Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2023 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-36865263

RESUMEN

The gut epithelium is subject to constant renewal, a process reliant upon intestinal stem cell (ISC) proliferation that is driven by Wnt/ß-catenin signaling. Despite the importance of Wnt signaling within ISCs, the relevance of Wnt signaling within other gut cell types and the underlying mechanisms that modulate Wnt signaling in these contexts remain incompletely understood. Using challenge of the Drosophila midgut with a non-lethal enteric pathogen, we examine the cellular determinants of ISC proliferation, harnessing kramer, a recently identified regulator of Wnt signaling pathways, as a mechanistic tool. We find that Wnt signaling within Prospero-positive cells supports ISC proliferation and that kramer regulates Wnt signaling in this context by antagonizing kelch, a Cullin-3 E3 ligase adaptor that mediates Dishevelled polyubiquitination. This work establishes kramer as a physiological regulator of Wnt/ß-catenin signaling in vivo and suggests enteroendocrine cells as a new cell type that regulates ISC proliferation via Wnt/ß-catenin signaling.

2.
Elife ; 112022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35471187

RESUMEN

Mosquitoes transmit numerous pathogens, but large gaps remain in our understanding of their physiology. To facilitate explorations of mosquito biology, we have created Aegypti-Atlas (http://aegyptiatlas.buchonlab.com/), an online resource hosting RNAseq profiles of Ae. aegypti body parts (head, thorax, abdomen, gut, Malpighian tubules, ovaries), gut regions (crop, proventriculus, anterior and posterior midgut, hindgut), and a gut time course of blood meal digestion. Using Aegypti-Atlas, we provide insights into regionalization of gut function, blood feeding response, and immune defenses. We find that the anterior and posterior midgut possess digestive specializations which are preserved in the blood-fed state. Blood feeding initiates the sequential induction and repression/depletion of multiple cohorts of peptidases. With respect to defense, immune signaling components, but not recognition or effector molecules, show enrichment in ovaries. Basal expression of antimicrobial peptides is dominated by holotricin and gambicin, which are expressed in carcass and digestive tissues, respectively, in a mutually exclusive manner. In the midgut, gambicin and other effectors are almost exclusively expressed in the anterior regions, while the posterior midgut exhibits hallmarks of immune tolerance. Finally, in a cross-species comparison between Ae. aegypti and Anopheles gambiae midguts, we observe that regional digestive and immune specializations are conserved, indicating that our dataset may be broadly relevant to multiple mosquito species. We demonstrate that the expression of orthologous genes is highly correlated, with the exception of a 'species signature' comprising a few highly/disparately expressed genes. With this work, we show the potential of Aegypti-Atlas to unlock a more complete understanding of mosquito biology.


Asunto(s)
Aedes , Anopheles , Aedes/genética , Animales , Anopheles/genética , Femenino , Ovario , Azúcares , Transcriptoma
3.
Cell Rep ; 38(13): 110572, 2022 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-35354023

RESUMEN

Gut microbes play important roles in host physiology; however, the mechanisms underlying their impact remain poorly characterized. Here, we demonstrate that microbes not only influence gut physiology but also alter its epithelial composition. The microbiota and pathogens both influence intestinal stem cell (ISC) differentiation. Intriguingly, while the microbiota promotes ISC differentiation into enterocytes (EC), pathogens stimulate enteroendocrine cell (EE) fate and long-term accumulation of EEs in the midgut epithelium. Importantly, the evolutionarily conserved Drosophila NFKB (Relish) pushes stem cell lineage specification toward ECs by directly regulating differentiation factors. Conversely, the JAK-STAT pathway promotes EE fate in response to infectious damage. We propose a model in which the balance of microbial pattern recognition pathways, such as Imd-Relish, and damage response pathways, such as JAK-STAT, influence ISC differentiation, epithelial composition, and gut physiology.


Asunto(s)
Proteínas de Drosophila , Diferenciación Celular/fisiología , Proteínas de Drosophila/metabolismo , Enterocitos/metabolismo , Intestinos , Quinasas Janus/metabolismo , Factores de Transcripción STAT/metabolismo , Transducción de Señal
4.
Elife ; 102021 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-34553686

RESUMEN

The gut is the primary interface between an animal and food, but how it adapts to qualitative dietary variation is poorly defined. We find that the Drosophila midgut plastically resizes following changes in dietary composition. A panel of nutrients collectively promote gut growth, which sugar opposes. Diet influences absolute and relative levels of enterocyte loss and stem cell proliferation, which together determine cell numbers. Diet also influences enterocyte size. A high sugar diet inhibits translation and uncouples intestinal stem cell proliferation from expression of niche-derived signals, but, surprisingly, rescuing these effects genetically was not sufficient to modify diet's impact on midgut size. However, when stem cell proliferation was deficient, diet's impact on enterocyte size was enhanced, and reducing enterocyte-autonomous TOR signaling was sufficient to attenuate diet-dependent midgut resizing. These data clarify the complex relationships between nutrition, epithelial dynamics, and cell size, and reveal a new mode of plastic, diet-dependent organ resizing.


Asunto(s)
Dieta , Drosophila melanogaster/crecimiento & desarrollo , Tracto Gastrointestinal/crecimiento & desarrollo , Animales , Animales Modificados Genéticamente , Proliferación Celular , Drosophila melanogaster/fisiología , Enterocitos/citología , Femenino , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/fisiología , Masculino , Nicho de Células Madre
5.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33443193

RESUMEN

Drosophila melanogaster females experience a large shift in energy homeostasis after mating to compensate for nutrient investment in egg production. To cope with this change in metabolism, mated females undergo widespread physiological and behavioral changes, including increased food intake and altered digestive processes. The mechanisms by which the female digestive system responds to mating remain poorly characterized. Here, we demonstrate that the seminal fluid protein Sex Peptide (SP) is a key modulator of female post-mating midgut growth and gene expression. SP is both necessary and sufficient to trigger post-mating midgut growth in females under normal nutrient conditions, and likely acting via its receptor, Sex Peptide Receptor (SPR). Moreover, SP is responsible for almost the totality of midgut transcriptomic changes following mating, including up-regulation of protein and lipid metabolism genes and down-regulation of carbohydrate metabolism genes. These changes in metabolism may help supply the female with the nutrients required to sustain egg production. Thus, we report a role for SP in altering female physiology to enhance reproductive output: Namely, SP triggers the switch from virgin to mated midgut state.


Asunto(s)
Sistema Digestivo/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Fertilidad/fisiología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Receptores de Péptidos/metabolismo , Reproducción/fisiología , Transcriptoma/genética , Animales , Copulación , Sistema Digestivo/anatomía & histología , Sistema Digestivo/crecimiento & desarrollo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Femenino , Fertilidad/genética , Ontología de Genes , Intestinos/anatomía & histología , Intestinos/crecimiento & desarrollo , Intestinos/fisiología , Masculino , Receptores de Péptidos/genética , Reproducción/genética , Semen/metabolismo , Conducta Sexual Animal/fisiología , Transcriptoma/fisiología
6.
Cell Mol Gastroenterol Hepatol ; 9(3): 447-464, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31756561

RESUMEN

BACKGROUND & AIMS: The enteroendocrine cell (EEC) lineage is important for intestinal homeostasis. It was recently shown that EEC progenitors contribute to intestinal epithelial growth and renewal, but the underlying mechanisms remain poorly understood. MicroRNAs are under-explored along the entire EEC lineage trajectory, and comparatively little is known about their contributions to intestinal homeostasis. METHODS: We leverage unbiased sequencing and eight different mouse models and sorting methods to identify microRNAs enriched along the EEC lineage trajectory. We further characterize the functional role of EEC progenitor-enriched miRNA, miR-7, by in vivo dietary study as well as ex vivo enteroid in mice. RESULTS: First, we demonstrate that miR-7 is highly enriched across the entire EEC lineage trajectory and is the most enriched miRNA in EEC progenitors relative to Lgr5+ intestinal stem cells. Next, we show in vivo that in EEC progenitors miR-7 is dramatically suppressed under dietary conditions that favor crypt division and suppress EEC abundance. We then demonstrate by functional assays in mouse enteroids that miR-7 exerts robust control of growth, as determined by budding (proxy for crypt division), EdU and PH3 staining, and likely regulates EEC abundance also. Finally, we show by single-cell RNA sequencing analysis that miR-7 regulates Xiap in progenitor/stem cells and we demonstrate in enteroids that the effects of miR-7 on mouse enteroid growth depend in part on Xiap and Egfr signaling. CONCLUSIONS: This study demonstrates for the first time that EEC progenitor cell-enriched miR-7 is altered by dietary perturbations and that it regulates growth in enteroids via intact Xiap and Egfr signaling.


Asunto(s)
Células Enteroendocrinas/fisiología , Proteínas Inhibidoras de la Apoptosis/genética , Mucosa Intestinal/fisiología , MicroARNs/metabolismo , Células Madre/fisiología , Animales , Linaje de la Célula/genética , Proliferación Celular/genética , Células Cultivadas , Biología Computacional , Receptores ErbB/metabolismo , Conducta Alimentaria/fisiología , Femenino , Proteínas Inhibidoras de la Apoptosis/metabolismo , Mucosa Intestinal/citología , Masculino , Ratones , Ratones Transgénicos , Modelos Animales , Organoides , Cultivo Primario de Células , RNA-Seq , Transducción de Señal/genética , Análisis de la Célula Individual
7.
Cell Host Microbe ; 26(3): 412-425.e5, 2019 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-31492656

RESUMEN

Surviving infection requires immune and repair mechanisms. Developing organisms face the additional challenge of integrating these mechanisms with tightly controlled developmental processes. The larval Drosophila midgut lacks dedicated intestinal stem cells. We show that, upon infection, larvae perform limited repair using adult midgut precursors (AMPs). AMPs differentiate in response to damage to generate new enterocytes, transiently depleting their pool. Developmental delay allows for AMP reconstitution, ensuring the completion of metamorphosis. Notch signaling is required for the differentiation of AMPs into the encasing, niche-like peripheral cells (PCs), but not to differentiate PCs into enterocytes. Dpp (TGF-ß) signaling is sufficient, but not necessary, to induce PC differentiation into enterocytes. Infection-induced JAK-STAT pathway is both required and sufficient for differentiation of AMPs and PCs into new enterocytes. Altogether, this work highlights the constraints imposed by development on an organism's response to infection and demonstrates the transient use of adult precursors for tissue repair.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila/crecimiento & desarrollo , Drosophila/metabolismo , Tracto Gastrointestinal/metabolismo , Larva/metabolismo , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Drosophila/microbiología , Drosophila/fisiología , Proteínas de Drosophila/genética , Enterocitos/metabolismo , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/patología , Infecciones por Bacterias Gramnegativas/metabolismo , Infecciones por Bacterias Gramnegativas/patología , Quinasas Janus/metabolismo , Larva/inmunología , Larva/microbiología , Metamorfosis Biológica , Pectobacterium carotovorum/patogenicidad , Factores de Transcripción STAT/metabolismo , Transducción de Señal/fisiología , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Transcriptoma
8.
J Gerontol A Biol Sci Med Sci ; 74(10): 1549-1556, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30252027

