Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38798336

RESUMEN

Pair bonds powerfully modulate health, which becomes particularly important when facing the detrimental effects of aging. To examine the impact of aging on relationship formation and response to loss, we examined behavior in 6-, 12-, and 18-month male and female prairie voles, a monogamous species that forms mating-based pair bonds. We found that older males (18-months) bonded quicker than younger voles, while similarly aged female voles increased partner directed affiliative behaviors. Supporting sex differences in bonding behaviors, we found that males were more likely to sample both partner and novel voles while females were more likely to display partner preference during the initial 20 minutes of the test. Using partner separation to study loss, we observed an erosion of partner preference only in 12-month females, but an overall decrease in partner-directed affiliation in females across all groups, but not in males. Finally, we found that the number of oxytocin, but not vasopressin, cells in the paraventricular hypothalamus increased during aging. These results establish prairie voles as a novel model to study the effects of normal and abnormal aging on pair bonding. Highlights: 18-month male voles demonstrate accelerated bond formation18-month female voles increase partner-directed huddling after 2 wksBonds erode faster in 12-month female voles after partner separationFemale behavior from partner preference tests is reflected in free interactionThe number of paraventricular hypothalamus oxytocin cells increase during aging.

3.
Ann N Y Acad Sci ; 1535(1): 20-30, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38594916

RESUMEN

Loss of a loved one is a painful event that substantially elevates the risk for physical and mental illness and impaired daily function. Socially monogamous prairie voles are laboratory-amenable rodents that form life-long pair bonds and exhibit distress upon partner separation, mirroring phenotypes seen in humans. These attributes make voles an excellent model for studying the biology of loss. In this review, we highlight parallels between humans and prairie voles, focusing on reward system engagement during pair bonding and loss. As yearning is a unique feature that differentiates loss from other negative mental states, we posit a model in which the homeostatic reward mechanisms that help to maintain bonds are disrupted upon loss, resulting in yearning and other negative impacts. Finally, we synthesize studies in humans and voles that delineate the remodeling of reward systems during loss adaptation. The stalling of these processes likely contributes to prolonged grief disorder, a diagnosis recently added to the Diagnostic and Statistical Manual for Psychiatry.


Asunto(s)
Arvicolinae , Recompensa , Animales , Arvicolinae/fisiología , Humanos , Adaptación Psicológica/fisiología , Pesar , Apareamiento , Modelos Animales
4.
Curr Biol ; 34(3): 519-530.e5, 2024 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-38218185

RESUMEN

In monogamous species, prosocial behaviors directed toward partners are dramatically different from those directed toward unknown individuals and potential threats. Dopamine release in the nucleus accumbens has a well-established role in social reward and motivation, but how this mechanism may be engaged to drive the highly divergent social behaviors directed at a partner or unfamiliar conspecific remains unknown. Using monogamous prairie voles, we first employed receptor pharmacology in partner preference and social operant tasks to show that dopamine is critical for the appetitive drive for social interaction but not for low-effort, unconditioned consummatory behaviors. We then leveraged the subsecond temporal resolution of the fluorescent biosensor, GRABDA, to ask whether differential dopamine release might distinguish between partner and novel social access and interaction. We found that partner seeking, anticipation, and interaction resulted in more accumbal dopamine release than the same events directed toward a novel vole. Further, partner-associated dopamine release decreased after prolonged partner separation. Our results are consistent with a model in which dopamine signaling plays a prominent role in the appetitive aspects of social interactions. Within this framework, differences in partner- and novel-associated dopamine release reflect the selective nature of pair bonds and may drive the partner- and novel-directed social behaviors that reinforce and cement bonds over time. This provides a potential mechanism by which highly conserved reward systems can enable selective, species-appropriate social behaviors.


Asunto(s)
Núcleo Accumbens , Apareamiento , Humanos , Animales , Dopamina , Conducta Social , Motivación , Arvicolinae
6.
Curr Protoc ; 3(5): e763, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37184156

RESUMEN

Photometry approaches detect sensor-mediated changes in fluorescence as a proxy for rapid molecular changes within the brain. As a flexible technique with a relatively low cost to implement, photometry is rapidly being incorporated into neuroscience laboratories. Yet, although multiple data acquisition systems for photometry now exist, robust analytical pipelines for the resulting data remain limited. Here we present the Photometry Analysis Toolkit (PhAT)-a free open-source analysis pipeline that provides options for signal normalization, incorporation of multiple data streams to align photometry data with behavior and other events, calculation of event-related changes in fluorescence, and comparison of similarity across fluorescent traces. A graphical user interface (GUI) enables use of this software without prior coding knowledge. In addition to providing foundational analytical tools, PhAT is designed to readily incorporate community-driven development of new modules for more bespoke analyses, and enables data to be easily exported to enable subsequent statistical testing and/or code-based analyses. In addition, we provide recommendations regarding technical aspects of photometry experiments, including sensor selection and validation, reference signal considerations, and best practices for experimental design and data collection. We hope that the distribution of this software and protocols will lower the barrier to entry for new photometry users and improve the quality of collected data, increasing transparency and reproducibility in photometry analyses. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Software and environment installation Alternate Protocol 1: Software and environment update Basic Protocol 2: GUI-driven fiber photometry analysis Support Protocol 1: Examining signal quality Support Protocol 2: Interacting with graphs Basic Protocol 3: Adding modules to PhAT Alternate Protocol 2: Creating functions for use in Jupyter Notebook.


Asunto(s)
Encéfalo , Programas Informáticos , Reproducibilidad de los Resultados
7.
bioRxiv ; 2023 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-36993180

RESUMEN

Photometry approaches detect sensor-mediated changes in fluorescence as a proxy for rapid molecular changes within the brain. As a flexible technique with a relatively low cost to implement, photometry is rapidly being incorporated into neuroscience laboratories. While multiple data acquisition systems for photometry now exist, robust analytical pipelines for the resulting data remain limited. Here we present the Ph otometry A nalysis T oolkit (PhAT) - a free open source analysis pipeline that provides options for signal normalization, incorporation of multiple data streams to align photometry data with behavior and other events, calculation of event-related changes in fluorescence, and comparison of similarity across fluorescent traces. A graphical user interface (GUI) enables use of this software without prior coding knowledge. In addition to providing foundational analytical tools, PhAT is designed to readily incorporate community-driven development of new modules for more bespoke analyses, and data can be easily exported to enable subsequent statistical testing and/or code-based analyses. In addition, we provide recommendations regarding technical aspects of photometry experiments including sensor selection and validation, reference signal considerations, and best practices for experimental design and data collection. We hope that the distribution of this software and protocol will lower the barrier to entry for new photometry users and improve the quality of collected data, increasing transparency and reproducibility in photometry analyses. Basic Protocol 1: Software Environment InstallationBasic Protocol 2: GUI-driven Fiber Photometry AnalysisBasic Protocol 3: Adding Modules.

8.
Elife ; 122023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36852906

RESUMEN

The loss of a spouse is often cited as the most traumatic event in a person's life. However, for most people, the severity of grief and its maladaptive effects subside over time via an understudied adaptive process. Like humans, socially monogamous prairie voles (Microtus ochrogaster) form opposite-sex pair bonds, and upon partner separation, show stress phenotypes that diminish over time. We test the hypothesis that extended partner separation diminishes pair bond-associated behaviors and causes pair bond transcriptional signatures to erode. Opposite-sex or same-sex paired males were cohoused for 2 weeks and then either remained paired or were separated for 48 hours or 4 weeks before collecting fresh nucleus accumbens tissue for RNAseq. In a separate cohort, we assessed partner-directed affiliation at these time points. We found that these behaviors persist despite prolonged separation in both same-sex and opposite-sex paired voles. Opposite-sex pair bonding led to changes in accumbal transcription that were stably maintained while animals remained paired but eroded following prolonged partner separation. Eroded genes are associated with gliogenesis and myelination, suggesting a previously undescribed role for glia in pair bonding and loss. Further, we pioneered neuron-specific translating ribosomal affinity purification in voles. Neuronally enriched transcriptional changes revealed dopaminergic-, mitochondrial-, and steroid hormone signaling-associated gene clusters sensitive to acute pair bond disruption and loss adaptation. Our results suggest that partner separation erodes transcriptomic signatures of pair bonding despite core behavioral features of the bond remaining intact, revealing potential molecular processes priming a vole to be able to form a new bond.


Losing a spouse or life partner is a deeply traumatic event that can have long-term repercussions. Given enough time, however, most surviving partners are able to process their grief. The neural processes that enable people to adapt to their loss remain unknown. To explore this question, scientists often turn to animals that form long-term mating based pair bonds and can be raised in the laboratory. Monogamous prairie voles enter lifelong partnerships where the two individuals live together, prefer to cuddle with each other, and take care of their pups as a team. After having lost their mate, they show signs of distress that eventually subside with time. Sadino et al. examined the biological impact of partner loss in these animals by focusing on the nucleus accumbens, a brain region important for social connections. This involved tracking gene expression ­ which genes were switched on and off in this area ­ as the voles established their pair bonds, and then at different time points after one of the partners had been removed. The experiments revealed that establishing a relationship leads to a stable shift in nucleus accumbens gene expression, which may help maintain bonds over time. In particular, genes related to glia (the non-neuronal cells which assist neurons in their tasks) see their expression levels increase, indicating a previously undescribed role for this cell type in regulating pair bonding. Having their partner removed led to an erosion of the gene expression pattern that had emerged during pair bonding; this may help the remaining vole adapt to its loss and go on to form a new bond. In addition, Sadino et al. explored the gene expression of only neurons in the nucleus accumbens and uncovered biological processes distinct from those that occur in glia after partner separation. Together, these results shed light on the genetic and neuronal mechanisms which underlie adaptation to loss; this knowledge could one day inform how to better support individuals during this time.


Asunto(s)
Pradera , Núcleo Accumbens , Animales , Humanos , Masculino , Apareamiento , Arvicolinae/genética , Proteínas de Unión al ADN , Conducta Social
9.
Trends Neurosci ; 46(4): 260-262, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36369029

RESUMEN

In monogamous species, pair bonding leads to striking changes in social behavior and neural circuitry. We outline the cognitive building blocks of monogamous pair bonding in prairie voles (Microtus ochrogaster), as well as opportunities afforded by the species to investigate diverse mechanisms underlying social experience-dependent plasticity and gain insights into the neurobiology of complex social behavior more generally.


Asunto(s)
Arvicolinae , Encéfalo , Plasticidad Neuronal , Apareamiento , Conducta Social , Animales , Arvicolinae/fisiología , Arvicolinae/psicología , Encéfalo/fisiología , Plasticidad Neuronal/fisiología , Masculino , Femenino
11.
Genes Brain Behav ; 21(3): e12790, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35044087

RESUMEN

The rewarding properties of social interactions facilitate relationship formation and maintenance. Prairie voles are one of the few laboratory species that form selective relationships, manifested as "partner preferences" for familiar partners versus strangers. While both sexes exhibit strong partner preferences, this similarity in outward behavior likely results from sex-specific neurobiological mechanisms. We recently demonstrated that in operant trials, females worked hardest for access to familiar conspecifics of either sex, while males worked equally hard for access to any female, indicating a sex difference in social motivation. As tests were performed with one social target at a time, males might have experienced a ceiling effect, and familiar females might be more relatively rewarding in a choice scenario. Here we performed an operant social choice task in which voles lever-pressed to gain temporary access to either the chamber containing their mate or one containing a novel opposite-sex vole. Females worked hardest to access their mate, while males pressed at similar rates for either female. Individual male behavior was heterogeneous, congruent with multiple mating strategies in the wild. Voles exhibited preferences for favorable over unfavorable environments in a non-social operant task, indicating that lack of social preference does not reflect lack of discrimination. Natural variation in oxytocin receptor genotype at the intronic single nucleotide polymorphism NT213739 was associated with oxytocin receptor density, and predicted individual variation in stranger-directed aggressive behavior. These findings suggest that convergent preference behavior in male and female voles results from sex-divergent pathways, particularly in the realm of social motivation.


Asunto(s)
Receptores de Oxitocina , Caracteres Sexuales , Animales , Arvicolinae/genética , Femenino , Pradera , Masculino , Receptores de Oxitocina/metabolismo , Recompensa , Conducta Sexual Animal , Conducta Social
12.
Genes Brain Behav ; 21(3): e12786, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35044090

RESUMEN

In pair bonding animals, coordinated behavior between partners is required for the pair to accomplish shared goals such as raising young. Despite this, experimental designs rarely assess the behavior of both partners within a bonded pair. Thus, we lack an understanding of the interdependent behavioral dynamics between partners that likely facilitate relationship success. To identify intra-pair behavioral correlates of pair bonding, we used socially monogamous prairie voles (Microtus ochrogaster) and tested both partners using social choice and non-choice tests at short- and long-term pairing timepoints. Females developed a preference for their partner more rapidly than males, with preference driven by different behaviors in each sex. Further, as bonds matured, intra-pair behavioral sex differences and organized behavior emerged-females consistently huddled more with their partner than males did regardless of overall intra-pair affiliation levels. When animals were allowed to freely interact with a partner or a novel vole in sequential free interaction tests, pairs spent more time interacting together than either animal did with a novel vole, consistent with partner preference in the more commonly employed choice test. Total pair interaction in freely moving voles was correlated with female, but not male, behavior. Via a social operant paradigm, we found that pair-bonded females, but not males, are more motivated to access and huddle with their partner than a novel vole. Together, our data indicate that as pair bonds mature, sex differences and organized behavior emerge within pairs, and that these intra-pair behavioral changes are likely organized and driven by the female animal.


Asunto(s)
Pradera , Caracteres Sexuales , Animales , Arvicolinae , Proteínas de Unión al ADN , Femenino , Masculino , Conducta Sexual Animal , Conducta Social
13.
Nat Neurosci ; 24(7): 1035-1045, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33972800

RESUMEN

Advanced technologies for controlled delivery of light to targeted locations in biological tissues are essential to neuroscience research that applies optogenetics in animal models. Fully implantable, miniaturized devices with wireless control and power-harvesting strategies offer an appealing set of attributes in this context, particularly for studies that are incompatible with conventional fiber-optic approaches or battery-powered head stages. Limited programmable control and narrow options in illumination profiles constrain the use of existing devices. The results reported here overcome these drawbacks via two platforms, both with real-time user programmability over multiple independent light sources, in head-mounted and back-mounted designs. Engineering studies of the optoelectronic and thermal properties of these systems define their capabilities and key design considerations. Neuroscience applications demonstrate that induction of interbrain neuronal synchrony in the medial prefrontal cortex shapes social interaction within groups of mice, highlighting the power of real-time subject-specific programmability of the wireless optogenetic platforms introduced here.


Asunto(s)
Optogenética/instrumentación , Conducta Social , Tecnología Inalámbrica/instrumentación , Animales , Ratones
14.
Endocrinology ; 162(2)2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33367612

RESUMEN

Pair bonds represent some of the strongest attachments we form as humans. These relationships positively modulate health and well-being. Conversely, the loss of a spouse is an emotionally painful event that leads to numerous deleterious physiological effects, including increased risk for cardiac dysfunction and mental illness. Much of our understanding of the neuroendocrine basis of pair bonding has come from studies of monogamous prairie voles (Microtus ochrogaster), laboratory-amenable rodents that, unlike laboratory mice and rats, form lifelong pair bonds. Specifically, research using prairie voles has delineated a role for multiple neuromodulatory and neuroendocrine systems in the formation and maintenance of pair bonds, including the oxytocinergic, dopaminergic, and opioidergic systems. However, while these studies have contributed to our understanding of selective attachment, few studies have examined how interactions among these 3 systems may be essential for expression of complex social behaviors, such as pair bonding. Therefore, in this review, we focus on how the social neuropeptide, oxytocin, interacts with classical reward system modulators, including dopamine and endogenous opioids, during bond formation and maintenance. We argue that an understanding of these interactions has important clinical implications and is required to understand the evolution and encoding of complex social behaviors more generally. Finally, we provide a brief consideration of future directions, including a discussion of the possible roles that glia, specifically microglia, may have in modulating social behavior by acting as a functional regulator of these 3 neuromodulatory systems.


Asunto(s)
Arvicolinae/metabolismo , Dopamina/metabolismo , Microglía/metabolismo , Oxitocina/metabolismo , Apareamiento , Receptores Opioides/metabolismo , Animales , Femenino , Humanos , Masculino , Recompensa , Conducta Social
16.
Horm Behav ; 126: 104847, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32910950

RESUMEN

Monogamous prairie voles (Microtus ochrogaster) form mating-based pair bonds. Although wild prairie voles rarely re-pair following loss of a partner, laboratory studies have shown that previous pairing and mating does not negate the ability to form a new partner preference. However, little is known about how prior bond experience may alter the trajectory and display of a new pair bond. In the present study, we disrupted an initial pair bond by separating partners and then varied the amount of time before a new partner was introduced. We assessed how separation time affected the stability of partner preference over time and influenced decision-making in male voles performing a head-to-head partner preference test in which they chose between the first and second partner. We found that the ability to consistently display a preference for the second partner, supplanting the initial pair bond, depended on how long the test animal was separated from their first partner. Prior bonding experience also shaped the subsequent effects of mating on partner preference. Partner preference strength was sensitive to latency to mate with the second partner but not the first partner, irrespective of separation time. These results suggest that the ability to form a consistent, strong preference for a new partner after an initial pair bond depends upon the amount of time that has passed since separation from the first partner. These results provide valuable insight into how social bonds are dynamically shaped by prior social experience and identify variables that contribute to recovery from partner loss and the ability to form a new pair bond. They also delineate a behavioral trajectory essential for future work examining the hormonal and genetic changes that enable recovery from partner loss.


Asunto(s)
Arvicolinae/fisiología , Apareamiento , Conducta Sexual Animal/fisiología , Animales , Conducta de Elección/fisiología , Femenino , Masculino , Conducta Social , Factores de Tiempo
17.
Proc Natl Acad Sci U S A ; 117(20): 11076-11084, 2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32381740

RESUMEN

Pair-bond formation depends vitally on neuromodulatory signaling within the nucleus accumbens, but the neuronal dynamics underlying this behavior remain unclear. Using 1-photon in vivo Ca2+ imaging in monogamous prairie voles, we found that pair bonding does not elicit differences in overall nucleus accumbens Ca2+ activity. Instead, we identified distinct ensembles of neurons in this region that are recruited during approach to either a partner or a novel vole. The partner-approach neuronal ensemble increased in size following bond formation, and differences in the size of approach ensembles for partner and novel voles predict bond strength. In contrast, neurons comprising departure ensembles do not change over time and are not correlated with bond strength, indicating that ensemble plasticity is specific to partner approach. Furthermore, the neurons comprising partner and novel-approach ensembles are nonoverlapping while departure ensembles are more overlapping than chance, which may reflect another key feature of approach ensembles. We posit that the features of the partner-approach ensemble and its expansion upon bond formation potentially make it a key neuronal substrate associated with bond formation and maturation.


Asunto(s)
Neuronas/fisiología , Núcleo Accumbens/fisiología , Apareamiento , Conducta Sexual Animal/fisiología , Animales , Arvicolinae/fisiología , Femenino , Masculino , Preferencia en el Apareamiento Animal/fisiología , Núcleo Accumbens/diagnóstico por imagen , Conducta Social
18.
Neuropsychopharmacology ; 45(6): 1068-1077, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32035426

RESUMEN

Social buffering occurs when the presence of a companion attenuates the physiological and/or behavioral effects of a stressful or fear-provoking event. It represents a way in which social interactions can immediately and potently modulate behavior. As such, social buffering is one mechanism by which strong social support increases resilience to mental illness. Although the behavioral and neuroendocrine impacts of social buffering are well studied in multiple species, including humans, the neuronal underpinnings of this behavioral phenomenon remain largely unexplored. Previous work has shown that the infralimbic prefrontal cortex (IL-PFC) is important for processing social information and, in separate studies, for modulating fear and anxiety. Thus, we hypothesized that socially active cells within the IL-PFC may integrate social information to modulate fear responsivity. To test this hypothesis, we employed social buffering paradigms in male and female mice. Similar to prior studies in rats, we found that the presence of a cagemate reduced freezing in fear- and anxiety-provoking contexts. In accordance with previous work, we demonstrated that interaction with a novel or familiar conspecific induces activity in the IL-PFC as evidenced by increased immediate early gene (IEG) expression. We then utilized an activity-dependent tagging murine line, the ArcCreERT2 mice, to express channelrhodopsin (ChR2) in neurons active during the social encoding of a new cagemate. We found that optogenetic reactivation of these socially active neuronal ensembles phenocopied the effects of cagemate presence in male and female mice in learned and innate fear contexts without being inherently rewarding or altering locomotion. These data suggest that a social neural ensemble within the IL-PFC may contribute to social buffering of fear. These neurons may represent a novel therapeutic target for fear and anxiety disorders.


Asunto(s)
Optogenética , Conducta Social , Animales , Miedo , Femenino , Masculino , Ratones , Neuronas , Corteza Prefrontal , Ratas
19.
ACS Chem Neurosci ; 10(7): 3197-3206, 2019 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-30694044

RESUMEN

The serotonin 1A receptor (5-HT1A) system has been extensively implicated in modulating mood and behavior. Notably, 5-HT1A levels in humans display remarkable variation, and differences in receptor levels have been linked with a variety of psychiatric disorders. Further, reduction of receptor levels by 30-50% in mice suggests that changes in receptor levels that model existing human variation are sufficient to drive behavioral alterations. As a result, genetic mechanisms that modulate human 5-HT1A levels may be important for explaining individual differences in mood and behavior, representing a potential source of psychiatric disease risk. One common genetic variant implicated in differential 5-HT1A levels is the G/C single nucleotide polymorphism (SNP) rs6295, located upstream of the human 5-HT1A gene. This SNP differentially binds the transcription factor, NUDR/Deaf1, leading to cell-type specific effects on transcription in vitro. To investigate the direct effects of this SNP in the heterogeneous cellular context of the brain, we generated humanized transgenic mice using a design that maximized the local transcriptional landscape of the human HTR1A gene while also controlling for effects of genomic insertion location. We integrated a 180 kb human bacteria artificial chromosome (BAC) transgene containing G- and C-alleles of rs6295 flanked by FRT or loxP sites. Subsequent deletion of each allele by Cre- or Flp-recombinase resulted in rs6295G and C alleles in the same genomic location. These alleles were bred onto a 5-HT1A null mouse such that the human BAC was the sole source of 5-HT1A in these mice. We generated three separate lines, two of which had detectable human 5-HT1A levels in the brain, although none displayed expression in the raphe. Of these, one line exhibited rs6295-dependent differences in 5-HT1A levels and differences in behavior, even though the overall levels were considerably lower than native expression levels. The line-dependent effect of rs6295 on protein levels and behavior may depend upon differences in background genetic factors or different insertion sites across each line. This work confirms that relatively subtle differences in 5-HT1A levels can contribute to differences in behavior and highlights the challenges of modeling human noncoding genetic variation in mice.


Asunto(s)
Encéfalo/metabolismo , Depresión/genética , Trastorno Depresivo/genética , Polimorfismo de Nucleótido Simple , Receptor de Serotonina 5-HT1A/genética , Alelos , Animales , Conducta Animal/fisiología , Depresión/metabolismo , Trastorno Depresivo/metabolismo , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Actividad Motora/fisiología , Receptor de Serotonina 5-HT1A/metabolismo
20.
Sci Rep ; 8(1): 5788, 2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29636529

RESUMEN

The 5-HT1A autoreceptor mediates feedback inhibition of serotonin (5-HT) neurons, and is implicated in major depression. The human 5-HT1A gene (HTR1A) rs6295 risk allele prevents Deaf1 binding to HTR1A, resulting in increased 5-HT1A autoreceptor transcription. Since chronic stress alters HTR1A methylation and expression, we addressed whether recruitment of methyl-binding protein MeCP2 may alter Deaf1 regulation at the HTR1A locus. We show that MeCP2 enhances Deaf1 binding to its HTR1A site and co-immunoprecipitates with Deaf1 in cells and brain tissue. Chromatin immunoprecipitation assays showed Deaf1-dependent recruitment of MeCP2 to the mouse HTR1A promoter, and MeCP2 modulated human and mouse HTR1A gene transcription in a Deaf1-dependent fashion, enhancing Deaf1-induced repression at the Deaf1 site. To address the role of MeCP2 in HTR1A regulation in vivo, mice with conditional knockout of MeCP2 in adult 5-HT neurons (MeCP2 cKO) were generated. These mice exhibited increased 5-HT1A autoreceptor levels and function, consistent with MeCP2 enhancement of Deaf1 repression in 5-HT neurons. Interestingly, female MeCP2-cKO mice displayed reduced anxiety, while males showed increased anxiety and reduced depression-like behaviors. These data uncover a novel role for MeCP2 in 5-HT neurons to repress HTR1A expression and drive adult anxiety- and depression-like behaviors in a sex-specific manner.


Asunto(s)
Ansiedad/metabolismo , Depresión/metabolismo , Proteína 2 de Unión a Metil-CpG/metabolismo , Receptor de Serotonina 5-HT1A/genética , Neuronas Serotoninérgicas/metabolismo , Animales , Ansiedad/genética , Encéfalo/metabolismo , Línea Celular , Proteínas de Unión al ADN , Depresión/genética , Femenino , Regulación de la Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones , Ratones Noqueados , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Factores Sexuales , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...