Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Cell Rep ; 41(3): 111495, 2022 10 18.
Article in English | MEDLINE | ID: mdl-36261011

ABSTRACT

Somatic adult stem cell lineages in high-turnover tissues are under tight gene regulatory control. Like its mammalian counterpart, the Drosophila intestine precisely adjusts the rate of stem cell division with the onset of differentiation based on physiological demand. Although Notch signaling is indispensable for these decisions, the regulation of Notch activity that drives the differentiation of stem cell progenies into functional, mature cells is not well understood. Here, we report that commitment to the terminally differentiated enterocyte (EC) cell fate is under microRNA control. We show that an intestinally enriched microRNA, miR-956, fine-tunes Notch signaling activity specifically in intermediate, enteroblast (EB) progenitor cells to control EC differentiation. We further identify insensitive mRNA as a target of miR-956 that regulates EB/EC ratios by repressing Notch activity in EBs. In summary, our study highlights a post-transcriptional gene-regulatory mechanism for controlling differentiation in an adult intestinal stem cell lineage.


Subject(s)
Drosophila Proteins , MicroRNAs , Animals , Drosophila/genetics , Drosophila Proteins/genetics , Receptors, Notch/genetics , Drosophila melanogaster/physiology , MicroRNAs/genetics , Intestines , RNA, Messenger , Mammals/genetics
2.
Methods Mol Biol ; 2540: 79-92, 2022.
Article in English | MEDLINE | ID: mdl-35980573

ABSTRACT

Since the widespread discovery of microRNAs (miRNAs) 20 years ago, the Drosophila melanogaster model system has made important contributions to understanding the biology of this class of noncoding RNAs. These contributions are based on the amenability of this model system not only for biochemical analysis but molecular, genetic, and cell biological analyses as well. Nevertheless, while the Drosophila genome is now known to encode 258 miRNA precursors, the function of only a small minority of these have been well characterized. In this review, we summarize the current resources and methods that are available to study miRNA function in Drosophila with a particular focus on the large-scale resources that enable systematic analysis. Application of these methods will accelerate the discovery of ways that miRNAs are embedded into genetic networks that control basic features of metazoan cells.


Subject(s)
Drosophila , MicroRNAs , Animals , Computational Biology/methods , Drosophila/genetics , Drosophila melanogaster/genetics , Gene Regulatory Networks , MicroRNAs/chemistry , MicroRNAs/genetics
3.
Front Cell Dev Biol ; 10: 909212, 2022.
Article in English | MEDLINE | ID: mdl-35784477

ABSTRACT

Differential processing is a hallmark of clustered microRNAs (miRNAs) and the role of position and order of miRNAs in a cluster together with the contribution of stem-base and terminal loops has not been explored extensively within the context of a polycistronic transcript. To elucidate the structural attributes of a polycistronic transcript that contribute towards the differences in efficiencies of processing of the co-transcribed miRNAs, we constructed a series of chimeric variants of Drosophila let-7-Complex that encodes three evolutionary conserved and differentially expressed miRNAs (miR-100, let-7 and miR-125) and examined the expression and biological activity of the encoded miRNAs. The kinetic effects of Drosha and Dicer processing on the chimeric precursors were examined by in vitro processing assays. Our results highlight the importance of stem-base and terminal loop sequences in differential expression of polycistronic miRNAs and provide evidence that processing of a particular miRNA in a polycistronic transcript is in part determined by the kinetics of processing of adjacent miRNAs in the same cluster. Overall, this analysis provides specific guidelines for achieving differential expression of a particular miRNA in a cluster by structurally induced changes in primary miRNA (pri-miRNA) sequences.

4.
Genetics ; 222(2)2022 09 30.
Article in English | MEDLINE | ID: mdl-35762963

ABSTRACT

The regulation of stem cell survival, self-renewal, and differentiation is critical for the maintenance of tissue homeostasis. Although the involvement of signaling pathways and transcriptional control mechanisms in stem cell regulation have been extensively investigated, the role of post-transcriptional control is still poorly understood. Here, we show that the nuclear activity of the RNA-binding protein Second Mitotic Wave Missing is critical for Drosophila melanogaster intestinal stem cells and their daughter cells, enteroblasts, to maintain their progenitor cell properties and functions. Loss of swm causes intestinal stem cells and enteroblasts to stop dividing and instead detach from the basement membrane, resulting in severe progenitor cell loss. swm loss is further characterized by nuclear accumulation of poly(A)+ RNA in progenitor cells. Second Mitotic Wave Missing associates with transcripts involved in epithelial cell maintenance and adhesion, and the loss of swm, while not generally affecting the levels of these Second Mitotic Wave Missing-bound mRNAs, leads to elevated expression of proteins encoded by some of them, including the fly ortholog of Filamin. Taken together, this study indicates a nuclear role for Second Mitotic Wave Missing in adult stem cell maintenance, raising the possibility that nuclear post-transcriptional regulation of mRNAs encoding cell adhesion proteins ensures proper attachment of progenitor cells.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Animals , Cell Differentiation/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Filamins/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Stem Cells/metabolism
5.
Curr Biol ; 32(2): 386-397.e6, 2022 01 24.
Article in English | MEDLINE | ID: mdl-34875230

ABSTRACT

The role of processing bodies (P-bodies), key sites of post-transcriptional control, in adult stem cells remains poorly understood. Here, we report that adult Drosophila intestinal stem cells, but not surrounding differentiated cells such as absorptive enterocytes (ECs), harbor P-bodies that contain Drosophila orthologs of mammalian P-body components DDX6, EDC3, EDC4, and LSM14A/B. A targeted RNAi screen in intestinal progenitor cells identified 39 previously known and 64 novel P-body regulators, including Patr-1, a gene necessary for P-body assembly. Loss of Patr-1-dependent P-bodies leads to a loss of stem cells that is associated with inappropriate expression of EC-fate gene nubbin. Transcriptomic analysis of progenitor cells identifies a cadre of such weakly transcribed pro-differentiation transcripts that are elevated after P-body loss. Altogether, this study identifies a P-body-dependent repression activity that coordinates with known transcriptional repression programs to maintain a population of in vivo stem cells in a state primed for differentiation.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Cell Differentiation/genetics , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Intestines , Mammals , Stem Cells/metabolism
6.
Elife ; 102021 06 08.
Article in English | MEDLINE | ID: mdl-34100717

ABSTRACT

Dietary restriction (DR) extends healthy lifespan in diverse species. Age and nutrient-related changes in the abundance of microRNAs (miRNAs) and their processing factors have been linked to organismal longevity. However, the mechanisms by which they modulate lifespan and the tissue-specific role of miRNA-mediated networks in DR-dependent enhancement of lifespan remains largely unexplored. We show that two neuronally enriched and highly conserved microRNAs, miR-125 and let-7 mediate the DR response in Drosophila melanogaster. Functional characterization of miR-125 demonstrates its role in neurons while its target chinmo acts both in neurons and the fat body to modulate fat metabolism and longevity. Proteomic analysis revealed that Chinmo exerts its DR effects by regulating the expression of FATP, CG2017, CG9577, CG17554, CG5009, CG8778, CG9527, and FASN1. Our findings identify miR-125 as a conserved effector of the DR pathway and open the avenue for this small RNA molecule and its downstream effectors to be considered as potential drug candidates for the treatment of late-onset diseases and biomarkers for healthy aging in humans.


Subject(s)
Caloric Restriction , Drosophila Proteins/metabolism , Longevity/physiology , MicroRNAs/metabolism , Nerve Tissue Proteins/metabolism , Animals , Cell Line , Drosophila , Drosophila Proteins/analysis , Drosophila Proteins/chemistry , Embryo, Nonmammalian , Female , Signal Transduction/physiology
7.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Article in English | MEDLINE | ID: mdl-34016750

ABSTRACT

Adult organisms must sense and adapt to environmental fluctuations. In high-turnover tissues such as the intestine, these adaptive responses require rapid changes in gene expression that, in turn, likely involve posttranscriptional gene control. However, intestinal-tissue-specific microRNA (miRNA)-mediated regulatory pathways remain unexplored. Here, we report the role of an intestinal-specific miRNA, miR-958, that non-cell autonomously regulates stem cell numbers during tissue homeostasis and regeneration in the Drosophila adult midgut. We identify its downstream target cabut, the Drosophila ortholog of mammalian KLF10/11 transcription factors, which mediates this miR-958 function by promoting paracrine enterocyte-to-stem-cell bone morphogenetic protein (BMP) signaling. We also show that mature miR-958 levels transiently decrease in response to stress and that this decrease is required for proper stem cell expansion during tissue regeneration. In summary, we have identified a posttranscriptional mechanism that modulates BMP signaling activity within Drosophila adult intestinal tissue during both normal homeostasis and tissue regeneration to regulate intestinal stem cell numbers.


Subject(s)
Bone Morphogenetic Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Enterocytes/metabolism , MicroRNAs/genetics , Stem Cells/metabolism , Transcription Factors/genetics , Animals , Bleomycin/pharmacology , Bone Morphogenetic Proteins/metabolism , Cell Count , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Enterocytes/cytology , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeostasis/genetics , MicroRNAs/metabolism , Regeneration/genetics , Signal Transduction , Stem Cells/cytology , Transcription Factors/metabolism
8.
Development ; 148(1)2021 01 07.
Article in English | MEDLINE | ID: mdl-33246929

ABSTRACT

The adult Drosophila intestinal epithelium is a model system for stem cell biology, but its utility is limited by current biochemical methods that lack cell type resolution. Here, we describe a new proximity-based profiling method that relies upon a GAL4 driver, termed intestinal-kickout-GAL4 (I-KCKT-GAL4), that is exclusively expressed in intestinal progenitor cells. This method uses UV crosslinked whole animal frozen powder as its starting material to immunoprecipitate the RNA cargoes of transgenic epitope-tagged RNA binding proteins driven by I-KCKT-GAL4 When applied to the general mRNA-binder, poly(A)-binding protein, the RNA profile obtained by this method identifies 98.8% of transcripts found after progenitor cell sorting, and has low background noise despite being derived from whole animal lysate. We also mapped the targets of the more selective RNA binder, Fragile X mental retardation protein (FMRP), using enhanced crosslinking and immunoprecipitation (eCLIP), and report for the first time its binding motif in Drosophila cells. This method will therefore enable the RNA profiling of wild-type and mutant intestinal progenitor cells from intact flies exposed to normal and altered environments, as well as the identification of RNA-protein interactions crucial for stem cell function.


Subject(s)
Aging/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Genetic Techniques , Intestines/cytology , RNA/metabolism , Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , Enhancer Elements, Genetic/genetics , Female , Gene Expression Regulation , Organ Specificity , Poly(A)-Binding Proteins/metabolism , RNA/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
9.
G3 (Bethesda) ; 10(11): 4271-4285, 2020 11 05.
Article in English | MEDLINE | ID: mdl-32972999

ABSTRACT

Balancers are rearranged chromosomes used in Drosophila melanogaster to maintain deleterious mutations in stable populations, preserve sets of linked genetic elements and construct complex experimental stocks. Here, we assess the phenotypes associated with breakpoint-induced mutations on commonly used third chromosome balancers and show remarkably few deleterious effects. We demonstrate that a breakpoint in p53 causes loss of radiation-induced apoptosis and a breakpoint in Fucosyltransferase A causes loss of fucosylation in nervous and intestinal tissue-the latter study providing new markers for intestinal cell identity and challenging previous conclusions about the regulation of fucosylation. We also describe thousands of potentially harmful mutations shared among X or third chromosome balancers, or unique to specific balancers, including an Ankyrin2 mutation present on most TM3 balancers, and reiterate the risks of using balancers as experimental controls. We used long-read sequencing to confirm or refine the positions of two inversions with breakpoints lying in repetitive sequences and provide evidence that one of the inversions, In(2L)Cy, arose by ectopic recombination between foldback transposon insertions and the other, In(3R)C, cleanly separates subtelomeric and telomeric sequences and moves the subtelomeric sequences to an internal chromosome position. In addition, our characterization of In(3R)C shows that balancers may be polymorphic for terminal deletions. Finally, we present evidence that extremely distal mutations on balancers can add to the stability of stocks whose purpose is to maintain homologous chromosomes carrying mutations in distal genes. Overall, these studies add to our understanding of the structure, diversity and effectiveness of balancer chromosomes.


Subject(s)
Chromosomes , Drosophila melanogaster , Animals , Chromosome Inversion , Drosophila melanogaster/genetics , Mutation , Phenotype
10.
Genetics ; 216(4): 891-903, 2020 12.
Article in English | MEDLINE | ID: mdl-32988987

ABSTRACT

The Drosophila adult midgut is a model epithelial tissue composed of a few major cell types with distinct regional identities. One of the limitations to its analysis is the lack of tools to manipulate gene expression based on these regional identities. To overcome this obstacle, we applied the intersectional split-GAL4 system to the adult midgut and report 653 driver combinations that label cells by region and cell type. We first identified 424 split-GAL4 drivers with midgut expression from ∼7300 drivers screened, and then evaluated the expression patterns of each of these 424 when paired with three reference drivers that report activity specifically in progenitor cells, enteroendocrine cells, or enterocytes. We also evaluated a subset of the drivers expressed in progenitor cells for expression in enteroblasts using another reference driver. We show that driver combinations can define novel cell populations by identifying a driver that marks a distinct subset of enteroendocrine cells expressing genes usually associated with progenitor cells. The regional cell type patterns associated with the entire set of driver combinations are documented in a freely available website, providing information for the design of thousands of additional driver combinations to experimentally manipulate small subsets of intestinal cells. In addition, we show that intestinal enhancers identified with the split-GAL4 system can confer equivalent expression patterns on other transgenic reporters. Altogether, the resource reported here will enable more precisely targeted gene expression for studying intestinal processes, epithelial cell functions, and diseases affecting self-renewing tissues.


Subject(s)
Drosophila Proteins/genetics , Enhancer Elements, Genetic , Gene Targeting/methods , Genetic Engineering/methods , Intestinal Mucosa/cytology , Transcription Factors/genetics , Animals , Drosophila melanogaster , Enteroendocrine Cells/metabolism , Intestinal Mucosa/metabolism , Promoter Regions, Genetic
11.
Trends Microbiol ; 28(6): 430-432, 2020 06.
Article in English | MEDLINE | ID: mdl-32396824

ABSTRACT

Interactions between the eukaryotic host, microbiome members, and invading pathogens help to shape disease outcomes. Using the Drosophila model, Fast et al. identified that Vibrio cholerae acts to inhibit epithelial renewal through complex interactions between the type VI secretion system of V. cholerae and the microbial community of the fly.


Subject(s)
Cholera , Microbiota , Type VI Secretion Systems , Vibrio cholerae , Animals , Drosophila
12.
J Cell Sci ; 133(10)2020 05 18.
Article in English | MEDLINE | ID: mdl-32265270

ABSTRACT

Stressed cells downregulate translation initiation and assemble membrane-less foci termed stress granules (SGs). Although SGs have been extensively characterized in cultured cells, the existence of such structures in stressed adult stem cell pools remains poorly characterized. Here, we report that the Drosophila orthologs of the mammalian SG components AGO1, ATX2, CAPRIN, eIF4E, FMRP, G3BP, LIN-28, PABP and TIAR are enriched in adult fly intestinal progenitor cells, where they accumulate in small cytoplasmic messenger ribonucleoprotein complexes (mRNPs). Treatment with sodium arsenite or rapamycin reorganized these mRNPs into large cytoplasmic granules. Formation of these intestinal progenitor stress granules (IPSGs) depended on polysome disassembly, led to translational downregulation and was reversible. Although the canonical SG nucleators ATX2 and G3BP were sufficient for IPSG formation in the absence of stress, neither of them, nor TIAR, either individually or collectively, were required for stress-induced IPSG formation. This work therefore finds that IPSGs do not assemble via a canonical mechanism, raising the possibility that other stem cell populations employ a similar stress-response mechanism.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Argonaute Proteins , Cell Line , Cells, Cultured , Cytoplasmic Granules , Drosophila Proteins/genetics , Polyribosomes , RNA-Binding Proteins
13.
G3 (Bethesda) ; 10(4): 1393-1401, 2020 04 09.
Article in English | MEDLINE | ID: mdl-32071070

ABSTRACT

During Drosophila melanogaster metamorphosis, arrested immature neurons born during larval development differentiate into their functional adult form. This differentiation coincides with the downregulation of two zinc-finger transcription factors, Chronologically Inappropriate Morphogenesis (Chinmo) and the Z3 isoform of Broad (Br-Z3). Here, we show that br-Z3 is regulated by two microRNAs, let-7 and miR-125, that are activated at the larval-to-pupal transition and are known to also regulate chinmo The br-Z3 3'UTR contains functional binding sites for both let-7 and miR-125 that confers sensitivity to both of these microRNAs, as determined by deletion analysis in reporter assays. Forced expression of let-7 and miR-125 miRNAs leads to early silencing of Br-Z3 and Chinmo and is associated with inappropriate neuronal sprouting and outgrowth. Similar phenotypes were observed by the combined but not separate depletion of br-Z3 and chinmo Because persistent Br-Z3 was not detected in let-7-C mutants, this work suggests a model in which let-7 and miR-125 activation at the onset of metamorphosis may act as a failsafe mechanism that ensures the coordinated silencing of both br-Z3 and chinmo needed for the timely outgrowth of neurons arrested during larval development. The let-7 and miR-125 binding site sequences are conserved across Drosophila species and possibly other insects as well, suggesting that this functional relationship is evolutionarily conserved.


Subject(s)
Drosophila Proteins , MicroRNAs , Neurons , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , MicroRNAs/genetics , Nerve Tissue Proteins/genetics , Neurons/metabolism , Transcription Factors
14.
Development ; 146(17)2019 09 12.
Article in English | MEDLINE | ID: mdl-31399469

ABSTRACT

The dramatic growth that occurs during Drosophila larval development requires rapid conversion of nutrients into biomass. Many larval tissues respond to these biosynthetic demands by increasing carbohydrate metabolism and lactate dehydrogenase (LDH) activity. The resulting metabolic program is ideally suited for synthesis of macromolecules and mimics the manner by which cancer cells rely on aerobic glycolysis. To explore the potential role of Drosophila LDH in promoting biosynthesis, we examined how Ldh mutations influence larval development. Our studies unexpectedly found that Ldh mutants grow at a normal rate, indicating that LDH is dispensable for larval biomass production. However, subsequent metabolomic analyses suggested that Ldh mutants compensate for the inability to produce lactate by generating excess glycerol-3-phosphate (G3P), the production of which also influences larval redox balance. Consistent with this possibility, larvae lacking both LDH and G3P dehydrogenase (GPDH1) exhibit growth defects, synthetic lethality and decreased glycolytic flux. Considering that human cells also generate G3P upon inhibition of lactate dehydrogenase A (LDHA), our findings hint at a conserved mechanism in which the coordinate regulation of lactate and G3P synthesis imparts metabolic robustness to growing animal tissues.


Subject(s)
Drosophila melanogaster/physiology , Glycerolphosphate Dehydrogenase/metabolism , L-Lactate Dehydrogenase/metabolism , Larva/growth & development , Larva/metabolism , Sugars/metabolism , Adenosine Triphosphate/metabolism , Animals , Animals, Genetically Modified , Female , Glycerolphosphate Dehydrogenase/genetics , Glycolysis/genetics , Homeostasis/genetics , L-Lactate Dehydrogenase/genetics , Lactic Acid/biosynthesis , Male , Mutation , NAD/metabolism , Oxidation-Reduction
15.
Cell Rep ; 21(10): 2671-2677, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-29212015

ABSTRACT

Although the intrinsic mechanisms that control whether stem cells divide symmetrically or asymmetrically underlie tissue growth and homeostasis, they remain poorly defined. We report that the RNA-binding protein fragile X mental retardation protein (FMRP) limits the symmetric division, and resulting expansion, of the stem cell population during adaptive intestinal growth in Drosophila. The elevated insulin sensitivity that FMRP-deficient progenitor cells display contributes to their accelerated expansion, which is suppressed by the depletion of insulin-signaling components. This FMRP activity is mediated solely via a second conserved RNA-binding protein, LIN-28, known to boost insulin signaling in stem cells. Via LIN-28, FMRP controls progenitor cell behavior by post-transcriptionally repressing the level of insulin receptor (InR). This study identifies the stem cell-based mechanism by which FMRP controls tissue adaptation, and it raises the possibility that defective adaptive growth underlies the accelerated growth, gastrointestinal, and other symptoms that affect fragile X syndrome patients.


Subject(s)
Drosophila Proteins/metabolism , Fragile X Mental Retardation Protein/metabolism , Intestines/cytology , RNA-Binding Proteins/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Drosophila Proteins/genetics , Female , Fragile X Mental Retardation Protein/genetics , RNA-Binding Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism
16.
Methods Mol Biol ; 1478: 79-94, 2016.
Article in English | MEDLINE | ID: mdl-27730576

ABSTRACT

MicroRNAs are short noncoding, ~22-nucleotide RNAs that regulate protein abundance. The growth in our understanding of this class of RNAs has been rapid since their discovery just over a decade ago. We now appreciate that miRNAs are deeply embedded within the genetic networks that control basic features of metazoan cells including their identity, metabolism, and reproduction. The Drosophila melanogaster model system has made and will continue to make important contributions to this analysis. Intended as an introductory overview, here we review the current methods and resources available for functional analysis of fly miRNAs for those interested in performing this type of analysis.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation , MicroRNAs/genetics , RNA-Binding Proteins/genetics , Ribonuclease III/genetics , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Profiling , Genes, Reporter , Genetic Techniques , MicroRNAs/antagonists & inhibitors , MicroRNAs/classification , MicroRNAs/metabolism , Nucleic Acid Conformation , Oligonucleotide Array Sequence Analysis , RNA Isoforms/antagonists & inhibitors , RNA Isoforms/genetics , RNA Isoforms/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Proteins/metabolism , Ribonuclease III/metabolism , Signal Transduction
17.
PLoS Genet ; 12(8): e1006247, 2016 08.
Article in English | MEDLINE | ID: mdl-27508495

ABSTRACT

Messenger RNAs (mRNAs) often contain binding sites for multiple, different microRNAs (miRNAs). However, the biological significance of this feature is unclear, since such co-targeting miRNAs could function coordinately, independently, or redundantly with one another. Here, we show that two co-transcribed Drosophila miRNAs, let-7 and miR-125, non-redundantly regulate a common target, the transcription factor Chronologically Inappropriate Morphogenesis (Chinmo). We first characterize novel adult phenotypes associated with loss of both let-7 and miR-125, which are derived from a common, polycistronic transcript that also encodes a third miRNA, miR-100. Consistent with the coordinate upregulation of all three miRNAs in aging flies, these phenotypes include brain degeneration and shortened lifespan. However, transgenic rescue analysis reveal separable roles for these miRNAs: adult miR-125 but not let-7 mutant phenotypes are associated with ectopic Chinmo expression in adult brains and are suppressed by chinmo reduction. In contrast, let-7 is predominantly responsible for regulating chinmo during nervous system formation. These results indicate that let-7 and miR-125 function during two distinct stages, development and adulthood, rather than acting at the same time. These different activities are facilitated by an increased rate of processing of let-7 during development and a lower rate of decay of the accumulated miR-125 in the adult nervous system. Thus, this work not only establishes a key role for the highly conserved miR-125 in aging. It also demonstrates that two co-transcribed miRNAs function independently during distinct stages to regulate a common target, raising the possibility that such biphasic control may be a general feature of clustered miRNAs.


Subject(s)
Drosophila Proteins/genetics , Longevity/genetics , MicroRNAs/genetics , Nerve Tissue Proteins/genetics , Aging/genetics , Aging/pathology , Animals , Binding Sites , Brain/growth & development , Brain/metabolism , Drosophila/genetics , Drosophila/growth & development , Drosophila Proteins/biosynthesis , Gene Expression Regulation, Developmental , MicroRNAs/biosynthesis , Morphogenesis/genetics , Nerve Tissue Proteins/biosynthesis , Nervous System/growth & development , Nervous System/pathology , Neurons/metabolism
18.
Elife ; 52016 06 14.
Article in English | MEDLINE | ID: mdl-27296804

ABSTRACT

Pediatric neural tumors are often initiated during early development and can undergo very rapid transformation. However, the molecular basis of this early malignant susceptibility remains unknown. During Drosophila development, neural stem cells (NSCs) divide asymmetrically and generate intermediate progenitors that rapidly differentiate in neurons. Upon gene inactivation, these progeny can dedifferentiate and generate malignant tumors. Here, we find that intermediate progenitors are prone to malignancy only when born during an early window of development while expressing the transcription factor Chinmo, and the mRNA-binding proteins Imp/IGF2BP and Lin-28. These genes compose an oncogenic module that is coopted upon dedifferentiation of early-born intermediate progenitors to drive unlimited tumor growth. In late larvae, temporal transcription factor progression in NSCs silences the module, thereby limiting mitotic potential and terminating the window of malignant susceptibility. Thus, this study identifies the gene regulatory network that confers malignant potential to neural tumors with early developmental origins.


Subject(s)
Carcinogenesis , Cell Differentiation , Cell Proliferation , Disease Susceptibility , Drosophila/embryology , Neural Stem Cells/physiology , Animals , Drosophila Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , RNA-Binding Proteins/biosynthesis , Time Factors
19.
Development ; 142(20): 3478-87, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26487778

ABSTRACT

Stem cells switch between asymmetric and symmetric division to expand in number as tissues grow during development and in response to environmental changes. The stem cell intrinsic proteins controlling this switch are largely unknown, but one candidate is the Lin-28 pluripotency factor. A conserved RNA-binding protein that is downregulated in most animals as they develop from embryos to adults, Lin-28 persists in populations of adult stem cells. Its function in these cells has not been previously characterized. Here, we report that Lin-28 is highly enriched in adult intestinal stem cells in the Drosophila intestine. lin-28 null mutants are homozygous viable but display defects in this population of cells, which fail to undergo a characteristic food-triggered expansion in number and have reduced rates of symmetric division as well as reduced insulin signaling. Immunoprecipitation of Lin-28-bound mRNAs identified Insulin-like Receptor (InR), forced expression of which completely rescues lin-28-associated defects in intestinal stem cell number and division pattern. Furthermore, this stem cell activity of lin-28 is independent of one well-known lin-28 target, the microRNA let-7, which has limited expression in the intestinal epithelium. These results identify Lin-28 as a stem cell intrinsic factor that boosts insulin signaling in intestinal progenitor cells and promotes their symmetric division in response to nutrients, defining a mechanism through which Lin-28 controls the adult stem cell division patterns that underlie tissue homeostasis and regeneration.


Subject(s)
Drosophila Proteins/physiology , Drosophila melanogaster/growth & development , Intestinal Mucosa/cytology , Intestinal Mucosa/growth & development , RNA-Binding Proteins/physiology , Stem Cells/cytology , Animals , Cell Division , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Female , Genotype , Green Fluorescent Proteins/metabolism , Homozygote , Insulin/metabolism , MicroRNAs/metabolism , Mutation , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Receptor Protein-Tyrosine Kinases/physiology , Regeneration , Signal Transduction , Temperature
20.
Fly (Austin) ; 9(4): 173-7, 2015.
Article in English | MEDLINE | ID: mdl-26934725

ABSTRACT

In this Extra View, we extend our recent work on the protein LIN-28 and its role in adult stem cell divisions. LIN-28 is an mRNA- and microRNA-binding protein that is conserved from worms to humans. When expressed ectopically, it promotes the reprogramming of differentiated vertebrate cells into pluripotent stem cells as well as the regeneration of vertebrate tissues after injury. However, its endogenous function in stem cell populations is less clear. We recently reported that LIN-28 is specifically expressed in progenitor cells in the adult Drosophila intestine and enhances insulin signaling within this population. Loss of lin-28 alters the division patterns of these progenitor cells, limiting the growth of the intestinal epithelium that is ordinarily caused by feeding. Thus, LIN-28 is part of an uncharacterized circuit used to remodel a tissue in response to environmental cues like nutrition. Here, we extend this analysis by reporting that the levels of LIN-28 in progenitor cells are sensitive to nutrient availability. In addition, we speculate about the role of LIN-28 in the translational control of target mRNAs such as Insulin Receptor (InR) and how such translational control may be an important mechanism that underlies the stem cell dynamics needed for tissue homeostasis and growth.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/physiology , RNA-Binding Proteins/metabolism , Animals , Drosophila/cytology , Intestinal Mucosa/metabolism , Intestines/cytology , Protein Biosynthesis , Receptor, Insulin/metabolism , Starvation , Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...