RESUMEN

Dietary supplementation of glucomannan has been shown to have multiple health benefits, but its effect on life span has not been investigated. Here, we show that glucomannan hydrolysate (GMH) treatment extends mean life span of the model organism Drosophila melanogaster. To unravel the underlying mechanisms, we first examined the effect of GMH on the gut microbiota. We found that GMH treatment is associated with an elevated bacterial load in aged flies but overall has limited effects on the relative microbiota composition. We also demonstrated that GMH inhibits age-associated hyperproliferation of intestinal stem cells and thus delays the deterioration of gut integrity. Further analysis of the midgut transcriptome revealed that both EGFR/MAPK and JAK/STAT signaling pathways are suppressed in GMH groups. Multiple key regulators or effectors of EGFR/MAPK pathway, Ets21c, Mkp3, and Rho, are downregulated by GMH treatment. In the JAK/STAT pathway, major ligands (eg, Upd2 and Upd3) and negative feedback inhibitors (eg, Socs36e) are all significantly downregulated. Additionally, the expression of genes encoding antimicrobial peptides is elevated by GMH treatment. Taken together, our study shows that dietary supplementation of GMH can prolong life span, possibly through regulating gut proliferative homeostasis.


Asunto(s)
Catárticos/farmacología , Longevidad/efectos de los fármacos , Mananos/farmacología , Animales , Drosophila melanogaster , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Homeostasis/efectos de los fármacos , Masculino , Modelos Animales
9.
PLoS Genet ; 13(11): e1007091, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29108021

RESUMEN

Cytokine signaling is responsible for coordinating conserved epithelial regeneration and immune responses in the digestive tract. In the Drosophila midgut, Upd3 is a major cytokine, which is induced in enterocytes (EC) and enteroblasts (EB) upon oral infection, and initiates intestinal stem cell (ISC) dependent tissue repair. To date, the genetic network directing upd3 transcription remains largely uncharacterized. Here, we have identified the key infection-responsive enhancers of the upd3 gene and show that distinct enhancers respond to various stresses. Furthermore, through functional genetic screening, bioinformatic analyses and yeast one-hybrid screening, we determined that the transcription factors Scalloped (Sd), Mothers against dpp (Mad), and D-Fos are principal regulators of upd3 expression. Our study demonstrates that upd3 transcription in the gut is regulated by the activation of multiple pathways, including the Hippo, TGF-ß/Dpp, and Src, as well as p38-dependent MAPK pathways. Thus, these essential pathways, which are known to control ISC proliferation cell-autonomously, are also activated in ECs to promote tissue turnover the regulation of upd3 transcription.


Asunto(s)
Infecciones Bacterianas/metabolismo , Drosophila/genética , Drosophila/microbiología , Transducción de Señal , Animales , Infecciones Bacterianas/genética , Proliferación Celular , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Enterocitos/metabolismo , Femenino , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Intestinos/citología , Intestinos/microbiología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Sistema de Señalización de MAP Quinasas , Masculino , Pectobacterium carotovorum/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Pseudomonas/metabolismo , Células Madre/microbiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
10.
Open Biol ; 7(4)2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28424321

RESUMEN

The developmental signalling protein Notch can be proteolytically activated following ligand-interaction at the cell surface, or can be activated independently of its ligands, following Deltex (Dx)-induced Notch endocytosis and trafficking to the lysosomal membrane. The means by which different pools of Notch are directed towards these alternative outcomes remains poorly understood. We found that the Drosophila ZO-1 protein Polychaetoid (Pyd) suppresses specifically the Dx-induced form of Notch activation both in vivo and in cell culture assays. In vivo we confirmed the physiological relevance and direction of the Pyd/Dx interaction by showing that the expanded ovary stem cell niche phenotypes of pyd mutants require the presence of functional Dx and other components that are specific to the Dx-induced Notch activation mechanism. In S2 cells we found that Pyd can form a complex with Dx and Notch at the cell surface and reduce Dx-induced Notch endocytosis. Similar to other known activities of ZO-1 family proteins, the action of Pyd on Dx-induced endocytosis and signalling was found to be cell density dependent. Thus, together, our results suggest an alternative means by which external cues can tune Notch signalling through Pyd regulation of Dx-induced Notch trafficking.


