Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 29(7): 1344-1359, 2023 04 03.
Article in English | MEDLINE | ID: mdl-36689560

ABSTRACT

PURPOSE: Cisplatin (CDDP)-based chemotherapy is a first-line treatment for patients with advanced head and neck squamous cell carcinomas (HNSCC), despite a high rate of treatment failures, acquired resistance, and subsequent aggressive behavior. The purpose of this study was to study the mechanism of CDDP resistance and metastasis in HNSCC. We investigated the role of NRF2 pathway activation as a driven event for tumor progression and metastasis of HNSCC. EXPERIMENTAL DESIGN: Human HNSCC cell lines that are highly resistant to CDDP were generated. Clonogenic survival assays and a mouse model of oral cancer were used to examine the impact of NRF2 activation in vitro and in vivo on CDDP sensitivity and development of metastasis. Western blotting, immunostaining, whole-exome sequencing, single-cell transcriptomic and epigenomic profiling platforms were performed to dissect clonal evolution and molecular mechanisms. RESULTS: Implantation of CDDP-resistant HNSCC cells into the tongues of nude mice resulted in a very high rate of distant metastases. The CDDP-resistant cells had significantly higher expression of NRF2 pathway genes in the presence of newly acquired KEAP1 mutations, or via epigenomic activation of target genes. Knockdown of NRF2 or restoration of the wild-type KEAP1 genes resensitized resistant cells to CDDP and decreased distant metastasis (DM). Finally, treatment with inhibitor of glutaminase-1, a NRF2 target gene, alleviated CDDP resistance. CONCLUSIONS: CDDP resistance and development of DM are associated with dysregulated and epigenetically reprogrammed KEAP1-NRF2 signaling pathway. A strategy targeting KEAP1/NRF2 pathway or glutamine metabolism deserves further clinical investigation in patients with CDDP-resistant head and neck tumors.


Subject(s)
Antineoplastic Agents , Head and Neck Neoplasms , NF-E2-Related Factor 2 , Squamous Cell Carcinoma of Head and Neck , Animals , Humans , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cisplatin/pharmacology , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Epigenomics , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Mice, Nude , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Signal Transduction , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/genetics
2.
Oncotarget ; 12(14): 1339-1353, 2021 Jul 06.
Article in English | MEDLINE | ID: mdl-34262645

ABSTRACT

The resistance of cancer cells to radiation-based treatment is a major clinical challenge confounding standard of care in cancer. This problem is particularly notable in many solid tumors where cancer cells are only partially responsive to radiation therapy. Combination of radiation with radiosensitizers is able to enhance tumor cell killing. However, currently available radiosensitizers are associated with significant normal tissue toxicity. Accordingly, there is an unmet need to develop safer and more effective radiosensitizers to improve tumor control. Here, we evaluated the radiosensitizing effect of the FDA-approved drug esomeprazole in normal and radioresistant human head and neck squamous cell carcinoma (HNSCC) cells in vitro, and in a mouse model of HNSCC. For the in vitro studies, we used cancer cell colony formation (clonogenicity) assay to compare cancer cell growth in the absence or presence of esomeprazole. To determine mechanism(s) of action, we assessed cell proliferation and profiled cell cycle regulatory proteins. In addition, we performed reverse phase protein array (RPPA) study to understand the global effect of esomeprazole on over 200 cancer-related proteins. For the in vivo study, we engrafted HNSCC in a mouse model and compared tumor growth in animals treated with radiation, esomeprazole, and combination of radiation with esomeprazole. We found that esomeprazole inhibits tumor growth and dose-dependently enhances the cell killing effect of ionizing radiation in wildtype and p53-mutant radioresistant cancer cells. Mechanistic studies demonstrate that esomeprazole arrests cancer cells in the G1 phase of the cell cycle through upregulation of p21 protein and inhibition of cyclin-dependent kinases (Cdks) type 1 (Cdk1) and type 2 (Cdk2). In vivo data showed greater tumor control in animals treated with combination of radiation and esomeprazole compared to either treatment alone, and that this was associated with inhibition of cell proliferation in vivo. In addition, combination of esomeprazole with radiation significantly impaired repair following radiation-induced DNA damage. Our studies indicate that esomeprazole sensitizes cancer cells to ionizing radiation, and is associated with upregulation of p21 to arrest cells in the G1 phase of the cell cycle. Our findings have significant therapeutic implications for the repurposing of esomeprazole as a radiosensitizer in HNSCC and other solid tumors.

3.
J Immunother Cancer ; 9(7)2021 07.
Article in English | MEDLINE | ID: mdl-34230113

ABSTRACT

BACKGROUND: Metformin is a commonly used antidiabetic medication which has demonstrated promise as an anticancer agent alone and in combination with conventional treatment regimens. There is increasing evidence that metformin can also generate immunomodulatory effects in solid tumors and is currently being investigated as an adjunct to immune checkpoint inhibitors (ICIs). We hypothesized that metformin would generate a shift in immunity unfavorable to tumor growth and tested this hypothesis in a preclinical model of head and neck cancer. METHODS: Using a syngeneic mouse model of human papillomavirus-associated head and neck cancer (mEER/MTEC), we tested the impact of metformin on systemic and local immunity and tumor growth velocity. We compared the effects of acute and chronic treatment regimens on immunocyte presence and activation using a combination of flow cytometry and targeted transcriptomic analysis. RESULTS: Acute metformin exposure generated measurable shifts in systemic myeloid and T-cell populations in non-tumor-bearing mice and decreased myeloid derived suppressor cell (MDSC) levels in tumor draining lymph nodes of tumor-bearing mice. Although metformin decreased regulatory T-cell (T-reg) and MDSC levels and increased CD8+ levels in murine tumors when combined with ICIs, acute metformin exposure was insufficient to generate substantial antitumor activity. Conversely, long-term metformin treatment significantly reduced tumor growth velocity, increased the CD8+/T-reg ratio, increased tumor infiltrating lymphocyte levels and upregulated component genes of the previously validated T-cell inflamed expression profile. CONCLUSIONS: Metformin generates complex systemic and local immune effects which vary as a function of treatment duration. Combinatorial strategies with ICIs must take into account both the complexity and variability of these effects in order to generate maximal antitumor activity in future clinical trials.


Subject(s)
Antineoplastic Agents/therapeutic use , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Hypoglycemic Agents/pharmacology , Male , Metformin/pharmacology , Mice , Neoplasms/immunology
4.
ACS Cent Sci ; 6(10): 1772-1788, 2020 Oct 28.
Article in English | MEDLINE | ID: mdl-33145414

ABSTRACT

The development of a fluorescent probe for a specific metal has required exquisite design, synthesis, and optimization of fluorogenic molecules endowed with chelating moieties with heteroatoms. These probes are generally chelation- or reactivity-based. Catalysis-based fluorescent probes have the potential to be more sensitive; however, catalytic methods with a biocompatible fluorescence turn-on switch are rare. Here, we have exploited ligand-accelerated metal catalysis to repurpose known fluorescent probes for different metals, a new approach in probe development. We used the cleavage of allylic and propargylic ethers as platforms that were previously designed for palladium. After a single experiment that combinatorially examined >800 reactions with two variables (metal and ligand) for each ether, we discovered a platinum- or copper-selective method with the ligand effect of specific phosphines. Both metal-ligand systems were previously unknown and afforded strong signals owing to catalytic turnover. The fluorometric technologies were applied to geological, pharmaceutical, serum, and live cell samples and were used to discover that platinum accumulates in lysosomes in cisplatin-resistant cells in a manner that appears to be independent of copper distribution. The use of ligand-accelerated catalysis may present a new blueprint for engineering metal selectivity in probe development.

5.
Cancers (Basel) ; 12(6)2020 Jun 24.
Article in English | MEDLINE | ID: mdl-32599707

ABSTRACT

Background: Cisplatin (CDDP) is commonly utilized in the treatment of advanced solid tumors including head and neck squamous cell carcinoma (HNSCC). Cisplatin response remains highly variable among individual tumors and development of cisplatin resistance is common. We hypothesized that development of cisplatin resistance is partially driven by metabolic reprogramming. Methods: Using a pre-clinical HNSCC model and an integrated approach to steady state metabolomics, metabolic flux and gene expression data we characterized the interaction between cisplatin resistance and metabolic reprogramming. Results: Cisplatin toxicity in HNSCC was driven by generation of intra-cellular oxidative stress. This was validated by demonstrating that acquisition of cisplatin resistance generates cross-resistance to ferroptosis agonists despite the fact that cisplatin itself does not trigger ferroptosis. Acquisition of cisplatin resistance dysregulated the expression of genes involved in amino acid, fatty acid metabolism and central carbon catabolic pathways, enhanced glucose catabolism and serine synthesis. Acute cisplatin exposure increased intra-tumoral levels of S-methyl-5-thiadenosine (MTA) precursors and metabotoxins indicative of generalized oxidative stress. Conclusions: Acquisition of cisplatin resistance is linked to metabolic recovery from oxidative stress. Although this portends poor effectiveness for directed metabolic targeting, it supports the potential for biomarker development of cisplatin effectiveness using an integrated approach.

6.
Proc Natl Acad Sci U S A ; 114(41): 10972-10977, 2017 10 10.
Article in English | MEDLINE | ID: mdl-28973907

ABSTRACT

The Drosophila circadian clock keeps time via transcriptional feedback loops. These feedback loops are initiated by CLOCK-CYCLE (CLK-CYC) heterodimers, which activate transcription of genes encoding the feedback repressors PERIOD and TIMELESS. Circadian clocks normally operate in ∼150 brain pacemaker neurons and in many peripheral tissues in the head and body, but can also be induced by expressing CLK in nonclock cells. These ectopic clocks also require cyc, yet CYC expression is restricted to canonical clock cells despite evidence that cyc mRNA is widely expressed. Here we show that CLK binds to and stabilizes CYC in cell culture and in nonclock cells in vivo. Ectopic clocks also require the blue light photoreceptor CRYPTOCHROME (CRY), which is required for both light entrainment and clock function in peripheral tissues. These experiments define the genetic architecture required to initiate circadian clock function in Drosophila, reveal mechanisms governing circadian activator stability that are conserved in perhaps all eukaryotes, and suggest that Clk, cyc, and cry expression is sufficient to drive clock expression in naive cells.


Subject(s)
ARNTL Transcription Factors/chemistry , Animals, Genetically Modified/metabolism , CLOCK Proteins/metabolism , Circadian Clocks , Cryptochromes/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Neurons/physiology , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Animals , Animals, Genetically Modified/genetics , CLOCK Proteins/genetics , Cells, Cultured , Circadian Rhythm , Drosophila Proteins/genetics , Neurons/cytology
7.
PLoS Genet ; 12(11): e1006430, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27814361

ABSTRACT

The Drosophila circadian oscillator controls daily rhythms in physiology, metabolism and behavior via transcriptional feedback loops. CLOCK-CYCLE (CLK-CYC) heterodimers initiate feedback loop function by binding E-box elements to activate per and tim transcription. PER-TIM heterodimers then accumulate, bind CLK-CYC to inhibit transcription, and are ultimately degraded to enable the next round of transcription. The timing of transcriptional events in this feedback loop coincide with, and are controlled by, rhythms in CLK-CYC binding to E-boxes. PER rhythmically binds CLK-CYC to initiate transcriptional repression, and subsequently promotes the removal of CLK-CYC from E-boxes. However, little is known about the mechanism by which CLK-CYC is removed from DNA. Previous studies demonstrated that the transcription repressor CLOCKWORK ORANGE (CWO) contributes to core feedback loop function by repressing per and tim transcription in cultured S2 cells and in flies. Here we show that CWO rhythmically binds E-boxes upstream of core clock genes in a reciprocal manner to CLK, thereby promoting PER-dependent removal of CLK-CYC from E-boxes, and maintaining repression until PER is degraded and CLK-CYC displaces CWO from E-boxes to initiate transcription. These results suggest a model in which CWO co-represses CLK-CYC transcriptional activity in conjunction with PER by competing for E-box binding once CLK-CYC-PER complexes have formed. Given that CWO orthologs DEC1 and DEC2 also target E-boxes bound by CLOCK-BMAL1, a similar mechanism may operate in the mammalian clock.


Subject(s)
ARNTL Transcription Factors/genetics , CLOCK Proteins/genetics , Circadian Rhythm/genetics , Drosophila Proteins/genetics , Period Circadian Proteins/genetics , ARNTL Transcription Factors/metabolism , Animals , CLOCK Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , E-Box Elements , Gene Expression Regulation , Period Circadian Proteins/metabolism , Repressor Proteins , Transcription, Genetic
8.
Methods Enzymol ; 551: 323-47, 2015.
Article in English | MEDLINE | ID: mdl-25662463

ABSTRACT

In eukaryotes, the circadian clock controls 24h rhythms in physiology, metabolism, and behavior via cell autonomous transcriptional feedback loops. These feedback loops keep circadian time and control rhythmic outputs by driving rhythms in transcription; thus, it is important to determine when clock transcription factors bind their target sequences in vivo to promote or repress transcription. Interactions between proteins and DNA can be measured in cells, tissue, or whole organisms using a technique called chromatin immunoprecipitation (ChIP). The principle underlying ChIP is that protein is cross-linked to associated chromatin to form a protein-DNA complex, the DNA is then sheared, and the protein of interest is immunoprecipitated. The cross-links are then removed from the antibody-protein-DNA complex, and the associated DNA fragments are purified. The DNA is then used to quantify specific targets by real-time quantitative PCR or to generate libraries for global analysis of protein target sites by high-throughput sequencing (ChIP-seq). ChIP has been widely used in circadian biology to assess rhythmic binding of clock components, RNA polymerase II, and rhythms in chromatin modifications such as histone acetylation and methylation. Here, we present a detailed method for ChIP analysis in Drosophila that can be used to assess protein-DNA-binding rhythms at specific genomic target sites. With minor modifications, this technique can be used to assess protein-DNA-binding rhythms at all target sites via ChIP-seq. ChIP analysis has revealed the relationship between clock factor binding, transcription, and chromatin modifications and promises to reveal circadian transcription networks that control phase and tissue specificity.


Subject(s)
Drosophila Proteins/isolation & purification , Animals , Chromatin Immunoprecipitation , DNA/genetics , DNA/isolation & purification , Drosophila , Drosophila Proteins/physiology , Real-Time Polymerase Chain Reaction
9.
Curr Biol ; 21(9): 756-61, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21514156

ABSTRACT

The Drosophila circadian oscillator is comprised of transcriptional feedback loops that are activated by CLOCK (CLK) and CYCLE (CYC) and repressed by PERIOD (PER) and TIMELESS (TIM) [1]. The timing of CLK-CYC activation and PER-TIM repression is regulated posttranslationally, in part through rhythmic phosphorylation of CLK, PER, and TIM [2-4]. Although kinases that control PER and TIM levels and subcellular localization have been identified [5-10], additional kinases are predicted to target PER, TIM, and/or CLK to promote time-specific transcriptional repression. We screened for kinases that alter circadian behavior via clock cell-directed RNA interference (RNAi) and identified the proline-directed kinase nemo (nmo) as a novel component of the circadian oscillator. Both nmo RNAi knockdown and a nmo hypomorphic mutant shorten circadian period, whereas nmo overexpression lengthens circadian period. CLK levels increase when nmo expression is knocked down in clock cells, whereas CLK levels decrease and PER and TIM accumulation are delayed when nmo is overexpressed in clock cells. These data suggest that nmo slows the pace of the circadian oscillator by altering CLK, PER, and TIM expression, thereby contributing to the generation of an ~24 hr circadian period.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , Drosophila Proteins/metabolism , Drosophila/physiology , Gene Expression Regulation/physiology , Mitogen-Activated Protein Kinases/metabolism , ARNTL Transcription Factors/metabolism , Animals , CLOCK Proteins/metabolism , Period Circadian Proteins/metabolism , Phosphorylation , RNA Interference
10.
Mol Cell Biol ; 29(6): 1452-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19139270

ABSTRACT

Circadian clocks keep time via gene expression feedback loops that are controlled by time-of-day-specific changes in the synthesis, activity, and degradation of transcription factors. Within the Drosophila melanogaster circadian clock, DOUBLETIME (DBT) kinase is necessary for the phosphorylation of PERIOD (PER), a transcriptional repressor, and CLOCK (CLK), a transcriptional activator, as CLK-dependent transcription is being repressed. PER- and DBT-containing protein complexes feed back to repress CLK-dependent transcription, but how DBT promotes PER and CLK phosphorylation and how PER and CLK phosphorylation contributes to transcriptional repression have not been defined. Here, we show that DBT catalytic activity is not required for CLK phosphorylation or transcriptional repression and that PER phosphorylation is dispensable for repressing CLK-dependent transcription. These results support a model in which DBT plays a novel noncatalytic role in recruiting additional kinases that phosphorylate CLK, thereby repressing transcription. A similar mechanism likely operates in mammals, given the conserved activities of PER, DBT, and CLK orthologs.


Subject(s)
Casein Kinase 1 epsilon/physiology , Circadian Rhythm/physiology , Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Drosophila/physiology , Transcription Factors/metabolism , Animals , CLOCK Proteins , Drosophila/metabolism , Nuclear Proteins/physiology , Period Circadian Proteins , Phosphorylation , Protein Binding , Transcription, Genetic
11.
Mol Cell Biol ; 27(13): 5014-28, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17452449

ABSTRACT

A common feature of animal circadian clocks is the progressive phosphorylation of PERIOD (PER) proteins from hypo- to hyperphosphorylated species, events that are highly dependent on casein kinase 1 epsilon (termed DOUBLETIME [DBT] in Drosophila melanogaster) and necessary for normal clock progression. Drosophila PER (dPER) functions in the negative limb of the clockworks by presumably binding to the transcription factor CLOCK (CLK) and inhibiting its transactivation activity. Here, we identify a small region on dPER that is conserved with mammalian PERs and contains the major in vivo DBT binding domain, termed dPDBD (for dPER DBT binding domain). This domain is required for the manifestation of molecular and behavioral rhythms in vivo. In the absence of the dPDBD, the dPER protein is present at constant high levels throughout a daily cycle, undergoes little phosphorylation, and is severely impaired in its ability to function as a transcriptional repressor. Our findings indicate that the binding of dPER to CLK is not sufficient for transcriptional inhibition, implicating a more indirect mode of action whereby dPER acts as a molecular bridge to "deliver" DBT and/or other factors that directly repress CLK-dependent gene expression.


Subject(s)
Biological Clocks , Casein Kinase 1 epsilon/metabolism , Circadian Rhythm , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Transcription, Genetic , Amino Acid Sequence , Animals , Cell Cycle Proteins/metabolism , Drosophila melanogaster/physiology , Feedback, Physiological , Models, Biological , Molecular Sequence Data , Motor Activity , Mutant Proteins/metabolism , Period Circadian Proteins , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , Protein Structure, Tertiary , Repressor Proteins/metabolism , Sequence Deletion , Structure-Activity Relationship , Transcriptional Activation/genetics , Ubiquitin-Protein Ligases/metabolism
12.
Methods Mol Biol ; 362: 465-80, 2007.
Article in English | MEDLINE | ID: mdl-17417035

ABSTRACT

Circadian rhythms in metabolic, physiological, and behavioral processes are regulated by biological clocks. Many of these rhythmic processes can be measured over many days or weeks using automated recording devices, thus making it possible to precisely calculate period, phase, and amplitude values. With the advent of luciferase reporter genes and machines capable of quantifying luciferase-generated bioluminescence over long time frames, it is now possible to precisely monitor the rhythms in gene expression that underlie circadian clock function. These assays can be used to monitor gene expression in large numbers of individual plants and animals, and/or various cultured tissues and cells. After acquiring bioluminescence data, rhythm analysis programs are used to calculate the period, phase, amplitude, and overall levels of gene expression for individuals or groups, and to measure their statistical significance. Here we will describe how luciferase assays are performed and analyzed to measure gene expression rhythms in Drosophila.


Subject(s)
Circadian Rhythm/genetics , Circadian Rhythm/physiology , Luciferases, Firefly/genetics , Luciferases, Firefly/metabolism , Animals , Animals, Genetically Modified , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Gene Expression , Genes, Reporter , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Tissue Culture Techniques
14.
Cell ; 125(3): 424-6, 2006 May 05.
Article in English | MEDLINE | ID: mdl-16678086

ABSTRACT

In animals, the circadian timekeeping mechanism relies on the coordinated activities of activators and repressors to control rhythmic transcription. In this issue of Cell, Doi et al. (2006) reveal that rhythms in histone acetylation are necessary for rhythmic transcription and that the histone acetyl transferase responsible is CLOCK, a key transcription factor that is essential for circadian oscillator function.


Subject(s)
Biological Clocks/physiology , Circadian Rhythm/physiology , Drosophila Proteins/genetics , Trans-Activators/physiology , Transcription Factors/genetics , Transcriptional Activation/physiology , ARNTL Transcription Factors , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , CLOCK Proteins , Drosophila Proteins/metabolism , Histone Acetyltransferases/genetics , Histones/metabolism , Humans , Repressor Proteins/genetics
15.
J Biol Rhythms ; 21(2): 93-103, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16603674

ABSTRACT

CLOCK (CLK) is a core component of the transcriptional feedback loops that comprise the circadian timekeeping mechanism in Drosophila. As a heterodimer with CYCLE (CYC), CLK binds E-boxes to activate the transcription of rhythmically expressed genes within and downstream of the circadian clock, but this activation unexpectedly occurs at times when CLK is at its lowest levels on Western blots. Recent studies demonstrate that CLK also regulates nonrhythmic gene expression and behaviors. Despite the critical roles CLK plays within and outside the circadian clock, its spatial expression pattern has not been characterized. Using a newly developed CLK antibody, the authors show that CLK is coexpressed with PERIOD (PER) in canonical oscillator cells throughout the head and body. In contrast to PER, however, the levels of CLK immunoreactivity do not cycle in intensity, CLK is detected primarily in the nucleus throughout the circadian cycle, and CLK is expressed in non-oscillator cells within the lateral and dorsal brain, including Kenyon cells, which mediate various forms of learning and memory. These results indicate that constitutive levels of nuclear CLK regulate rhythmic transcription in circadian oscillator cells and suggest that CLK contributes to other behavioral processes by regulating gene expression in non-oscillator cells.


Subject(s)
Drosophila Proteins/physiology , Gene Expression Regulation , Transcription Factors/physiology , Animals , Blotting, Western , Brain/metabolism , CLOCK Proteins , Cell Nucleus/metabolism , Circadian Rhythm , Drosophila , Drosophila Proteins/metabolism , Immunoblotting , Immunohistochemistry , Microscopy, Confocal , Microscopy, Fluorescence , Models, Biological , Nuclear Proteins/metabolism , Oscillometry , Period Circadian Proteins , Protein Binding , Time Factors , Transcription Factors/metabolism , Transcription, Genetic
16.
Genes Dev ; 20(6): 723-33, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16543224

ABSTRACT

Transcriptional activation by CLOCK-CYCLE (CLK-CYC) heterodimers and repression by PERIOD-TIMELESS (PER-TIM) heterodimers are essential for circadian oscillator function in Drosophila. PER-TIM was previously found to interact with CLK-CYC to repress transcription, and here we show that this interaction inhibits binding of CLK-CYC to E-box regulatory elements in vivo. Coincident with the interaction between PER-TIM and CLK-CYC is the hyperphosphorylation of CLK. This hyperphosphorylation occurs in parallel with the PER-dependent entry of DOUBLE-TIME (DBT) kinase into a complex with CLK-CYC, where DBT destabilizes both CLK and PER. Once PER and CLK are degraded, a novel hypophosphorylated form of CLK accumulates in parallel with E-box binding and transcriptional activation. These studies suggest that PER-dependent rhythms in CLK phosphorylation control rhythms in E-box-dependent transcription and CLK stability, thus linking PER and CLK function during the circadian cycle and distinguishing the transcriptional feedback mechanism in flies from that in mammals.


Subject(s)
Circadian Rhythm , Drosophila Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Base Sequence , CLOCK Proteins , Cells, Cultured , DNA Primers , Drosophila , Phosphorylation , Polymerase Chain Reaction
17.
Biochem Biophys Res Commun ; 290(3): 933-41, 2002 Jan 25.
Article in English | MEDLINE | ID: mdl-11798163

ABSTRACT

Transcriptional regulation appears to be fundamental to circadian oscillations of clock gene expression. These oscillations are believed to control output rhythms. The transcriptional feedback loop and a model of interlocked loops have been proposed as the basis for these oscillations. We characterized the genomic structure of the mouse Bmal1 gene (mBmal1) and defined the mBmal1 promoter region. Transcription of mBmal1 was activated by CRY1, CRY2, and PER2, and was repressed by BMAL1-CLOCK dimers. Therefore, CRY, PER2, and BMAL1-CLOCK play bidirectional roles in transcription when they are at high levels by late day and midnight, respectively. This underlies the opposite phase of BMAL1 compared to CRY and PER. We propose that a BMAL1 negative feedback loop interlocks with the CRY and PER2 negative feedback loop by inter-activation, forming a third positive forward loop. This transcriptional model suggests a molecular basis for the maintenance of stability, persistence, and period of circadian rhythms. The transcriptional potency of CRY is predominant within the mammalian clock, suggesting a clearance mechanism for CRY in period maintenance. (c)2002 Elsevier Science (USA).


Subject(s)
Biological Clocks , Circadian Rhythm , Drosophila Proteins , Eye Proteins , Feedback, Physiological , Flavoproteins/physiology , Nuclear Proteins/physiology , Photoreceptor Cells, Invertebrate , Transcription Factors/genetics , 3T3 Cells , ARNTL Transcription Factors , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors , CLOCK Proteins , Cell Cycle Proteins , Cryptochromes , Down-Regulation , Genes, Reporter , Mice , Models, Genetic , Molecular Sequence Data , Period Circadian Proteins , Promoter Regions, Genetic , RNA, Messenger/biosynthesis , Receptors, G-Protein-Coupled , Trans-Activators/physiology , Transcription Factors/metabolism , Transcription, Genetic , Transcriptional Activation , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...