Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Nature ; 617(7962): 677-678, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37138060
3.
Nat Commun ; 14(1): 3076, 2023 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-37248237

RESUMEN

Coupling the release of pituitary hormones to the developmental stage of the oocyte is essential for female fertility. It requires estrogen to restrain kisspeptin (KISS1)-neuron pulsatility in the arcuate hypothalamic nucleus, while also exerting a surge-like effect on KISS1-neuron activity in the AVPV hypothalamic nucleus. However, a mechanistic basis for this region-specific effect has remained elusive. Our genomic analysis in female mice demonstrate that some processes, such as restraint of KISS1-neuron activity in the arcuate nucleus, may be explained by region-specific estrogen receptor alpha (ERα) DNA binding at gene regulatory regions. Furthermore, we find that the Kiss1-locus is uniquely regulated in these hypothalamic nuclei, and that the nuclear receptor co-repressor NR0B1 (DAX1) restrains its transcription specifically in the arcuate nucleus. These studies provide mechanistic insight into how ERα may control the KISS1-neuron, and Kiss1 gene expression, to couple gonadotropin release to the developmental stage of the oocyte.


Asunto(s)
Receptor Nuclear Huérfano DAX-1 , Receptor alfa de Estrógeno , Hipotálamo , Kisspeptinas , Animales , Femenino , Ratones , Núcleo Arqueado del Hipotálamo/metabolismo , Estradiol/metabolismo , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Estrógenos/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Receptor Nuclear Huérfano DAX-1/genética , Receptor Nuclear Huérfano DAX-1/metabolismo
4.
Dev Cell ; 57(24): 2675-2678, 2022 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-36538892

RESUMEN

Researchers are exploring sex differences in experimental models of both development and disease-but are we doing enough? In this collection of Voices, we celebrate researchers who are asking this question and starting to offer mechanistic clues on sexually dimorphic differences seen in interorgan communication, metabolic disease, neurological disorders, and more.


Asunto(s)
Caracteres Sexuales , Voz , Masculino , Humanos , Femenino
5.
Nat Metab ; 4(11): 1444-1458, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36396854

RESUMEN

The small intestine displays marked anatomical and functional plasticity that includes adaptive alterations in adult gut morphology, enteroendocrine cell profile and their hormone secretion, as well as nutrient utilization and storage. In this Perspective, we examine how shifts in dietary and environmental conditions bring about changes in gut size, and describe how the intestine adapts to changes in internal state, bowel resection and gastric bypass surgery. We highlight the critical importance of these intestinal remodelling processes in maintaining energy balance of the organism, and in protecting the metabolism of other organs. The intestinal resizing is supported by changes in the microbiota composition, and by activation of carbohydrate and fatty acid metabolism, which govern the intestinal stem cell proliferation, intestinal cell fate, as well as survivability of differentiated epithelial cells. The discovery that intestinal remodelling is part of the normal physiological adaptation to various triggers, and the potential for harnessing the reversible gut plasticity, in our view, holds extraordinary promise for developing therapeutic approaches against metabolic and inflammatory diseases.


Asunto(s)
Metabolismo Energético , Intestinos , Homeostasis , Intestinos/fisiología , Nutrientes , Dieta
6.
Nat Rev Mol Cell Biol ; 23(4): 227-228, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35197611
7.
Curr Biol ; 32(1): 149-163.e8, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-34798050

RESUMEN

Animals display selective escape behaviors when faced with environmental threats. Selection of the appropriate response by the underlying neuronal network is key to maximizing chances of survival, yet the underlying network mechanisms are so far not fully understood. Using synapse-level reconstruction of the Drosophila larval network paired with physiological and behavioral readouts, we uncovered a circuit that gates selective escape behavior for noxious light through acute and input-specific neuropeptide action. Sensory neurons required for avoidance of noxious light and escape in response to harsh touch, each converge on discrete domains of neuromodulatory hub neurons. We show that acute release of hub neuron-derived insulin-like peptide 7 (Ilp7) and cognate relaxin family receptor (Lgr4) signaling in downstream neurons are required for noxious light avoidance, but not harsh touch responses. Our work highlights a role for compartmentalized circuit organization and neuropeptide release from regulatory hubs, acting as central circuit elements gating escape responses.


Asunto(s)
Proteínas de Drosophila , Neuropéptidos , Animales , Drosophila/fisiología , Proteínas de Drosophila/genética , Drosophila melanogaster/fisiología , Larva/fisiología , Neuropéptidos/genética , Nociceptores/fisiología , Células Receptoras Sensoriales/fisiología
8.
Dev Cell ; 56(23): 3185-3191, 2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34875224

RESUMEN

In our 20th anniversary year, we reflect on how fields have changed since our first issue and here look to the future. In this collection of Voices, our writers speculate on the future: in terms of philosophy, cell states, cell processes, and then how to model cell systems.


Asunto(s)
Biología Celular , Biología Evolutiva , Publicaciones Periódicas como Asunto/estadística & datos numéricos , Humanos , Factores de Tiempo
9.
Cell Metab ; 33(12): 2307-2309, 2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34879236

RESUMEN

There are dramatic disparities in infection susceptibility within populations. In this issue of Cell Metabolism, Zhao and Karpac uncover a muscle-adipose-gut axis in Drosophila that explains variability in pathogen susceptibility. They show that the degree of intramuscular NF-κB activation accounts for differences in circulating glutamate, which enhances infection resistance at the expense of reproduction.


Asunto(s)
Ácido Glutámico , Reproducción , Tejido Adiposo , Humanos , Músculos , Transducción de Señal
10.
Ann N Y Acad Sci ; 1506(1): 55-73, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34414571

RESUMEN

There is an increasing appreciation for the role of metabolism in cell signaling and cell decision making. Precise metabolic control is essential in development, as evident by the disorders caused by mutations in metabolic enzymes. The metabolic profile of cells is often cell-type specific, changing as cells differentiate or during tumorigenesis. Recent evidence has shown that changes in metabolism are not merely a consequence of changes in cell state but that metabolites can serve to promote and/or inhibit these changes. Metabolites can link metabolic pathways with cell signaling pathways via several mechanisms, for example, by serving as substrates for protein post-translational modifications, by affecting enzyme activity via allosteric mechanisms, or by altering epigenetic markers. Unraveling the complex interactions governing metabolism, gene expression, and protein activity that ultimately govern a cell's fate will require new tools and interactions across disciplines. On March 24 and 25, 2021, experts in cell metabolism, developmental biology, and human disease met virtually for the Keystone eSymposium, "Metabolic Decisions in Development and Disease." The discussions explored how metabolites impact cellular and developmental decisions in a diverse range of model systems used to investigate normal development, developmental disorders, dietary effects, and cancer-mediated changes in metabolism.


Asunto(s)
Congresos como Asunto/tendencias , Desarrollo Humano/fisiología , Enfermedades Metabólicas/fisiopatología , Redes y Vías Metabólicas/fisiología , Neoplasias/fisiopatología , Informe de Investigación , Animales , Epigénesis Genética/fisiología , Humanos , Enfermedades Metabólicas/genética , Neoplasias/genética , Transducción de Señal/fisiología
11.
Cell Metab ; 33(7): 1279-1292, 2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34139200

RESUMEN

There has been rapid growth in the use of Drosophila and other invertebrate systems to dissect mechanisms governing metabolism. New assays and approaches to physiology have aligned with superlative genetic tools in fruit flies to provide a powerful platform for posing new questions, or dissecting classical problems in metabolism and disease genetics. In multiple examples, these discoveries exploit experimental advantages as-yet unavailable in mammalian systems. Here, we illustrate how fly studies have addressed long-standing questions in three broad areas-inter-organ signaling through hormonal or neural mechanisms governing metabolism, intestinal interoception and feeding, and the cellular and signaling basis of sexually dimorphic metabolism and physiology-and how these findings relate to human (patho)physiology. The imaginative application of integrative physiology and related approaches in flies to questions in metabolism is expanding, and will be an engine of discovery, revealing paradigmatic features of metabolism underlying human diseases and physiological equipoise in health.


Asunto(s)
Modelos Animales de Enfermedad , Drosophila melanogaster , Metabolismo Energético , Enfermedades Metabólicas/etiología , Animales , Animales Modificados Genéticamente , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Metabolismo Energético/genética , Humanos , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/patología , Modelos Biológicos , Transducción de Señal/fisiología
12.
Elife ; 102021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33448263

RESUMEN

Nutrient-dependent body size plasticity differs between the sexes in most species, including mammals. Previous work in Drosophila showed that body size plasticity was higher in females, yet the mechanisms underlying increased female body size plasticity remain unclear. Here, we discover that a protein-rich diet augments body size in females and not males because of a female-biased increase in activity of the conserved insulin/insulin-like growth factor signaling pathway (IIS). This sex-biased upregulation of IIS activity was triggered by a diet-induced increase in stunted mRNA in females, and required Drosophila insulin-like peptide 2, illuminating new sex-specific roles for these genes. Importantly, we show that sex determination gene transformer promotes the diet-induced increase in stunted mRNA via transcriptional coactivator Spargel to regulate the male-female difference in body size plasticity. Together, these findings provide vital insight into conserved mechanisms underlying the sex difference in nutrient-dependent body size plasticity.


Asunto(s)
Tamaño Corporal , Drosophila melanogaster/fisiología , Insulina/metabolismo , Transducción de Señal , Animales , Femenino , Masculino , Caracteres Sexuales , Procesos de Determinación del Sexo , Regulación hacia Arriba
13.
Nat Protoc ; 16(1): 239-262, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33247285

RESUMEN

Advances in light-sheet and confocal microscopy now allow imaging of cleared large biological tissue samples and enable the 3D appreciation of cell and protein localization in their native organ environment. However, the sample preparations for such imaging are often onerous, and their capability for antigen detection is limited. Here, we describe FLASH (fast light-microscopic analysis of antibody-stained whole organs), a simple, rapid, fully customizable technique for molecular phenotyping of intact tissue volumes. FLASH utilizes non-degradative epitope recovery and membrane solubilization to enable the detection of a multitude of membranous, cytoplasmic and nuclear antigens in whole mouse organs and embryos, human biopsies, organoids and Drosophila. Retrieval and immunolabeling of epithelial markers, an obstacle for previous clearing techniques, can be achieved with FLASH. Upon volumetric imaging, FLASH-processed samples preserve their architecture and integrity and can be paraffin-embedded for subsequent histopathological analysis. The technique can be performed by scientists trained in light microscopy and yields results in <1 week.


Asunto(s)
Antígenos/análisis , Técnica del Anticuerpo Fluorescente/métodos , Imagenología Tridimensional/métodos , Microscopía Confocal/métodos , Animales , Drosophila , Epítopos/análisis , Femenino , Humanos , Riñón/ultraestructura , Aparato Lagrimal/ultraestructura , Hígado/ultraestructura , Pulmón/ultraestructura , Masculino , Glándulas Mamarias Humanas/ultraestructura , Ratones , Organoides/ultraestructura , Páncreas/ultraestructura , Estómago/ultraestructura
15.
Nature ; 587(7834): 455-459, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33116314

RESUMEN

Reproduction induces increased food intake across females of many animal species1-4, providing a physiologically relevant paradigm for the exploration of appetite regulation. Here, by examining the diversity of enteric neurons in Drosophila melanogaster, we identify a key role for gut-innervating neurons with sex- and reproductive state-specific activity in sustaining the increased food intake of mothers during reproduction. Steroid and enteroendocrine hormones functionally remodel these neurons, which leads to the release of their neuropeptide onto the muscles of the crop-a stomach-like organ-after mating. Neuropeptide release changes the dynamics of crop enlargement, resulting in increased food intake, and preventing the post-mating remodelling of enteric neurons reduces both reproductive hyperphagia and reproductive fitness. The plasticity of enteric neurons is therefore key to reproductive success. Our findings provide a mechanism to attain the positive energy balance that sustains gestation, dysregulation of which could contribute to infertility or weight gain.


Asunto(s)
Drosophila melanogaster/citología , Drosophila melanogaster/fisiología , Ingestión de Alimentos/fisiología , Ingestión de Energía/fisiología , Madres , Neuronas/metabolismo , Reproducción/fisiología , Estructuras Animales/citología , Estructuras Animales/inervación , Estructuras Animales/metabolismo , Animales , Regulación del Apetito/fisiología , Femenino , Hiperfagia/metabolismo , Masculino , Neuropéptidos/metabolismo
16.
Nature ; 580(7802): 263-268, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32269334

RESUMEN

In cells, organs and whole organisms, nutrient sensing is key to maintaining homeostasis and adapting to a fluctuating environment1. In many animals, nutrient sensors are found within the enteroendocrine cells of the digestive system; however, less is known about nutrient sensing in their cellular siblings, the absorptive enterocytes1. Here we use a genetic screen in Drosophila melanogaster to identify Hodor, an ionotropic receptor in enterocytes that sustains larval development, particularly in nutrient-scarce conditions. Experiments in Xenopus oocytes and flies indicate that Hodor is a pH-sensitive, zinc-gated chloride channel that mediates a previously unrecognized dietary preference for zinc. Hodor controls systemic growth from a subset of enterocytes-interstitial cells-by promoting food intake and insulin/IGF signalling. Although Hodor sustains gut luminal acidity and restrains microbial loads, its effect on systemic growth results from the modulation of Tor signalling and lysosomal homeostasis within interstitial cells. Hodor-like genes are insect-specific, and may represent targets for the control of disease vectors. Indeed, CRISPR-Cas9 genome editing revealed that the single hodor orthologue in Anopheles gambiae is an essential gene. Our findings highlight the need to consider the instructive contributions of metals-and, more generally, micronutrients-to energy homeostasis.


Asunto(s)
Canales de Cloruro/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Ingestión de Alimentos/fisiología , Intestinos/fisiología , Zinc/metabolismo , Animales , Drosophila melanogaster/genética , Enterocitos/metabolismo , Femenino , Preferencias Alimentarias , Homeostasis , Insectos Vectores , Insulina/metabolismo , Activación del Canal Iónico , Larva/genética , Larva/crecimiento & desarrollo , Larva/metabolismo , Lisosomas/metabolismo , Masculino , Oocitos/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal , Xenopus
17.
Curr Opin Neurobiol ; 62: 83-91, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32028080

RESUMEN

The gastrointestinal tract is innervated by its own enteric nervous system and by extrinsic neurons that connect it with the central nervous system. Innervation allows the gastrointestinal tract to sense and respond to diverse stimuli, adjusting motility and secretion, but also affecting our physiology, behaviour and immunity. The mechanisms underlying the formation of gastrointestinal neurons are beginning to be elucidated; those that keep them plastic over an organism's lifetime remain to be explored. Here, we review the effects of microbiota, nutrients, sex and ageing on the morphology and function of gastrointestinal innervation in mammals, and discuss how this plasticity shapes gut-brain crosstalk and whole-body physiology. We also highlight insights gained by nascent studies of the enteric innervation of Drosophila melanogaster.


Asunto(s)
Sistema Nervioso Entérico , Tracto Gastrointestinal , Fenómenos Fisiológicos del Sistema Nervioso , Animales , Drosophila melanogaster , Neuronas
18.
Mol Metab ; 30: 221-229, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31767173

RESUMEN

OBJECTIVE: Coupling metabolic and reproductive pathways is essential for the survival of species. However, the functions of steroidogenic enzymes expressed in metabolic tissues are largely unknown. METHODS AND RESULTS: Here, we show that in the liver, the classical steroidogenic enzyme Cyp17a1 forms an essential nexus for glucose and ketone metabolism during feed-fast cycles. Both gain- and loss-of-function approaches are used to show that hepatic Cyp17a1 is induced by fasting, catalyzes the production of at least one hormone-ligand (DHEA) for the nuclear receptor PPARα, and is ultimately required for maintaining euglycemia and ketogenesis during nutrient deprivation. The feedback-loop that terminates Cyp17a1-PPARα activity, and re-establishes anabolic liver metabolism during re-feeding is mapped to postprandial bile acid-signaling, involving the receptors FXR, SHP and LRH-1. CONCLUSIONS: Together, these findings represent a novel paradigm of homeostatic control in which nutritional cues feed-forward on to metabolic pathways by influencing extragonadal steroidogenesis.


Asunto(s)
Hígado/metabolismo , PPAR alfa/metabolismo , Esteroide 17-alfa-Hidroxilasa/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Glucosa/metabolismo , Células HEK293 , Hepatocitos/metabolismo , Homeostasis , Humanos , Cetonas/metabolismo , Lipogénesis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oxidación-Reducción , Receptores Citoplasmáticos y Nucleares , Transducción de Señal , Esteroide 17-alfa-Hidroxilasa/fisiología
19.
Cell ; 178(4): 901-918.e16, 2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31398343

RESUMEN

Physiology and metabolism are often sexually dimorphic, but the underlying mechanisms remain incompletely understood. Here, we use the intestine of Drosophila melanogaster to investigate how gut-derived signals contribute to sex differences in whole-body physiology. We find that carbohydrate handling is male-biased in a specific portion of the intestine. In contrast to known sexual dimorphisms in invertebrates, the sex differences in intestinal carbohydrate metabolism are extrinsically controlled by the adjacent male gonad, which activates JAK-STAT signaling in enterocytes within this intestinal portion. Sex reversal experiments establish roles for this male-biased intestinal metabolic state in controlling food intake and sperm production through gut-derived citrate. Our work uncovers a male gonad-gut axis coupling diet and sperm production, revealing that metabolic communication across organs is physiologically important. The instructive role of citrate in inter-organ communication might be significant in more biological contexts than previously recognized.


Asunto(s)
Metabolismo de los Hidratos de Carbono/fisiología , Drosophila melanogaster/metabolismo , Ingestión de Alimentos/fisiología , Mucosa Intestinal/metabolismo , Caracteres Sexuales , Maduración del Esperma/fisiología , Animales , Ácido Cítrico/metabolismo , Proteínas de Drosophila/metabolismo , Femenino , Expresión Génica , Quinasas Janus/metabolismo , Masculino , RNA-Seq , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Azúcares/metabolismo , Testículo/metabolismo
20.
Genetics ; 210(2): 357-396, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30287514

RESUMEN

The gastrointestinal tract has recently come to the forefront of multiple research fields. It is now recognized as a major source of signals modulating food intake, insulin secretion and energy balance. It is also a key player in immunity and, through its interaction with microbiota, can shape our physiology and behavior in complex and sometimes unexpected ways. The insect intestine had remained, by comparison, relatively unexplored until the identification of adult somatic stem cells in the Drosophila intestine over a decade ago. Since then, a growing scientific community has exploited the genetic amenability of this insect organ in powerful and creative ways. By doing so, we have shed light on a broad range of biological questions revolving around stem cells and their niches, interorgan signaling and immunity. Despite their relatively recent discovery, some of the mechanisms active in the intestine of flies have already been shown to be more widely applicable to other gastrointestinal systems, and may therefore become relevant in the context of human pathologies such as gastrointestinal cancers, aging, or obesity. This review summarizes our current knowledge of both the formation and function of the Drosophila melanogaster digestive tract, with a major focus on its main digestive/absorptive portion: the strikingly adaptable adult midgut.


Asunto(s)
Drosophila melanogaster/fisiología , Tracto Gastrointestinal/fisiología , Animales , Drosophila melanogaster/anatomía & histología , Drosophila melanogaster/embriología , Tracto Gastrointestinal/anatomía & histología , Tracto Gastrointestinal/embriología , Morfogénesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...