Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Cell Stem Cell ; 30(1): 20-37.e9, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36493780

ABSTRACT

Variation in lung alveolar development is strongly linked to disease susceptibility. However, underlying cellular and molecular mechanisms are difficult to study in humans. We have identified an alveolar-fated epithelial progenitor in human fetal lungs, which we grow as self-organizing organoids that model key aspects of cell lineage commitment. Using this system, we have functionally validated cell-cell interactions in the developing human alveolar niche, showing that Wnt signaling from differentiating fibroblasts promotes alveolar-type-2 cell identity, whereas myofibroblasts secrete the Wnt inhibitor, NOTUM, providing spatial patterning. We identify a Wnt-NKX2.1 axis controlling alveolar differentiation. Moreover, we show that differential binding of NKX2.1 coordinates alveolar maturation, allowing us to model the effects of human genetic variation in NKX2.1 on alveolar differentiation. Our organoid system recapitulates key aspects of human fetal lung stem cell biology allowing mechanistic experiments to determine the cellular and molecular regulation of human development and disease.


Subject(s)
Cell Differentiation , Lung , Organoids , Humans , Infant, Newborn , Alveolar Epithelial Cells/metabolism , Cell Differentiation/physiology , Cell Lineage , Lung/embryology , Respiratory Tract Diseases/embryology , Respiratory Tract Diseases/metabolism
2.
STAR Protoc ; 3(4): 101788, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36345375

ABSTRACT

NanoDam is a technique for genome-wide profiling of the binding targets of any endogenously tagged chromatin-binding protein in vivo, without the need for antibodies, crosslinking, or immunoprecipitation. Here, we explain the procedure for NanoDam experiments in Drosophila, starting from a genetic cross, to the generation of sequencing libraries and, finally, bioinformatic analysis. This protocol can be readily adapted for use in other model systems after simple modifications. For complete details on the use and execution of this protocol, please refer to Tang et al. (2022).


Subject(s)
Chromatin , Drosophila , Animals , Chromatin/genetics , Chromatin Immunoprecipitation/methods , Drosophila/genetics , Carrier Proteins/genetics , High-Throughput Nucleotide Sequencing/methods
3.
Development ; 149(17)2022 09 01.
Article in English | MEDLINE | ID: mdl-36069896

ABSTRACT

In the developing nervous system, neural stem cells (NSCs) use temporal patterning to generate a wide variety of different neuronal subtypes. In Drosophila, the temporal transcription factors, Hunchback, Kruppel, Pdm and Castor, are sequentially expressed by NSCs to regulate temporal identity during neurogenesis. Here, we identify a new temporal transcription factor that regulates the transition from the Pdm to Castor temporal windows. This factor, which we call Chronophage (or 'time-eater'), is homologous to mammalian CTIP1 (Bcl11a) and CTIP2 (Bcl11b). We show that Chronophage binds upstream of the castor gene and regulates its expression. Consistent with Chronophage promoting a temporal switch, chronophage mutants generate an excess of Pdm-specified neurons and are delayed in generating neurons associated with the Castor temporal window. In addition to promoting the Pdm to Castor transition, Chronophage also represses the production of neurons generated during the earlier Hunchback and Kruppel temporal windows. Genetic interactions with Hunchback and Kruppel indicate that Chronophage regulates NSC competence to generate Hunchback- and Kruppel-specified neurons. Taken together, our results suggest that Chronophage has a conserved role in temporal patterning and neuronal subtype specification.


Subject(s)
Drosophila Proteins , Neural Stem Cells , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Mammals/metabolism , Neural Stem Cells/metabolism , Receptors, Cell Surface/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
4.
EMBO J ; 41(21): e111338, 2022 11 02.
Article in English | MEDLINE | ID: mdl-36121125

ABSTRACT

The balance between self-renewal and differentiation in human foetal lung epithelial progenitors controls the size and function of the adult organ. Moreover, progenitor cell gene regulation networks are employed by both regenerating and malignant lung cells, where modulators of their effects could potentially be of therapeutic value. Details of the molecular networks controlling human lung progenitor self-renewal remain unknown. We performed the first CRISPRi screen in primary human lung organoids to identify transcription factors controlling progenitor self-renewal. We show that SOX9 promotes proliferation of lung progenitors and inhibits precocious airway differentiation. Moreover, by identifying direct transcriptional targets using Targeted DamID, we place SOX9 at the centre of a transcriptional network, which amplifies WNT and RTK signalling to stabilise the progenitor cell state. In addition, the proof-of-principle CRISPRi screen and Targeted DamID tools establish a new workflow for using primary human organoids to elucidate detailed functional mechanisms underlying normal development and disease.


Subject(s)
Lung , SOX9 Transcription Factor , Stem Cells , Humans , Cell Differentiation/physiology , Lung/embryology , Signal Transduction , SOX9 Transcription Factor/metabolism , Stem Cells/metabolism
5.
Dev Cell ; 57(9): 1193-1207.e7, 2022 05 09.
Article in English | MEDLINE | ID: mdl-35483359

ABSTRACT

Temporal patterning of neural progenitors is an evolutionarily conserved strategy for generating neuronal diversity. Type II neural stem cells in the Drosophila central brain produce transit-amplifying intermediate neural progenitors (INPs) that exhibit temporal patterning. However, the known temporal factors cannot account for the neuronal diversity in the adult brain. To search for missing factors, we developed NanoDam, which enables rapid genome-wide profiling of endogenously tagged proteins in vivo with a single genetic cross. Mapping the targets of known temporal transcription factors with NanoDam revealed that Homeobrain and Scarecrow (ARX and NKX2.1 orthologs) are also temporal factors. We show that Homeobrain and Scarecrow define middle-aged and late INP temporal windows and play a role in cellular longevity. Strikingly, Homeobrain and Scarecrow have conserved functions as temporal factors in the developing visual system. NanoDam enables rapid cell-type-specific genome-wide profiling with temporal resolution and is easily adapted for use in higher organisms.


Subject(s)
Drosophila Proteins , Neural Stem Cells , Animals , Brain/metabolism , Cell Lineage , Drosophila/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Gene Expression Regulation, Developmental , Neural Stem Cells/metabolism
6.
Cell Rep ; 39(3): 110679, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35443165

ABSTRACT

Adult stem cells coordinate intrinsic and extrinsic, local and systemic, cues to maintain the proper balance between self-renewal and differentiation. However, the precise mechanisms stem cells use to integrate these signals remain elusive. Here, we show that Escargot (Esg), a member of the Snail family of transcription factors, regulates the maintenance of somatic cyst stem cells (CySCs) in the Drosophila testis by attenuating the activity of the pro-differentiation insulin receptor (InR) pathway. Esg positively regulates the expression of an antagonist of insulin signaling, ImpL2, while also attenuating the expression of InR. Furthermore, Esg-mediated repression of the InR pathway is required to suppress CySC loss in response to starvation. Given the conservation of Snail-family transcription factors, characterizing the mechanisms by which Esg regulates cell-fate decisions during homeostasis and a decline in nutrient availability is likely to provide insight into the metabolic regulation of stem cell behavior in other tissues and organisms.


Subject(s)
Adult Stem Cells , Drosophila Proteins , Adult Stem Cells/metabolism , Animals , Cell Differentiation , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Insulin-Like Growth Factor Binding Proteins/metabolism , Male , Receptor, Insulin/metabolism , Testis , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Nat Commun ; 13(1): 2210, 2022 04 25.
Article in English | MEDLINE | ID: mdl-35468895

ABSTRACT

The Notch signalling pathway is a master regulator of cell fate transitions in development and disease. In the brain, Notch promotes neural stem cell (NSC) proliferation, regulates neuronal migration and maturation and can act as an oncogene or tumour suppressor. How NOTCH and its transcription factor RBPJ activate distinct gene regulatory networks in closely related cell types in vivo remains to be determined. Here we use Targeted DamID (TaDa), requiring only thousands of cells, to identify NOTCH and RBPJ binding in NSCs and their progeny in the mouse embryonic cerebral cortex in vivo. We find that NOTCH and RBPJ associate with a broad network of NSC genes. Repression of NSC-specific Notch target genes in intermediate progenitors and neurons correlates with decreased chromatin accessibility, suggesting that chromatin compaction may contribute to restricting NOTCH-mediated transactivation.


Subject(s)
Chromatin , Neural Stem Cells , Animals , Cell Differentiation/physiology , Mice , Neural Stem Cells/metabolism , Receptors, Notch/metabolism , Signal Transduction
8.
iScience ; 24(11): 103234, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34746699

ABSTRACT

Genetic studies of autism have revealed causal roles for chromatin remodeling gene mutations. Chromodomain helicase DNA binding protein 8 (CHD8) encodes a chromatin remodeler with significant de novo mutation rates in sporadic autism. However, relationships between CHD8 genomic function and autism-relevant biology remain poorly elucidated. Published studies utilizing ChIP-seq to map CHD8 protein-DNA interactions have high variability, consistent with technical challenges and limitations associated with this method. Thus, complementary approaches are needed to establish CHD8 genomic targets and regulatory functions in developing brain. We used in utero CHD8 Targeted DamID followed by sequencing (TaDa-seq) to characterize CHD8 binding in embryonic mouse cortex. CHD8 TaDa-seq reproduced interaction patterns observed from ChIP-seq and further highlighted CHD8 distal interactions associated with neuronal loci. This study establishes TaDa-seq as a useful alternative for mapping protein-DNA interactions in vivo and provides insights into the regulatory targets of CHD8 and autism-relevant pathophysiology associated with CHD8 mutations.

9.
PLoS Biol ; 19(11): e3001255, 2021 11.
Article in English | MEDLINE | ID: mdl-34748544

ABSTRACT

The discovery of human obesity-associated genes can reveal new mechanisms to target for weight loss therapy. Genetic studies of obese individuals and the analysis of rare genetic variants can identify novel obesity-associated genes. However, establishing a functional relationship between these candidate genes and adiposity remains a significant challenge. We uncovered a large number of rare homozygous gene variants by exome sequencing of severely obese children, including those from consanguineous families. By assessing the function of these genes in vivo in Drosophila, we identified 4 genes, not previously linked to human obesity, that regulate adiposity (itpr, dachsous, calpA, and sdk). Dachsous is a transmembrane protein upstream of the Hippo signalling pathway. We found that 3 further members of the Hippo pathway, fat, four-jointed, and hippo, also regulate adiposity and that they act in neurons, rather than in adipose tissue (fat body). Screening Hippo pathway genes in larger human cohorts revealed rare variants in TAOK2 associated with human obesity. Knockdown of Drosophila tao increased adiposity in vivo demonstrating the strength of our approach in predicting novel human obesity genes and signalling pathways and their site of action.


Subject(s)
Drosophila melanogaster/genetics , Genetic Association Studies , Genetic Testing , Obesity/genetics , Age of Onset , Animals , Case-Control Studies , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Female , Homozygote , Humans , Male , Mutation/genetics , Pedigree , Signal Transduction/genetics
10.
J Neurosci ; 41(30): 6430-6448, 2021 07 28.
Article in English | MEDLINE | ID: mdl-34210781

ABSTRACT

The adaptable transcriptional response to changes in food availability not only ensures animal survival but also lets embryonic development progress. Interestingly, the CNS is preferentially protected from periods of malnutrition, a phenomenon known as "brain sparing." However, the mechanisms that mediate this response remain poorly understood. To get a better understanding of this, we used Drosophila melanogaster as a model, analyzing the transcriptional response of neural stem cells (neuroblasts) and glia of the blood-brain barrier (BBB) from larvae of both sexes during nutrient restriction using targeted DamID. We found differentially expressed genes in both neuroblasts and glia of the BBB, although the effect of nutrient deficiency was primarily observed in the BBB. We characterized the function of a nutritional sensitive gene expressed in the BBB, the serine protease homolog, scarface (scaf). Scaf is expressed in subperineurial glia in the BBB in response to nutrition. Tissue-specific knockdown of scaf increases subperineurial glia endoreplication and proliferation of perineurial glia in the blood-brain barrier. Furthermore, neuroblast proliferation is diminished on scaf knockdown in subperineurial glia. Interestingly, reexpression of Scaf in subperineurial glia is able to enhance neuroblast proliferation and brain growth of animals in starvation. Finally, we show that loss of scaf in the blood-brain barrier increases sensitivity to drugs in adulthood, suggesting a physiological impairment. We propose that Scaf integrates the nutrient status to modulate the balance between neurogenesis and growth of the BBB, preserving the proper equilibrium between the size of the barrier and the brain.SIGNIFICANCE STATEMENT The Drosophila BBB separates the CNS from the open circulatory system. The BBB glia are not only acting as a physical segregation of tissues but participate in the regulation of the metabolism and neurogenesis during development. Here we analyze the transcriptional response of the BBB glia to nutrient deprivation during larval development, a condition in which protective mechanisms are switched on in the brain. Our findings show that the gene scarface reduces growth in the BBB while promoting the proliferation of neural stem, assuring the balanced growth of the larval brain. Thus, Scarface would link animal nutrition with brain development, coordinating neurogenesis with the growth of the BBB.


Subject(s)
Blood-Brain Barrier/metabolism , Drosophila Proteins/metabolism , Neural Stem Cells/metabolism , Neurogenesis/physiology , Neuroglia/metabolism , Serine Proteases/metabolism , Animals , Blood-Brain Barrier/growth & development , Drosophila melanogaster , Female , Male , Malnutrition
11.
Curr Biol ; 31(8): 1744-1753.e5, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33621481

ABSTRACT

Stem cells reside in specialized microenvironments or niches that balance stem cell proliferation and differentiation.1,2 The extracellular matrix (ECM) is an essential component of most niches, because it controls niche homeostasis, provides physical support, and conveys extracellular signals.3-11 Basement membranes (BMs) are thin ECM sheets that are constituted mainly by Laminins, Perlecan, Collagen IV, and Entactin/Nidogen and surround epithelia and other tissues.12 Perlecans are secreted proteoglycans that interact with ECM proteins, ligands, receptors, and growth factors such as FGF, PDGF, VEGF, Hedgehog, and Wingless.13-18 Thus, Perlecans have structural and signaling functions through the binding, storage, or sequestering of specific ligands. We have used the Drosophila ovary to assess the importance of Perlecan in the functioning of a stem cell niche. Ovarioles in the adult ovary are enveloped by an ECM sheath and possess a tapered structure at their anterior apex termed the germarium. The anterior tip of the germarium hosts the germline niche, where two to four germline stem cells (GSCs) reside together with a few somatic cells: terminal filament cells (TFCs), cap cells (CpCs), and escort cells (ECs).19 We report that niche architecture in the developing gonad requires trol, that niche cells secrete an isoform-specific Perlecan-rich interstitial matrix, and that DE-cadherin-dependent stem cell-niche adhesion necessitates trol. Hence, we provide evidence to support a structural role for Perlecan in germline niche establishment during larval stages and in the maintenance of a normal pool of stem cells in the adult niche.


Subject(s)
Drosophila , Stem Cell Niche , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Female , Heparan Sulfate Proteoglycans , Ligands , Ovary
12.
Methods Mol Biol ; 2161: 255-264, 2020.
Article in English | MEDLINE | ID: mdl-32681518

ABSTRACT

Long-noncoding RNAs (lncRNAs) are emerging as regulators of development and disease. lncRNAs are expressed in exquisitely precise expression patterns in vivo and many interact with chromatin to regulate gene expression. However, the limited sensitivity of RNA-purification techniques has precluded the identification of genomic targets of cell-type specific lncRNAs. RNA-DamID is a powerful new approach to understand the mechanisms by which lncRNAs act in vivo. RNA-DamID is highly sensitive and accurate, and can resolve cell-type-specific chromatin binding patterns without cell isolation. The determinants of RNA-chromatin interactions can be identified with RNA-DamID by analyzing RNA and protein cofactor mutants. Here we describe how to implement RNA-DamID and the design considerations to take into account to accurately identify lncRNA-chromatin interactions in vivo.


Subject(s)
Chromatin/metabolism , RNA, Long Noncoding/chemistry , Sequence Analysis/methods , Animals , Cell Line , Cells, Cultured , Chromatin/chemistry , DNA Methylation , Humans , Protein Binding , Protein Interaction Mapping , RNA, Long Noncoding/metabolism
13.
Trends Neurosci ; 43(4): 213-226, 2020 04.
Article in English | MEDLINE | ID: mdl-32209453

ABSTRACT

Neural stem cells (NSCs) are multipotent progenitors that are responsible for producing all of the neurons and macroglia in the nervous system. In adult mammals, NSCs reside predominantly in a mitotically dormant, quiescent state, but they can proliferate in response to environmental inputs such as feeding or exercise. It is hoped that quiescent NSCs could be activated therapeutically to contribute towards repair in humans. This will require an understanding of quiescent NSC heterogeneities and regulation during normal physiology and following brain injury. Non-mammalian vertebrates (zebrafish and salamanders) and invertebrates (Drosophila) offer insights into brain repair and quiescence regulation that are difficult to obtain using rodent models alone. We review conceptual progress from these various models, a first step towards harnessing quiescent NSCs for therapeutic purposes.


Subject(s)
Brain , Cell- and Tissue-Based Therapy , Neural Stem Cells , Animals , Cell- and Tissue-Based Therapy/trends , Models, Biological , Neural Stem Cells/physiology
14.
Elife ; 92020 02 19.
Article in English | MEDLINE | ID: mdl-32073402

ABSTRACT

Understanding the sequence of events leading to cancer relies in large part upon identifying the tumour cell of origin. Glioblastoma is the most malignant brain cancer but the early stages of disease progression remain elusive. Neural lineages have been implicated as cells of origin, as have glia. Interestingly, high levels of the neural stem cell regulator TLX correlate with poor patient prognosis. Here we show that high levels of the Drosophila TLX homologue, Tailless, initiate tumourigenesis by reverting intermediate neural progenitors to a stem cell state. Strikingly, we could block tumour formation completely by re-expressing Asense (homologue of human ASCL1), which we show is a direct target of Tailless. Our results predict that expression of TLX and ASCL1 should be mutually exclusive in glioblastoma, which was verified in single-cell RNA-seq of human glioblastoma samples. Counteracting high TLX is a potential therapeutic strategy for suppressing tumours originating from intermediate progenitor cells.


Subject(s)
Carcinogenesis/metabolism , Drosophila Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Repressor Proteins/metabolism , Stem Cells/metabolism , Animals , Cell Differentiation , Cell Lineage , Cell Transformation, Neoplastic/metabolism , Drosophila melanogaster/metabolism , Female , Glioblastoma/metabolism , Humans , Male
15.
Nat Cell Biol ; 21(11): 1321-1333, 2019 11.
Article in English | MEDLINE | ID: mdl-31685987

ABSTRACT

Following severe or chronic liver injury, adult ductal cells (cholangiocytes) contribute to regeneration by restoring both hepatocytes and cholangiocytes. We recently showed that ductal cells clonally expand as self-renewing liver organoids that retain their differentiation capacity into both hepatocytes and ductal cells. However, the molecular mechanisms by which adult ductal-committed cells acquire cellular plasticity, initiate organoids and regenerate the damaged tissue remain largely unknown. Here, we describe that ductal cells undergo a transient, genome-wide, remodelling of their transcriptome and epigenome during organoid initiation and in vivo following tissue damage. TET1-mediated hydroxymethylation licences differentiated ductal cells to initiate organoids and activate the regenerative programme through the transcriptional regulation of stem-cell genes and regenerative pathways including the YAP-Hippo signalling. Our results argue in favour of the remodelling of genomic methylome/hydroxymethylome landscapes as a general mechanism by which differentiated cells exit a committed state in response to tissue damage.


Subject(s)
DNA-Binding Proteins/genetics , Epigenesis, Genetic , Epigenome , Liver Regeneration/genetics , Liver/metabolism , Organoids/metabolism , Proto-Oncogene Proteins/genetics , Transcriptome , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Bile Ducts/cytology , Bile Ducts/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA Methylation , DNA-Binding Proteins/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Gene Expression Profiling , Hippo Signaling Pathway , Liver/cytology , Male , Mice, Transgenic , Organoids/cytology , Primary Cell Culture , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , YAP-Signaling Proteins
16.
Elife ; 82019 09 12.
Article in English | MEDLINE | ID: mdl-31513013

ABSTRACT

Translating advances in cancer research to clinical applications requires better insight into the metabolism of normal cells and tumour cells in vivo. Much effort has focused on understanding how glycolysis and oxidative phosphorylation (OxPhos) support proliferation, while their impact on other aspects of development and tumourigenesis remain largely unexplored. We found that inhibition of OxPhos in neural stem cells (NSCs) or tumours in the Drosophila brain not only decreases proliferation, but also affects many different aspects of stem cell behaviour. In NSCs, OxPhos dysfunction leads to a protracted G1/S-phase and results in delayed temporal patterning and reduced neuronal diversity. As a consequence, NSCs fail to undergo terminal differentiation, leading to prolonged neurogenesis into adulthood. Similarly, in brain tumours inhibition of OxPhos slows proliferation and prevents differentiation, resulting in reduced tumour heterogeneity. Thus, in vivo, highly proliferative stem cells and tumour cells require OxPhos for efficient growth and generation of diversity.


Subject(s)
Brain Neoplasms/pathology , Cell Proliferation , Neural Stem Cells/pathology , Oxidative Phosphorylation , Animals , Cell Differentiation , Disease Models, Animal , Drosophila , Glycolysis
17.
Curr Opin Cell Biol ; 60: 131-138, 2019 10.
Article in English | MEDLINE | ID: mdl-31330360

ABSTRACT

Determining the premalignant lesions that develop into malignant tumours remains a daunting task. Brain tumours are frequently characterised by a block in differentiation, implying that normal developmental pathways become hijacked during tumourigenesis. However, the heterogeneity of stem cells and their progenitors in the brain suggests there are many potential routes to tumour initiation. Studies in Drosophila melanogaster have enhanced our understanding of the tumourigenic potential of distinct cell types in the brain. Here we review recent studies that have improved our knowledge of neural stem cell behaviour during development and in brain tumour models.


Subject(s)
Brain Neoplasms/pathology , Neural Stem Cells/metabolism , Animals , Cell Lineage , Drosophila melanogaster/cytology , Humans , Models, Biological , Neural Stem Cells/cytology , Time Factors
18.
Dev Cell ; 49(4): 556-573.e6, 2019 05 20.
Article in English | MEDLINE | ID: mdl-31112698

ABSTRACT

Chromatin remodeling accompanies differentiation, however, its role in self-renewal is less well understood. We report that in Drosophila, the chromatin remodeler Kismet/CHD7/CHD8 limits intestinal stem cell (ISC) number and proliferation without affecting differentiation. Stem-cell-specific whole-genome profiling of Kismet revealed its enrichment at transcriptionally active regions bound by RNA polymerase II and Brahma, its recruitment to the transcription start site of activated genes and developmental enhancers and its depletion from regions bound by Polycomb, Histone H1, and heterochromatin Protein 1. We demonstrate that the Trithorax-related/MLL3/4 chromatin modifier regulates ISC proliferation, colocalizes extensively with Kismet throughout the ISC genome, and co-regulates genes in ISCs, including Cbl, a negative regulator of Epidermal Growth Factor Receptor (EGFR). Loss of kismet or trr leads to elevated levels of EGFR protein and signaling, thereby promoting ISC self-renewal. We propose that Kismet with Trr establishes a chromatin state that limits EGFR proliferative signaling, preventing tumor-like stem cell overgrowths.


Subject(s)
Chromatin/metabolism , DNA Helicases/metabolism , Drosophila Proteins/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Homeodomain Proteins/metabolism , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Chromatin Assembly and Disassembly/physiology , DNA Helicases/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/metabolism , ErbB Receptors/metabolism , Histone-Lysine N-Methyltransferase/physiology , Histones/metabolism , Homeodomain Proteins/physiology , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Receptors, Invertebrate Peptide/metabolism , Signal Transduction/physiology , Stem Cells/metabolism , Transcription Factors/metabolism
19.
Curr Opin Neurobiol ; 56: 160-166, 2019 06.
Article in English | MEDLINE | ID: mdl-30844670

ABSTRACT

The emergence of neuronal diversity during development of the nervous system relies on dynamic changes in the epigenetic landscape of neural stem cells and their progeny. Targeted DamID (TaDa) is proving invaluable in identifying the genome-wide binding sites of chromatin-associated proteins in vivo, without fixation, cell isolation, or immunoprecipitation. The simplicity and efficiency of the technique have led to an ever-expanding TaDa toolbox. These tools enable profiling of gene expression and chromatin accessibility, as well as the identification of the genome-wide binding sites of chromatin complexes, transcription factors and RNAs. Here, we review these new developments, with particular emphasis on the use of TaDa in studying neuronal specification.


Subject(s)
Chromatin , Binding Sites , DNA Methylation , Genome , Transcription Factors
20.
Dev Cell ; 49(2): 293-300.e3, 2019 04 22.
Article in English | MEDLINE | ID: mdl-30905769

ABSTRACT

Quiescent neural stem cells (NSCs) in the adult brain are regenerative cells that could be activated therapeutically to repair damage. It is becoming apparent that quiescent NSCs exhibit heterogeneity in their propensity for activation and in the progeny that they generate. We discovered recently that NSCs undergo quiescence in either G0 or G2 in the Drosophila brain, challenging the notion that all quiescent stem cells are G0 arrested. We found that G2-quiescent NSCs become activated prior to G0 NSCs. Here, we show that the cyclin-dependent kinase inhibitor Dacapo (Dap; ortholog of p57KIP2) determines whether NSCs enter G0 or G2 quiescence during embryogenesis. We demonstrate that the dorsal patterning factor, Muscle segment homeobox (Msh; ortholog of MSX1/2/3) binds directly to the Dap locus and induces Dap expression in dorsal NSCs, resulting in G0 arrest, while more ventral NSCs undergo G2 quiescence. Our results reveal region-specific regulation of stem cell quiescence.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p57/metabolism , Drosophila Proteins/metabolism , Neural Stem Cells/metabolism , Nuclear Proteins/metabolism , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Cell Cycle/physiology , Cell Division/physiology , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , G2 Phase/physiology , Homeodomain Proteins/metabolism , Neural Stem Cells/cytology , Neurogenesis/physiology , Resting Phase, Cell Cycle/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...