Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 21(4): 568-581, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35149549

ABSTRACT

Clinical use of doxorubicin (Dox) is limited by cumulative myelo- and cardiotoxicity. This research focuses on the detailed characterization of PhAc-ALGP-Dox, a targeted tetrapeptide prodrug with a unique dual-step activation mechanism, designed to circumvent Dox-related toxicities and is ready for upcoming clinical investigation. Coupling Dox to a phosphonoacetyl (PhAc)-capped tetrapeptide forms the cell-impermeable, inactive compound, PhAc-ALGP-Dox. After extracellular cleavage by tumor-enriched thimet oligopeptidase-1 (THOP1), a cell-permeable but still biologically inactive dipeptide-conjugate is formed (GP-Dox), which is further processed intracellularly to Dox by fibroblast activation protein-alpha (FAPα) and/or dipeptidyl peptidase-4 (DPP4). In vitro, PhAc-ALGP-Dox is effective in various 2D- and 3D-cancer models, while showing improved safety toward normal epithelium, hematopoietic progenitors, and cardiomyocytes. In vivo, these results translate into a 10-fold higher tolerability and 5-fold greater retention of Dox in the tumor microenvironment compared with the parental drug. PhAc-ALGP-Dox demonstrates 63% to 96% tumor growth inhibition in preclinical models, an 8-fold improvement in efficacy in patient-derived xenograft (PDX) models, and reduced metastatic burden in a murine model of experimental lung metastasis, improving survival by 30%. The current findings highlight the potential clinical benefit of PhAc-ALGP-Dox, a targeted drug-conjugate with broad applicability, favorable tissue biodistribution, significantly improved tolerability, and tumor growth inhibition at primary and metastatic sites in numerous solid tumor models.


Subject(s)
Antineoplastic Agents , Lung Neoplasms , Prodrugs , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Doxorubicin/chemistry , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Humans , Lung Neoplasms/drug therapy , Mice , Prodrugs/pharmacology , Prodrugs/therapeutic use , Therapeutic Index , Tissue Distribution , Tumor Microenvironment
2.
Genome Biol ; 22(1): 302, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34724962

ABSTRACT

BACKGROUND: Precise gene dosage of the X chromosomes is critical for normal development and cellular function. In mice, XX female somatic cells show transcriptional X chromosome upregulation of their single active X chromosome, while the other X chromosome is inactive. Moreover, the inactive X chromosome is reactivated during development in the inner cell mass and in germ cells through X chromosome reactivation, which can be studied in vitro by reprogramming of somatic cells to pluripotency. How chromatin processes and gene regulatory networks evolved to regulate X chromosome dosage in the somatic state and during X chromosome reactivation remains unclear. RESULTS: Using genome-wide approaches, allele-specific ATAC-seq and single-cell RNA-seq, in female embryonic fibroblasts and during reprogramming to pluripotency, we show that chromatin accessibility on the upregulated mammalian active X chromosome is increased compared to autosomes. We further show that increased accessibility on the active X chromosome is erased by reprogramming, accompanied by erasure of transcriptional X chromosome upregulation and the loss of increased transcriptional burst frequency. In addition, we characterize gene regulatory networks during reprogramming and X chromosome reactivation, revealing changes in regulatory states. Our data show that ZFP42/REX1, a pluripotency-associated gene that evolved specifically in placental mammals, targets multiple X-linked genes, suggesting an evolutionary link between ZFP42/REX1, X chromosome reactivation, and pluripotency. CONCLUSIONS: Our data reveal the existence of intrinsic compensatory mechanisms that involve modulation of chromatin accessibility to counteract X-to-Autosome gene dosage imbalances caused by evolutionary or in vitro X chromosome loss and X chromosome inactivation in mammalian cells.


Subject(s)
Chromatin/metabolism , X Chromosome Inactivation , Alleles , Aneuploidy , Animals , Cellular Reprogramming/genetics , Gene Regulatory Networks , Induced Pluripotent Stem Cells/metabolism , Mice , RNA-Seq , Single-Cell Analysis , Transcription Factors/metabolism , Transcription, Genetic , X Chromosome
3.
Nat Cell Biol ; 23(1): 49-60, 2021 01.
Article in English | MEDLINE | ID: mdl-33420491

ABSTRACT

Totipotency is the ability of a single cell to give rise to all of the differentiated cell types that build the conceptus, yet how to capture this property in vitro remains incompletely understood. Defining totipotency relies on a variety of assays of variable stringency. Here, we describe criteria to define totipotency. We explain how distinct criteria of increasing stringency can be used to judge totipotency by evaluating candidate totipotent cell types in mice, including early blastomeres and expanded or extended pluripotent stem cells. Our data challenge the notion that expanded or extended pluripotent states harbour increased totipotent potential relative to conventional embryonic stem cells under in vitro and in vivo conditions.


Subject(s)
Blastomeres/cytology , Cell Differentiation , Cell Lineage/genetics , Embryo, Mammalian/cytology , Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Totipotent Stem Cells/cytology , Animals , Blastomeres/metabolism , Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Female , Gene Expression Profiling , Gene Regulatory Networks , Male , Mice , Pluripotent Stem Cells/metabolism , Single-Cell Analysis , Totipotent Stem Cells/metabolism
4.
Genome Res ; 29(10): 1659-1672, 2019 10.
Article in English | MEDLINE | ID: mdl-31515287

ABSTRACT

Induction and reversal of chromatin silencing is critical for successful development, tissue homeostasis, and the derivation of induced pluripotent stem cells (iPSCs). X-Chromosome inactivation (XCI) and reactivation (XCR) in female cells represent chromosome-wide transitions between active and inactive chromatin states. Although XCI has long been studied, providing important insights into gene regulation, the dynamics and mechanisms underlying the reversal of stable chromatin silencing of X-linked genes are much less understood. Here, we use allele-specific transcriptomics to study XCR during mouse iPSC reprogramming in order to elucidate the timing and mechanisms of chromosome-wide reversal of gene silencing. We show that XCR is hierarchical, with subsets of genes reactivating early, late, and very late during reprogramming. Early genes are activated before the onset of late pluripotency genes activation. Early genes are located genomically closer to genes that escape XCI, unlike genes reactivating late. Early genes also show increased pluripotency transcription factor (TF) binding. We also reveal that histone deacetylases (HDACs) restrict XCR in reprogramming intermediates and that the severe hypoacetylation state of the inactive X Chromosome (Xi) persists until late reprogramming stages. Altogether, these results reveal the timing of transcriptional activation of monoallelically repressed genes during iPSC reprogramming, and suggest that allelic activation involves the combined action of chromatin topology, pluripotency TFs, and chromatin regulators. These findings are important for our understanding of gene silencing, maintenance of cell identity, reprogramming, and disease.


Subject(s)
Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/cytology , RNA, Long Noncoding/genetics , X Chromosome Inactivation/genetics , Animals , Chromatin/genetics , Female , Gene Silencing , Genes, X-Linked/genetics , Histone Deacetylases/genetics , Mice , Transcriptional Activation/genetics , X Chromosome/genetics
5.
J Cell Sci ; 132(20)2019 10 22.
Article in English | MEDLINE | ID: mdl-31519808

ABSTRACT

Reprogramming to induced pluripotency induces the switch of somatic cell identity to induced pluripotent stem cells (iPSCs). However, the mediators and mechanisms of reprogramming remain largely unclear. To elucidate the mediators and mechanisms of reprogramming, we used a siRNA-mediated knockdown approach for selected candidate genes during the conversion of somatic cells into iPSCs. We identified Tox4 as a novel factor that modulates cell fate through an assay that determined the efficiency of iPSC reprogramming. We found that Tox4 is needed early in reprogramming to efficiently generate early reprogramming intermediates, irrespective of the reprogramming conditions used. Tox4 enables proper exogenous reprogramming factor expression, and the closing and opening of putative somatic and pluripotency enhancers early during reprogramming, respectively. We show that the TOX4 protein assembles into a high molecular form. Moreover, Tox4 is also required for the efficient conversion of fibroblasts towards the neuronal fate, suggesting a broader role of Tox4 in modulating cell fate. Our study reveals Tox4 as a novel transcriptional modulator of cell fate that mediates reprogramming from the somatic state to the pluripotent and neuronal fate.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Cellular Reprogramming , Fibroblasts/metabolism , High Mobility Group Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/metabolism , Animals , Cell Line , Fibroblasts/cytology , High Mobility Group Proteins/genetics , Induced Pluripotent Stem Cells/cytology , Mice , Neural Stem Cells/cytology
6.
Stem Cell Reports ; 12(2): 333-350, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30639215

ABSTRACT

Reprogramming female mouse somatic cells into induced pluripotent stem cells (iPSCs) leads to X-chromosome reactivation. The extent to which increased X-chromosome dosage (X-dosage) in female iPSCs compared with male iPSCs leads to differences in the properties of iPSCs is still unclear. We show that chromatin accessibility in mouse iPSCs is modulated by X-dosage. Specific sets of transcriptional regulator motifs are enriched in chromatin with increased accessibility in XX or XY iPSCs. The transcriptome, growth and pluripotency exit are also modulated by X-dosage in iPSCs. To understand how increased X-dosage modulates the properties of mouse pluripotent stem cells, we used heterozygous deletions of the X-linked gene Dusp9. We show that X-dosage regulates the transcriptome, open chromatin landscape, growth, and pluripotency exit largely independently of global DNA methylation. Our results provide insights into how gene dosage modulates the epigenetic and genetic mechanisms that regulate cell identity.


Subject(s)
DNA Methylation/genetics , Gene Dosage/genetics , Pluripotent Stem Cells/physiology , X Chromosome/genetics , Animals , Cell Line , Cellular Reprogramming/genetics , Chromatin/genetics , Dual-Specificity Phosphatases/genetics , Epigenesis, Genetic/genetics , Epigenomics/methods , Female , Induced Pluripotent Stem Cells/physiology , Male , Mice , Mitogen-Activated Protein Kinase Phosphatases/genetics , Transcriptome/genetics
7.
Epigenet Insights ; 11: 2516865718802931, 2018.
Article in English | MEDLINE | ID: mdl-30443643

ABSTRACT

How the epigenome of one cell type is remodeled during reprogramming into another unrelated type of cell remains unclear. Overexpression of transcription factors in somatic cells enables the induction of induced pluripotent stem cells (iPSCs). This process entails genome-wide remodeling of DNA methylation, chromatin, and transcription. Recent work suggests that the number of active X chromosomes present in a cell influences remodeling of DNA methylation during somatic cell reprogramming to mouse iPSCs. Female iPSCs with 2 active X chromosomes display global DNA hypomethylation, whereas male XY iPSCs show DNA methylation levels similar to the somatic cells they are derived from. Global DNA methylation erasure in female iPSCs takes place genome-wide and involves repression of DNA methyltransferases. However, on loss of one X chromosome, female iPSCs acquire a DNA methylation landscape resembling that of XY iPSCs. Therefore, it is the X chromosome dosage that dictates global DNA methylation levels in iPSCs. Here, we discuss the evidence that links X chromosome dosage with the regulation of DNA methylation in pluripotent stem cells. We focus on iPSCs reprogramming studies, where X chromosome status is a novel factor impacting our understanding of epigenetic remodeling.

8.
Dev Cell ; 45(1): 53-66.e5, 2018 04 09.
Article in English | MEDLINE | ID: mdl-29576424

ABSTRACT

The importance of producing the correct numbers of neurons during development is illustrated by both evolutionary enhancement of cognitive capacities in larger brains, and developmental disorders of brain size. In humans, increased neuronal numbers during development is speculated to partly derive from a unique subtype of neural stem cells (NSCs) that undergo a phase of expansion through symmetric self-amplifying divisions before generating neurons. Symmetric amplification also appears to underlie adult neural stem maintenance in the mouse. However, the mechanisms regulating this behavior are unclear. We report the discovery of self-amplifying NSCs in Drosophila and show that they arise by a spatiotemporal conversion of classical self-renewing NSCs. This conversion is regulated by a temporal transition in the expression of proneural transcription factors prior to cell division. We find a causal link between stem cell self-amplification and increased neuronal numbers. We further show that the temporal transcriptional switch controls both stem cell division and subsequent neuronal differentiation.


Subject(s)
Cell Differentiation , Cell Proliferation , Drosophila melanogaster/growth & development , Neural Stem Cells/cytology , Neurogenesis/physiology , Neurons/cytology , Animals , Cell Count , Cell Self Renewal , Cells, Cultured , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Female , Male , Neural Stem Cells/metabolism , Neurons/metabolism , Signal Transduction , Transcription, Genetic
9.
Front Cell Neurosci ; 11: 416, 2017.
Article in English | MEDLINE | ID: mdl-29472843

ABSTRACT

Injury to the adult central nervous systems (CNS) can result in severe long-term disability because damaged CNS connections fail to regenerate after trauma. Identification of regulators that enhance the intrinsic growth capacity of severed axons is a first step to restore function. Here, we conducted a gain-of-function genetic screen in Drosophila to identify strong inducers of axonal growth after injury. We focus on a novel axis the Down Syndrome Cell Adhesion Molecule (Dscam1), the de-ubiquitinating enzyme Fat Facets (Faf)/Usp9x and the Jun N-Terminal Kinase (JNK) pathway transcription factor Kayak (Kay)/Fos. Genetic and biochemical analyses link these genes in a common signaling pathway whereby Faf stabilizes Dscam1 protein levels, by acting on the 3'-UTR of its mRNA, and Dscam1 acts upstream of the growth-promoting JNK signal. The mammalian homolog of Faf, Usp9x/FAM, shares both the regenerative and Dscam1 stabilizing activities, suggesting a conserved mechanism.

10.
Dev Cell ; 39(2): 267-278, 2016 10 24.
Article in English | MEDLINE | ID: mdl-27780041

ABSTRACT

The axonal wiring molecule Slit and its Round-About (Robo) receptors are conserved regulators of nerve cord patterning. Robo receptors also contribute to wiring brain circuits. Whether molecular mechanisms regulating these signals are modified to fit more complex brain wiring processes is unclear. We investigated the role of Slit and Robo receptors in wiring Drosophila higher-order brain circuits and identified differences in the cellular and molecular mechanisms of Robo/Slit function. First, we find that signaling by Robo receptors in the brain is regulated by the Receptor Protein Tyrosine Phosphatase RPTP69d. RPTP69d increases membrane availability of Robo3 without affecting its phosphorylation state. Second, we detect no midline localization of Slit during brain development. Instead, Slit is enriched in the mushroom body, a neuronal structure covering large areas of the brain. Thus, a divergent molecular mechanism regulates neuronal circuit wiring in the Drosophila brain, partly in response to signals from the mushroom body.


Subject(s)
Brain/metabolism , Drosophila Proteins/metabolism , Nerve Net/metabolism , Nerve Tissue Proteins/metabolism , Neuropil/metabolism , Receptor-Like Protein Tyrosine Phosphatases/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Animals , Axons/metabolism , Cell Membrane/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Epistasis, Genetic , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Larva/metabolism , Multiprotein Complexes/metabolism , Mushroom Bodies/metabolism , Nerve Tissue Proteins/genetics , Phenotype
11.
Development ; 143(15): 2760-6, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27385016

ABSTRACT

The neurogenin (Ngn) transcription factors control early neurogenesis and neurite outgrowth in mammalian cortex. In contrast to their proneural activity, their function in neurite growth is poorly understood. Drosophila has a single predicted Ngn homolog, Tap, of unknown function. Here we show that Tap is not a proneural protein in Drosophila but is required for proper axonal growth and guidance of neurons of the mushroom body, a neuropile required for associative learning and memory. Genetic and expression analyses suggest that Tap inhibits excessive axonal growth by fine regulation of the levels of the Wnt signaling adaptor protein Dishevelled.


Subject(s)
Cell Polarity/physiology , Drosophila Proteins/metabolism , Neuropeptides/metabolism , Transcription Factors/metabolism , Wnt Signaling Pathway/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Axon Guidance/genetics , Axon Guidance/physiology , Axons/metabolism , Cell Polarity/genetics , Drosophila , Drosophila Proteins/genetics , Mushroom Bodies/metabolism , Neuropeptides/genetics , Protein Binding , Transcription Factors/genetics , Wnt Signaling Pathway/genetics
12.
Cell ; 164(3): 460-75, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26824657

ABSTRACT

Neurogenesis is initiated by the transient expression of the highly conserved proneural proteins, bHLH transcriptional regulators. Here, we discover a conserved post-translational switch governing the duration of proneural protein activity that is required for proper neuronal development. Phosphorylation of a single Serine at the same position in Scute and Atonal proneural proteins governs the transition from active to inactive forms by regulating DNA binding. The equivalent Neurogenin2 Threonine also regulates DNA binding and proneural activity in the developing mammalian neocortex. Using genome editing in Drosophila, we show that Atonal outlives its mRNA but is inactivated by phosphorylation. Inhibiting the phosphorylation of the conserved proneural Serine causes quantitative changes in expression dynamics and target gene expression resulting in neuronal number and fate defects. Strikingly, even a subtle change from Serine to Threonine appears to shift the duration of Atonal activity in vivo, resulting in neuronal fate defects.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , Neurogenesis , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Drosophila , Drosophila Proteins , Eye/growth & development , Eye/ultrastructure , Imaginal Discs/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , Phosphorylation , Retina/growth & development , Sequence Alignment
13.
Elife ; 3: e01699, 2014 Apr 22.
Article in English | MEDLINE | ID: mdl-24755286

ABSTRACT

Axonal branching allows a neuron to connect to several targets, increasing neuronal circuit complexity. While axonal branching is well described, the mechanisms that control it remain largely unknown. We find that in the Drosophila CNS branches develop through a process of excessive growth followed by pruning. In vivo high-resolution live imaging of developing brains as well as loss and gain of function experiments show that activation of Epidermal Growth Factor Receptor (EGFR) is necessary for branch dynamics and the final branching pattern. Live imaging also reveals that intrinsic asymmetry in EGFR localization regulates the balance between dynamic and static filopodia. Elimination of signaling asymmetry by either loss or gain of EGFR function results in reduced dynamics leading to excessive branch formation. In summary, we propose that the dynamic process of axon branch development is mediated by differential local distribution of signaling receptors. DOI: http://dx.doi.org/10.7554/eLife.01699.001.


Subject(s)
Axons/physiology , Neuronal Plasticity , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Animals , Drosophila , Drosophila Proteins/metabolism , ErbB Receptors/metabolism , Optical Imaging , Receptors, Invertebrate Peptide/metabolism
14.
Neural Dev ; 9: 4, 2014 Feb 26.
Article in English | MEDLINE | ID: mdl-24571981

ABSTRACT

BACKGROUND: Vertebrates and invertebrates obtain visual motion information by channeling moving visual cues perceived by the retina through specific motion sensitive synaptic relays in the brain. In Drosophila, the series of synaptic relays forming the optic lobe are known as the lamina, medulla, lobula and lobula plate neuropiles. The fly's motion detection output neurons, called the T4 and T5 cells, reside in the lobula plate. Adult optic lobe neurons are derived from larval neural progenitors in two proliferating compartments known as the outer and inner proliferation centers (OPC and IPC). Important insight has been gained into molecular mechanisms involved in the development of the lamina and medulla from the OPC, though less is known about the development of the lobula and lobula plate. RESULTS: Here we show that the proneural gene Atonal is expressed in a subset of IPC progenitors that give rise to the higher order motion detection neurons, T4 and T5, of the lobula plate. We also show that Atonal does not act as a proneural gene in this context. Rather, it is required specifically in IPC neural progenitors to regulate neurite outgrowth in the neuronal progeny. CONCLUSIONS: Our findings reveal that a proneural gene is expressed in progenitors but is required for neurite development of their progeny neurons. This suggests that transcriptional programs initiated specifically in progenitors are necessary for subsequent neuronal morphogenesis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Nerve Tissue Proteins/genetics , Neural Stem Cells/metabolism , Neurites/metabolism , Neurons/metabolism , Optic Lobe, Nonmammalian/metabolism , Animals , Drosophila Proteins , Drosophila melanogaster , Motion Perception/physiology
15.
Elife ; 2: e00337, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23471010

ABSTRACT

Brain connectivity maps display a delicate balance between individual variation and stereotypy, suggesting the existence of dedicated mechanisms that simultaneously permit and limit individual variation. We show that during the development of the Drosophila central nervous system, mutual inhibition among groups of neighboring postmitotic neurons during development regulates the robustness of axon target choice in a nondeterministic neuronal circuit. Specifically, neighboring postmitotic neurons communicate through Notch signaling during axonal targeting, to ensure balanced alternative axon target choices without a corresponding change in cell fate. Loss of Notch in postmitotic neurons modulates an axon's target choice. However, because neighboring axons respond by choosing the complementary target, the stereotyped connectivity pattern is preserved. In contrast, loss of Notch in clones of neighboring postmitotic neurons results in erroneous coinnervation by multiple axons. Our observations establish mutual inhibition of axonal target choice as a robustness mechanism for brain wiring and unveil a novel cell fate independent function for canonical Notch signaling. DOI:http://dx.doi.org/10.7554/eLife.00337.001.


Subject(s)
Brain/physiology , Drosophila/physiology , Mitosis , Neural Inhibition , Neurons/physiology , Visual Pathways/physiology , Animals , Axons/physiology , Brain/metabolism , Cell Line , Computer Simulation , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Neurons/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Time Factors , Transfection , Visual Pathways/metabolism , p21-Activated Kinases/metabolism
16.
Nat Commun ; 1: 18, 2010 May 17.
Article in English | MEDLINE | ID: mdl-20975679

ABSTRACT

Intestinal cells are constantly produced from a stem cell reservoir that gives rise to proliferating transient amplifying cells, which subsequently differentiate into one of the four principal cell types. Signalling pathways, including the Notch signalling pathway, coordinate these differentiation processes and their deregulation may cause cancer. Pharmacological inhibition through γ-secretase inhibitors or genetic inactivation of the Notch signalling pathway results in the complete loss of proliferating crypt progenitors due to their conversion into post-mitotic goblet cells. The basic helix-loop-helix transcription factor Math1 is essential for intestinal secretory cell differentiation. Because of the critical roles of both Math1 and Notch signalling in intestinal homeostasis and neoplastic transformation, we sought to determine the genetic hierarchy regulating the differentiation of intestinal stem cells into secretory cells. In this paper, we demonstrate that the conversion of intestinal stem cells into goblet cells upon inhibition of the Notch signalling pathway requires Math1.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Intestines/cytology , Stem Cells/cytology , Stem Cells/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Dibenzazepines/pharmacology , Goblet Cells/cytology , Goblet Cells/metabolism , Immunohistochemistry , In Situ Hybridization , In Vitro Techniques , Mice , Receptors, Notch/genetics , Receptors, Notch/metabolism , Stem Cells/drug effects
17.
Biochemistry ; 49(8): 1577-89, 2010 Mar 02.
Article in English | MEDLINE | ID: mdl-20102160

ABSTRACT

Neuronal specification is regulated by the activity of transcription factors containing the basic helix-loop-helix motif (bHLH); these regulating proteins include, among others, the neurogenin (Ngn) family, related to the atonal family of genes. Neurogenin 1 (NGN1) is a 237-residue protein that contains a bHLH domain and is involved in neuronal differentiation. In this work, we synthesized the bHLH region of NGN1 (bHLHN) comprising residues 90-150 of the full-length NGN1. The domain is a monomeric natively unfolded protein with a pH-dependent premolten globule conformation, as shown by several spectroscopic techniques (namely, NMR, fluorescence, FTIR, and circular dichroism). The unfolded character of the domain also explains, first, the impossibility of its overexpression in several Escherichia coli strains and, second, its insolubility in aqueous buffers. To the best of our knowledge, this is the first extensive study of the conformational preferences of a bHLH domain under different solution conditions. Upon binding to two DNA E-boxes, the protein forms "fuzzy" complexes (that is, the complexes were not fully folded). The affinities of bHLHN for both DNA boxes were smaller than those of other bHLH domains, which might explain why the protein-DNA complexes were not fully folded.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Circular Dichroism , Helix-Loop-Helix Motifs/genetics , Helix-Loop-Helix Motifs/physiology , Humans , Nerve Tissue Proteins/genetics , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Folding , Spectroscopy, Fourier Transform Infrared
18.
J Cell Biol ; 187(1): 91-100, 2009 Oct 05.
Article in English | MEDLINE | ID: mdl-19786578

ABSTRACT

Merkel cells (MCs) are located in the touch-sensitive area of the epidermis and mediate mechanotransduction in the skin. Whether MCs originate from embryonic epidermal or neural crest progenitors has been a matter of intense controversy since their discovery >130 yr ago. In addition, how MCs are maintained during adulthood is currently unknown. In this study, using lineage-tracing experiments, we show that MCs arise through the differentiation of epidermal progenitors during embryonic development. In adults, MCs undergo slow turnover and are replaced by cells originating from epidermal stem cells, not through the proliferation of differentiated MCs. Conditional deletion of the Atoh1/Math1 transcription factor in epidermal progenitors results in the absence of MCs in all body locations, including the whisker region. Our study demonstrates that MCs arise from the epidermis by an Atoh1-dependent mechanism and opens new avenues for study of MC functions in sensory perception, neuroendocrine signaling, and MC carcinoma.


Subject(s)
Epidermal Cells , Homeostasis , Merkel Cells/cytology , Merkel Cells/physiology , Aging , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/metabolism , Cadherins/metabolism , Cell Differentiation , Cell Lineage , Epidermis/metabolism , Epidermis/ultrastructure , Fluorescent Antibody Technique, Direct , Immunohistochemistry , Integrases/genetics , Integrases/metabolism , Merkel Cells/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Neural Crest/cytology , Neural Crest/embryology , Neurofilament Proteins/genetics , Neurofilament Proteins/metabolism , Skin/cytology , Skin/embryology , Skin/metabolism , Skin/ultrastructure , Stem Cells/cytology , Time Factors , Vibrissae/cytology , Vibrissae/embryology , Vibrissae/metabolism
19.
Science ; 324(5923): 54, 2009 Apr 03.
Article in English | MEDLINE | ID: mdl-19342580

ABSTRACT

Most genes function at multiple stages of metazoan development, in dividing and nondividing cells. Generating mouse conditional knock-outs (cKO), where a gene can be eliminated in a temporally and spatially controlled manner, is a valuable technique because it allows study of gene function at any stage of life. In contrast and despite the development of many other powerful genetic tools, cKO has thus far been lacking in Drosophila. We combined several recent molecular and genetic technical advances in an approach termed integrase-mediated approach for gene knock-out (IMAGO). IMAGO allows the replacement of any genomic sequence, such as a gene, with another desired sequence, including cKO alleles that can be used to create positively marked mutant cells. IMAGO should also be applicable to other genetic model organisms.


Subject(s)
Drosophila melanogaster/genetics , Gene Knockout Techniques , Mutagenesis , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Drosophila Proteins , Drosophila melanogaster/cytology , Genes, Insect , Integrases/metabolism , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Photoreceptor Cells, Invertebrate/cytology , Photoreceptor Cells, Invertebrate/physiology , Recombination, Genetic , Sense Organs/cytology , Sense Organs/physiology
20.
PLoS Biol ; 7(2): e39, 2009 Feb 24.
Article in English | MEDLINE | ID: mdl-19243219

ABSTRACT

Colon cancer accounts for more than 10% of all cancer deaths annually. Our genetic evidence from Drosophila and previous in vitro studies of mammalian Atonal homolog 1 (Atoh1, also called Math1 or Hath1) suggest an anti-oncogenic function for the Atonal group of proneural basic helix-loop-helix transcription factors. We asked whether mouse Atoh1 and human ATOH1 act as tumor suppressor genes in vivo. Genetic knockouts in mouse and molecular analyses in the mouse and in human cancer cell lines support a tumor suppressor function for ATOH1. ATOH1 antagonizes tumor formation and growth by regulating proliferation and apoptosis, likely via activation of the Jun N-terminal kinase signaling pathway. Furthermore, colorectal cancer and Merkel cell carcinoma patients show genetic and epigenetic ATOH1 loss-of-function mutations. Our data indicate that ATOH1 may be an early target for oncogenic mutations in tissues where it instructs cellular differentiation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Carcinoma, Merkel Cell/genetics , Colorectal Neoplasms/genetics , Genes, Tumor Suppressor/physiology , Skin Neoplasms/genetics , Animals , Apoptosis/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Merkel Cell/metabolism , Carcinoma, Merkel Cell/pathology , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , DNA Mutational Analysis , Gene Expression Regulation, Neoplastic , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , JNK Mitogen-Activated Protein Kinases , Male , Mice , Mice, Knockout , Mutation , Signal Transduction , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...