Asunto(s)
Proteínas de Drosophila/metabolismo , Células Germinativas/metabolismo , Proteínas de la Membrana/metabolismo , Receptores Notch/metabolismo , Nicho de Células Madre , Proteínas de Uniones Estrechas/metabolismo , Proteínas de Drosophila/genética , Expresión Génica , Proteínas de la Membrana/genética , Mutación , Transporte de Proteínas , Transducción de Señal
12.
Dev Comp Immunol ; 64: 22-38, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-26855015

RESUMEN

The intestine acts as one of the interfaces between an organism and its external environment. As the primary digestive organ, it is constantly exposed to a multitude of stresses as it processes and absorbs nutrients. Among these is the recurring damage induced by ingested pathogenic and commensal microorganisms. Both the bacterial activity and immune response itself can result in the loss of epithelial cells, which subsequently requires replacement. In the Drosophila midgut, this regenerative role is fulfilled by intestinal stem cells (ISCs). Microbes not only trigger cell loss and replacement, but also modify intestinal and whole organism physiology, thus modulating ISC activity. Regulation of ISCs is integrated through a complex network of signaling pathways initiated by other gut cell populations, including enterocytes, enteroblasts, enteroendocrine and visceral muscles cells. The gut also receives signals from circulating immune cells, the hemocytes, to properly respond against infection. This review summarizes the types of gut microbes found in Drosophila, mechanisms for their elimination, and provides an integrated view of the signaling pathways that regulate tissue renewal in the midgut.


Asunto(s)
Drosophila melanogaster/inmunología , Mucosa Intestinal/inmunología , Intestinos/fisiología , Microbiota , Células Madre/fisiología , Animales , Antígenos Bacterianos/inmunología , Interacciones Huésped-Patógeno , Regeneración , Transducción de Señal
13.
BMC Dev Biol ; 15: 8, 2015 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-25637382

RESUMEN

BACKGROUND: Stem cells can respond to environmental and physiological inputs to adaptively remodel tissues. Little is known about whether stem cell niches are similarly responsive. The Drosophila ovary germline stem cell (GSC) niche is a well-studied model, which is comprised of cap cells that provide anchorage and maintenance signals for GSCs to maintain oogenesis. Previous studies have shown a strong link between diet and the regulation of oogenesis, making this a useful model system in which to investigate dietary regulation of the niche and its associated stem cells. RESULTS: We show that the Drosophila ovary GSC cap cell niche is a dynamic structure, which can contract and expand in fluctuating dietary conditions. Cap cells are lost when adult flies are shifted to nutrient poor diet and are restored after returning flies to nutrient-rich medium. Notch signalling in cap and escort cells is similarly reduced and restored by dietary shifts to nutrient poor and rich media. In old flies decreased Notch signalling is associated with decreased robustness of the niche to dietary changes. We demonstrated using a Notch temperature sensitive allele that removal and restoration of Notch signalling also leads to a reduction and re-expansion of the niche. Changes in niche size were not associated with apoptosis or cell division. We identified two distinct roles for Notch in the adult germarium. Notch can act in cap cells to prevent their loss while activation of Notch in the flanking escort cells results in expansion of the niche. CONCLUSIONS: We provide evidence that dietary changes alone are sufficient to alter Notch signalling and reversibly change niche size in the adult in wild type flies. We show Notch acts in different cells to maintain and re-expand the niche and propose a model in which cell fate transitions between cap cells and flanking somatic cells accounts for niche dynamics. These findings reveal an unexpected reversible plasticity of the GSC niche whose responses provide an integrated read out of the physiological status of the fly that is modulated by diet and age.


Asunto(s)
Dieta , Receptores Notch/metabolismo , Transducción de Señal , Células Madre/citología , Animales , Drosophila
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA