Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Cell ; 187(6): 1354-1357, 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38490178

RESUMO

Our understanding of sex and gender evolves. We asked scientists about their work and the future of sex and gender research. They discuss, among other things, interdisciplinary collaboration, moving beyond binary conceptualizations, accounting for intersecting factors, reproductive strategies, expanding research on sex-related differences, and sex's dynamic nature.


Assuntos
Pesquisa Biomédica , Identidade de Gênero , Sexo , Feminino , Humanos , Masculino , Caracteres Sexuais
2.
Nature ; 599(7883): 131-135, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34646010

RESUMO

Oestrogen depletion in rodents and humans leads to inactivity, fat accumulation and diabetes1,2, underscoring the conserved metabolic benefits of oestrogen that inevitably decrease with age. In rodents, the preovulatory surge in 17ß-oestradiol (E2) temporarily increases energy expenditure to coordinate increased physical activity with peak sexual receptivity. Here we report that a subset of oestrogen-sensitive neurons in the ventrolateral ventromedial hypothalamic nucleus (VMHvl)3-7 projects to arousal centres in the hippocampus and hindbrain, and enables oestrogen to rebalance energy allocation in female mice. Surges in E2 increase melanocortin-4 receptor (MC4R) signalling in these VMHvl neurons by directly recruiting oestrogen receptor-α (ERα) to the Mc4r gene. Sedentary behaviour and obesity in oestrogen-depleted female mice were reversed after chemogenetic stimulation of VMHvl neurons expressing both MC4R and ERα. Similarly, a long-term increase in physical activity is observed after CRISPR-mediated activation of this node. These data extend the effect of MC4R signalling - the most common cause of monogenic human obesity8 - beyond the regulation of food intake and rationalize reported sex differences in melanocortin signalling, including greater disease severity of MC4R insufficiency in women9. This hormone-dependent node illuminates the power of oestrogen during the reproductive cycle in motivating behaviour and maintaining an active lifestyle in women.


Assuntos
Encéfalo/fisiologia , Estrogênios/metabolismo , Esforço Físico/fisiologia , Receptor Tipo 4 de Melanocortina/metabolismo , Transdução de Sinais , Animais , Sistemas CRISPR-Cas , Metabolismo Energético , Receptor alfa de Estrogênio/metabolismo , Estrogênios/deficiência , Feminino , Edição de Genes , Hipocampo/metabolismo , Masculino , Melanocortinas/metabolismo , Camundongos , Neurônios/metabolismo , Obesidade/metabolismo , Rombencéfalo/metabolismo , Comportamento Sedentário , Caracteres Sexuais , Núcleo Hipotalâmico Ventromedial/citologia , Núcleo Hipotalâmico Ventromedial/fisiologia
3.
Nat Rev Genet ; 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38778132
5.
Horm Behav ; 158: 105463, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37995608

RESUMO

The socially monogamous prairie vole (Microtus ochrogaster) and promiscuous meadow vole (Microtus pennsylvanicus) are closely related, but only prairie voles display long-lasting pair bonds, biparental care, and selective aggression towards unfamiliar individuals after pair bonding. These social behaviors in mammals are largely mediated by steroid hormone signaling in the social behavior network (SBN) of the brain. Hormone receptors are reproducible markers of sex differences that can provide more information than anatomy alone and can even be at odds with anatomical dimorphisms. We reasoned that behaviors associated with social monogamy in prairie voles may emerge in part from unique expression patterns of steroid hormone receptors in this species, and that these expression patterns would be more similar across males and females in prairie than in meadow voles or the laboratory mouse. To obtain insight into steroid hormone signaling in the developing prairie vole brain, we assessed expression of estrogen receptor alpha (Esr1), estrogen receptor beta (Esr2), and androgen receptor (Ar) within the SBN, using in situ hybridization at postnatal day 14 in mice, meadow, and prairie voles. We found species-specific patterns of hormone receptor expression in the hippocampus and ventromedial hypothalamus, as well as species differences in the sex bias of these markers in the principal nucleus of the bed nucleus of the stria terminalis. These findings suggest the observed differences in gonadal hormone receptor expression may underlie species differences in the display of social behaviors.


Assuntos
Encéfalo , Pradaria , Feminino , Animais , Masculino , Camundongos , Encéfalo/metabolismo , Comportamento Social , Arvicolinae/metabolismo , Hormônios/metabolismo , Hormônios Gonadais/metabolismo , Esteroides/metabolismo
6.
Cell ; 139(1): 61-72, 2009 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-19804754

RESUMO

Sex hormones are essential for neural circuit development and sex-specific behaviors. Male behaviors require both testosterone and estrogen, but it is unclear how the two hormonal pathways intersect. Circulating testosterone activates the androgen receptor (AR) and is also converted into estrogen in the brain via aromatase. We demonstrate extensive sexual dimorphism in the number and projections of aromatase-expressing neurons. The masculinization of these cells is independent of AR but can be induced in females by either testosterone or estrogen, indicating a role for aromatase in sexual differentiation of these neurons. We provide evidence suggesting that aromatase is also important in activating male-specific aggression and urine marking because these behaviors can be elicited by testosterone in males mutant for AR and in females subjected to neonatal estrogen exposure. Our results suggest that aromatization of testosterone into estrogen is important for the development and activation of neural circuits that control male territorial behaviors.


Assuntos
Encéfalo/metabolismo , Estrogênios/metabolismo , Vias Neurais , Caracteres Sexuais , Animais , Animais Recém-Nascidos , Aromatase/metabolismo , Sobrevivência Celular , Estrogênios/biossíntese , Feminino , Masculino , Camundongos , Neurônios/metabolismo , Receptores Androgênicos/metabolismo , Comportamento Sexual Animal , Territorialidade
7.
Genome Res ; 30(1): 49-61, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31727682

RESUMO

We show the use of 5'-Acrydite oligonucleotides to copolymerize single-cell DNA or RNA into balls of acrylamide gel (BAGs). Combining this step with split-and-pool techniques for creating barcodes yields a method with advantages in cost and scalability, depth of coverage, ease of operation, minimal cross-contamination, and efficient use of samples. We perform DNA copy number profiling on mixtures of cell lines, nuclei from frozen prostate tumors, and biopsy washes. As applied to RNA, the method has high capture efficiency of transcripts and sufficient consistency to clearly distinguish the expression patterns of cell lines and individual nuclei from neurons dissected from the mouse brain. By using varietal tags (UMIs) to achieve sequence error correction, we show extremely low levels of cross-contamination by tracking source-specific SNVs. The method is readily modifiable, and we will discuss its adaptability and diverse applications.


Assuntos
Acrilamida , Ácidos Nucleicos , Análise de Célula Única/métodos , Acrilamida/química , DNA , Contaminação por DNA , Variações do Número de Cópias de DNA , Dosagem de Genes , Perfilação da Expressão Gênica/métodos , Perfilação da Expressão Gênica/normas , Biblioteca Gênica , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Ácidos Nucleicos/química , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Análise de Sequência com Séries de Oligonucleotídeos/normas , Polimerização , RNA , Análise de Célula Única/normas
8.
Horm Behav ; 143: 105203, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35636023

RESUMO

Oxytocin is a neuropeptide that can produce anxiolytic effects and promote social approach. However, emerging evidence shows that under some conditions, oxytocin can instead induce anxiety-related behaviors. These diverse effects of oxytocin appear to be mediated by circuit-specific actions. Recent data showed that inhibition of oxytocin receptors (OTRs) in the bed nucleus of the stria terminalis (BNST) was sufficient to increase social approach and decrease social vigilance in female California mice (Peromyscus californicus) exposed to social defeat stress. As a member of the G-protein coupled receptor family, OTRs can induce distinct downstream pathways by coupling to different G-protein isoforms. We show that infusion of carbetocin, a biased OTR-Gq agonist, in the BNST reduced social approach in both female and male California mice. In both females and males, carbetocin also increased social vigilance. To gain insight into cell types that could be mediating this effect, we analyzed previously published single-cell RNAseq data from the BNST and nucleus accumbens (NAc). In the NAc, we and others showed that OTR activation promotes social approach behaviors. In the BNST, Oxtr was expressed in over 40 cell types, that span both posterior and anterior subregions of the BNST. The majority of Oxtr-expressing neurons were GABAergic. In the anterior regions of BNST targeted in our carbetocin experiments, Cyp26b1-expressing neurons had high average Oxtr expression. In the NAc, most Oxtr+ cells were D1 dopamine receptor-expressing neurons and interneurons. These differences in Oxtr cell type distribution may help explain how activation of OTR in BNST versus NAc can have different effects on social approach and social vigilance.


Assuntos
Núcleos Septais , Animais , Feminino , Masculino , Núcleo Accumbens/metabolismo , Ocitocina/metabolismo , Ocitocina/farmacologia , Receptores de Dopamina D1/metabolismo , Receptores de Ocitocina/metabolismo , Núcleos Septais/metabolismo , Comportamento Social
9.
Horm Behav ; 127: 104882, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33121994

RESUMO

There has been rapid growth in the use of transcriptomic analyses to study the interplay between gene expression and behavior. Experience can modify gene expression in the brain, leading to changes in internal state and behavioral displays, while gene expression variation between species is thought to specify many innate behavior differences. However, providing a causal association between a gene and a given behavior remains challenging as it is difficult to determine when and where a gene contributes to the function of a behaviorally-relevant neuronal population. Moreover, given that there are fewer genetic tools available for non-traditional model organisms, transcriptomic approaches have been largely limited to profiling of bulk tissue, which can obscure the contributions of subcortical brain regions implicated in multiple behaviors. Here, we discuss how emerging single cell technologies combined with methods offering additional spatial and connectivity information can give us insight about the genetic profile of specific cells involved in the neural circuit of target social behaviors. We also emphasize how these techniques are broadly adaptable to non-traditional model organisms. We propose that, ultimately, a combination of these approaches applied throughout development will be key to discerning how genes shape the formation of social behavior circuits.


Assuntos
Encéfalo/metabolismo , Genômica , Comportamento Social , Animais , Regulação da Expressão Gênica/fisiologia , Interação Gene-Ambiente , Genômica/métodos , Genômica/tendências , Rede Nervosa/metabolismo , Rede Nervosa/fisiologia , Neurônios/fisiologia , Personalidade/genética , Análise de Célula Única/métodos , Análise de Célula Única/tendências
10.
Nature ; 514(7521): 257-61, 2014 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-25119036

RESUMO

Homeodomain proteins, described 30 years ago, exert essential roles in development as regulators of target gene expression; however, the molecular mechanisms underlying transcriptional activity of homeodomain factors remain poorly understood. Here investigation of a developmentally required POU-homeodomain transcription factor, Pit1 (also known as Pou1f1), has revealed that, unexpectedly, binding of Pit1-occupied enhancers to a nuclear matrin-3-rich network/architecture is a key event in effective activation of the Pit1-regulated enhancer/coding gene transcriptional program. Pit1 association with Satb1 (ref. 8) and ß-catenin is required for this tethering event. A naturally occurring, dominant negative, point mutation in human PIT1(R271W), causing combined pituitary hormone deficiency, results in loss of Pit1 association with ß-catenin and Satb1 and therefore the matrin-3-rich network, blocking Pit1-dependent enhancer/coding target gene activation. This defective activation can be rescued by artificial tethering of the mutant R271W Pit1 protein to the matrin-3 network, bypassing the pre-requisite association with ß-catenin and Satb1 otherwise required. The matrin-3 network-tethered R271W Pit1 mutant, but not the untethered protein, restores Pit1-dependent activation of the enhancers and recruitment of co-activators, exemplified by p300, causing both enhancer RNA transcription and target gene activation. These studies have thus revealed an unanticipated homeodomain factor/ß-catenin/Satb1-dependent localization of target gene regulatory enhancer regions to a subnuclear architectural structure that serves as an underlying mechanism by which an enhancer-bound homeodomain factor effectively activates developmental gene transcriptional programs.


