Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 2656, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38531872

RESUMO

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.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila/genética , Fatores de Transcrição/metabolismo , Proteínas de Drosophila/metabolismo , Transdução de Sinais/fisiologia , Intestinos , Diferenciação Celular/genética , Cromatina
2.
Nat Commun ; 13(1): 6525, 2022 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-36316343

RESUMO

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.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila/genética , Drosophila/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Células Enteroendócrinas/metabolismo , Regulação da Expressão Gênica , Mamíferos/metabolismo
3.
Methods Cell Biol ; 170: 169-187, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35811098

RESUMO

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.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Diferenciação Celular/genética , Proliferação de Células/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Mucosa Intestinal , Intestinos , Células-Tronco
4.
FEBS J ; 289(16): 4773-4796, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-34115929

RESUMO

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.


Assuntos
Drosophila melanogaster , Drosophila , Animais , Drosophila melanogaster/genética , Células Enteroendócrinas , Mamíferos
5.
Stem Cell Rev Rep ; 17(6): 2304-2313, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34561772

RESUMO

Polycomb group protein Bmi1 is essential for hematopoietic stem cell (HSC) self-renewal and terminal differentiation. However, its target genes in hematopoietic stem and progenitor cells are largely unknown. We performed gene expression profiling assays and found that genes of the Wnt signaling pathway are significantly elevated in Bmi1 null hematopoietic stem and progenitor cells (HSPCs). Bmi1 is associated with several genes of the Wnt signaling pathway in hematopoietic cells. Further, we found that Bmi1 represses Wnt gene expression in HSPCs. Importantly, loss of ß-catenin, which reduces Wnt activation, partially rescues the HSC self-renewal and differentiation defects seen in the Bmi1 null mice. Thus, we have identified Bmi1 as a novel regulator of Wnt signaling pathway in HSPCs. Given that Wnt signaling pathway plays an important role in hematopoiesis, our studies suggest that modulating Wnt signaling may hold potential for enhancing HSC self-renewal, thereby improving the outcomes of HSC transplantation.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Via de Sinalização Wnt , Animais , Hematopoese/genética , Células-Tronco Hematopoéticas , Camundongos , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Via de Sinalização Wnt/genética
6.
Cell Regen ; 10(1): 24, 2021 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-34080092

RESUMO

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).

7.
Cell Regen ; 10(1): 1, 2021 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-33385259

RESUMO

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.

8.
Curr Biol ; 31(4): 840-852.e5, 2021 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-33340458

RESUMO

In the germarium of the Drosophila ovary, developing germline cysts are surrounded by a population of somatic escort cells that are known to function as the niche cells for germline differentiation;1 however, the underlying molecular mechanisms of this niche function remain poorly understood. Through single-cell gene expression profiling combined with genetic analyses, we here demonstrate that the escort cells can be spatially and functionally divided into two successive domains. The anterior escort cells (aECs) specifically produce ecdysone, which acts on the cystoblast to promote synchronous cell division, whereas the posterior escort cells (pECs) respond to ecdysone signaling and regulate soma-germline cell adhesion to promote the transition from 16-cell cyst-to-egg chamber formation. The patterning of the aEC and pEC domains is independent of the germline but is dependent on JAK/STAT signaling activity, which emanates from the posterior. Thus, a heterogeneous population of escort cells constitutes a stepwise niche environment to orchestrate cystoblast division and differentiation toward egg chamber formation.


Assuntos
Cistos , Proteínas de Drosophila , Ecdisona , Animais , Diferenciação Celular , Drosophila , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Ecdisona/metabolismo , Feminino , Células Germinativas , Ovário , Células-Tronco
10.
Cell Rep ; 31(8): 107683, 2020 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-32460025

RESUMO

Balanced stem cell self-renewal and differentiation is essential for maintaining tissue homeostasis, but the underlying mechanisms are poorly understood. Here, we identified the transcription factor SRY-related HMG-box (Sox) 100B, which is orthologous to mammalian Sox8/9/10, as a common target and central mediator of the EGFR/Ras and JAK/STAT signaling pathways that coordinates intestinal stem cell (ISC) proliferation and differentiation during both normal epithelial homeostasis and stress-induced intestinal repair in Drosophila. The two stress-responsive pathways directly regulate Sox100B transcription via two separate enhancers. Interestingly, an appropriate level of Sox100B is critical for its function, as its depletion inhibits ISC proliferation via cell cycle arrest, while its overexpression also inhibits ISC proliferation by directly suppressing EGFR expression and additionally promotes ISC differentiation by activating a differentiation-promoting regulatory circuitry composed of Sox100B, Sox21a, and Pdm1. Thus, our study reveals a Sox family transcription factor that functions as a stress-responsive signaling nexus that ultimately controls tissue homeostasis and regeneration.


Assuntos
Proteínas de Drosophila/metabolismo , Intestinos/fisiopatologia , Fatores de Transcrição SOX9/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Drosophila , Homeostase , Regeneração
11.
J Immunol ; 204(12): 3262-3272, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32332108

RESUMO

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.


Assuntos
Subpopulações de Linfócitos B/metabolismo , Complexo Repressor Polycomb 1/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Subpopulações de Linfócitos B/fisiologia , Medula Óssea/metabolismo , Linhagem da Célula/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Microambiente Celular/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Linfócitos/metabolismo , Linfócitos/fisiologia , Linfopoese/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID
12.
Cell Rep ; 30(6): 1724-1734.e4, 2020 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-32049006

RESUMO

Intestinal stem cells (ISCs) are able to generate gut-specific enterocytes, as well as neural-like enteroendocrine cells. It is unclear how the tissue identity of the ISC lineage is regulated to confer cell-lineage fidelity. Here, we show that, in adult Drosophila midgut, loss of the transcriptional repressor Tramtrack in ISCs causes a self-renewal program switch to neural stem cell (NSC)-like, and that switch drives neuroendocrine tumor development. In Tramtrack-depleted ISCs, the ectopically expressed Deadpan acts as a major self-renewal factor for cell propagation, and Sequoia acts as a differentiation factor for the neuroendocrine phenotype. In addition, the expression of Sequoia renders NSC-specific self-renewal genes responsive to Notch in ISCs, thus inverting the differentiation-promoting function of Notch into a self-renewal role as in normal NSCs. These results suggest an active maintenance mechanism for the gut identity of ISCs, whose disruption may lead to an improper acquisition of NSC-like traits and tumorigenesis.

13.
Cell Rep ; 29(12): 4172-4185.e5, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31851941

RESUMO

Enteroendocrine cells (EEs) in the intestinal epithelium have important endocrine functions, yet this cell lineage exhibits great local and regional variations that have hampered detailed characterization of EE subtypes. Through single-cell RNA-sequencing analysis, combined with a collection of peptide hormone and receptor knockin strains, here we provide a comprehensive analysis of cellular diversity, spatial distribution, and transcription factor (TF) code of EEs in adult Drosophila midgut. We identify 10 major EE subtypes that totally produced approximately 14 different classes of hormone peptides. Each EE on average co-produces approximately 2-5 different classes of hormone peptides. Functional screen with subtype-enriched TFs suggests a combinatorial TF code that controls EE cell diversity; class-specific TFs Mirr and Ptx1 respectively define two major classes of EEs, and regional TFs such as Esg, Drm, Exex, and Fer1 further define regional EE identity. Our single-cell data should greatly facilitate Drosophila modeling of EE differentiation and function.


Assuntos
Proteínas de Drosophila/metabolismo , Mucosa Intestinal/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Drosophila , Proteínas de Drosophila/genética , Células Enteroendócrinas , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Masculino , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Fatores de Transcrição/genética
14.
Stem Cell Reports ; 12(5): 1007-1023, 2019 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-30982741

RESUMO

Intestinal stem cell (ISC) differentiation in the Drosophila midgut requires Delta/Notch-mediated lateral inhibition, which separates the fate of ISCs from differentiating enteroblasts (EBs). Although a canonical Notch signaling cascade is involved in the lateral inhibition, its regulation at the transcriptional level is still unclear. Here we show that the establishment of lateral inhibition between ISC-EB requires two evolutionarily conserved transcriptional co-repressors Groucho (Gro) and C-terminal binding protein (CtBP) that act differently. Gro functions in EBs with E(spl)-C proteins to suppress Delta expression, inhibit cell-cycle re-entry, and promote cell differentiation, whereas CtBP functions specifically in ISCs to mediate transcriptional repression of Su(H) targets and maintain ISC fate. Interestingly, several E(spl)-C genes are also expressed in ISCs that cooperate with Gro to inhibit cell proliferation. Collectively, our study demonstrates separable and cell-type-specific functions of Gro and CtBP in a lateral inhibition process that controls the proliferation and differentiation of tissue stem cells.


Assuntos
Oxirredutases do Álcool/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/genética , Proteínas de Ligação a DNA/genética , Drosophila melanogaster/genética , Receptores Notch/genética , Proteínas Repressoras/genética , Células-Tronco/metabolismo , Oxirredutases do Álcool/metabolismo , Animais , Animais Geneticamente Modificados , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Divisão Celular/genética , Proliferação de Células/genética , Proteínas de Ligação a DNA/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Intestinos/citologia , Interferência de RNA , Receptores Notch/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais/genética , Células-Tronco/citologia
15.
Sci Rep ; 9(1): 3226, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30824860

RESUMO

Hbs1, which is homologous to the GTPase eRF3, is a small G protein implicated in mRNA quality control. It interacts with a translation-release factor 1-like protein Dom34/Pelota to direct decay of mRNAs with ribosomal stalls. Although both proteins are evolutionarily conserved in eukaryotes, the biological function of Hbs1 in multicellular organisms is yet to be characterized. In Drosophila, pelota is essential for the progression through meiosis during spermatogenesis and germline stem cell maintenance. Here we show that homozygous Hbs1 mutant flies are viable, female-fertile, but male-sterile, which is due to defects in meiosis and spermatid individualization, phenotypes that are also observed in pelota hypomorphic mutants. In contrast, Hbs1 mutants have no obvious defects in germline stem cell maintenance. We show that Hbs1 genetically interacts with pelota during spermatid individualization. Furthermore, Pelota with a point mutation on the putative Hbs1-binding site cannot substitute the wild type protein for normal spermatogenesis. These data suggest that Pelota forms a complex with Hbs1 to regulate multiple processes during spermatogenesis. Our results reveal a specific requirement of Hbs1 in male gametogenesis in Drosophila and indicate an essential role for the RNA surveillance complex Pelota-Hbs1 in spermatogenesis, a function that could be conserved in mammals.


Assuntos
Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Proteínas de Ligação ao GTP/genética , Proteínas de Choque Térmico HSP70/genética , Proteínas Nucleares/genética , Fatores de Alongamento de Peptídeos/genética , Espermatogênese/genética , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Feminino , Proteínas de Ligação ao GTP/metabolismo , Proteínas de Choque Térmico HSP70/metabolismo , Infertilidade Masculina/genética , Masculino , Meiose/genética , Mutação , Proteínas Nucleares/metabolismo , Fatores de Alongamento de Peptídeos/metabolismo , Ligação Proteica , Homologia de Sequência de Aminoácidos , Espermátides/metabolismo
16.
Dev Cell ; 48(3): 406-419.e5, 2019 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-30595536

RESUMO

Replication-dependent histone genes often reside in tandemly arrayed gene clusters, hindering systematic loss-of-function analyses. Here, we used CRISPR/Cas9 and the attP/attB double-integration system to alter numbers and sequences of histone genes in their original genomic context in Drosophila melanogaster. As few as 8 copies of the histone gene unit supported embryo development and adult viability, whereas flies with 20 copies were indistinguishable from wild-types. By hierarchical assembly, 40 alanine-substitution mutations (covering all known modified residues in histones H3 and H4) were introduced and characterized. Mutations at multiple residues compromised viability, fertility, and DNA-damage responses. In particular, H4K16 was necessary for expression of male X-linked genes, male viability, and maintenance of ovarian germline stem cells, whereas H3K27 was essential for late embryogenesis. Simplified mosaic analysis showed that H3R26 is required for H3K27 trimethylation. We have developed a powerful strategy and valuable reagents to systematically probe histone functions in D. melanogaster.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Biblioteca Gênica , Histonas/genética , Mutação/genética , Acetilação , Animais , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Histonas/metabolismo , Lisina/metabolismo
17.
Elife ; 82019 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-30648969

RESUMO

Heterochromatin Protein 1 (HP1) is a conserved chromosomal protein in eukaryotic cells that has a major role in directing heterochromatin formation, a process that requires co-transcriptional gene silencing mediated by small RNAs and their associated argonaute proteins. Heterochromatin formation requires erasing the active epigenetic mark, such as H3K4me2, but the molecular link between HP1 and H3K4 demethylation remains unclear. In a fertility screen in female Drosophila, we identified ovaries absent (ova), which functions in the stem cell niche, downstream of Piwi, to support germline stem cell differentiation. Moreover, ova acts as a suppressor of position effect variegation, and is required for silencing telomeric transposons in the germline. Biochemically, Ova acts to link the H3K4 demethylase dLsd1 to HP1a for local histone modifications. Therefore, our study provides a molecular connection between HP1a and local H3K4 demethylation during HP1a-mediated gene silencing that is required for ovary development, transposon silencing, and heterochromatin formation. Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Inativação Gênica , Heterocromatina/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Oxirredutases N-Desmetilantes/metabolismo , Fatores Genéricos de Transcrição/metabolismo , Animais , Homólogo 5 da Proteína Cromobox , Desmetilação , Feminino , Células Germinativas/citologia , Ovário/crescimento & desenvolvimento , Ovário/metabolismo , Ligação Proteica , Células-Tronco/citologia , Células-Tronco/metabolismo
18.
Oncotarget ; 9(81): 35283, 2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30443295

RESUMO

[This corrects the article DOI: 10.18632/oncotarget.5221.].

19.
Nat Cell Biol ; 20(8): 991, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29674680

RESUMO

In the version of this Article originally published, the author had misnumbered the reference citations in the Methods, using numbers 1-14 instead of 46-59. These errors have now been corrected in all online versions of the Article.

20.
Nat Cell Biol ; 20(2): 152-161, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29335529

RESUMO

The process through which multiple types of cell-lineage-restricted progenitor cells are specified from multipotent stem cells is unclear. Here we show that, in intestinal stem cell lineages in adult Drosophila, in which the Delta-Notch-signalling-guided progenitor cell differentiation into enterocytes is the default mode, the specification of enteroendocrine cells (EEs) is initiated by transient Scute activation in a process driven by transcriptional self-stimulation combined with a negative feedback regulation between Scute and Notch targets. Scute activation induces asymmetric intestinal stem cell divisions that generate EE progenitor cells. The mitosis-inducing and fate-inducing activities of Scute guide each EE progenitor cell to divide exactly once prior to its terminal differentiation, yielding a pair of EEs. The transient expression of a fate inducer therefore specifies both type and numbers of committed progenitor cells originating from stem cells, which could represent a general mechanism used for diversifying committed progenitor cells from multipotent stem cells.


Assuntos
Autorrenovação Celular/genética , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Receptores Notch/genética , Fatores de Transcrição/genética , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Células Enteroendócrinas/citologia , Células Enteroendócrinas/metabolismo , Intestinos/citologia , Transdução de Sinais/genética , Células-Tronco/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...