-adrenal (HPA) stress hormone response is important for well-being. One neurochemical implicated in inhibitory control of the HPA axis is serotonin. In this study we show that activation of serotonin receptors, specifically inhibitory 5-HT1A receptors in the dorsomedial hypothalamus, is sufficient to inhibit stress-induced HPA axis activity in rats.


Asunto(s)
8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Agonistas de Receptores de Serotonina/farmacología , Animales , Corticosterona/sangre , Sistema Hipotálamo-Hipofisario/fisiopatología , Hipotálamo/fisiopatología , Masculino , Sistema Hipófiso-Suprarrenal/fisiopatología , Ratas , Ratas Sprague-Dawley , Serotonina/farmacología
5.
Stress ; 18(1): 76-87, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25556980

RESUMEN

Previous studies suggest that multiple corticolimbic and hypothalamic structures are involved in glucocorticoid-mediated feedback inhibition of the hypothalamic-pituitary-adrenal (HPA) axis, including the dorsomedial hypothalamus (DMH), but a potential role of the DMH has not been directly tested. To investigate the role of the DMH in glucocorticoid-mediated negative feedback, adult male Sprague Dawley rats were implanted with jugular cannulae and bilateral guide cannulae directed at the DMH, and finally were either adrenalectomized (ADX) or were subjected to sham-ADX. ADX rats received corticosterone (CORT) replacement in the drinking water (25 µg/mL), which, based on initial studies, restored a rhythm of plasma CORT concentrations in ADX rats that was similar in period and amplitude to the diurnal rhythm of plasma CORT concentrations in sham-ADX rats, but with a significant phase delay. Following recovery from surgery, rats received microinjections of either CORT (10 ng, 0.5 µL, 0.25 µL/min, per side) or vehicle (aCSF containing 0.2% EtOH), bilaterally, directly into the DMH, prior to a 40-min period of restraint stress. In sham-ADX rats, bilateral intra-DMH microinjections of CORT, relative to bilateral intra-DMH microinjections of vehicle, decreased restraint stress-induced elevation of endogenous plasma CORT concentrations 60 min after the onset of intra-DMH injections. Intra-DMH CORT decreased the overall area under the curve for plasma CORT concentrations during the intermediate time frame of glucocorticoid negative feedback, from 0.5 to 2 h following injection. These data are consistent with the hypothesis that the DMH is involved in feedback inhibition of HPA axis activity at the intermediate time frame.


Asunto(s)
Corticosterona/administración & dosificación , Núcleo Hipotalámico Dorsomedial/efectos de los fármacos , Glucocorticoides/administración & dosificación , Terapia de Reemplazo de Hormonas , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Adrenalectomía , Animales , Ritmo Circadiano/efectos de los fármacos , Modelos Animales de Enfermedad , Núcleo Hipotalámico Dorsomedial/metabolismo , Núcleo Hipotalámico Dorsomedial/fisiopatología , Retroalimentación Fisiológica , Hidrocortisona/sangre , Sistema Hipotálamo-Hipofisario/metabolismo , Sistema Hipotálamo-Hipofisario/fisiopatología , Masculino , Sistema Hipófiso-Suprarrenal/metabolismo , Sistema Hipófiso-Suprarrenal/fisiopatología , Ratas Sprague-Dawley , Restricción Física/psicología , Transducción de Señal/efectos de los fármacos , Estrés Psicológico/sangre , Estrés Psicológico/fisiopatología , Estrés Psicológico/psicología , Factores de Tiempo
6.
Eur J Neurosci ; 40(11): 3684-92, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25234335

RESUMEN

Withdrawal from amphetamine increases anxiety and reduces the ability to cope with stress, which are factors that are believed to contribute to drug relapse. Stress-induced serotonergic transmission in the central nucleus of the amygdala is associated with anxiety states and fear. Conversely, stress-induced increases in ventral hippocampal serotonin (5-HT) levels have been linked to coping mechanisms. The goal of this study was to investigate the neurobiological changes induced by amphetamine that contribute to stress sensitivity during withdrawal. We tested the hypothesis that limbic serotonergic responses to restraint stress would be altered in male Sprague-Dawley rats chronically pretreated with amphetamine (2.5 mg/kg, intraperitoneal) and then subjected to 2 weeks of withdrawal. Amphetamine withdrawal resulted in increased stress-induced behavioral arousal relative to control treatment, suggesting that drug withdrawal induced greater sensitivity to the stressor. When microdialysis was used to determine the effects of restraint on extracellular 5-HT, stress-induced increases in 5-HT levels were abolished in the ventral hippocampus and augmented in the central amygdala during amphetamine withdrawal. Reverse dialysis of the glucocorticoid receptor antagonist mifepristone into the ventral hippocampus blocked the stress-induced increase in 5-HT levels in saline-pretreated rats, suggesting that glucocorticoid receptors mediate stress-induced increases in 5-HT levels in the ventral hippocampus. However, mifepristone had no effect on stress-induced increases in 5-HT levels in the central amygdala, indicating that stress increases 5-HT levels in this region independently of glucocorticoid receptors. During amphetamine withdrawal, the absence of stress-induced increases in ventral hippocampal 5-HT levels combined with enhanced stress-induced serotonergic responses in the central amygdala may contribute to drug relapse by decreasing stress-coping ability and heightening stress responsiveness.


Asunto(s)
Trastornos Relacionados con Anfetaminas/metabolismo , Núcleo Amigdalino Central/metabolismo , Hipocampo/metabolismo , Serotonina/metabolismo , Estrés Psicológico/metabolismo , Síndrome de Abstinencia a Sustancias/metabolismo , Anfetamina/efectos adversos , Anfetamina/farmacología , Animales , Núcleo Amigdalino Central/efectos de los fármacos , Fármacos del Sistema Nervioso Central/farmacología , Estimulantes del Sistema Nervioso Central/efectos adversos , Estimulantes del Sistema Nervioso Central/farmacología , Cromatografía Líquida de Alta Presión , Hipocampo/efectos de los fármacos , Masculino , Microdiálisis , Mifepristona/farmacología , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Ratas Sprague-Dawley , Receptores de Glucocorticoides/antagonistas & inhibidores , Receptores de Glucocorticoides/metabolismo , Restricción Física , Estrés Psicológico/tratamiento farmacológico
7.
Eur J Neurosci ; 37(3): 479-90, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23157166

RESUMEN

Amphetamine withdrawal in both humans and rats is associated with increased anxiety states, which are thought to contribute to drug relapse. Serotonin in the ventral hippocampus mediates affective behaviors, and reduced serotonin levels in this region are observed in rat models of high anxiety, including during withdrawal from chronic amphetamine. This goal of this study was to understand the mechanisms by which reduced ventral hippocampus serotonergic neurotransmission occurs during amphetamine withdrawal. Serotonin synthesis (assessed by accumulation of serotonin precursor as a measure of the capacity of in vivo tryptophan hydroxylase activity), expression of serotonergic transporters, and in vivo serotonergic clearance using in vivo microdialysis were assessed in the ventral hippocampus in adult male Sprague Dawley rats at 24 h withdrawal from chronic amphetamine. Overall, results showed that diminished extracellular serotonin at 24 h withdrawal from chronic amphetamine was not accompanied by a change in capacity for serotonin synthesis (in vivo tryptophan hydroxylase activity), or serotonin transporter expression or function in the ventral hippocampus, but instead was associated with increased expression and function of organic cation transporters (low-affinity, high-capacity serotonin transporters). These findings suggest that 24 h withdrawal from chronic amphetamine reduces the availability of extracellular serotonin in the ventral hippocampus by increasing organic cation transporter-mediated serotonin clearance, which may represent a future pharmacological target for reversing anxiety states during drug withdrawal.


Asunto(s)
Anfetamina/toxicidad , Hipocampo/metabolismo , Serotonina/biosíntesis , Síndrome de Abstinencia a Sustancias/metabolismo , Anfetamina/efectos adversos , Animales , Expresión Génica , Masculino , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/metabolismo , Ratas , Ratas Sprague-Dawley , Serotonina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Transmisión Sináptica , Triptófano Hidroxilasa/metabolismo
8.
Brain Res ; 1800: 148189, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36462646

RESUMEN

Organic cation transporter-3 (OCT3) is widely distributed in the brain with high expression in portions of the stress axis. These high capacity, polyspecific transporters function in monoamine clearance and are sensitive to the stress hormone corticosterone. In rats, withdrawal from chronic amphetamine increases OCT3 expression in specific limbic brain regions involved anxiety and stress responses, including the ventral hippocampus, central nucleus of amygdala (CeA) and dorsomedial hypothalamus. (DMH). Previous studies show that glucocorticoid receptor (GR) agonists increase OCT1 mRNA and OCT2 mRNA expression in non-neural tissues. Thus, we hypothesized that corticosterone increases OCT3 expression in the brain by activating GRs. Male Sprague-Dawley rats were pre-treated daily with the GR antagonist mifepristone (20 mg/kg; sc.) or vehicle followed 45 min later by injections of corticosterone or vehicle for 2 weeks. Corticosterone treatment significantly increased OCT3 expression in the ventral hippocampus and increased anxiety-like behavior. However, these effects were not blocked by mifepristone. Interestingly, treatment with mifepristone alone reduced plasma corticosterone levels and increased serotonin transporter and GR expression in the ventral hippocampus but did not significantly affect OCT3 expression or behavior. No treatment effects on OCT3, serotonin transporter or GR expression were observed in the DMH, CeA or dorsal hippocampus. Our findings suggest that corticosterone increases OCT3 expression in the ventral hippocampus by a mechanism independent of GRs, and that mifepristone and corticosterone can act in an independent manner to affect HPA axis-related physiological and behavioral parameters.


Asunto(s)
Corticosterona , Receptores de Glucocorticoides , Ratas , Masculino , Animales , Receptores de Glucocorticoides/metabolismo , Ratas Sprague-Dawley , Serotonina/metabolismo , Sistema Hipotálamo-Hipofisario/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Mifepristona/farmacología , Sistema Hipófiso-Suprarrenal/metabolismo , Hipocampo/metabolismo , Anfetamina/farmacología , Anfetamina/metabolismo , Ansiedad
9.
Front Behav Neurosci ; 17: 1148292, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37064300

RESUMEN

Corticotropin-releasing factor (CRF) is essential for coordinating endocrine and neural responses to stress, frequently facilitated by vasopressin (AVP). Previous work has linked CRF hypersecretion, binding site changes, and dysfunctional serotonergic transmission with anxiety and affective disorders, including clinical depression. Crucially, CRF can alter serotonergic activity. In the dorsal raphé nucleus and serotonin (5-HT) terminal regions, CRF effects can be stimulatory or inhibitory, depending on the dose, site, and receptor type activated. Prior stress alters CRF neurotransmission and CRF-mediated behaviors. Lateral, medial, and ventral subdivisions of the central nucleus of the amygdala (CeA) produce CRF and coordinate stress responsiveness. The purpose of these experiments was to determine the effect of intracerebroventricular (icv) administration of CRF and AVP on extracellular 5-HT as an index of 5-HT release in the CeA, using in vivo microdialysis in freely moving rats and high performance liquid chromatography (HPLC) analysis. We also examined the effect of prior stress (1 h restraint, 24 h prior) on CRF- and AVP-mediated release of 5-HT within the CeA. Our results show that icv CRF infusion in unstressed animals had no effect on 5-HT release in the CeA. Conversely, in rats with prior stress, CRF caused a profound dose-dependent decrease in 5-HT release within the CeA. This effect was long-lasting (240 min) and was mimicked by CRF plus AVP infusion without stress. Thus, prior stress and AVP functionally alter CRF-mediated neurotransmission and sensitize CRF-induced inhibition of 5-HT release, suggesting that this is a potential mechanism underlying stress-induced affective reactivity in humans.

10.
Biol Psychiatry ; 91(9): 841-852, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35279280

RESUMEN

BACKGROUND: Stress produces differential behavioral responses through select molecular modifications to specific neurocircuitry elements. The orexin (Orx) system targets key components of this neurocircuitry in the basolateral amygdala (BLA). METHODS: We assessed the contribution of intra-BLA Orx1 receptors (Orx1Rs) in the expression of stress-induced phenotypes of mice. Using the Stress Alternatives Model, a social stress paradigm that produces two behavioral phenotypes, we characterized the role of intra-BLA Orx1R using acute pharmacological inhibition (SB-674042) and genetic knockdown (AAV-U6-Orx1R-shRNA) strategies. RESULTS: In the BLA, we observed that Orx1R (Hcrtr1) messenger RNA is predominantly expressed in CamKIIα+ glutamatergic neurons and rarely in GABAergic (gamma-aminobutyric acidergic) cells. While there is a slight overlap in Hcrtr1 and Orx2 receptor (Hcrtr2) messenger RNA expression in the BLA, we find that these receptors are most often expressed in separate cells. Antagonism of intra-BLA Orx1R after phenotype formation shifted behavioral expression from stress-sensitive (Stay) to stress-resilient (Escape) responses, an effect that was mimicked by genetic knockdown. Acute inhibition of Orx1R in the BLA also reduced contextual and cued fear freezing responses in Stay animals. This phenotype-specific behavioral change was accompanied by biased molecular transcription favoring Hcrtr2 over Hcrtr1 and Mapk3 over Plcb1 cell signaling cascades and enhanced Bdnf messenger RNA. CONCLUSIONS: Functional reorganization of intra-BLA gene expression is produced by antagonism of Orx1R, which promotes elevated Hcrtr2, greater Mapk3, and increased Bdnf expression. Together, these results provide evidence for a receptor-driven mechanism that balances pro- and antistress responses within the BLA.


Asunto(s)
Complejo Nuclear Basolateral , Receptores de Orexina , Animales , Ansiedad/metabolismo , Complejo Nuclear Basolateral/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Ratones , Receptores de Orexina/genética , ARN Mensajero/metabolismo , Transducción de Señal
11.
Fish Physiol Biochem ; 37(3): 425-32, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20963632

RESUMEN

The concentrations of monoamines in the hypothalamus were determined in yellow perch Perca flavescens before and after injection with lipopolysaccharide (LPS; 3 mg kg⁻¹ fish weight) or saline to test for the presence of neurochemical changes potentially associated with changes in plasma cortisol characteristic of intraperitoneal (ip) challenge with LPS. In the first experiment, yellow perch were injected with saline or LPS and the hypothalamus removed and plasma sampled before and at 0.5, 1.5, 3.0, and 6 h after injection. Plasma cortisol was elevated in both saline- and LPS-injected fish through 1.5 h after injection and returned to levels resembling pre-injection by 3 h after injection. Significantly higher amounts of cortisol in plasma from LPS-injected relative to saline-injected fish were observed 6 h following injection. A significant decrease relative to levels observed 0.5-3 h after handling was observed in serotonin concentrations at 6 h following LPS and saline injection with a concomitant increase in the ratio of 5-hydroxyindoleacetic acid:serotonin. In the second experiment, hypothalamic monoamines were sampled before and at 3, 6, 9, 12, and 24 h after injection with LPS or saline. Significant increases from pre-injection levels were observed in the ratio 5-hydroxyindoleacetic acid:serotonin at 9, 12, and 24 h after injection, but no differences were detected between LPS- and saline-injected fish. These results support a model linking serotonergic system activation following handling stress, but no correlations with the sustained elevations of plasma cortisol associated with inflammatory challenge were observed.


Asunto(s)
Monoaminas Biogénicas/metabolismo , Hidrocortisona/sangre , Hipotálamo/metabolismo , Lipopolisacáridos/toxicidad , Neurotransmisores/metabolismo , Percas/sangre , Animales , Hipotálamo/efectos de los fármacos , Inyecciones Intraperitoneales , Factores de Tiempo
12.
Curr Biol ; 31(16): 3694-3701.e4, 2021 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-34293332

RESUMEN

Adaptation to novel environments often involves the evolution of multiple morphological, physiological, and behavioral traits. One striking example of multi-trait evolution is the suite of traits that has evolved repeatedly in cave animals, including regression of eyes, loss of pigmentation, and enhancement of non-visual sensory systems.1,2 The Mexican tetra, Astyanax mexicanus, consists of fish that inhabit at least 30 caves in Mexico and ancestral-like surface fish that inhabit the rivers of Mexico and southern Texas.3 Cave A. mexicanus are interfertile with surface fish and have evolved a number of traits, including reduced pigmentation, eye loss, and alterations to behavior.4-6 To define relationships between different cave-evolved traits, we phenotyped 208 surface-cave F2 hybrid fish for numerous morphological and behavioral traits. We found differences in sleep between pigmented and albino hybrid fish, raising the possibility that these traits share a genetic basis. In cavefish and other species, mutations in oculocutaneous albinism 2 (oca2) cause albinism.7-12 Surface fish with mutations in oca2 displayed both albinism and reduced sleep. Further, this mutation in oca2 fails to complement sleep loss when surface fish harboring this engineered mutation are crossed to independently evolved populations of albino cavefish with naturally occurring mutations in oca2. Analysis of the oca2 locus in wild-caught cave and surface fish suggests that oca2 is under positive selection in 3 cave populations. Taken together, these findings identify oca2 as a novel regulator of sleep and suggest that a pleiotropic function of oca2 underlies the adaptive evolution of albinism and sleep loss.


Asunto(s)
Albinismo , Characidae , Proteínas de Peces/genética , Sueño , Animales , Evolución Biológica , Characidae/genética , Ojo , Pigmentación/genética
13.
Elife ; 92020 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-32314737

RESUMEN

A widely accepted model for the evolution of cave animals posits colonization by surface ancestors followed by the acquisition of adaptations over many generations. However, the speed of cave adaptation in some species suggests mechanisms operating over shorter timescales. To address these mechanisms, we used Astyanax mexicanus, a teleost with ancestral surface morphs (surface fish, SF) and derived cave morphs (cavefish, CF). We exposed SF to completely dark conditions and identified numerous altered traits at both the gene expression and phenotypic levels. Remarkably, most of these alterations mimicked CF phenotypes. Our results indicate that many cave-related traits can appear within a single generation by phenotypic plasticity. In the next generation, plasticity can be further refined. The initial plastic responses are random in adaptive outcome but may determine the subsequent course of evolution. Our study suggests that phenotypic plasticity contributes to the rapid evolution of cave-related traits in A. mexicanus.


The Mexican tetra is a fish that has two forms: a surface-dwelling form, which has eyes and silvery grey appearance, and a cave-dwelling form, which is blind and has lost its pigmentation. Recent studies have shown that the cave-dwelling form evolved rapidly within the last 200,000 years from an ancestor that lived at the surface. The recent evolution of the cave-dwelling form of the tetra poses an interesting evolutionary question: how did the surface-dwelling ancestor of the tetra quickly adapt to the new and challenging environment found in the caves? 'Phenotypic plasticity' is a phenomenon through which a single set of genes can produce different observable traits depending on the environment. An example of phenotypic plasticity occurs in response to diet: in animals, poor diets can lead to an increase in the size of the digestive organs and to the animals eating more. To see if surface-dwelling tetras can quickly adapt to cave environments through phenotypic plasticity, Bilandzija et al. have exposed these fish to complete darkness (the major feature of the cave environment) for two years. After spending up to two years in the dark, these fish were compared to normal surface-dwelling and cave-dwelling tetras. Results revealed that surface-dwelling tetras raised in the dark exhibited traits associated with cave-dwelling tetras. These traits included changes in the activity of many genes involved in diverse processes, resistance to starvation, metabolism, and levels of hormones and molecules involved in neural signaling, which could lead to changes in behavior. However, the fish also exhibited traits, including an increase in the cells responsible for pigmentation, that would have no obvious benefit in the darkness. Even though the changes observed require no genetic mutations, they can help or hinder the fish's survival once they occur, possibly determining subsequent evolution. Thus, a trait beneficial for surviving in the dark that appears simply through phenotypic plasticity may eventually be selected for and genetic mutations that encode it more reliably may appear too. These results shed light on how species may quickly adapt to new environments without accumulating genetic mutations, which can take hundreds of thousands of years. They also may help to explain how colonizer species succeed in challenging environments. The principles described by Bilandzija et al. can be applied to different organisms adapting to new environments, and may help understand the role of phenotypic plasticity in evolution.


Asunto(s)
Adaptación Fisiológica/fisiología , Cuevas , Characidae/fisiología , Animales , Evolución Biológica , Fenotipo
14.
Behav Neurosci ; 123(3): 564-76, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19485563

RESUMEN

Social stress in adolescence is correlated with emergence of psychopathologies during early adulthood. In this study, the authors investigated the impact of social defeat stress during mid-adolescence on adult male brain and behavior. Adolescent male Sprague-Dawley rats were exposed to repeated social defeat for 5 days while controls were placed in a novel empty cage. When exposed to defeat-associated cues as adults, previously defeated rats showed increased risk assessment and behavioral inhibition, demonstrating long-term memory for the defeat context. However, previously defeated rats exhibited increased locomotion in both elevated plus-maze and open field tests, suggesting heightened novelty-induced behavior. Adolescent defeat also affected adult monoamine levels in stress-responsive limbic regions, causing decreased medial prefrontal cortex dopamine, increased norepinephrine and serotonin in the ventral dentate gyrus, and decreased norepinephrine in the dorsal raphe. Our results suggest that adolescent social defeat produces both deficits in anxiety responses and altered monoaminergic function in adulthood. This model offers potential for identifying specific mechanisms induced by severe adolescent social stress that may contribute to increased adult male vulnerability to psychopathology.


Asunto(s)
Ansiedad/psicología , Monoaminas Biogénicas/metabolismo , Dominación-Subordinación , Sistema Límbico/crecimiento & desarrollo , Sistema Límbico/fisiopatología , Estrés Psicológico/fisiopatología , Animales , Condicionamiento Psicológico , Corticosterona/sangre , Giro Dentado/crecimiento & desarrollo , Giro Dentado/fisiopatología , Dopamina/metabolismo , Masculino , Actividad Motora , Norepinefrina/metabolismo , Corteza Prefrontal/crecimiento & desarrollo , Corteza Prefrontal/fisiopatología , Núcleos del Rafe/crecimiento & desarrollo , Núcleos del Rafe/fisiopatología , Ratas , Ratas Sprague-Dawley , Serotonina/metabolismo , Sustancia Negra/crecimiento & desarrollo , Sustancia Negra/fisiopatología
15.
Stress ; 12(3): 225-39, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-18951247

RESUMEN

Corticotropin-releasing factor (CRF) is a potent neuromodulator of stress-related behaviour but the neural mechanisms underlying these effects are not clear. Studies were designed to test the hypothesis that CRF-induced behavioural arousal involves interactions with brainstem serotonergic systems. To examine interactions between CRF and serotonergic systems in the regulation of behaviour, CRF (1 microg, intracerebroventricular (i.c.v.)) or vehicle was infused in the presence or absence of the selective serotonin re-uptake inhibitor fluoxetine (0, 0.1, 1 or 10 mg/kg, intravenous (i.v.)). Fluoxetine was used at these doses because it is known to decrease serotonin cell firing rates while increasing extracellular serotonin concentrations in select forebrain regions. We then measured behavioural, neurochemical and endocrine responses. CRF increased locomotion and spontaneous non-ambulatory motor activity (SNAMA) in the home cages. Fluoxetine decreased tissue 5-hydroxyindoleacetic acid concentrations, a measure of serotonin metabolism, in specific limbic brain regions of CRF-treated rats (nucleus accumbens shell region, entorhinal cortex, central nucleus of the amygdala). Furthermore, fluoxetine inhibited CRF-induced SNAMA. CRF and fluoxetine independently increased plasma corticosterone concentrations, but the responses had distinct temporal profiles. Overall, these data are consistent with the hypothesis that CRF-induced facilitation of behavioural activity is dependent on brainstem serotonergic systems. Therefore, fluoxetine may attenuate or alleviate some behavioural responses to stress by interfering with CRF-induced responses.


Asunto(s)
Conducta Animal/efectos de los fármacos , Hormona Liberadora de Corticotropina/fisiología , Fluoxetina/farmacología , Animales , Nivel de Alerta/efectos de los fármacos , Corticosterona/sangre , Hormona Liberadora de Corticotropina/antagonistas & inhibidores , Fluoxetina/administración & dosificación , Ácido Hidroxiindolacético/metabolismo , Inyecciones Intraventriculares , Locomoción/efectos de los fármacos , Masculino , Actividad Motora/efectos de los fármacos , Prosencéfalo/efectos de los fármacos , Prosencéfalo/metabolismo , Ratas , Ratas Wistar , Serotonina/metabolismo
16.
Horm Behav ; 56(1): 177-84, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19409390

RESUMEN

The anxiety- and stress-related neuropeptide corticotropin-releasing factor (CRF) elicits behavioral changes in vertebrates including increases in behavioral arousal and locomotor activity. Intracerebroventricular injections of CRF in an amphibian, the roughskin newt (Taricha granulosa), induces rapid increases in locomotor activity in both intact and hypophysectomized animals. We hypothesized that this CRF-induced increase in locomotor activity involves a central effect of CRF on serotonergic neurons, based on known stimulatory actions of serotonin (5-hydroxytryptamine, 5-HT) on spinal motor neurons and the central pattern generator for locomotor activity in vertebrates. In Experiment 1, we found that neither intracerebroventricular injections of low doses of CRF (25 ng) nor the selective serotonin reuptake inhibitor fluoxetine (10, 100 ng), by themselves, altered locomotor activity. In contrast, newts treated concurrently with CRF and fluoxetine responded with marked increases in locomotor activity. In Experiment 2, we found that increases in locomotor activity following co-administration of CRF (25 ng) and fluoxetine (100 ng) were associated with decreased 5-HT concentrations in a number of forebrain structures involved in regulation of emotional behavior and emotional states, including the ventral striatum, amygdala pars lateralis, and dorsal hypothalamus, measured 37 min after treatment. These results are consistent with the hypothesis that CRF stimulates locomotor activity through activation of serotonergic systems.


