Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell ; 186(18): 3862-3881.e28, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37572660

RESUMEN

Male sexual behavior is innate and rewarding. Despite its centrality to reproduction, a molecularly specified neural circuit governing innate male sexual behavior and reward remains to be characterized. We have discovered a developmentally wired neural circuit necessary and sufficient for male mating. This circuit connects chemosensory input to BNSTprTac1 neurons, which innervate POATacr1 neurons that project to centers regulating motor output and reward. Epistasis studies demonstrate that BNSTprTac1 neurons are upstream of POATacr1 neurons, and BNSTprTac1-released substance P following mate recognition potentiates activation of POATacr1 neurons through Tacr1 to initiate mating. Experimental activation of POATacr1 neurons triggers mating, even in sexually satiated males, and it is rewarding, eliciting dopamine release and self-stimulation of these cells. Together, we have uncovered a neural circuit that governs the key aspects of innate male sexual behavior: motor displays, drive, and reward.


Asunto(s)
Vías Nerviosas , Conducta Sexual Animal , Animales , Masculino , Neuronas/fisiología , Recompensa , Conducta Sexual Animal/fisiología , Ratones
2.
Cell ; 186(6): 1195-1211.e19, 2023 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-36796363

RESUMEN

Social interactions require awareness and understanding of the behavior of others. Mirror neurons, cells representing an action by self and others, have been proposed to be integral to the cognitive substrates that enable such awareness and understanding. Mirror neurons of the primate neocortex represent skilled motor tasks, but it is unclear if they are critical for the actions they embody, enable social behaviors, or exist in non-cortical regions. We demonstrate that the activity of individual VMHvlPR neurons in the mouse hypothalamus represents aggression performed by self and others. We used a genetically encoded mirror-TRAP strategy to functionally interrogate these aggression-mirroring neurons. We find that their activity is essential for fighting and that forced activation of these cells triggers aggressive displays by mice, even toward their mirror image. Together, we have discovered a mirroring center in an evolutionarily ancient region that provides a subcortical cognitive substrate essential for a social behavior.


Asunto(s)
Agresión , Hipotálamo , Neuronas Espejo , Animales , Ratones , Agresión/fisiología , Hipotálamo/citología , Conducta Social
3.
Neuron ; 111(6): 787-796.e4, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36708707

RESUMEN

Prairie voles are among a small group of mammals that display long-term social attachment between mating partners. Many pharmacological studies show that signaling via the oxytocin receptor (Oxtr) is critical for the display of social monogamy in these animals. We used CRISPR mutagenesis to generate three different Oxtr-null mutant prairie vole lines. Oxtr mutants displayed social attachment such that males and females showed a behavioral preference for their mating partners over a stranger of the opposite sex, even when assayed using different experimental setups. Mothers lacking Oxtr delivered viable pups, and parents displayed care for their young and raised them to the weanling stage. Together, our studies unexpectedly reveal that social attachment, parturition, and parental behavior can occur in the absence of Oxtr signaling in prairie voles.


Asunto(s)
Pradera , Receptores de Oxitocina , Animales , Masculino , Femenino , Receptores de Oxitocina/genética , Oxitocina , Mamíferos , Arvicolinae , Conducta Social
4.
Cell ; 185(4): 654-671.e22, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-35065713

RESUMEN

Sex hormones exert a profound influence on gendered behaviors. How individual sex hormone-responsive neuronal populations regulate diverse sex-typical behaviors is unclear. We performed orthogonal, genetically targeted sequencing of four estrogen receptor 1-expressing (Esr1+) populations and identified 1,415 genes expressed differentially between sexes or estrous states. Unique subsets of these genes were distributed across all 137 transcriptomically defined Esr1+ cell types, including estrous stage-specific ones, that comprise the four populations. We used differentially expressed genes labeling single Esr1+ cell types as entry points to functionally characterize two such cell types, BNSTprTac1/Esr1 and VMHvlCckar/Esr1. We observed that these two cell types, but not the other Esr1+ cell types in these populations, are essential for sex recognition in males and mating in females, respectively. Furthermore, VMHvlCckar/Esr1 cell type projections are distinct from those of other VMHvlEsr1 cell types. Together, projection and functional specialization of dimorphic cell types enables sex hormone-responsive populations to regulate diverse social behaviors.


Asunto(s)
Ciclo Estral/genética , Regulación de la Expresión Génica , Caracteres Sexuales , Conducta Sexual Animal/fisiología , Agresión , Animales , Aromatasa/metabolismo , Trastorno Autístico/genética , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neuronas/metabolismo , Conducta Social
5.
Cell ; 179(6): 1393-1408.e16, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31735496

RESUMEN

Behaviors are inextricably linked to internal state. We have identified a neural mechanism that links female sexual behavior with the estrus, the ovulatory phase of the estrous cycle. We find that progesterone-receptor (PR)-expressing neurons in the ventromedial hypothalamus (VMH) are active and required during this behavior. Activating these neurons, however, does not elicit sexual behavior in non-estrus females. We show that projections of PR+ VMH neurons to the anteroventral periventricular (AVPV) nucleus change across the 5-day mouse estrous cycle, with ∼3-fold more termini and functional connections during estrus. This cyclic increase in connectivity is found in adult females, but not males, and regulated by estrogen signaling in PR+ VMH neurons. We further show that these connections are essential for sexual behavior in receptive females. Thus, estrogen-regulated structural plasticity of behaviorally salient connections in the adult female brain links sexual behavior to the estrus phase of the estrous cycle.


Asunto(s)
Red Nerviosa/fisiología , Conducta Sexual Animal/fisiología , Animales , Estrógenos/metabolismo , Ciclo Estral/efectos de los fármacos , Femenino , Hormonas Esteroides Gonadales/farmacología , Hipotálamo Anterior/fisiología , Masculino , Ratones Endogámicos C57BL , Red Nerviosa/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ovario/metabolismo , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/metabolismo , Receptores de Progesterona/metabolismo , Conducta Sexual Animal/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
6.
Cell Rep ; 27(9): 2527-2536.e4, 2019 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-31141679

RESUMEN

Genetically wired neural mechanisms inhibit mating between species because even naive animals rarely mate with other species. These mechanisms can evolve through changes in expression or function of key genes in sensory pathways or central circuits. Gr32a is a gustatory chemoreceptor that, in D. melanogaster, is essential to inhibit interspecies courtship and sense quinine. Similar to D. melanogaster, we find that D. simulans Gr32a is expressed in foreleg tarsi, sensorimotor appendages that inhibit interspecies courtship, and it is required to sense quinine. Nevertheless, Gr32a is not required to inhibit interspecies mating by D. simulans males. However, and similar to its function in D. melanogaster, Ppk25, a member of the Pickpocket family, promotes conspecific courtship in D. simulans. Together, we have identified distinct evolutionary mechanisms underlying chemosensory control of taste and courtship in closely related Drosophila species.


Asunto(s)
Evolución Biológica , Cortejo/psicología , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/fisiología , Drosophila simulans/fisiología , Receptores de Superficie Celular/metabolismo , Conducta Sexual Animal , Gusto/fisiología , Animales , Comunicación Celular , Células Quimiorreceptoras , Proteínas de Drosophila/genética , Femenino , Masculino , Feromonas , Receptores de Superficie Celular/genética , Reproducción
7.
Cell ; 176(5): 1190-1205.e20, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30712868

RESUMEN

Sexually naive animals have to distinguish between the sexes because they show species-typical interactions with males and females without meaningful prior experience. However, central neural pathways in naive mammals that recognize sex of other individuals remain poorly characterized. We examined the role of the principal component of the bed nucleus of stria terminalis (BNSTpr), a limbic center, in social interactions in mice. We find that activity of aromatase-expressing BNSTpr (AB) neurons appears to encode sex of other animals and subsequent displays of mating in sexually naive males. Silencing these neurons in males eliminates preference for female pheromones and abrogates mating success, whereas activating them even transiently promotes male-male mating. Surprisingly, female AB neurons do not appear to control sex recognition, mating, or maternal aggression. In summary, AB neurons represent sex of other animals and govern ensuing social behaviors in sexually naive males.


Asunto(s)
Sistema Límbico/metabolismo , Núcleos Septales/fisiología , Conducta Sexual Animal/fisiología , Amígdala del Cerebelo/fisiología , Animales , Aromatasa/metabolismo , Encéfalo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Vías Nerviosas/metabolismo , Neuronas/metabolismo , Feromonas/metabolismo , Caracteres Sexuales , Conducta Social
8.
Curr Opin Neurobiol ; 53: 192-197, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30316066

RESUMEN

A long-standing goal in developmental neuroscience is to understand the mechanisms by which steroid sex hormones pattern the mammalian central nervous system along male or female pathways to enable subsequent displays of sexually dimorphic behaviors. In this article, we review recent advances in understanding the epigenetic and transcriptional mechanisms mediating sexual differentiation of the brain in mammals, flies, and worms. These studies suggest a model of sexual differentiation wherein master regulators of sex determination initiate a cascade of sexually dimorphic gene expression that controls development of neural pathways and behavioral displays in a strikingly modular manner. With these advances in molecular genetics, it is now feasible to disassemble different components of sexually dimorphic social behaviors without disrupting other behavioral interactions. Such experimental tractability promises rapid advances in this exciting field.


Asunto(s)
Conducta Animal/fisiología , Encéfalo/fisiología , Epigénesis Genética/fisiología , Hormonas Esteroides Gonadales/fisiología , Células Receptoras Sensoriales/fisiología , Diferenciación Sexual/fisiología , Conducta Social , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Hormonas Esteroides Gonadales/metabolismo
9.
Curr Opin Physiol ; 6: 89-95, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31535059

RESUMEN

Sexually reproducing animals display sex differences in behavior. Although many of these sex differences in behavior are acquired with experience, sexually dimorphic behaviors such as mating and aggression are innate in the sense that they can be displayed without prior training or experience. In this review, we present recent advances in our understanding of the neural control of such innate sexually dimorphic social behaviors, with a focus on sexual behavior and aggression in flies and mice. We provide a brief overview of fundamental processes that regulate sexual differentiation in these animals to provide a framework within which more recent advances can be understood. We discuss advances in sensory, neuromodulatory, neural circuit, and experiential regulation of sexually dimorphic social behaviors.

10.
Neuron ; 95(4): 955-970.e4, 2017 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-28757304

RESUMEN

How environmental and physiological signals interact to influence neural circuits underlying developmentally programmed social interactions such as male territorial aggression is poorly understood. We have tested the influence of sensory cues, social context, and sex hormones on progesterone receptor (PR)-expressing neurons in the ventromedial hypothalamus (VMH) that are critical for male territorial aggression. We find that these neurons can drive aggressive displays in solitary males independent of pheromonal input, gonadal hormones, opponents, or social context. By contrast, these neurons cannot elicit aggression in socially housed males that intrude in another male's territory unless their pheromone-sensing is disabled. This modulation of aggression cannot be accounted for by linear integration of environmental and physiological signals. Together, our studies suggest that fundamentally non-linear computations enable social context to exert a dominant influence on developmentally hard-wired hypothalamus-mediated male territorial aggression.


Asunto(s)
Agresión/fisiología , Hipotálamo/citología , Hipotálamo/fisiología , Neuronas/fisiología , Conducta Social , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Adenoviridae/genética , Animales , Antipsicóticos/farmacología , Clozapina/análogos & derivados , Clozapina/farmacología , Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Femenino , Técnicas In Vitro , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Técnicas de Placa-Clamp , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo , Factores Sexuales , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo
11.
Nat Neurosci ; 20(7): 978-986, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28504672

RESUMEN

Learning to vocalize depends on the ability to adaptively modify the temporal and spectral features of vocal elements. Neurons that convey motor-related signals to the auditory system are theorized to facilitate vocal learning, but the identity and function of such neurons remain unknown. Here we identify a previously unknown neuron type in the songbird brain that transmits vocal motor signals to the auditory cortex. Genetically ablating these neurons in juveniles disrupted their ability to imitate features of an adult tutor's song. Ablating these neurons in adults had little effect on previously learned songs but interfered with their ability to adaptively modify the duration of vocal elements and largely prevented the degradation of songs' temporal features that is normally caused by deafening. These findings identify a motor to auditory circuit essential to vocal imitation and to the adaptive modification of vocal timing.


Asunto(s)
Corteza Auditiva/fisiología , Vías Auditivas/fisiología , Aprendizaje/fisiología , Telencéfalo/fisiología , Vocalización Animal/fisiología , Envejecimiento/fisiología , Animales , Animales Modificados Genéticamente , Recuento de Células , Sordera/fisiopatología , Pinzones , Masculino , Vías Nerviosas/fisiología , Técnicas de Trazados de Vías Neuroanatómicas , Neuronas/fisiología
12.
Curr Opin Neurobiol ; 38: 89-95, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27162162

RESUMEN

Sexually reproducing animals exhibit sex differences in behavior. Sexual dimorphisms in mating, aggression, and parental care directly contribute to reproductive success of the individual and survival of progeny. In this review, we discuss recent advances in our understanding of the molecular and neural network mechanisms underlying these behaviors in mice. Notable advances include novel insights into the sensory control of social interactions and the identification of molecularly-specified neuronal populations in the brain that control mating, aggression, and parental behaviors. In the case of the latter, these advances mark a watershed because scientists can now focus on discrete neural pathways in an effort to understand how the brain encodes these fundamental social behaviors.


Asunto(s)
Fenómenos Fisiológicos del Sistema Nervioso , Conducta Social , Agresión/fisiología , Animales , Ratones , Caracteres Sexuales , Conducta Sexual Animal/fisiología
13.
PLoS One ; 11(2): e0149501, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26895233

RESUMEN

To understand visual functions mediated by intrinsically photosensitive melanopsin-expressing retinal ganglion cells (mRGCs), it is important to elucidate axonal projections from these cells into the brain. Initial studies reported that melanopsin is expressed only in retinal ganglion cells within the eye. However, recent studies in Opn4-Cre mice revealed Cre-mediated marker expression in multiple brain areas. These discoveries complicate the use of melanopsin-driven genetic labeling techniques to identify retinofugal projections specifically from mRGCs. To restrict labeling to mRGCs, we developed a recombinant adeno-associated virus (AAV) carrying a Cre-dependent reporter (human placental alkaline phosphatase) that was injected into the vitreous of Opn4-Cre mouse eyes. The labeling observed in the brain of these mice was necessarily restricted specifically to retinofugal projections from mRGCs in the injected eye. We found that mRGCs innervate multiple nuclei in the basal forebrain, hypothalamus, amygdala, thalamus and midbrain. Midline structures tended to be bilaterally innervated, whereas the lateral structures received mostly contralateral innervation. As validation of our approach, we found projection patterns largely corresponded with previously published results; however, we have also identified a few novel targets. Our discovery of projections to the central amygdala suggests a possible direct neural pathway for aversive responses to light in neonates. In addition, projections to the accessory optic system suggest that mRGCs play a direct role in visual tracking, responses that were previously attributed to other classes of retinal ganglion cells. Moreover, projections to the zona incerta raise the possibility that mRGCs could regulate visceral and sensory functions. However, additional studies are needed to investigate the actual photosensitivity of mRGCs that project to the different brain areas. Also, there is a concern of "overlabeling" with very sensitive reporters that uncover low levels of expression. Light-evoked signaling from these cells must be shown to be of sufficient sensitivity to elicit physiologically relevant responses.


Asunto(s)
Retina/metabolismo , Células Ganglionares de la Retina/metabolismo , Opsinas de Bastones/biosíntesis , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Dependovirus/genética , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Genes Reporteros , Humanos , Inyecciones Intraoculares , Integrasas/genética , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Ratones Endogámicos C57BL , Retina/citología
14.
Philos Trans R Soc Lond B Biol Sci ; 371(1688): 20150109, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26833830

RESUMEN

The unique hormonal, genetic and epigenetic environments of males and females during development and adulthood shape the neural circuitry of the brain. These differences in neural circuitry result in sex-typical displays of social behaviours such as mating and aggression. Like other neural circuits, those underlying sex-typical social behaviours weave through complex brain regions that control a variety of diverse behaviours. For this reason, the functional dissection of neural circuits underlying sex-typical social behaviours has proved to be difficult. However, molecularly discrete neuronal subpopulations can be identified in the heterogeneous brain regions that control sex-typical social behaviours. In addition, the actions of oestrogens and androgens produce sex differences in gene expression within these brain regions, thereby highlighting the neuronal subpopulations most likely to control sexually dimorphic social behaviours. These conditions permit the implementation of innovative genetic approaches that, in mammals, are most highly advanced in the laboratory mouse. Such approaches have greatly advanced our understanding of the functional significance of sexually dimorphic neural circuits in the brain. In this review, we discuss the neural circuitry of sex-typical social behaviours in mice while highlighting the genetic technical innovations that have advanced the field.


Asunto(s)
Encéfalo/citología , Encéfalo/fisiología , Hormonas Esteroides Gonadales/fisiología , Neuronas/fisiología , Conducta Social , Animales , Femenino , Humanos , Masculino , Factores Sexuales
15.
Mol Metab ; 4(11): 857-66, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26629409

RESUMEN

OBJECTIVES: The ventromedial hypothalamic nucleus (VMH) regulates energy homeostasis as well as social and emotional behaviors. Nearly all VMH neurons, including those in the sexually dimorphic ventrolateral VMH (VMHvl) subregion, release the excitatory neurotransmitter glutamate and use the vesicular glutamate transporter 2 (Vglut2). Here, we asked how glutamatergic signaling contributes to the collective metabolic and behavioral responses attributed to the VMH and VMHvl. METHODS: Using Sf1-Cre and a Vglut2 floxed allele, Vglut2 was knocked-out in SF-1 VMH neurons (Vglut2 (Sf1-Cre) ). Metabolic and neurobehavioral assays were carried out initially on Vglut2 (fl/fl) and Vglut2 (Sf1-Cre) mice in a mixed, and then in the C57BL/6 genetic background, which is prone to hyperglycemia and diet induced obesity (DIO). RESULTS: Several phenotypes observed in Vglut2 (Sf1-Cre) mice were largely unexpected based on prior studies that have perturbed VMH development or VMH glutamate signaling. In our hands, Vglut2 (Sf1-Cre) mice failed to exhibit the anticipated increase in body weight after high fat diet (HFD) or the impaired glucose homeostasis after fasting. Instead, there was a significant sex-dependent attenuation of DIO in Vglut2 (Sf1-Cre) females. Vglut2 (Sf1-Cre) males also display a sex-specific loss of conditioned-fear responses and aggression accompanied by more novelty-associated locomotion. Finally, unlike the higher anxiety noted in Sf1 (Nestin-Cre) mice that lack a fully formed VMH, both male and female Vglut2 (Sf1-Cre) mice were less anxious. CONCLUSIONS: Loss of VMH glutamatergic signaling sharply decreased DIO in females, attenuated aggression and learned fear in males, and was anxiolytic in males and females. Collectively, our findings demonstrate that while glutamatergic output from the VMH appears largely dispensable for counter regulatory responses to hypoglycemia, it drives sex-dependent differences in metabolism and social behaviors and is essential for adaptive responses to anxiety-provoking stimuli in both sexes.

16.
Neuron ; 86(2): 403-16, 2015 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-25864637

RESUMEN

The hypothalamus integrates information required for the production of a variety of innate behaviors such as feeding, mating, aggression, and predator avoidance. Despite an extensive knowledge of hypothalamic function, how embryonic genetic programs specify circuits that regulate these behaviors remains unknown. Here, we find that in the hypothalamus the developmentally regulated homeodomain-containing transcription factor Dbx1 is required for the generation of specific subclasses of neurons within the lateral hypothalamic area/zona incerta (LH) and the arcuate (Arc) nucleus. Consistent with this specific developmental role, Dbx1 hypothalamic-specific conditional-knockout mice display attenuated responses to predator odor and feeding stressors but do not display deficits in other innate behaviors such as mating or conspecific aggression. Thus, activity of a single developmentally regulated gene, Dbx1, is a shared requirement for the specification of hypothalamic nuclei governing a subset of innate behaviors. VIDEO ABSTRACT.


Asunto(s)
Conducta Animal/fisiología , Proteínas de Homeodominio/genética , Hipotálamo/embriología , Hipotálamo/fisiología , Instinto , Animales , Tipificación del Cuerpo/genética , Conducta Alimentaria/fisiología , Femenino , Expresión Génica , Proteínas de Homeodominio/metabolismo , Hipotálamo/citología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Neuropéptidos/metabolismo , Orexinas
17.
Int J Oral Sci ; 7(1): 23-6, 2015 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-25634121

RESUMEN

Continuously growing incisors are common to all rodents, which include the Microtus genus of voles. However, unlike many rodents, voles also possess continuously growing molars. Here, we report spontaneous molar defects in a population of Prairie voles (Microtus ochrogaster). We identified bilateral protuberances on the ventral surface of the mandible in several voles in our colony. In some cases, the protuberances broke through the cortical bone. The mandibular molars became exposed and infected, and the maxillary molars entered the cranial vault. Visualisation upon soft tissue removal and microcomputed tomography (microCT) analyses confirmed that the protuberances were caused by the overgrowth of the apical ends of the molar teeth. We speculate that the unrestricted growth of the molars was due to the misregulation of the molar dental stem cell niche. Further study of this molar phenotype may yield additional insight into stem cell regulation and the evolution and development of continuously growing teeth.


Asunto(s)
Arvicolinae/anatomía & histología , Diente Molar/crecimiento & desarrollo , Animales , Arvicolinae/genética , Femenino , Humanos , Masculino , Diente Molar/diagnóstico por imagen , Linaje , Microtomografía por Rayos X
18.
Cell Rep ; 10(4): 453-62, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25620703

RESUMEN

Aromatase-expressing neuroendocrine neurons in the vertebrate male brain synthesize estradiol from circulating testosterone. This locally produced estradiol controls neural circuits underlying courtship vocalization, mating, aggression, and territory marking in male mice. How aromatase-expressing neuronal populations control these diverse estrogen-dependent male behaviors is poorly understood, and the function, if any, of aromatase-expressing neurons in females is unclear. Using targeted genetic approaches, we show that aromatase-expressing neurons within the male posterodorsal medial amygdala (MeApd) regulate components of aggression, but not other estrogen-dependent male-typical behaviors. Remarkably, aromatase-expressing MeApd neurons in females are specifically required for components of maternal aggression, which we show is distinct from intermale aggression in pattern and execution. Thus, aromatase-expressing MeApd neurons control distinct forms of aggression in the two sexes. Moreover, our findings indicate that complex social behaviors are separable in a modular manner at the level of genetically identified neuronal populations.


Asunto(s)
Agresión/fisiología , Aromatasa/metabolismo , Neuronas/enzimología , Neuronas/metabolismo , Animales , Conducta Animal/fisiología , Femenino , Masculino , Ratones , Conducta Sexual Animal/fisiología
19.
Methods Enzymol ; 544: 179-213, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24974291

RESUMEN

Caspases, aspartate-specific cysteine proteases, have fate-determining roles in many cellular processes including apoptosis, differentiation, neuronal remodeling, and inflammation (for review, see Yuan & Kroemer, 2010). There are a dozen caspases in humans alone, yet their individual contributions toward these phenotypes are not well understood. Thus, there has been considerable interest in activating individual caspases or using their activity to drive these processes in cells and animals. We envision that such experimental control of caspase activity can not only afford novel insights into fundamental biological problems but may also enable new models for disease and suggest possible routes to therapeutic intervention. In particular, localized, genetic, and small-molecule-controlled caspase activation has the potential to target the desired cell type in a tissue. Suppression of caspase activation is one of the hallmarks of cancer and thus there has been significant enthusiasm for generating selective small-molecule activators that could bypass upstream mutational events that prevent apoptosis. Here, we provide a practical guide that investigators have devised, using genetics or small molecules, to activate specific caspases in cells or animals. Additionally, we show genetically controlled activation of an executioner caspase to target the function of a defined group of neurons in the adult mammalian brain.


Asunto(s)
Caspasas/genética , Caspasas/metabolismo , Activación Enzimática/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Apoptosis/efectos de los fármacos , Caspasas/análisis , Ingeniería Celular/métodos , Clonación Molecular/métodos , Humanos , Modelos Moleculares , Neuronas/citología , Neuronas/enzimología , Neuronas/metabolismo , Multimerización de Proteína/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química
20.
Neuron ; 82(2): 261-78, 2014 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-24742456

RESUMEN

Sexually dimorphic behaviors, qualitative or quantitative differences in behaviors between the sexes, result from the activity of a sexually differentiated nervous system. Sensory cues and sex hormones control the entire repertoire of sexually dimorphic behaviors, including those commonly thought to be charged with emotion such as courtship and aggression. Such overarching control mechanisms regulate distinct genes and neurons that in turn specify the display of these behaviors in a modular manner. How such modular control is transformed into cohesive internal states that correspond to sexually dimorphic behavior is poorly understood. We summarize current understanding of the neural circuit control of sexually dimorphic behaviors from several perspectives, including how neural circuits in general, and sexually dimorphic neurons in particular, can generate sexually dimorphic behaviors, and how molecular mechanisms and evolutionary constraints shape these behaviors. We propose that emergent themes such as the modular genetic and neural control of dimorphic behavior are broadly applicable to the neural control of other behaviors.


Asunto(s)
Encéfalo/fisiología , Caracteres Sexuales , Conducta Sexual/fisiología , Animales , Encéfalo/citología , Humanos , Vías Nerviosas/fisiología , Neuronas/fisiología , Conducta Sexual Animal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...