Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
iScience ; 26(10): 107918, 2023 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-37817932

RESUMO

Balance between metabolic and reproductive processes is important for survival, particularly in mammals that gestate their young. How the nervous system coordinates this balance is an active area of study. Herein, we demonstrate that somatostatin (SST) neurons of the tuberal hypothalamus alter feeding in a manner sensitive to metabolic and reproductive states in mice. Whereas chemogenetic activation of SST neurons increased food intake across sexes, ablation decreased food intake only in female mice during proestrus. This ablation effect was only apparent in animals with low body mass. Fat transplantation and bioinformatics analysis of SST neuronal transcriptomes revealed white adipose as a key modulator of these effects. These studies indicate that SST hypothalamic neurons integrate metabolic and reproductive cues by responding to varying levels of circulating estrogens to modulate feeding differentially based on energy stores. Thus, gonadal steroid modulation of neuronal circuits can be context dependent and gated by metabolic status.

2.
Ecol Evol ; 13(9): e10476, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37706165

RESUMO

While cross-species comparisons of birds suggest that as latitude decreases or elevation increases, clutch size decreases and the duration of developmental stages and parental attentiveness increases, studies comparing populations of the same species are rare. We studied populations of house wrens, Troglodytes aedon, at high and low elevations in California and Costa Rica, collecting data on clutch size, the duration of incubation and nestling periods, parental attentiveness, nestling growth rate, and nesting success. Our data support results from cross-species comparisons, but also revealed unanticipated results from low elevation temperate zone house wrens in the southwest. This population had prolonged incubation and nestling periods similar to those found in the tropics. We also found that temperate zone females, especially those at our higher elevation site, spent more of their day incubating than did tropical females. Nest temperature at our high elevation temperate zone site was higher than that at all other tropical sites. Age at fledging did not differ between sites. Total feeding rates per chick and male feedings per chick did not vary between sites. Nest success rates showed the predicted effect of latitude, but not the predicted effects of elevation. Our results extend low elevation house wren research into the southwestern US and contribute the first intraspecific elevational comparison in the Neotropics. Data from our low elevation southwestern site present a unique suite of life history traits that align more with tropical house wrens, although with a larger clutch size, and point to food limitation and/or high predation pressure as being possible drivers of some of these differences. These results highlight the need for additional studies of house wrens and other broadly distributed species at a more diverse array of sites to better understand which forces drive the evolution of different life history strategies across major biogeographical gradients.

3.
Prev Med Rep ; 35: 102292, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37449004

RESUMO

In the United States, adherence to follow up medical appointments among patients discharged from the emergency department varies between 26% and 56%, depending on the population. It is well known that patients face significant barriers to care within an increasingly complicated system of care. In an effort to better support patients, in 2020, NewYork-Presbyterian Queens implemented a Patient Navigator Program with 7 bilingual Patient Navigators who were trained to deliver culturally sensitive education and support, and to schedule follow up appointments for patients experiencing barriers to care. Between February 2020 and December 2022, 30,164 patients were supported by the 7 Patient Navigators. Ninety-four percent of patients without a primary care provider had a new provider and appointment upon discharge, and 81% of patients attended the appointment scheduled by the Patient Navigator. This study demonstrates that Patient Navigators can work alongside clinical colleagues, and as members of emergency department health care teams, to support patients to connect to care and to attend follow up appointments. It also highlights that Patient Navigators are uniquely qualified to build trust and to support patients to achieve appropriate, continuous care within a rapidly evolving health care system.

4.
bioRxiv ; 2023 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-36747631

RESUMO

Trade-offs between metabolic and reproductive processes are important for survival, particularly in mammals that gestate their young. Puberty and reproduction, as energetically taxing life stages, are often gated by metabolic availability in animals with ovaries. How the nervous system coordinates these trade-offs is an active area of study. We identify somatostatin neurons of the tuberal nucleus (TNSST) as a node of the feeding circuit that alters feeding in a manner sensitive to metabolic and reproductive states in mice. Whereas chemogenetic activation of TNSST neurons increased food intake across sexes, selective ablation decreased food intake only in female mice during proestrus. Interestingly, this ablation effect was only apparent in animals with a low body mass. Fat transplantation and bioinformatics analysis of TNSST neuronal transcriptomes revealed white adipose as a key modulator of the effects of TNSST neurons on food intake. Together, these studies point to a mechanism whereby TNSST hypothalamic neurons modulate feeding by responding to varying levels of circulating estrogens differentially based on energy stores. This research provides insight into how neural circuits integrate reproductive and metabolic signals, and illustrates how gonadal steroid modulation of neuronal circuits can be context-dependent and gated by metabolic status.

5.
Mol Psychiatry ; 28(5): 1857-1867, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36765131

RESUMO

Antipsychotic (AP) drugs are efficacious treatments for various psychiatric disorders, but excessive weight gain and subsequent development of metabolic disease remain serious side effects of their use. Increased food intake leads to AP-induced weight gain, but the underlying molecular mechanisms remain unknown. In previous studies, we identified the neuropeptide Agrp and the transcription factor nuclear receptor subfamily 5 group A member 2 (Nr5a2) as significantly upregulated genes in the hypothalamus following AP-induced hyperphagia. While Agrp is expressed specifically in the arcuate nucleus of the hypothalamus and plays a critical role in appetite stimulation, Nr5a2 is expressed in both the CNS and periphery, but its role in food intake behaviors remains unknown. In this study, we investigated the role of hypothalamic Nr5a2 in AP-induced hyperphagia and weight gain. In hypothalamic cell lines, olanzapine treatment resulted in a dose-dependent increase in gene expression of Nr5a2 and Agrp. In mice, the pharmacological inhibition of NR5A2 decreased olanzapine-induced hyperphagia and weight gain, while the knockdown of Nr5a2 in the arcuate nucleus partially reversed olanzapine-induced hyperphagia. Chromatin-immunoprecipitation studies showed for the first time that NR5A2 directly binds to the Agrp promoter region. Lastly, the analysis of single-cell RNA seq data confirms that Nr5a2 and Agrp are co-expressed in a subset of neurons in the arcuate nucleus. In summary, we identify Nr5a2 as a key mechanistic driver of AP-induced food intake. These findings can inform future clinical development of APs that do not activate hyperphagia and weight gain.


Assuntos
Hiperfagia , Animais , Humanos , Camundongos , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Proteína Relacionada com Agouti/farmacologia , Antipsicóticos/efeitos adversos , Ingestão de Alimentos , Hiperfagia/induzido quimicamente , Hiperfagia/genética , Hiperfagia/metabolismo , Hipotálamo/metabolismo , Olanzapina/efeitos adversos , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores Citoplasmáticos e Nucleares/farmacologia , Receptores Citoplasmáticos e Nucleares/uso terapêutico , Aumento de Peso
6.
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
7.
Endocrinology ; 162(8)2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-33939822

RESUMO

Declining and variable levels of estrogens around the time of menopause are associated with a suite of metabolic, vascular, and neuroendocrine changes. The archetypal adverse effects of perimenopause are vasomotor symptoms, which include hot flashes and night sweats. Although vasomotor symptoms are routinely treated with hormone therapy, the risks associated with these treatments encourage us to seek alternative treatment avenues. Understanding the mechanisms underlying the effects of estrogens on temperature regulation is a first step toward identifying novel therapeutic targets. Here we outline findings in rodents that reveal neural and molecular targets of estrogens within brain regions that control distinct components of temperature homeostasis. These insights suggest that estrogens may alter the function of multiple specialized neural circuits to coordinate the suite of changes after menopause. Thus, defining the precise cells and neural circuits that mediate the effects of estrogens on temperature has promise to identify strategies that would selectively counteract hot flashes or other negative side effects without the health risks that accompany systemic hormone therapies.


Assuntos
Regulação da Temperatura Corporal , Encéfalo/fisiologia , Estrogênios/fisiologia , Animais , Fogachos/etiologia , Humanos , Neurônios/metabolismo , Receptores de Estrogênio/metabolismo , Torpor
9.
Elife ; 102021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33647234

RESUMO

Adjuvant tamoxifen therapy improves survival in breast cancer patients. Unfortunately, long-term treatment comes with side effects that impact health and quality of life, including hot flashes, changes in bone density, and fatigue. Partly due to a lack of proven animal models, the tissues and cells that mediate these negative side effects are unclear. Here, we show that mice undergoing tamoxifen treatment experience changes in temperature, bone, and movement. Single-cell RNA sequencing reveals that tamoxifen treatment induces widespread gene expression changes in the hypothalamus and preoptic area (hypothalamus-POA). These expression changes are dependent on estrogen receptor alpha (ERα), as conditional knockout of ERα in the hypothalamus-POA ablates or reverses tamoxifen-induced gene expression. Accordingly, ERα-deficient mice do not exhibit tamoxifen-induced changes in temperature, bone, or movement. These findings provide mechanistic insight into the effects of tamoxifen on the hypothalamus-POA and indicate that ERα mediates several physiological effects of tamoxifen treatment in mice.


Estrogen is a hormone often known for its role in female development and reproduction. Yet, it also has an impact on many biological processes such as immunity and the health of bones, the heart, or the brain. It usually works by attaching to receptor proteins in specific cells. For instance, estrogen-responsive cells are present in the hypothalamus, the brain area that controls energy levels as well as the body's temperature and internal clock. Breast cancer cells are also often sensitive to estrogen, with the hormone fuelling the growth of tumors. The drug tamoxifen blocks estrogen receptors, stopping cells from responding to the hormone. As such, it is often used to reduce the likelihood that estrogen-dependent breast cancer will come back after treatment. However, its use can induce hot flashes, changes in bone density, fatigue and other life-altering side effects. Here, Zhang et al. investigated how estrogen receptors in the hypothalamus and a related region known as the preoptic area could be responsible for these side effects in mice. When the rodents were given tamoxifen for 28 days, they experienced changes in temperature, bone density and movement similar to those found in humans. In fact, genetic analyses revealed that the drug altered the way genes were turned on and off in certain cells types in the hypothalamus. Crucially, mice whose hypothalamus and preoptic area lacked estrogen receptors did not experience these behavioral and biological alterations. The findings by Zhang et al. help to understand how the side effects of tamoxifen emerge, singling out estrogen receptors in particular brain regions. This result could help to develop new therapies so that breast cancer can be treated with a better quality of life.


Assuntos
Antineoplásicos Hormonais/farmacologia , Hipotálamo/metabolismo , Área Pré-Óptica/metabolismo , Tamoxifeno/farmacologia , Animais , Temperatura Corporal/efeitos dos fármacos , Densidade Óssea/efeitos dos fármacos , Receptor alfa de Estrogênio/deficiência , Feminino , Regulação da Expressão Gênica , Camundongos , Movimento/efeitos dos fármacos
10.
Nat Commun ; 11(1): 6378, 2020 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-33311503

RESUMO

Homeotherms maintain a stable internal body temperature despite changing environments. During energy deficiency, some species can cease to defend their body temperature and enter a hypothermic and hypometabolic state known as torpor. Recent advances have revealed the medial preoptic area (MPA) as a key site for the regulation of torpor in mice. The MPA is estrogen-sensitive and estrogens also have potent effects on both temperature and metabolism. Here, we demonstrate that estrogen-sensitive neurons in the MPA can coordinate hypothermia and hypometabolism in mice. Selectively activating estrogen-sensitive MPA neurons was sufficient to drive a coordinated depression of metabolic rate and body temperature similar to torpor, as measured by body temperature, physical activity, indirect calorimetry, heart rate, and brain activity. Inducing torpor with a prolonged fast revealed larger and more variable calcium transients from estrogen-sensitive MPA neurons during bouts of hypothermia. Finally, whereas selective ablation of estrogen-sensitive MPA neurons demonstrated that these neurons are required for the full expression of fasting-induced torpor in both female and male mice, their effects on thermoregulation and torpor bout initiation exhibit differences across sex. Together, these findings suggest a role for estrogen-sensitive MPA neurons in directing the thermoregulatory and metabolic responses to energy deficiency.


Assuntos
Temperatura Corporal/fisiologia , Estrogênios/metabolismo , Neurônios/fisiologia , Área Pré-Óptica/metabolismo , Torpor/fisiologia , Animais , Temperatura Corporal/genética , Regulação da Temperatura Corporal/fisiologia , Metabolismo Energético/fisiologia , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Jejum , Feminino , Hipotermia/genética , Hipotermia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
11.
Biol Sex Differ ; 11(1): 28, 2020 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-32398044

RESUMO

BACKGROUND: The commonly used laboratory rat, Rattus norvegicus, is unique in having multiple Sry gene copies found on the Y chromosome, with different copies encoding amino acid variations that influence the resulting protein function. It is not clear which Sry genes are expressed at the onset of testis differentiation or how their expression correlates with that of other genes in testis-determination pathways. METHODS: Here, two independent E11-E14 developmental RNAseq datasets show that multiple Sry genes are expressed at E12-E13. RESULTS: The identified copies expressed during testis initiation include Sry4A, Sry1, and Sry3C, which are conserved in every strain of Rattus norvegicus with genomes sequenced to date. CONCLUSIONS: This work represents a first step in defining the complex environment of rat testis differentiation that can open the door for generating sex reversal model systems using embryo manipulation techniques that have been available in the mouse but not the rat.


Assuntos
Genes sry , Testículo/crescimento & desenvolvimento , Animais , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Ratos Sprague-Dawley , Transcrição Gênica
12.
Nat Metab ; 2(4): 351-363, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32377634

RESUMO

Estrogen receptor a (ERa) signaling in the ventromedial hypothalamus (VMH) contributes to energy homeostasis by modulating physical activity and thermogenesis. However, the precise neuronal populations involved remain undefined. Here, we describe six neuronal populations in the mouse VMH by using single-cell RNA transcriptomics and in situ hybridization. ERa is enriched in populations showing sex biased expression of reprimo (Rprm), tachykinin 1 (Tac1), and prodynorphin (Pdyn). Female biased expression of Tac1 and Rprm is patterned by ERa-dependent repression during male development, whereas male biased expression of Pdyn is maintained by circulating testicular hormone in adulthood. Chemogenetic activation of ERa positive VMH neurons stimulates heat generation and movement in both sexes. However, silencing Rprm gene function increases core temperature selectively in females and ectopic Rprm expression in males is associated with reduced core temperature. Together these findings reveal a role for Rprm in temperature regulation and ERa in the masculinization of neuron populations that underlie energy expenditure.


Assuntos
Metabolismo Energético , Receptor alfa de Estrogênio/metabolismo , Hipotálamo/metabolismo , Caracteres Sexuais , Animais , Feminino , Corantes Fluorescentes/química , Marcadores Genéticos , Hipotálamo/citologia , Masculino , Camundongos , Neurônios/metabolismo
13.
Biochim Biophys Acta Mol Basis Dis ; 1866(10): 165840, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32428559

RESUMO

Neuronal interactions at the level of vagal, homeostatic, and hedonic circuitry work to regulate the neuronal control of feeding. This integrative system appears to vary across sex and gender in the animal and human worlds. Most feeding research investigating these variations across sex and gender focus on how the organizational and activational mechanisms of hormones contribute to these differences. However, in limited studies spanning both the central and peripheral nervous systems, sex differences in feeding have been shown to manifest not just at the level of the hormonal, but also at the chromosomal, epigenetic, cellular, and even circuitry levels to alter food intake. In this review, we provide a brief orientation to the current understanding of how these neuronal systems interact before dissecting selected studies from the recent literature to exemplify how feeding physiology at all levels can be affected by the various components of sex.


Assuntos
Encéfalo/fisiologia , Ingestão de Alimentos/fisiologia , Comportamento Alimentar/fisiologia , Neurônios/fisiologia , Caracteres Sexuais , Animais , Ingestão de Alimentos/genética , Epigenômica , Feminino , Homeostase/fisiologia , Humanos , Masculino , Cromossomos Sexuais
14.
J Neuroendocrinol ; 32(1): e12801, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31605642

RESUMO

Sex differences among neurones in the ventrolateral region of the ventromedial hypothalamic nucleus (VMHvl) allow for the display of a diversity of sex-typical behaviours and physiological responses, ranging from mating behaviour to metabolism. Here, we review recent studies that interrogate the relationship between sex-typical responses and changes in cellular phenotypes. We discuss technologies that increase the resolution of molecular profiling or targeting of cell populations, including single-cell transcriptional profiling and conditional viral genetic approaches to manipulate neurone survival or activity. Overall, emerging studies indicate that sex-typical functions of the VMH may be mediated by phenotypically distinct and sexually differentiated neurone populations within the VMHvl. Future studies in this and other brain regions could exploit cell-type-specific tools to reveal the cell populations and molecular mediators that modulate sex-typical responses. Furthermore, cell-type-specific analyses of the effects of sexually differentiating factors, including sex hormones, can test the hypothesis that distinct cell types within a single brain region vary with respect to sexual differentiation.


Assuntos
Neurônios/metabolismo , Caracteres Sexuais , Diferenciação Sexual/fisiologia , Núcleo Hipotalâmico Ventromedial/fisiologia , Animais , Feminino , Humanos , Masculino , Comportamento Sexual Animal/fisiologia
15.
Nat Commun ; 10(1): 163, 2019 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-30635563

RESUMO

Central estrogen signaling coordinates energy expenditure, reproduction, and in concert with peripheral estrogen impacts skeletal homeostasis in females. Here, we ablate estrogen receptor alpha (ERα) in the medial basal hypothalamus and find a robust bone phenotype only in female mice that results in exceptionally strong trabecular and cortical bones, whose density surpasses other reported mouse models. Stereotaxic guided deletion of ERα in the arcuate nucleus increases bone mass in intact and ovariectomized females, confirming the central role of estrogen signaling in this sex-dependent bone phenotype. Loss of ERα in kisspeptin (Kiss1)-expressing cells is sufficient to recapitulate the bone phenotype, identifying Kiss1 neurons as a critical node in this powerful neuroskeletal circuit. We propose that this newly-identified female brain-to-bone pathway exists as a homeostatic regulator diverting calcium and energy stores from bone building when energetic demands are high. Our work reveals a previously unknown target for treatment of age-related bone disease.


Assuntos
Núcleo Arqueado do Hipotálamo/fisiologia , Densidade Óssea , Receptor alfa de Estrogênio/fisiologia , Kisspeptinas/metabolismo , Animais , Metabolismo Energético , Feminino , Homeostase , Masculino , Camundongos Transgênicos , Osteogênese , Fenótipo , Caracteres Sexuais
16.
Nat Metab ; 1(11): 1089-1100, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-32072135

RESUMO

Liver X receptors limit cellular lipid uptake by stimulating the transcription of Inducible Degrader of the LDL Receptor (IDOL), an E3 ubiquitin ligase that targets lipoprotein receptors for degradation. The function of IDOL in systemic metabolism is incompletely understood. Here we show that loss of IDOL in mice protects against the development of diet-induced obesity and metabolic dysfunction by altering food intake and thermogenesis. Unexpectedly, analysis of tissue-specific knockout mice revealed that IDOL affects energy balance, not through its actions in peripheral metabolic tissues (liver, adipose, endothelium, intestine, skeletal muscle), but by controlling lipoprotein receptor abundance in neurons. Single-cell RNA sequencing of the hypothalamus demonstrated that IDOL deletion altered gene expression linked to control of metabolism. Finally, we identify VLDLR rather than LDLR as the primary mediator of IDOL effects on energy balance. These studies identify a role for the neuronal IDOL-VLDLR pathway in metabolic homeostasis and diet-induced obesity.


Assuntos
Metabolismo Energético/fisiologia , Neurônios/metabolismo , Receptores de LDL/metabolismo , Ubiquitina-Proteína Ligases/fisiologia , Animais , Glicemia/metabolismo , Dieta , Metabolismo Energético/genética , Hipotálamo/metabolismo , Resistência à Insulina , Camundongos , Camundongos Knockout , Obesidade/metabolismo , Obesidade/prevenção & controle , Ubiquitina-Proteína Ligases/genética
17.
Cell Rep ; 10(1): 62-74, 2015 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-25543145

RESUMO

Estrogen-receptor alpha (ERα) neurons in the ventrolateral region of the ventromedial hypothalamus (VMHVL) control an array of sex-specific responses to maximize reproductive success. In females, these VMHVL neurons are believed to coordinate metabolism and reproduction. However, it remains unknown whether specific neuronal populations control distinct components of this physiological repertoire. Here, we identify a subset of ERα VMHVL neurons that promotes hormone-dependent female locomotion. Activating Nkx2-1-expressing VMHVL neurons via pharmacogenetics elicits a female-specific burst of spontaneous movement, which requires ERα and Tac1 signaling. Disrupting the development of Nkx2-1(+) VMHVL neurons results in female-specific obesity, inactivity, and loss of VMHVL neurons coexpressing ERα and Tac1. Unexpectedly, two responses controlled by ERα(+) neurons, fertility and brown adipose tissue thermogenesis, are unaffected. We conclude that a dedicated subset of VMHVL neurons marked by ERα, NKX2-1, and Tac1 regulates estrogen-dependent fluctuations in physical activity and constitutes one of several neuroendocrine modules that drive sex-specific responses.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Locomoção/genética , Proteínas Nucleares/biossíntese , Obesidade/metabolismo , Taquicininas/genética , Fatores de Transcrição/biossíntese , Animais , Receptor alfa de Estrogênio/genética , Estrogênios/metabolismo , Feminino , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Proteínas Nucleares/genética , Obesidade/genética , Obesidade/fisiopatologia , Caracteres Sexuais , Taquicininas/metabolismo , Fator Nuclear 1 de Tireoide , Fatores de Transcrição/genética , Núcleo Hipotalâmico Ventromedial/metabolismo , Núcleo Hipotalâmico Ventromedial/patologia
18.
Gen Comp Endocrinol ; 190: 34-41, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23499787

RESUMO

Maternal effects are influences of parents on offspring phenotype occurring through pathways other than inherited DNA. In birds, two important routes for such transmission are parental behavior and non-DNA egg constituents such as yolk hormones. Offspring traits subject to parental effects include behavior and endocrine function. Research from the Adkins-Regan lab has used three avian species to investigate maternal effects related to hormones and behavior. Experiments with chickens and Japanese quail have shown that maternal sex steroids can influence sex determination to produce biased offspring sex ratios. Because all birds have a ZZ/ZW chromosomal sex determining system in which the female parent determines the sex of the offspring, these results raise the possibility that maternal steroids can influence the outcome of sex chromosome meiosis. Learning has been shown to influence egg investment by female quail in ways that are likely to alter offspring phenotype. In quail, embryonic and exogenous sex steroids have well established and long-lasting effects on sexual differentiation of behavior during a critical period in ovo, but elevated yolk testosterone has long-term effects on behavior that do not seem to be occurring through an alteration in sexual differentiation. In biparental zebra finches, removal of mothers alters not only later behavior, but also the adult response of the hypothalamic-pituitary-adrenal (HPA) axis to an environmental stressor, as indicated by plasma corticosterone. Birds raised only by fathers have lower levels of mRNA for both glucocorticoid receptors in several brain regions as adults. These studies add to the evidence that one generation influences the behavioral or endocrine phenotype of the next through routes other than transmission of DNA. Additional research will be required to understand the adaptive significance of these effects.


Assuntos
Tentilhões/metabolismo , Codorniz/metabolismo , Animais , Aves , Hormônios Esteroides Gonadais/metabolismo , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Receptores de Glucocorticoides/metabolismo
19.
PLoS Genet ; 8(4): e1002569, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22496664

RESUMO

Sex reversal can occur in XY humans with only a single functional WT1 or SF1 allele or a duplication of the chromosome region containing WNT4. In contrast, XY mice with only a single functional Wt1, Sf1, or Wnt4 allele, or mice that over-express Wnt4 from a transgene, reportedly are not sex-reversed. Because genetic background plays a critical role in testis differentiation, particularly in C57BL/6J (B6) mice, we tested the hypothesis that Wt1, Sf1, and Wnt4 are dosage sensitive in B6 XY mice. We found that reduced Wt1 or Sf1 dosage in B6 XY(B6) mice impaired testis differentiation, but no ovarian tissue developed. If, however, a Y(AKR) chromosome replaced the Y(B6) chromosome, these otherwise genetically identical B6 XY mice developed ovarian tissue. In contrast, reduced Wnt4 dosage increased the amount of testicular tissue present in Sf1+/- B6 XY(AKR), Wt1+/- B6 XY(AKR), B6 XY(POS), and B6 XY(AKR) fetuses. We propose that Wt1(B6) and Sf1(B6) are hypomorphic alleles of testis-determining pathway genes and that Wnt4(B6) is a hypermorphic allele of an ovary-determining pathway gene. The latter hypothesis is supported by the finding that expression of Wnt4 and four other genes in the ovary-determining pathway are elevated in normal B6 XX E12.5 ovaries. We propose that B6 mice are sensitive to XY sex reversal, at least in part, because they carry Wt1(B6) and/or Sf1(B6) alleles that compromise testis differentiation and a Wnt4(B6) allele that promotes ovary differentiation and thereby antagonizes testis differentiation. Addition of a "weak" Sry allele, such as the one on the Y(POS) chromosome, to the sensitized B6 background results in inappropriate development of ovarian tissue. We conclude that Wt1, Sf1, and Wnt4 are dosage-sensitive in mice, this dosage-sensitivity is genetic background-dependant, and the mouse strains described here are good models for the investigation of human dosage-sensitive XY sex reversal.


Assuntos
Ovário/metabolismo , Processos de Determinação Sexual , Fator Esteroidogênico 1/metabolismo , Testículo/metabolismo , Proteínas WT1/metabolismo , Proteína Wnt4/metabolismo , Alelos , Animais , Feminino , Dosagem de Genes , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hibridização In Situ , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Ovário/crescimento & desenvolvimento , Fatores de Transcrição SOXB1/genética , Fator Esteroidogênico 1/genética , Testículo/crescimento & desenvolvimento , Proteínas WT1/genética , Proteína Wnt4/genética , Cromossomo X/genética , Cromossomo Y/genética
20.
Horm Behav ; 59(4): 556-64, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21376051

RESUMO

Environmental cues and social interactions are known to influence reproductive physiology and behavior in vertebrates. In female birds, male courtship displays can result in the growth of ovarian follicles, the production of reproductive hormones, and stimulation of oviduct development, all of which have the potential to influence maternal investment. Male Japanese quail follow a typical sequence of copulatory behaviors during a mating interaction and often force copulations with unreceptive females. We hypothesized that female Japanese quail could adjust maternal investment in response to male copulatory behaviors during a single mating interaction. We investigated the relationships between 1) male copulatory behaviors and post-mating concentrations of steroids in the female, 2) female steroid concentrations and fertilization success of inseminations and 3) female steroid concentrations and the offspring sex ratio. We found that male condition and copulatory behaviors predicted female steroid concentrations and maternal investment in eggs laid after a mating trial. The body condition of one or both mates was a significant predictor of the changes in female corticosterone and testosterone concentrations after mating, whereas specific male copulatory behaviors significantly predicted changes in female progesterone concentrations. Male and female body condition, male neck grabs and post-mating concentrations of female corticosterone, progesterone, and testosterone were all significant predictors of egg fertilization rates. Female body condition, male copulation efficiency, and female testosterone concentrations were significant predictors of offspring sex ratios. Our results show that phenotypic and behavioral characteristics of male Japanese quail modulate female steroid concentrations and result in changes in maternal investment.


Assuntos
Copulação/fisiologia , Corticosterona/sangue , Coturnix/fisiologia , Fertilização/fisiologia , Progesterona/sangue , Testosterona/sangue , Animais , Feminino , Masculino , Radioimunoensaio , Razão de Masculinidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...