Asunto(s)
Hormona Liberadora de Corticotropina/metabolismo , Fluoxetina/administración & dosificación , Actividad Motora/efectos de los fármacos , Salamandridae/fisiología , Inhibidores Selectivos de la Recaptación de Serotonina/administración & dosificación , Serotonina/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Corticosterona/sangre , Relación Dosis-Respuesta a Droga , Ácido Hidroxiindolacético/metabolismo , Masculino , Actividad Motora/fisiología , Prosencéfalo/efectos de los fármacos , Prosencéfalo/metabolismo , Radioinmunoensayo
17.
Horm Behav ; 56(1): 149-57, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19371745

RESUMEN

The dorsomedial hypothalamus (DMH) plays an important role in coordinating physiological and behavioral responses to stress-related stimuli. In vertebrates, DMH serotonin (5-HT) concentrations increase rapidly in response to acute stressors or corticosterone (CORT). Recent studies suggest that CORT inhibits postsynaptic clearance of 5-HT from the extracellular fluid in the DMH by blocking organic cation transporter 3 (OCT3), a polyspecific CORT-sensitive transport protein. Because OCTs are low-affinity, high-capacity transporters, we hypothesized that CORT effects on extracellular 5-HT are most pronounced in the presence of elevated 5-HT release. We predicted that local application of CORT into the DMH would potentiate the effects of d-fenfluramine, a 5-HT-releasing agent, on extracellular 5-HT. These experiments were conducted using in vivo microdialysis in freely-moving male Sprague-Dawley rats implanted with a microdialysis probe into the medial hypothalamus (MH), which includes the DMH. In Experiment 1, rats simultaneously received intraperitoneal (i.p.) injections of 1 mg/kg D-fenfluramine or saline and either 200 ng/mL CORT or dilute ethanol (EtOH) vehicle delivered to the MH by reverse-dialysis for 40 min. In Experiment 2, 5 microM D-fenfluramine and either 200 ng/mL CORT or EtOH vehicle were concurrently delivered to the MH for 40 min using reverse-dialysis. CORT potentiated the increases in extracellular 5-HT concentrations induced by either i.p. or intra-MH administration of D-fenfluramine. Furthermore, CORT and D-fenfluramine interacted to alter home cage behaviors. Our results support the hypothesis that CORT inhibition of OCT3-mediated 5-HT clearance from the extracellular fluid contributes to stress-induced increases in extracellular 5-HT and 5-HT signaling.


Asunto(s)
Corticosterona/metabolismo , Fenfluramina/administración & dosificación , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Serotoninérgicos/administración & dosificación , Serotonina/metabolismo , Análisis de Varianza , Animales , Cromatografía Líquida de Alta Presión , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/metabolismo , Aseo Animal/efectos de los fármacos , Aseo Animal/fisiología , Inyecciones Intraperitoneales , Masculino , Microdiálisis/métodos , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Ratas , Ratas Sprague-Dawley
18.
Physiol Behav ; 96(3): 493-504, 2009 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-19103211

RESUMEN

Repeated amphetamine treatment results in behavioral sensitization in a high percentage of rats. Alterations to plasma corticosterone, neural monoamines and stress behavior can accompany amphetamine sensitization. Whether these changes occur following repeated amphetamine treatment in the absence of behavioral sensitization is not known. Male Sprague-Dawley rats were treated with amphetamine (2.5 mg/kg, i.p.) or saline once daily for 6 days. Amphetamine-induced locomotion and stereotypy, open-field anxiety behavior, plasma corticosterone and limbic monoamines were measured during withdrawal. Sixty-two percent of amphetamine-treated rats showed behavioral sensitization over the test periods. Only amphetamine-sensitized rats showed increased latency to enter the center of the open-field, as well as increased plasma corticosterone when compared to saline-treated controls. Amphetamine-sensitized rats showed increased dopamine concentrations in the shell of the nucleus accumbens and increased serotonin concentrations in the dorsal hippocampus, which were not observed in amphetamine-treated non-sensitized rats. These findings suggest that anxiety behavior, plasma corticosterone and limbic monoamines concentrations are altered by repeated amphetamine (2.5 mg/kg) treatment, and that these neuroendocrine and behavioral changes are often associated with sensitization to the psychostimulant effects of amphetamine.


Asunto(s)
Anfetamina/farmacología , Conducta Animal/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/farmacología , Actividad Motora/efectos de los fármacos , Serotonina/metabolismo , Análisis de Varianza , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Corticosterona/sangre , Conducta Exploratoria/efectos de los fármacos , Ácido Hidroxiindolacético/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Conducta Estereotipada/efectos de los fármacos
19.
Physiol Behav ; 96(2): 218-32, 2009 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-18957299

RESUMEN

Male Anolis carolinensis that win aggressive interactions mobilize neuroendocrine responses to social stress more rapidly than defeated lizards. We initially examined temporal patterns of neuroendocrine response to restraint stress in lizards of unknown status, and then investigated whether winning males respond more rapidly to this non-social stressor. Size-matched male pairs interacted to establish social status, and then were returned to individual home cages for 3 days. Plasma and brains were collected from non-restrained dominants and subordinates, and from a non-interacting control group. Additional groups of dominants and subordinates underwent 90 s restraint stress, with plasma and brains collected either immediately or 300 s after restraint. In lizards of unknown social status restraint stimulated rapid monoaminergic responses in nucleus accumbens, hippocampus, amygdala, and locus ceruleus, with delayed responses seen in VTA and raphé. Non-restrained dominants and subordinates had lower levels of raphé serotonergic activity and lower hippocampal dopaminergic activity 3 days after interacting, compared to controls. Dominants had higher corticosterone levels, both immediately and 300 s after restraint, than either non-restrained dominants or restrained subordinates. Restraint induced higher raphé serotonergic activity in dominants. However, subordinates also showed rapid responses to restraint; exhibiting lower hippocampal dopamine (DA) levels than non-restrained subordinates. At 300 s after the stress, amygdalar serotonin levels increased in dominants, while subordinates showed higher amygdalar DA levels. These results suggest that stressful aggressive interactions will not only alter basal neurochemical activity, but also influence neuroendocrine responses to non-social stressors according to individual social status.


Asunto(s)
Sistemas Neurosecretores/fisiopatología , Medio Social , Estrés Psicológico/fisiopatología , Estrés Psicológico/psicología , Análisis de Varianza , Animales , Conducta Animal/fisiología , Monoaminas Biogénicas/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Química Encefálica/fisiología , Corticosterona/sangre , Lagartos , Masculino , Restricción Física/métodos , Estrés Psicológico/patología , Factores de Tiempo
20.
PLoS One ; 14(1): e0203980, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30695038

RESUMEN

Despite the conserved function of aggression across taxa in obtaining critical resources such as food and mates, serotonin's (5-HT) modulatory role on aggressive behavior appears to be largely inhibitory for vertebrates but stimulatory for invertebrates. However, critical gaps exist in our knowledge of invertebrates that need to be addressed before definitively stating opposing roles for 5-HT and aggression. Specifically, the role of 5-HT receptor subtypes are largely unknown, as is the potential interactive role of 5-HT with other neurochemical systems known to play a critical role in aggression. Similarly, the influence of these systems in driving sex differences in aggressive behavior of invertebrates is not well understood. Here, we investigated these questions by employing complementary approaches in a novel invertebrate model of aggression, the stalk-eyed fly. A combination of altered social conditions, pharmacological manipulation and 5-HT2 receptor knockdown by siRNA revealed an inhibitory role of this receptor subtype on aggression. Additionally, we provide evidence for 5-HT2's involvement in regulating neuropeptide F activity, a suspected inhibitor of aggression. However, this function appears to be stage-specific, altering only the initiation stage of aggressive conflicts. Alternatively, pharmacologically increasing systemic concentrations of 5-HT significantly elevated the expression of the neuropeptide tachykinin, which did not affect contest initiation but instead promoted escalation via production of high intensity aggressive behaviors. Notably, these effects were limited solely to males, with female aggression and neuropeptide expression remaining unaltered by any manipulation that affected 5-HT. Together, these results demonstrate a more nuanced role for 5-HT in modulating aggression in invertebrates, revealing an important interactive role with neuropeptides that is more reminiscent of vertebrates. The sex-differences described here also provide valuable insight into the evolutionary contexts of this complex behavior.


Asunto(s)
Agresión/fisiología , Conducta Animal/fisiología , Dípteros/fisiología , Caracteres Sexuales , 5-Hidroxitriptófano/administración & dosificación , 5-Hidroxitriptófano/farmacología , Agresión/efectos de los fármacos , Animales , Técnicas de Observación Conductual/métodos , Conducta Animal/efectos de los fármacos , Femenino , Técnicas de Silenciamiento del Gen , Masculino , Modelos Animales , Neuropéptidos/metabolismo , ARN Interferente Pequeño/metabolismo , Receptores de Serotonina 5-HT2/genética , Receptores de Serotonina 5-HT2/metabolismo , Serotonina/metabolismo , Taquicininas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA