Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
J Clin Endocrinol Metab ; 108(8): 2065-2077, 2023 Jul 14.
Article in English | MEDLINE | ID: mdl-36683389

ABSTRACT

CONTEXT: Gut bacteria can influence host immune responses but little is known about their role in tolerance-loss mechanisms in Graves disease (GD; hyperthyroidism caused by autoantibodies, TRAb, to the thyrotropin receptor, TSHR) and its progression to Graves orbitopathy (GO). OBJECTIVE: This work aimed to compare the fecal microbiota in GD patients, with GO of varying severity, and healthy controls (HCs). METHODS: Patients were recruited from 4 European countries (105 GD patients, 41 HCs) for an observational study with cross-sectional and longitudinal components. RESULTS: At recruitment, when patients were hyperthyroid and TRAb positive, Actinobacteria were significantly increased and Bacteroidetes significantly decreased in GD/GO compared with HCs. The Firmicutes to Bacteroidetes (F:B) ratio was significantly higher in GD/GO than in HCs. Differential abundance of 15 genera was observed in patients, being most skewed in mild GO. Bacteroides displayed positive and negative correlations with TSH and free thyroxine, respectively, and was also significantly associated with smoking in GO; smoking is a risk factor for GO but not GD. Longitudinal analyses revealed that the presence of certain bacteria (Clostridiales) at diagnosis correlated with the persistence of TRAb more than 200 days after commencing antithyroid drug treatment. CONCLUSION: The increased F:B ratio observed in GD/GO mirrors our finding in a murine model comparing TSHR-immunized with control mice. We defined a microbiome signature and identified changes associated with autoimmunity as distinct from those due to hyperthyroidism. Persistence of TRAb is predictive of relapse; identification of these patients at diagnosis, via their microbiome, could improve management with potential to eradicate Clostridiales.


Subject(s)
Gastrointestinal Microbiome , Graves Disease , Graves Ophthalmopathy , Hyperthyroidism , Humans , Mice , Animals , Indigo Carmine/therapeutic use , Cross-Sectional Studies , Autoantibodies , Receptors, Thyrotropin , Hyperthyroidism/complications
2.
Sci Signal ; 12(565)2019 01 22.
Article in English | MEDLINE | ID: mdl-30670636

ABSTRACT

In medulloblastomas (MBs), the expression and activity of RE1-silencing transcription factor (REST) is increased in tumors driven by the sonic hedgehog (SHH) pathway, specifically the SHH-α (children 3 to 16 years) and SHH-ß (infants) subgroups. Neuronal maturation is greater in SHH-ß than SHH-α tumors, but both correlate with poor overall patient survival. We studied the contribution of REST to MB using a transgenic mouse model (RESTTG ) wherein conditional NeuroD2-controlled REST transgene expression in lineage-committed Ptch1 +/- cerebellar granule neuron progenitors (CGNPs) accelerated tumorigenesis and increased penetrance and infiltrative disease. This model revealed a neuronal maturation context-specific antagonistic interplay between the transcriptional repressor REST and the activator GLI1 at Ptch1 Expression of Arrb1, which encodes ß-arrestin1 (a GLI1 inhibitor), was substantially reduced in proliferating and, to a lesser extent, lineage-committed RESTTG cells compared with wild-type proliferating CGNPs. Lineage-committed RESTTG cells also had decreased GLI1 activity and increased histone H3K9 methylation at the Ptch1 locus, which correlated with premature silencing of Ptch1 These cells also had decreased expression of Pten, which encodes a negative regulator of the kinase AKT. Expression of PTCH1 and GLI1 were less, and ARRB1 was somewhat greater, in patient SHH-ß than SHH-α MBs, whereas that of PTEN was similarly lower in both subtypes than in others. Inhibition of histone modifiers or AKT reduced proliferation and induced apoptosis, respectively, in cultured REST-high MB cells. Our findings linking REST to differentiation-specific chromatin remodeling, PTCH1 silencing, and AKT activation in MB tissues reveal potential subgroup-specific therapeutic targets for MB patients.


Subject(s)
Cerebellar Neoplasms/genetics , Chromatin/genetics , Hedgehog Proteins/genetics , Medulloblastoma/genetics , Patched-1 Receptor/genetics , Proto-Oncogene Proteins c-akt/genetics , Repressor Proteins/genetics , Adult , Animals , Cell Line, Tumor , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/pathology , Child , Chromatin/metabolism , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/metabolism , Humans , Infant , Male , Medulloblastoma/metabolism , Medulloblastoma/pathology , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Neoplasm Staging , Patched-1 Receptor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Repressor Proteins/metabolism , Signal Transduction/genetics , Transplantation, Heterologous
3.
Mol Cancer Res ; 15(8): 1073-1084, 2017 08.
Article in English | MEDLINE | ID: mdl-28483947

ABSTRACT

The deubiquitylase (DUB) USP37 is a component of the ubiquitin system and controls cell proliferation by regulating the stability of the cyclin-dependent kinase inhibitor 1B, (CDKN1B/p27Kip1). The expression of USP37 is downregulated in human medulloblastoma tumor specimens. In the current study, we show that USP37 prevents medulloblastoma growth in mouse orthotopic models, suggesting that it has tumor-suppressive properties in this neural cancer. Here, we also report on the mechanism underlying USP37 loss in medulloblastoma. Previously, we observed that the expression of USP37 is transcriptionally repressed by the RE1 silencing transcription factor (REST), which requires chromatin remodeling factors for its activity. Genetic and pharmacologic approaches were employed to identify a specific role for G9a, a histone methyltransferase (HMT), in promoting methylation of histone H3 lysine-9 (H3K9) mono- and dimethylation, and surprisingly trimethylation, at the USP37 promoter to repress its gene expression. G9a inhibition also blocked the tumorigenic potential of medulloblastoma cells in vivo Using isogenic low- and high-REST medulloblastoma cells, we further showed a REST-dependent elevation in G9a activity, which further increased mono- and trimethylation of histone H3K9, accompanied by downregulation of USP37 expression. Together, these findings reveal a role for REST-associated G9a and histone H3K9 methylation in the repression of USP37 expression in medulloblastoma.Implications: Reactivation of USP37 by G9a inhibition has the potential for therapeutic applications in REST-expressing medulloblastomas. Mol Cancer Res; 15(8); 1073-84. ©2017 AACR.


Subject(s)
Endopeptidases/genetics , Histocompatibility Antigens/genetics , Histone-Lysine N-Methyltransferase/genetics , Medulloblastoma/genetics , Repressor Proteins/genetics , Animals , Carcinogenesis/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Histones/genetics , Humans , Medulloblastoma/pathology , Methylation , Methyltransferases/genetics , Mice , Ubiquitin/genetics , Xenograft Model Antitumor Assays
4.
Genome Res ; 26(11): 1600-1611, 2016 11.
Article in English | MEDLINE | ID: mdl-27803194

ABSTRACT

Assaying in vivo accrual of DNA damage and DNA mutations by stem cells and pinpointing sources of damage and mutations would further our understanding of aging and carcinogenesis. Two main hurdles must be overcome. First, in vivo mutation rates are orders of magnitude lower than raw sequencing error rates. Second, stem cells are vastly outnumbered by differentiated cells, which have a higher mutation rate-quantification of stem cell DNA damage and DNA mutations is thus best performed from small, well-defined cell populations. Here we report a mutation detection technique, based on the "duplex sequencing" principle, with an error rate below ∼10-10 and that can start from as little as 50 pg DNA. We validate this technique, which we call SIP-HAVA-seq, by characterizing Caenorhabditis elegans germline stem cell mutation accrual and asking how mating affects that accrual. We find that a moderate mating-induced increase in cell cycling correlates with a dramatic increase in accrual of mutations. Intriguingly, these mutations consist chiefly of deletions in nonexpressed genes. This contrasts with results derived from mutation accumulation lines and suggests that mutation spectrum and genome distribution change with replicative age, chronological age, cell differentiation state, and/or overall worm physiological state. We also identify single-stranded gaps as plausible deletion precursors, providing a starting point to identify the molecular mechanisms of mutagenesis that are most active. SIP-HAVA-seq provides the first direct, genome-wide measurements of in vivo mutation accrual in stem cells and will enable further characterization of underlying mechanisms and their dependence on age and cell state.


Subject(s)
Mutation Accumulation , Sequence Analysis, DNA/methods , Animals , Caenorhabditis elegans , Cell Cycle , DNA Damage , Germ Cells/cytology , Germ Cells/metabolism , Mutation Rate , Sequence Analysis, DNA/standards
5.
Dev Cell ; 35(4): 405-17, 2015 Nov 23.
Article in English | MEDLINE | ID: mdl-26609956

ABSTRACT

Positional information derived from local morphogen concentration plays an important role in patterning. A key question is how morphogen diffusion and gene expression regulation shape positional information into an appropriate profile with suitably low noise. We address this question using a model system--the C. elegans germline--whose regulatory network has been well characterized genetically but whose spatiotemporal dynamics are poorly understood. We show that diffusion within the germline syncytium is a critical control of stem cell differentiation and that semi-permeable diffusion barriers present at key locations make it possible--in combination with a feedback loop in the germline regulatory network--for mitotic zone size to be robust against spatial noise in Notch signaling. Spatial averaging within compartments defined by diffusion barriers is an advantageous patterning strategy, which attenuates noise while still allowing for sharp transitions between compartments. This strategy could apply to other organs.


Subject(s)
Body Patterning/genetics , Caenorhabditis elegans/embryology , Caenorhabditis elegans/genetics , Cell Differentiation , Gene Expression Regulation, Developmental , Germ Cells/cytology , Stem Cells/cytology , Animals , Caenorhabditis elegans/metabolism , Feedback, Physiological , Germ Cells/metabolism , Models, Biological , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Stem Cells/metabolism
6.
Mol Cancer Ther ; 11(8): 1713-1723, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22848092

ABSTRACT

Medulloblastoma is a malignant pediatric brain tumor. Current treatment following patient stratification into standard and high-risk groups using clinical features has improved survival. However, a subset of patients with standard risk features have unanticipated aggressive disease, underscoring the need for a better understanding of tumor biology and the development of novel treatments. Poor differentiation, a hallmark of medulloblastomas is associated with elevated expression levels of the repressor of neuronal differentiation called repressor element 1-silencing transcription factor (REST). Here, we assessed whether elevated REST expression levels had prognostic significance and whether its pharmacologic manipulation would promote neurogenesis and block tumor cell growth. REST levels in patient tumors were measured by immunohistochemistry and stratified into negative, low/moderate- (+/++/+++), and high-REST (+++++) groups. Kaplan-Meier curves revealed that patients with high-REST tumors had worse overall and event-free survival compared with patients with REST-negative or REST-low tumors. Because histone deacetylases (HDAC) are required for REST-dependent repression of neurogenesis, we evaluated a panel of HDAC inhibitors (HDACI) for their effects on growth and differentiation of established and primary REST-positive cell lines. MS-275, trichostatin-A (TSA), valproic acid (VPA), and suberoylanilide hydroxamic acid (SAHA) upregulated expression of the REST-target neuronal differentiation gene, Syn1, suggesting a potential effect of these HDACIs on REST function. Interestingly, VPA and TSA substantially increased histone acetylation at the REST promoter and activated its transcription, whereas SAHA unexpectedly promoted its proteasomal degradation. A REST-dependent decrease in cell growth was also observed following SAHA treatment. Thus, our studies suggest that HDACIs may have therapeutic potential for patients with REST-positive tumors. This warrants further investigation.


Subject(s)
Cerebellar Neoplasms/genetics , Medulloblastoma/genetics , Repressor Proteins/genetics , Adolescent , Antineoplastic Agents/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/mortality , Child , Child, Preschool , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Male , Medulloblastoma/metabolism , Medulloblastoma/mortality , Prognosis , Repressor Proteins/metabolism , Transcription, Genetic/drug effects , Vorinostat
7.
Clin Cancer Res ; 16(24): 6060-70, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21169257

ABSTRACT

PURPOSE: Multiple developmental pathways including Notch, Hedgehog, and Wnt are active in malignant brain tumors such as medulloblastoma and glioblastoma (GBM). This raises the possibility that tumors might compensate for therapy directed against one pathway by upregulating a different one. We investigated whether brain tumors show resistance to therapies against Notch, and whether targeting multiple pathways simultaneously would kill brain tumor cells more effectively than monotherapy. EXPERIMENTAL DESIGN: We used GBM neurosphere lines to investigate the effects of a gamma-secretase inhibitor (MRK-003) on tumor growth, and chromatin immunoprecipitation to study the regulation of other genes by Notch targets. We also evaluated the effect of combined therapy with a Hedgehog inhibitor (cyclopamine) in GBM and medulloblastoma lines, and in primary human GBM cultures. RESULTS: GBM cells are at least partially resistant to long-term MRK-003 treatment, despite ongoing Notch pathway suppression, and show concomitant upregulation of Wnt and Hedgehog activity. The Notch target Hes1, a repressive transcription factor, bound the Gli1 first intron, and may inhibit its expression. Similar results were observed in a melanoma-derived cell line. Targeting Notch and Hedgehog simultaneously induced apoptosis, decreased cell growth, and inhibited colony-forming ability more dramatically than monotherapy. Low-passage neurospheres isolated from freshly resected human GBMs were also highly susceptible to coinhibition of the two pathways, indicating that targeting multiple developmental pathways can be more effective than monotherapy at eliminating GBM-derived cells. CONCLUSIONS: Notch may directly suppress Hedgehog via Hes1 mediated inhibition of Gli1 transcription, and targeting both pathways simultaneously may be more effective at eliminating GBMs cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Drug Resistance, Neoplasm/genetics , Hedgehog Proteins/metabolism , Homeodomain Proteins/physiology , Transcription Factors/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cyclic S-Oxides/administration & dosage , Drug Evaluation, Preclinical , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Hedgehog Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Receptors, Notch/metabolism , Receptors, Notch/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Thiadiazoles/administration & dosage , Transcription Factor HES-1 , Transcription Factors/metabolism , U937 Cells , Veratrum Alkaloids/administration & dosage , Zinc Finger Protein GLI1
8.
Eur J Cancer ; 46(15): 2771-80, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20886683

ABSTRACT

Etoposide, an inhibitor of topoisomerase II, promotes DNA damage and apoptosis of cancer cells and is a component of standard therapy for neuroblastoma. Resistance to etoposide has been observed in neural tumour cells expressing lower levels of topoisomerase II. In the present study, we have examined the contribution of epigenetic modulation of gene expression in the potentiation of etoposide-mediated cytotoxicity in neuroblastoma cells. Specifically, we studied the effects of histone deacetylase inhibition with valproic acid on topoisomerase II gene expression and apoptosis in response to etoposide. Using human neuroblastoma cell lines SK-N-AS and SK-N-SH, we show that although the combination of valproic acid and etoposide promoted a reduction in growth compared to either drug alone in both cells, the effect was substantially enhanced in SK-N-AS compared to SK-N-SH cells. An increase in histone H3 acetylation and p21 expression was observed in both cell lines, however, upregulation of topoisomerase II-beta gene expression and an increase in PARP cleavage was observed in SK-N-AS cells only. Furthermore, chromatin immunoprecipitation assays revealed an increase in acetylation of histone H3 at the cognate topoisomerase II-beta gene after treatment with valproic acid in SK-N-AS cells. These results suggest a potential epigenetic mechanism of regulation of the topoisomerase II-beta gene and a possible role for its increased expression in the sensitivity of SK-N-AS neuroblastoma cells to etoposide.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Chromatin Assembly and Disassembly/drug effects , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Etoposide/therapeutic use , Neuroblastoma/drug therapy , Valproic Acid/therapeutic use , Cell Cycle , Cell Line, Tumor , Cell Survival , Chromatin Immunoprecipitation , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins/genetics , Drug Synergism , Epigenomics , Histone Deacetylase Inhibitors/therapeutic use , Humans , Inhibitory Concentration 50 , Neuroblastoma/enzymology
9.
J Neurooncol ; 93(3): 303-18, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19148581

ABSTRACT

OBJECT: Apoptosis, a key cellular response to therapeutic agents is often inactivated in tumor cells. In this study, we evaluated the expression of the tumor necrosis family of death receptors, DR4 and DR5, in medulloblastoma tumor samples and cell lines to determine if epigenetic modulation of gene expression could sensitize tumor cell lines to TRAIL-mediated apoptosis. METHODS: Human medulloblastoma samples and cell lines were analyzed for DR4 and DR5 expression by quantitative PCR and immunofluorescence assays. Cell lines with downregulated expression of one or both genes were treated with the histone deacetylase inhibitor, MS-275, and the expression of DR4 and DR5 measured by quantitative PCR, Western blotting, flow cytometry and chromatin immunoprecipitation assays. Induction of apoptosis in the presence of MS-275 was evaluated by TUNEL assay and its ability to augment TRAIL-mediated cytotoxicity was determined by MTT assays, Western blotting and flow cytometry. RESULTS: Compared to normal cerebellum, DR4, but not DR5 expression was consistently downregulated in medulloblastoma tumor samples and in Daoy and D283 cell lines. Interestingly, MS-275 decreased cell growth and induced apoptosis in Daoy and D283 cells. In Daoy cells, this coincided with increased histone H3 and H4 acetylation at the DR4 promoter and enhanced DR4 gene and protein expression as well as elevated Caspase-8 activity. The involvement of DR4 in the cellular response to MS-275 was further confirmed by the observation that knockdown of DR4 and FADD abrogated apoptosis. Further, addition of TRAIL to MS-275 treated cells resulted in an enhancement of apoptosis, suggesting that the upregulated death receptors were functional. CONCLUSION: Our study provides an understanding of the role of DR4 in apoptosis of medulloblastoma cell lines and suggests a potential contribution of aberrant histone deacetylation to the resistance of medulloblastoma cells to therapeutic death.


Subject(s)
Apoptosis/physiology , Cerebellar Neoplasms/metabolism , Medulloblastoma/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Apoptosis/drug effects , Benzamides/pharmacology , Blotting, Western , Caspase 8/metabolism , Cell Line, Tumor , Cerebellar Neoplasms/genetics , Enzyme Inhibitors/pharmacology , Fas-Associated Death Domain Protein/drug effects , Fas-Associated Death Domain Protein/metabolism , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression , Histone Deacetylase Inhibitors , Humans , Immunoprecipitation , In Situ Nick-End Labeling , Medulloblastoma/genetics , Pyridines/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Reverse Transcriptase Polymerase Chain Reaction , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/pharmacology
10.
BMC Neurosci ; 9: 119, 2008 Dec 18.
Article in English | MEDLINE | ID: mdl-19094242

ABSTRACT

BACKGROUND: The Drosophila circadian oscillator is composed of transcriptional feedback loops in which CLOCK-CYCLE (CLK-CYC) heterodimers activate their feedback regulators period (per) and timeless (tim) via E-box mediated transcription. These feedback loop oscillators are present in distinct clusters of dorsal and lateral neurons in the adult brain, but how this pattern of expression is established during development is not known. Since CLK is required to initiate feedback loop function, defining the pattern of CLK expression in embryos and larvae will shed light on oscillator neuron development. RESULTS: A novel CLK antiserum is used to show that CLK expression in the larval CNS and adult brain is limited to circadian oscillator cells. CLK is initially expressed in presumptive small ventral lateral neurons (s-LNvs), dorsal neurons 2 s (DN2s), and dorsal neuron 1 s (DN1s) at embryonic stage (ES) 16, and this CLK expression pattern persists through larval development. PER then accumulates in all CLK-expressing cells except presumptive DN2s during late ES 16 and ES 17, consistent with the delayed accumulation of PER in adult oscillator neurons and antiphase cycling of PER in larval DN2s. PER is also expressed in non-CLK-expressing cells in the embryonic CNS starting at ES 12. Although PER expression in CLK-negative cells continues in ClkJrk embryos, PER expression in cells that co-express PER and CLK is eliminated. CONCLUSION: These data demonstrate that brain oscillator neurons begin development during embryogenesis, that PER expression in non-oscillator cells is CLK-independent, and that oscillator phase is an intrinsic characteristic of brain oscillator neurons. These results define the temporal and spatial coordinates of factors that initiate Clk expression, imply that circadian photoreceptors are not activated until the end of embryogenesis, and suggest that PER functions in a different capacity before oscillator cell development is initiated.


Subject(s)
Biological Clocks/physiology , Brain/embryology , Drosophila Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Biological Clocks/genetics , Biological Clocks/radiation effects , CLOCK Proteins , Circadian Rhythm , Drosophila , Drosophila Proteins/genetics , Gene Expression , Genes, Insect , Larva/genetics , Larva/metabolism , Microscopy, Confocal , Neurogenesis , Neurons/metabolism , Period Circadian Proteins , Photoreceptor Cells, Invertebrate/physiology , Transcription Factors/genetics
11.
Mol Cell Biol ; 28(14): 4642-52, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18474612

ABSTRACT

The Drosophila melanogaster circadian oscillator comprises interlocked per/tim and Clk transcriptional feedback loops. In the per/tim loop, CLK-CYC-dependent transcriptional activation is rhythmically repressed by PER or PER-TIM to control circadian gene expression that peaks around dusk. Here we show that rhythmic transcription of per and tim involves time-of-day-specific binding of CLK-CYC and associated cycles in chromatin modifications. Activation of per and tim transcription occurs in concert with CLK-CYC binding to upstream and/or intronic E-boxes, acetylation of histone H3-K9, and trimethylation of histone H3-K4. These events are associated with RNA polymerase II (Pol II) binding to the tim promoter and transcriptional elongation by Pol II that is constitutively bound to the per promoter. Repression of per and tim transcription is associated with PER-dependent reversal of these events. Rhythms in H3-K9 acetylation and H3-K4 trimethylation are also associated with CLOCK-BMAL1-dependent transcription in mammals, indicating that the mechanism that controls rhythmic transcription is a conserved feature of the circadian clock even though feedback repression is mediated by different proteins.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Chromatin/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Nuclear Proteins/genetics , Transcription Factors/metabolism , Transcription, Genetic , ARNTL Transcription Factors , Animals , CLOCK Proteins , Circadian Rhythm , Gene Expression Regulation , Histones/metabolism , Period Circadian Proteins , RNA Polymerase II/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...