Assuntos
Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Proteínas Associadas à Matriz Nuclear/metabolismo , Proteínas de Ligação a RNA/metabolismo , Transcrição Gênica , Animais , Células Cultivadas , Proteínas de Homeodomínio/genética , Humanos , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Camundongos , Hipófise/embriologia , Hipófise/metabolismo , Ligação Proteica , Fator de Transcrição Pit-1/genética , Fator de Transcrição Pit-1/metabolismo , Transcrição Gênica/genética , beta Catenina/metabolismo
11.
J Neurosci ; 38(24): 5567-5583, 2018 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-29844022

RESUMO

Both the amygdala and the bed nucleus of the stria terminalis (BNST) have been implicated in maladaptive anxiety characteristics of anxiety disorders. However, the underlying circuit and cellular mechanisms have remained elusive. Here we show that mice with Erbb4 gene deficiency in somatostatin-expressing (SOM+) neurons exhibit heightened anxiety as measured in the elevated plus maze test and the open field test, two assays commonly used to assess anxiety-related behaviors in rodents. Using a combination of electrophysiological, molecular, genetic, and pharmacological techniques, we demonstrate that the abnormal anxiety in the mutant mice is caused by enhanced excitatory synaptic inputs onto SOM+ neurons in the central amygdala (CeA), and the resulting reduction in inhibition onto downstream SOM+ neurons in the BNST. Notably, our results indicate that an increase in dynorphin signaling in SOM+ CeA neurons mediates the paradoxical reduction in inhibition onto SOM+ BNST neurons, and that the consequent enhanced activity of SOM+ BNST neurons is both necessary for and sufficient to drive the elevated anxiety. Finally, we show that the elevated anxiety and the associated synaptic dysfunctions and increased dynorphin signaling in the CeA-BNST circuit of the Erbb4 mutant mice can be recapitulated by stress in wild-type mice. Together, our results unravel previously unknown circuit and cellular processes in the central extended amygdala that can cause maladaptive anxiety.SIGNIFICANCE STATEMENT The central extended amygdala has been implicated in anxiety-related behaviors, but the underlying mechanisms are unclear. Here we found that somatostatin-expressing neurons in the central amygdala (CeA) controls anxiety through modulation of the stria terminalis, a process that is mediated by an increase in dynorphin signaling in the CeA. Our results reveal circuit and cellular dysfunctions that may account for maladaptive anxiety.


Assuntos
Ansiedade/fisiopatologia , Núcleo Central da Amígdala/fisiopatologia , Vias Neurais/fisiologia , Núcleos Septais/fisiopatologia , Animais , Núcleo Central da Amígdala/metabolismo , Dinorfinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/fisiologia , Receptor ErbB-4/deficiência , Núcleos Septais/metabolismo , Somatostatina/metabolismo
12.
Proc Natl Acad Sci U S A ; 113(47): 13408-13413, 2016 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-27810956

RESUMO

As a key component of the vertebrate neuroendocrine system, the pituitary gland relies on the progressive and coordinated development of distinct hormone-producing cell types and an invading vascular network. The molecular mechanisms that drive formation of the pituitary vasculature, which is necessary for regulated synthesis and secretion of hormones that maintain homeostasis, metabolism, and endocrine function, remain poorly understood. Here, we report that expression of integrin ß1 in embryonic pituitary epithelial cells is required for angiogenesis in the developing mouse pituitary gland. Deletion of pituitary epithelial integrin ß1 before the onset of angiogenesis resulted in failure of invading endothelial cells to recruit pericytes efficiently, whereas deletion later in embryogenesis led to decreased vascular density and lumen formation. In both cases, lack of epithelial integrin ß1 was associated with a complete absence of vasculature in the pituitary gland at birth. Within pituitary epithelial cells, integrin ß1 directs a large transcriptional program that includes components of the extracellular matrix and associated signaling factors that are linked to the observed non-cell-autonomous effects on angiogenesis. We conclude that epithelial integrin ß1 functions as a critical and canonical regulator of developmental angiogenesis in the pituitary gland, thus providing insight into the long-standing systems biology conundrum of how vascular invasion is coordinated with tissue development.


Assuntos
Desenvolvimento Embrionário , Células Epiteliais/metabolismo , Integrina beta1/metabolismo , Neovascularização Fisiológica , Hipófise/citologia , Hipófise/embriologia , Animais , Animais Recém-Nascidos , Contagem de Células , Diferenciação Celular , Desenvolvimento Embrionário/genética , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Matriz Extracelular/metabolismo , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Marcação de Genes , Integrases/metabolismo , Camundongos , Neovascularização Fisiológica/genética , Fatores de Transcrição Box Pareados/metabolismo , Pericitos/citologia , Pericitos/metabolismo , Fenótipo , Hipófise/metabolismo , Análise de Sequência de RNA , Fatores de Tempo , Fator C de Crescimento do Endotélio Vascular/metabolismo
13.
Horm Behav ; 95: 3-12, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28734725

RESUMO

Masculinization of the altricial rodent brain is driven by estrogen signaling during a perinatal critical period. Genetic deletion of estrogen receptor alpha (Esr1/ERα) results in altered hypothalamic-pituitary-gonadal (HPG) axis signaling and a dramatic reduction of male sexual and territorial behaviors. However, the role of ERα in masculinizing distinct classes of neurons remains unexplored. We deleted ERα in excitatory or inhibitory neurons using either a Vglut2 or Vgat driver and assessed male behaviors. We find that Vglut2-Cre;Esr1lox/lox mutant males lack ERα in the ventrolateral region of the ventromedial hypothalamus (VMHvl) and posterior ventral portion of the medial amygdala (MePV). These mutants recapitulate the increased serum testosterone levels seen with constitutive ERα deletion, but have none of the behavioral deficits. In contrast, Vgat-Cre;Esr1lox/lox males with substantial ERα deletion in inhibitory neurons, including those of the principal nucleus of the bed nucleus of the stria terminalis (BNSTpr), posterior dorsal MeA (MePD), and medial preoptic area (MPOA) have normal testosterone levels, but display alterations in mating and territorial behaviors. These mutants also show dysmasculinized expression of androgen receptor (AR) and estrogen receptor beta (Esr2). Our results demonstrate that ERα masculinizes GABAergic neurons that gate the display of male-typical behaviors.


Assuntos
Receptor alfa de Estrogênio/fisiologia , Neurônios GABAérgicos/metabolismo , Ácido Glutâmico/metabolismo , Neurônios/metabolismo , Comportamento Sexual Animal/fisiologia , Virilismo/genética , Agressão/fisiologia , Animais , Encéfalo/metabolismo , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Estrogênios/metabolismo , Feminino , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Territorialidade , Virilismo/metabolismo
14.
Nat Commun ; 15(1): 4682, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38824130

RESUMO

Interleukin-6 (IL-6) has been long considered a key player in cancer cachexia. It is believed that sustained elevation of IL-6 production during cancer progression causes brain dysfunctions, which ultimately result in cachexia. However, how peripheral IL-6 influences the brain remains poorly understood. Here we show that neurons in the area postrema (AP), a circumventricular structure in the hindbrain, is a critical mediator of IL-6 function in cancer cachexia in male mice. We find that circulating IL-6 can rapidly enter the AP and activate neurons in the AP and its associated network. Peripheral tumor, known to increase circulating IL-6, leads to elevated IL-6 in the AP, and causes potentiated excitatory synaptic transmission onto AP neurons and AP network hyperactivity. Remarkably, neutralization of IL-6 in the brain of tumor-bearing mice with an anti-IL-6 antibody attenuates cachexia and the hyperactivity in the AP network, and markedly prolongs lifespan. Furthermore, suppression of Il6ra, the gene encoding IL-6 receptor, specifically in AP neurons with CRISPR/dCas9 interference achieves similar effects. Silencing Gfral-expressing AP neurons also attenuates cancer cachectic phenotypes and AP network hyperactivity. Our study identifies a central mechanism underlying the function of peripheral IL-6, which may serve as a target for treating cancer cachexia.


Assuntos
Caquexia , Interleucina-6 , Neurônios , Receptores de Interleucina-6 , Animais , Caquexia/metabolismo , Caquexia/etiologia , Interleucina-6/metabolismo , Masculino , Neurônios/metabolismo , Camundongos , Receptores de Interleucina-6/metabolismo , Camundongos Endogâmicos C57BL , Neoplasias/metabolismo , Neoplasias/complicações , Linhagem Celular Tumoral , Humanos
15.
Psychoneuroendocrinology ; 161: 106920, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38128260

RESUMO

Mood disorders, like major depressive disorder, can be precipitated by chronic stress and are more likely to be diagnosed in cisgender women than in cisgender men. This suggests that stress signaling in the brain is sexually dimorphic. We used a chronic variable mild stress paradigm to stress female and male mice for 6 weeks, followed by an assessment of avoidance behavior: the open field test, the elevated plus maze, the light/dark box emergence test, and the novelty suppressed feeding test. Additional cohorts were used for bulk RNA-Sequencing of the anterodorsal bed nucleus of the stria terminalis (adBNST) and whole-cell patch clamp electrophysiology in NPY-expressing neurons of the adBNST to record stress-sensitive M-currents. Our results indicate that females are more affected by chronic stress as indicated by an increase in avoidance behaviors, but that this is also dependent on the estrous stage of the animals such that diestrus females show more avoidant behaviors regardless of stress treatment. Results also indicate that NPY-expressing neurons of the adBNST are not major mediators of chronic stress as the M-current was not affected by treatment. RNA-Sequencing data suggests sex differences in estrogen signaling, serotonin signaling, and orexin signaling in the adBNST. Our results indicate that chronic stress influences behavior in a sex- and estrous stage-dependent manner but NPY-expressing neurons in the BNST are not the mediators of these effects.


Assuntos
Transtorno Depressivo Maior , Núcleos Septais , Humanos , Camundongos , Feminino , Masculino , Animais , Núcleos Septais/fisiologia , Transtorno Depressivo Maior/metabolismo , Neurônios/metabolismo , Transdução de Sinais/fisiologia , RNA/metabolismo
16.
J Neurosci Methods ; 401: 110003, 2024 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-37918446

RESUMO

Recently, many funding agencies have released guidelines on the importance of considering sex as a biological variable (SABV) as an experimental factor, aiming to address sex differences and avoid possible sex biases to enhance the reproducibility and translational relevance of preclinical research. In neuroscience and pharmacology, the female sex is often omitted from experimental designs, with researchers generalizing male-driven outcomes to both sexes, risking a biased or limited understanding of disease mechanisms and thus potentially ineffective therapeutics. Herein, we describe key methodological aspects that should be considered when sex is factored into in vitro and in vivo experiments and provide practical knowledge for researchers to incorporate SABV into preclinical research. Both age and sex significantly influence biological and behavioral processes due to critical changes at different timepoints of development for males and females and due to hormonal fluctuations across the rodent lifespan. We show that including both sexes does not require larger sample sizes, and even if sex is included as an independent variable in the study design, a moderate increase in sample size is sufficient. Moreover, the importance of tracking hormone levels in both sexes and the differentiation between sex differences and sex-related strategy in behaviors are explained. Finally, the lack of robust data on how biological sex influences the pharmacokinetic (PK), pharmacodynamic (PD), or toxicological effects of various preclinically administered drugs to animals due to the exclusion of female animals is discussed, and methodological strategies to enhance the rigor and translational relevance of preclinical research are proposed.


Assuntos
Projetos de Pesquisa , Caracteres Sexuais , Animais , Masculino , Feminino , Reprodutibilidade dos Testes , Fatores Sexuais , Tamanho da Amostra
17.
bioRxiv ; 2023 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-36711916

RESUMO

Interleukin-6 (IL-6) has been long considered a key player in cancer-associated cachexia 1-15 . It is believed that sustained elevation of IL-6 production during cancer progression causes brain dysfunctions, which ultimately result in cachexia 16-20 . However, how peripheral IL-6 influences the brain remains poorly understood. Here we show that neurons in the area postrema (AP), a circumventricular structure in the hindbrain, mediate the function of IL-6 in cancer-associated cachexia in mice. We found that circulating IL-6 can rapidly enter the AP and activate AP neurons. Peripheral tumor, known to increase circulating IL-6 1-5,15,18,21-23 , leads to elevated IL-6 and neuronal hyperactivity in the AP, and causes potentiated excitatory synaptic transmission onto AP neurons. Remarkably, neutralization of IL-6 in the brain of tumor-bearing mice with an IL-6 antibody prevents cachexia, reduces the hyperactivity in an AP network, and markedly prolongs lifespan. Furthermore, suppression of Il6ra , the gene encoding IL-6 receptor, specifically in AP neurons with CRISPR/dCas9 interference achieves similar effects. Silencing of Gfral-expressing AP neurons also ameliorates the cancer-associated cachectic phenotypes and AP network hyperactivity. Our study identifies a central mechanism underlying the function of peripheral IL-6, which may serve as a target for treating cancer-associated cachexia.

18.
Artigo em Inglês | MEDLINE | ID: mdl-35817509

RESUMO

Across vertebrate species, gonadal hormones coordinate physiology with behavior to facilitate social interactions essential for reproduction and survival. In adulthood, these hormones activate neural circuits that regulate behaviors presenting differently in females and males, such as parenting and territorial aggression. Yet long before sex-typical behaviors emerge at puberty, transient hormone production during sensitive periods of neurodevelopment establish the circuits upon which adult hormones act. How transitory waves of early-life hormone signaling exert lasting effects on the brain remains a central question. Here we discuss how perinatal estradiol signaling organizes cellular and molecular sex differences in the rodent brain. We review classic anatomic studies revealing sex differences in cell number, volume, and neuronal projections, and consider how single-cell sequencing methods enable distinction between sex-biased cell-type abundance and gene expression. Finally, we highlight the recent discovery of a gene regulatory program activated by estrogen receptor α (ERα) following the perinatal hormone surge. A subset of this program displays sustained sex-biased gene expression and chromatin accessibility throughout the postnatal sensitive period, demonstrating a bona fide epigenetic mechanism. We propose that ERα-expressing neurons throughout the social behavior network use similar gene regulatory programs to coordinate brain sexual differentiation.


Assuntos
Receptor alfa de Estrogênio , Diferenciação Sexual , Feminino , Masculino , Animais , Diferenciação Sexual/genética , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Epigênese Genética , Encéfalo/metabolismo , Caracteres Sexuais , Hormônios/metabolismo
19.
Wiley Interdiscip Rev Dev Biol ; 9(1): e348, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31106965

RESUMO

Women and men differ in disease prevalence, symptoms, and progression rates for many psychiatric and neurological disorders. As more preclinical studies include both sexes in experimental design, an increasing number of sex differences in physiology and behavior have been reported. In the brain, sex-typical behaviors are thought to result from sex-specific patterns of neural activity in response to the same sensory stimulus or context. These differential firing patterns likely arise as a consequence of underlying anatomic or molecular sex differences. Accordingly, gene expression in the brains of females and males has been extensively investigated, with the goal of identifying biological pathways that specify or modulate sex differences in brain function. However, there is surprisingly little consensus on sex-biased genes across studies and only a handful of robust candidates have been pursued in the follow-up experiments. Furthermore, it is not known how or when sex-biased gene expression originates, as few studies have been performed in the developing brain. Here we integrate molecular genetic and neural circuit perspectives to provide a conceptual framework of how sex differences in gene expression can arise in the brain. We detail mechanisms of gene regulation by steroid hormones, highlight landmark studies in rodents and humans, identify emerging themes, and offer recommendations for future research. This article is categorized under: Nervous System Development > Vertebrates: General Principles Gene Expression and Transcriptional Hierarchies > Regulatory Mechanisms Gene Expression and Transcriptional Hierarchies > Sex Determination.


Assuntos
Encéfalo/fisiologia , Regulação da Expressão Gênica/fisiologia , Expressão Gênica/fisiologia , Vertebrados/fisiologia , Animais , Humanos , Caracteres Sexuais
20.
Curr Opin Genet Dev ; 15(3): 332-40, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15917210

RESUMO

The pituitary gland functions as a relay between the hypothalamus and peripheral target organs that regulate basic physiological functions, including growth, the stress response, reproduction, metabolism and lactation. The development of the pituitary gland has been studied extensively in mice, and has begun to be explored in zebrafish, an animal model system amenable to forward genetics. Multiple signaling molecules and transcription factors, expressed in overlapping but distinct spatial and temporal patterns, are required at various stages of pituitary development. Defects in this precisely regulated genetic program lead to diverse pituitary dysfunction. The animal models have greatly enhanced our understanding of molecular mechanisms underlying pituitary development in addition to congenital pituitary disorders in humans.


Assuntos
Hipopituitarismo/genética , Hipófise/embriologia , Hipófise/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hipopituitarismo/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA