Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Wangjie Yu is active.

Publication


Featured researches published by Wangjie Yu.


Journal of Cell Science | 2006

Circadian oscillators of Drosophila and mammals

Wangjie Yu; Paul E. Hardin

Animals, plants, fungi and even some prokaryotic organisms display daily rhythms in behavior, physiology, metabolic activity and gene expression. These rhythms are not passively driven by environmental cycles (e.g. light and temperature) but are controlled by endogenous circadian clocks that keep


Molecular and Cellular Biology | 2007

A DOUBLETIME Kinase Binding Domain on the Drosophila PERIOD Protein Is Essential for Its Hyperphosphorylation, Transcriptional Repression, and Circadian Clock Function

Eun Young Kim; Hyuk Wan Ko; Wangjie Yu; Paul E. Hardin; Isaac Edery

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 1ε (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.


Journal of Biological Rhythms | 2006

Drosophila CLOCK Is Constitutively Expressed in Circadian Oscillator and Non-Oscillator Cells

Jerry H. Houl; Wangjie Yu; Scott M. Dudek; Paul E. Hardin

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.


Molecular and Cellular Biology | 2009

DOUBLETIME Plays a Noncatalytic Role To Mediate CLOCK Phosphorylation and Repress CLOCK-Dependent Transcription within the Drosophila Circadian Clock

Wangjie Yu; Hao Zheng; Jeffrey L. Price; Paul E. Hardin

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.


Current Biology | 2011

NEMO kinase contributes to core period determination by slowing the pace of the Drosophila circadian oscillator.

Wangjie Yu; Jerry H. Houl; Paul E. Hardin

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.


Methods of Molecular Biology | 2007

Use of Firefly Luciferase Activity Assays to Monitor Circadian Molecular Rhythms In Vivo and In Vitro

Wangjie Yu; Paul E. Hardin

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.


PLOS Genetics | 2016

CLOCKWORK ORANGE Enhances PERIOD Mediated Rhythms in Transcriptional Repression by Antagonizing E-box Binding by CLOCK-CYCLE

Jian Zhou; Wangjie Yu; Paul E. Hardin

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.


Methods in Enzymology | 2015

ChIPping away at the Drosophila clock.

Jian Zhou; Wangjie Yu; Paul E. Hardin

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.


Proceedings of the National Academy of Sciences of the United States of America | 2017

CLOCK stabilizes CYCLE to initiate clock function in Drosophila

Tianxin Liu; Guruswamy Mahesh; Wangjie Yu; Paul E. Hardin

Significance The Drosophila circadian clock controls daily rhythms in physiology, metabolism, and behavior via ∼24-h transcriptional feedback loops. CLOCK requires its heterodimeric partner CYCLE to initiate clock function in canonical groups of brain neurons and peripheral tissues, but Clock expression can also induce clocks in ectopic locations. Here, we show that CLOCK stabilizes CYCLE in canonical and ectopic clock cells, where CYCLE is normally rapidly degraded, and that ectopic clocks also require CRYPTOCHROME. This work defines the genetic architecture and molecular mechanisms required for clock initiation that are likely conserved in other eukaryotes and suggests that ectopic clocks can be generated by Clock, cycle, and cryptochrome expression in naive cells. 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.


Genes & Development | 2006

PER-dependent rhythms in CLK phosphorylation and E-box binding regulate circadian transcription

Wangjie Yu; Hao Zheng; Jerry H. Houl; Brigitte Dauwalder; Paul E. Hardin

Collaboration


Dive into the Wangjie Yu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hao Zheng

University of Houston

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Isaac Edery

Center for Advanced Biotechnology and Medicine

View shared research outputs
Top Co-Authors

Avatar

Jeffrey L. Price

University of Missouri–Kansas City

View shared research outputs
Top Co-Authors

Avatar

Scott M. Dudek

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge