Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
J Immunol ; 204(12): 3262-3272, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32332108

RESUMEN

The self-renewal ability is a unique property of fetal-derived innate-like B-1a lymphocytes, which survive and function without being replenished by bone marrow (BM) progenitors. However, the mechanism by which IgM-secreting mature B-1a lymphocytes self-renew is poorly understood. In this study, we showed that Bmi1 was critically involved in this process. Although Bmi1 is considered essential for lymphopoiesis, the number of mature conventional B cells was not altered when Bmi1 was deleted in the B cell lineage. In contrast, the number of peritoneal B-1a cells was significantly reduced. Peritoneal cell transfer assays revealed diminished self-renewal ability of Bmi1-deleted B-1a cells, which was restored by additional deletion of Ink4-Arf, the well-known target of Bmi1 Fetal liver cells with B cell-specific Bmi1 deletion failed to repopulate peritoneal B-1a cells, but not other B-2 lymphocytes after transplantation assays, suggesting that Bmi1 may be involved in the developmental process of B-1 progenitors to mature B-1a cells. Although Bmi1 deletion has also been shown to alter the microenvironment for hematopoietic stem cells, fat-associated lymphoid clusters, the reported niche for B-1a cells, were not impaired in Bmi1 -/- mice. RNA expression profiling suggested lysine demethylase 5B (Kdm5b) as another possible target of Bmi1, which was elevated in Bmi1-/- B-1a cells in a stress setting and might repress B-1a cell proliferation. Our work has indicated that Bmi1 plays pivotal roles in self-renewal and maintenance of fetal-derived B-1a cells.


Asunto(s)
Subgrupos de Linfocitos B/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Subgrupos de Linfocitos B/fisiología , Médula Ósea/metabolismo , Linaje de la Célula/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Microambiente Celular/fisiología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/fisiología , Linfocitos/metabolismo , Linfocitos/fisiología , Linfopoyesis/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID
2.
Development ; 143(19): 3449-3458, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27510973

RESUMEN

Polycomb and Trithorax group (PcG and TrxG) genes function to regulate gene transcription by maintaining a repressive or active chromatin state, respectively. This antagonistic activity is important for body patterning during embryonic development, but whether this function module has a role in adult tissues is unclear. Here, we report that in the Drosophila ovary, disruption of the Polycomb repressive complex 1 (PRC1), specifically in the supporting escort cells, causes blockage of cystoblast differentiation and germline stem cell-like tumor formation. Tumors are caused by derepression of decapentaplegic (dpp), which prevents cystoblast differentiation. Interestingly, activation of dpp in escort cells requires the function of the TrxG gene brahma (brm), suggesting that loss of PRC1 in escort cells causes Brm-dependent dpp expression. Our study suggests a requirement for balanced activity between PcG and TrxG in an adult stem cell niche, and disruption of this balance could lead to the loss of tissue homeostasis and tumorigenesis.


Asunto(s)
Proteínas de Drosophila/metabolismo , Células Germinativas/citología , Complejo Represivo Polycomb 1/metabolismo , Células Madre/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proteínas de Drosophila/genética , Drosophila melanogaster , Complejo Represivo Polycomb 1/genética , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo , Proteínas Smad/genética , Proteínas Smad/metabolismo , Células Madre/citología
3.
Gastroenterology ; 153(6): 1607-1620, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28780076

RESUMEN

BACKGROUND & AIMS: Polycomb group proteins are epigenetic factors that silence gene expression; they are dysregulated in cancer cells and contribute to carcinogenesis by unclear mechanisms. We investigated whether BMI1 proto-oncogene, polycomb ring finger (BMI1), and polycomb group ring finger 2 (PCGF2, also called MEL18) are involved in the initiation and progression of colitis-associated cancer (CAC) in mice. METHODS: We generated mice containing floxed alleles of Bmi1 and/or Mel18 and/or Reg3b using the villin-Cre promoter (called Bmi1ΔIEC, Mel18ΔIEC, DKO, and TKO mice). We also disrupted Bmi1 and/or Mel18 specifically in intestinal epithelial cells (IECs) using the villin-CreERT2-inducible promoter. CAC was induced in cre-negative littermate mice (control) and mice with conditional disruption of Bmi1 and/or Mel18 by intraperitoneal injection of azoxymethane (AOM) followed by addition of dextran sulfate sodium (DSS) to drinking water. Colon tissues were collected from mice and analyzed by histology and immunoblots; IECs were isolated and used in cDNA microarray analyses. RESULTS: Following administration of AOM and DSS, DKO mice developed significantly fewer polyps than control, Bmi1ΔIEC, Mel18ΔIEC, Reg3bΔIEC, or TKO mice. Adenomas in the colons of DKO mice were low-grade dysplasias, whereas adenomas in control, Bmi1ΔIEC, Mel18ΔIEC, Reg3bΔIEC, or TKO mice were high-grade dysplasias with aggressive invasion of the muscularis mucosa. Disruption of Bmi1 and Mel18 (DKO mice) during late stages of carcinogenesis significantly reduced the numbers of large adenomas and the load of total adenomas, reduced proliferation, and increased apoptosis in colon tissues. IECs isolated from DKO mice after AOM and DSS administration had increased expression of Reg3b compared with control, Bmi1ΔIEC, or Mel18ΔIEC mice. Expression of REG3B was sufficient to inhibit cytokine-induced activation of STAT3 in IECs. The human REG3ß protein, the functional counterpart of mouse REG3B, inhibited STAT3 activity in human 293T cells, and its expression level in colorectal tumors correlated inversely with pSTAT3 level and survival times of patients. CONCLUSIONS: BMI1 and MEL18 contribute to the development of CAC in mice by promoting proliferation and reducing apoptosis via suppressing expression of Reg3b. REG3B negatively regulates cytokine-induced activation of STAT3 in colon epithelial cells. This pathway might be targeted in patients with colitis to reduce carcinogenesis.


Asunto(s)
Pólipos Adenomatosos/etiología , Transformación Celular Neoplásica/metabolismo , Colitis/complicaciones , Colon/enzimología , Neoplasias del Colon/etiología , Pólipos del Colon/etiología , Mucosa Intestinal/enzimología , Proteínas Asociadas a Pancreatitis/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Factor de Transcripción STAT3/metabolismo , Pólipos Adenomatosos/enzimología , Pólipos Adenomatosos/genética , Pólipos Adenomatosos/patología , Animales , Apoptosis , Factores de Coagulación Sanguínea/genética , Factores de Coagulación Sanguínea/metabolismo , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Colitis/enzimología , Colitis/genética , Colitis/patología , Colon/patología , Neoplasias del Colon/enzimología , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Pólipos del Colon/enzimología , Pólipos del Colon/genética , Pólipos del Colon/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Mucosa Intestinal/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Fosforilación , Complejo Represivo Polycomb 1/deficiencia , Complejo Represivo Polycomb 1/genética , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Proteínas de Unión al ARN , Proteínas Ribosómicas , Transducción de Señal , Factores de Tiempo
4.
Development ; 142(19): 3321-31, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26293304

RESUMEN

In adult Drosophila midgut, intestinal stem cells (ISCs) periodically produce progenitor cells that undergo a binary fate choice determined primarily by the levels of Notch activity that they receive, before terminally differentiating into enterocytes (ECs) or enteroendocrine (EE) cells. Here we identified Ttk69, a BTB domain-containing transcriptional repressor, as a master repressor of EE cell specification in the ISC lineages. Depletion of ttk69 in progenitor cells induced ISC proliferation and caused all committed progenitor cells to adopt EE fate, leading to the production of supernumerary EE cells in the intestinal epithelium. Conversely, forced expression of Ttk69 in progenitor cells was sufficient to prevent EE cell specification. The expression of Ttk69 was not regulated by Notch signaling, and forced activation of Notch, which is sufficient to induce EC specification of normal progenitor cells, failed to prevent EE cell specification of Ttk69-depleted progenitors. Loss of Ttk69 led to derepression of the acheate-scute complex (AS-C) genes scute and asense, which then induced prospero expression to promote EE cell specification. These studies suggest that Ttk69 functions in parallel with Notch signaling and acts as a master repressor of EE cell specification in Drosophila ISC lineages primarily by suppressing AS-C genes.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas de Drosophila/metabolismo , Drosophila/embriología , Células Madre Embrionarias/citología , Células Enteroendocrinas/citología , Intestinos/citología , Proteínas Represoras/metabolismo , Animales , Cartilla de ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Enteroendocrinas/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Intestinos/embriología , Proteínas del Tejido Nervioso/metabolismo , Receptores Notch/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo
5.
Cell Mol Life Sci ; 74(3): 435-448, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27600679

RESUMEN

Transposable elements or transposons are DNA pieces that can move around within the genome and are, therefore, potential threat to genome stability and faithful transmission of the genetic information in the germline. Accordingly, self-defense mechanisms have evolved in the metazoan germline to silence transposons, and the primary mechanism requires the germline-specific non-coding small RNAs, named Piwi-interacting RNA (piRNAs), which are in complex with Argonaute family of PIWI proteins (the piRNA-RISC complexes), to silence transposons. piRNA-mediated transposon silencing occurs at both transcriptional and post-transcriptional levels. With the advantages of genetic manipulation and advances of sequencing technology, much progress has been made on the molecular mechanisms of piRNA-mediated transposon silencing in Drosophila melanogaster, which will be the focus of this review. Because piRNA-mediated transposon silencing is evolutionarily conserved in metazoan, model organisms, such as Drosophila, will continue to be served as pioneer systems towards the complete understanding of transposon silencing in the metazoan germline.


Asunto(s)
Elementos Transponibles de ADN , Drosophila/genética , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Animales , Proteínas Argonautas/genética , Drosophila/crecimiento & desarrollo , Proteínas de Drosophila/genética , Células Germinativas/citología , ARN Interferente Pequeño/análisis , ARN Interferente Pequeño/metabolismo , Transcripción Genética
6.
Genes Dev ; 24(9): 933-46, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20439432

RESUMEN

Stem cells are critical for maintaining tissue homeostasis and are commonly governed by their niche microenvironment, although the intrinsic mechanisms controlling their multipotency are poorly understood. Polycomb group (PcG) genes are epigenetic silencers, and have emerged recently as important players in maintaining stem cell multipotency by preventing the initiation of differentiation programs. Here we describe an unexpected role of specific PcG genes in allowing adult stem cell differentiation and preventing stem cell-derived tumor development. We show that Posterior sex combs (Psc), which encodes a core Polycomb-repressive complex 1 (PRC1) component, functions redundantly with a similar gene, Suppressor of zeste two [Su(z)2], to restrict follicle stem cell (FSC) self-renewal in the Drosophila ovary. FSCs carrying deletion mutations of both genes extrude basally from the epithelium and continue to self-propagate at ectopic sites, leading to the development of FSC-like tumors. Furthermore, we show that the propagation of the mutant cells is driven by sustained activation of the canonical Wnt signaling pathway, which is essential for FSC self-renewal, whereas the epithelial extrusion is mediated through the planar cell polarity pathway. This study reveals a novel mechanism of epithelial extrusion, and indicates a novel role of polycomb function in allowing adult stem cell differentiation by antagonizing self-renewal programs. Given evolutionary conservation of PcG genes from Drosophila to mammals, they could have similar functions in mammalian stem cells and cancer.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/fisiología , Glicoproteínas/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , Animales , Diferenciación Celular , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Femenino , Eliminación de Gen , Ovario/citología , Ovario/patología , Complejo Represivo Polycomb 1 , Células Madre/citología , Células Madre/patología
7.
EMBO Rep ; 16(8): 965-74, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26124316

RESUMEN

Silencing of transposable elements (TEs) in the metazoan germline is critical for genome integrity and is primarily dependent on Piwi proteins and associated RNAs, which exert their function through both transcriptional and posttranscriptional mechanisms. Here, we report that the evolutionarily conserved Pelo (Dom34)-Hbs1 mRNA surveillance complex is required for transposon silencing in the Drosophila germline. In pelo mutant gonads, mRNAs and proteins of some selective TEs are up-regulated. Pelo is not required for piRNA biogenesis, and our studies suggest that Pelo may function at the translational level to silence TEs: This function requires interaction with Hbs1, and overexpression of RpS30a partially reverts TE-silencing defects in pelo mutants. Interestingly, TE silencing and spermatogenesis defects in pelo mutants can also effectively be rescued by expressing the mammalian ortholog of Pelo. We propose that the Pelo-Hbs1 surveillance complex provides another level of defense against the expression of TEs in the germline of Drosophila and possibly all metazoa.


Asunto(s)
Elementos Transponibles de ADN/genética , Drosophila melanogaster/genética , Silenciador del Gen , ARN/genética , Animales , Proteínas de Drosophila/genética , Femenino , Proteínas de Unión al GTP/genética , Células Germinativas , Proteínas HSP70 de Choque Térmico/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Ovario/metabolismo , Factores de Elongación de Péptidos/genética , ARN Interferente Pequeño/genética
8.
J Cell Sci ; 126(Pt 17): 3884-92, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23843608

RESUMEN

Tubular sclerosis complex gene products TSC1 and TSC2 have evolutionarily conserved roles in cell growth from Drosophila to mammals. Here we reveal important roles for TSC1/2 in regulating intestinal stem cell (ISC) maintenance and differentiation of the enteroendocrine cell lineage in the Drosophila midgut. Loss of either the Tsc1 or Tsc2 gene in ISCs causes rapid ISC loss through TORC1 hyperactivation, because ISCs can be efficiently rescued by mutation of S6k or by rapamycin treatment. In addition, overexpression of Rheb, which triggers TORC1 activation, recapitulates the phenotype caused by TSC1/2 disruption. Genetic studies suggest that TSC1/2 maintains ISCs independently of nutritional status or Notch regulation, probably by inhibiting cell delamination. We show that Tsc1/Tsc2 mutant ISCs can efficiently produce enterocytes but not enteroendocrine cells, and this altered differentiation potential is also caused by hyperactivation of TORC1. Reduced TORC1-S6K signaling by mutation of S6k, however, has no effect on ISC maintenance or cell lineage differentiation. Our studies demonstrate that hyperactivation of TORC1 following the loss of TSC1/2 is detrimental to stem cell maintenance and multiple lineage differentiation in the Drosophila ISC lineage, a mechanism that could be conserved in other stem cell lineages, including that in humans.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Dieta , Drosophila melanogaster , Enterocitos/metabolismo , Células Enteroendocrinas/metabolismo , Intestinos/citología , Proteínas de Unión al GTP Monoméricas/metabolismo , Neuropéptidos/metabolismo , Proteína Homóloga de Ras Enriquecida en el Cerebro , Receptores Notch/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Transducción de Señal/genética , Sirolimus/farmacología
9.
Dev Biol ; 377(1): 177-87, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23410794

RESUMEN

Tissue-specific stem cells are maintained by both local secreted signals and cell adhesion molecules that position the stem cells in the niche microenvironment. In the Drosophila midgut, multipotent intestinal stem cells (ISCs) are located basally along a thin layer of basement membrane that composed of extracellular matrix (ECM), which separates ISCs from the surrounding visceral musculature: the muscle cells constitute a regulatory niche for ISCs by producing multiple secreted signals that directly regulate ISC maintenance and proliferation. Here we show that integrin-mediated cell adhesion, which connects the ECM and intracellular cytoskeleton, is required for ISC anchorage to the basement membrane. Specifically, the α-integrin subunits including αPS1 encoded by mew and αPS3 encoded by scb, and the ß-integrin subunit encoded by mys are richly expressed in ISCs and are required for the maintenance, rather than their survival or multiple lineage differentiation. Furthermore, ISC maintenance also requires the intercellular and intracellular integrin signaling components including Talin, Integrin-linked kinase (Ilk), and the ligand, Laminin A. Notably, integrin mutant ISCs are also less proliferative, and genetic interaction studies suggest that proper integrin signaling is a pre-requisite for ISC proliferation in response to various proliferative signals and for the initiation of intestinal hyperplasia after loss of adenomatous polyposis coli (Apc). Our studies suggest that integrin not only functions to anchor ISCs to the basement membrane, but also serves as an essential element for ISC proliferation during normal homeostasis and in response to oncogenic mutations.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Integrinas/metabolismo , Intestinos/citología , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo , Animales , Adhesión Celular , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Supervivencia Celular , Proteínas del Citoesqueleto/metabolismo , Drosophila melanogaster/metabolismo , Hiperplasia
10.
Dev Biol ; 378(2): 122-40, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23570874

RESUMEN

Adenomatous polyposis coli (APC) and K-ras are the two most frequently mutated genes found in human colorectal cancers. In human colorectal cancers, Wnt signaling activation after the loss of APC is hypothesized to be the key event for adenoma initiation, whereas additional mutations such as Ras activation are required for the progression from adenoma to carcinoma. However, accumulating data have led to conflicting views regarding the precise role of Ras in APC loss-induced tumorigenesis. Here, using Drosophila midgut as a model system, we show that in the absence of Ras, APC mutant epithelial cells cannot initiate hyperplasia, suggesting that Ras plays an essential role in tumor initiation. Conversely, activating Ras by expressing oncogenic Ras or Raf in APC-deficient cells led to a blockage of cell differentiation and to preinvasive tumor outgrowth, characteristics that are shared by advanced colorectal carcinoma in humans. Mechanistically, we find that Ras is not required for Wnt signaling activation after APC loss, although Ras hyperactivation is able to potentiate Wnt signaling by increasing the cytoplasmic and nuclear accumulation of Armadillo/ß-catenin via mechanisms independent of JNK/Rac1 or PI3K-Akt signaling, partly owing to the downregulation of DE-cadherin. Together with the data from gene expression analyses, our results indicate that both parallel and cooperative mechanisms of Wnt and Ras signaling are responsible for the initiation and progression of intestinal tumorigenesis after APC loss.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteínas del Citoesqueleto/genética , Proteínas de Drosophila/genética , Mucosa Intestinal/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas del Dominio Armadillo/genética , Proteínas del Dominio Armadillo/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Núcleo Celular/metabolismo , Transformación Celular Neoplásica/metabolismo , Citoplasma/metabolismo , Proteínas del Citoesqueleto/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Perfilación de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Neoplasias Intestinales/genética , Neoplasias Intestinales/metabolismo , Intestinos/patología , Microscopía Confocal , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
11.
Nature ; 455(7216): 1119-23, 2008 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-18806781

RESUMEN

In the Drosophila midgut, multipotent intestinal stem cells (ISCs) that are scattered along the epithelial basement membrane maintain tissue homeostasis by their ability to steadily produce daughters that differentiate into either enterocytes or enteroendocrine cells, depending on the levels of Notch activity. However, the mechanisms controlling ISC self-renewal remain elusive. Here we show that a canonical Wnt signalling pathway controls ISC self-renewal. The ligand Wingless (Wg) is specifically expressed in the circular muscles next to ISCs, separated by a thin layer of basement membrane. Reduced function of wg causes ISC quiescence and differentiation, whereas wg overexpression produces excessive ISC-like cells that express high levels of the Notch ligand, Delta. Clonal analysis shows that the main downstream components of the Wg pathway, including Frizzled, Dishevelled and Armadillo, are autonomously required for ISC self-renewal. Furthermore, epistatic analysis suggests that Notch acts downstream of the Wg pathway and a hierarchy of Wg/Notch signalling pathways controls the balance between self-renewal and differentiation of ISCs. These data suggest that the underlying circular muscle constitutes the ISC niche, which produce Wg signals that act directly on ISCs to promote ISC self-renewal. This study demonstrates markedly conserved mechanisms regulating ISCs from Drosophila to mammals. The identification of the Drosophila ISC niche and the principal self-renewal signal will facilitate further understanding of intestinal homeostasis control and tumorigenesis.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Comunicación Paracrina , Células Madre/citología , Células Madre/metabolismo , Proteína Wnt1/metabolismo , Animales , Proliferación Celular , Drosophila melanogaster/metabolismo , Femenino , Regulación de la Expresión Génica , Intestinos/citología , Músculos/metabolismo , Receptores Notch/metabolismo , Transducción de Señal
12.
Nat Commun ; 15(1): 2656, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38531872

RESUMEN

The manipulation of cell identity by reprograming holds immense potential in regenerative medicine, but is often limited by the inefficient acquisition of fully functional cells. This problem can potentially be resolved by better understanding the reprogramming process using in vivo genetic models, which are currently scarce. Here we report that both enterocytes (ECs) and enteroendocrine cells (EEs) in adult Drosophila midgut show a surprising degree of cell plasticity. Depleting the transcription factor Tramtrack in the differentiated ECs can initiate Prospero-mediated cell transdifferentiation, leading to EE-like cells. On the other hand, depletion of Prospero in the differentiated EEs can lead to the loss of EE-specific transcription programs and the gain of intestinal progenitor cell identity, allowing cell cycle re-entry or differentiation into ECs. We find that intestinal progenitor cells, ECs, and EEs have a similar chromatin accessibility profile, supporting the concept that cell plasticity is enabled by pre-existing chromatin accessibility with switchable transcription programs. Further genetic analysis with this system reveals that the NuRD chromatin remodeling complex, cell lineage confliction, and age act as barriers to EC-to-EE transdifferentiation. The establishment of this genetically tractable in vivo model should facilitate mechanistic investigation of cell plasticity at the molecular and genetic level.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Drosophila/genética , Factores de Transcripción/metabolismo , Proteínas de Drosophila/metabolismo , Transducción de Señal/fisiología , Intestinos , Diferenciación Celular/genética , Cromatina
13.
Development ; 137(15): 2461-9, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20573703

RESUMEN

Tuberous sclerosis complex human disease gene products TSC1 and TSC2 form a functional complex that negatively regulates target of rapamycin (TOR), an evolutionarily conserved kinase that plays a central role in cell growth and metabolism. Here, we describe a novel role of TSC1/2 in controlling stem cell maintenance. We show that in the Drosophila ovary, disruption of either the Tsc1 or Tsc2 gene in germline stem cells (GSCs) leads to precocious GSC differentiation and loss. The GSC loss can be rescued by treatment with TORC1 inhibitor rapamycin, or by eliminating S6K, a TORC1 downstream effecter, suggesting that precocious differentiation of Tsc1/2 mutant GSC is due to hyperactivation of TORC1. One well-studied mechanism for GSC maintenance is that BMP signals from the niche directly repress the expression of a differentiation-promoting gene bag of marbles (bam) in GSCs. In Tsc1/2 mutant GSCs, BMP signalling activity is downregulated, but bam expression is still repressed. Moreover, Tsc1 bam double mutant GSCs could differentiate into early cystocytes, suggesting that TSC1/2 controls GSC differentiation via both BMP-Bam-dependent and -independent pathways. Taken together, these results suggest that TSC prevents precocious GSC differentiation by inhibiting TORC1 activity and subsequently differentiation-promoting programs. As TSC1/2-TORC1 signalling is highly conserved from Drosophila to mammals, it could have a similar role in controlling stem cell behaviour in mammals, including humans.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Proteínas de Drosophila/fisiología , Drosophila melanogaster/fisiología , Células Germinativas/citología , Células Madre/citología , Animales , Apoptosis , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Cruzamientos Genéticos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Microscopía Fluorescente/métodos , Modelos Biológicos , Mutación , Proteínas Serina-Treonina Quinasas/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR
14.
Dev Dyn ; 241(3): 455-64, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22241697

RESUMEN

BACKGROUND: The p55 family WD40 repeat-containing histone chaperone proteins are components of several chromatin regulatory complexes (such as PRC2, NURF and CAF-1) and interact with histone H4, yet their functional relevance in vivo is unclear. RESULTS: Here we use Drosophila as a genetic model to dissect the function of p55/Caf1 during development. In agree with a recent report, we find that p55 is essential for Drosophila development and is required for cell proliferation and viability. However, our data further demonstrate that histone H3K27 di-/tri-methylation and PRC2-mediated gene silencing still occur normally when p55 is missing. p55 is also implicated in bridging chromatin regulatory complexes to the chromatin by binding to histone H4, but we find that a transgene of p55 whose binding pocket is disrupted could still functionally substitute the wild-type p55 for the survival. CONCLUSIONS: Our studies suggest that p55 is not crucial for PRC2-mediated gene silencing in vivo, and the vital function of p55 is probably not dependent on its interaction with histone H4.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Regulación del Desarrollo de la Expresión Génica , Silenciador del Gen , Histonas/metabolismo , Proteína 4 de Unión a Retinoblastoma/metabolismo , Alelos , Animales , Proliferación Celular , Supervivencia Celular , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , N-Metiltransferasa de Histona-Lisina/metabolismo , Proteína 4 de Unión a Retinoblastoma/genética
15.
Dev Biol ; 354(1): 31-43, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21440535

RESUMEN

Tissue-specific adult stem cells are commonly associated with local niche for their maintenance and function. In the adult Drosophila midgut, the surrounding visceral muscle maintains intestinal stem cells (ISCs) by stimulating Wingless (Wg) and JAK/STAT pathway activities, whereas cytokine production in mature enterocytes also induces ISC division and epithelial regeneration, especially in response to stress. Here we show that EGFR/Ras/ERK signaling is another important participant in promoting ISC maintenance and division in healthy intestine. The EGFR ligand Vein is specifically expressed in muscle cells and is important for ISC maintenance and proliferation. Two additional EGFR ligands, Spitz and Keren, function redundantly as possible autocrine signals to promote ISC maintenance and proliferation. Notably, over-activated EGFR signaling could partially replace Wg or JAK/STAT signaling for ISC maintenance and division, and vice versa. Moreover, although disrupting any single one of the three signaling pathways shows mild and progressive ISC loss over time, simultaneous disruption of them all leads to rapid and complete ISC elimination. Taken together, our data suggest that Drosophila midgut ISCs are maintained cooperatively by multiple signaling pathway activities and reinforce the notion that visceral muscle is a critical component of the ISC niche.


Asunto(s)
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Transducción de Señal , Células Madre/metabolismo , Animales , Proliferación Celular , Proteínas de Drosophila/genética , Drosophila melanogaster/citología , Drosophila melanogaster/genética , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Inmunohistoquímica , Hibridación Fluorescente in Situ , Intestinos/citología , Quinasas Janus/genética , Quinasas Janus/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína Wnt1/genética , Proteína Wnt1/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
16.
Methods Cell Biol ; 170: 169-187, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35811098

RESUMEN

The intestinal epithelium in the anterior and posterior of the Drosophila midgut, which is maintained by intestinal stem cells (ISCs), represents a genetic tractable system for the study of stem cell biology, epithelial homeostasis and intestinal physiology and function. The ISCs self-renew and periodically generate absorptive enterocyte (EC) and secretory enteroendocrine cell (EE) via a committed progenitor stage termed as enteroblast (EB) or enteroendocrine progenitor (EEP), respectively. The progenitors in adult midgut are commonly referred to as all of the undifferentiated cells, including ISCs, EBs and EEPs. Under normal conditions, each of the above-mentioned specific type of cells can be reliably identified by a single cell marker or a combination of several cell markers. However, in aged or stressed gut, the increased proliferation and differentiation of ISCs may render many cell markers to be no longer strictly-specific to certain cell types. The self-renewal and differentiation abilities of ISCs or a particular cell of interest can be determined by cell lineage tracing analyses. Here, we provide detailed methods for the identification of ISC, EB and EEP in adult Drosophila gut, as well as methods for tracing the progenies of ISCs.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Diferenciación Celular/genética , Proliferación Celular/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Mucosa Intestinal , Intestinos , Células Madre
17.
Nat Commun ; 13(1): 6525, 2022 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-36316343

RESUMEN

Enteroendocrine cells (EEs) represent a heterogeneous cell population in intestine and exert endocrine functions by secreting a diverse array of neuropeptides. Although many transcription factors (TFs) required for specification of EEs have been identified in both mammals and Drosophila, it is not understood how these TFs work together to generate this considerable subtype diversity. Here we show that EE diversity in adult Drosophila is generated via an "additive hierarchical TF cascade". Specifically, a combination of a master TF, a secondary-level TF and a tertiary-level TF constitute a "TF code" for generating EE diversity. We also discover a high degree of post-specification plasticity of EEs, as changes in the code-including as few as one distinct TF-allow efficient switching of subtype identities. Our study thus reveals a hierarchically-organized TF code that underlies EE diversity and plasticity in Drosophila, which can guide investigations of EEs in mammals and inform their application in medicine.


Asunto(s)
Proteínas de Drosophila , Drosophila , Animales , Drosophila/genética , Drosophila/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Células Enteroendocrinas/metabolismo , Regulación de la Expresión Génica , Mamíferos/metabolismo
18.
FEBS J ; 289(16): 4773-4796, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-34115929

RESUMEN

Enteroendocrine cells (EECs) in both invertebrates and vertebrates derive from intestinal stem cells (ISCs) and are scattered along the digestive tract, where they function in sensing various environmental stimuli and subsequently secrete neurotransmitters or neuropeptides to regulate diverse biological and physiological processes. To fulfill these functions, EECs are specified into multiple subtypes that occupy specific gut regions. With advances in single-cell technology, organoid culture experimental systems, and CRISPR/Cas9-mediated genomic editing, rapid progress has been made toward characterization of EEC subtypes in mammals. Additionally, studies of genetic model organisms-especially Drosophila melanogaster-have also provided insights about the molecular processes underlying EEC specification from ISCs and about the establishment of diverse EEC subtypes. In this review, we compare the regulation of EEC specification and function in mammals and Drosophila, with a focus on EEC subtype characterization, on how internal and external regulators mediate EEC subtype specification, and on how EEC-mediated intra- and interorgan communications affect gastrointestinal physiology and pathology.


Asunto(s)
Drosophila melanogaster , Drosophila , Animales , Drosophila melanogaster/genética , Células Enteroendocrinas , Mamíferos
19.
Cell Regen ; 10(1): 24, 2021 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-34080092

RESUMEN

Increasing evidence suggest functional roles of subepithelial mesenchymal niche cells in maintaining intestinal stem cells and in modulating the pathogenesis of various intestinal diseases in mammals. A recent study reported the discovery of a new population of stromal cells in mice termed MAP3K2-Regulated Intestinal Stromal Cells (MRISCs); these cells reside at the base of colonic crypt and function to protect colonic stem cells during colonic inflammation by expressing the Wnt agonist R-spondin1 (Rspo1).

20.
Cell Regen ; 10(1): 1, 2021 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-33385259

RESUMEN

The rapidly self-renewing epithelium in the mammalian intestine is maintained by multipotent intestinal stem cells (ISCs) located at the bottom of the intestinal crypt that are interspersed with Paneth cells in the small intestine and Paneth-like cells in the colon. The ISC compartment is also closely associated with a sub-epithelial compartment that contains multiple types of mesenchymal stromal cells. With the advances in single cell and gene editing technologies, rapid progress has been made for the identification and characterization of the cellular components of the niche microenvironment that is essential for self-renewal and differentiation of ISCs. It has become increasingly clear that a heterogeneous population of mesenchymal cells as well as the Paneth cells collectively provide multiple secreted niche signals to promote ISC self-renewal. Here we review and summarize recent advances in the regulation of ISCs with a main focus on the definition of niche cells that sustain ISCs.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA