timeless REGULATION Protein Interactions and Post-transcriptional 
Regulationtimeless was cloned by chromosomal walking and subsequently used, 
in yeast, to identify Per as a physical partner. Timeless and Period 
interact, and both are required for production of circadian rhythms. The 
tim gene encodes a protein of 1389 amino acids, and possibly another 
protein of 1122 amino acids. The arrhythmic mutation tim01 is a 64-base 
pair deletion that truncates TIM to 749 amino acids. Absence of sequence 
similarity to the Per dimerization motif (PAS) indicates that direct 
interaction between Per andTim would require a heterotypic protein 
association (Meyers, 1995).Tim was isolated based on its ability to 
physically interact with the Per protein. A restricted segment of Tim binds 
directly to PAS, a part of the Per dimerization domain. PerL, a mutation in 
Per that causes a temperature-sensitive lengthening of circadian period and 
a temperature-sensitive delay in Per nuclear entry, exhibits a 
temperature-sensitive defect in binding to Tim (Gekakis, 1995).Tim and Per 
accumulate in the cytoplasm when independently expressed in cultured (S2) 
Drosophila cells. If coexpressed, however, the proteins move to the nuclei 
of these cells. Domains of Per and Tim have been identified that block 
nuclear localization of the monomeric proteins. These regions of Per and 
Tim interaction consist of the PAS domain of Per and an adjacent domain 
also required for cytoplasmic localization (CLD). The sequence of Tim 
involved in interaction with Per resides between amino acids 505 to 578. 
Tim and Per both contain domains required for cytoplasmic localization. The 
site in Per required for nuclear localization is a sequence between amino 
acids 453 and 511. The sequence of Tim required for cytoplasmic 
localization (the Tim CLD) is C-terminal. It is thought that the CLD 
interacts with a cytoplasmic factor that inhibits nuclear localization. The 
results indicate a mechanism for controlled nuclear localization in which 
suppression of cytoplasmic localization is accomplished by direct 
interaction of Per and Tim. No other clock functions are required for 
nuclear localization. The findings suggest that a checkpoint in the 
circadian cycle is established by requiring cytoplasmic assembly of a 
Per/Tim complex as a condition for nuclear transport of either protein 
(Saez, 1996). To investigate the mechanism of phase shifing of circadian 
clocks by light stimulation, the effects of light pulses on the protein and 
messenger RNA products of the Drosophilaclock gene period (per) were 
measured. Photic stimuli perturb the timing of the Per protein andmessenger 
RNA cycles in a manner consistent with the direction and magnitude of the 
phase shift.The recently identified clock protein Timeless interacts with 
Per in vivo, andthis association is rapidly decreased by light. This 
disruption of the Per-Tim complex in thecytoplasm is accompanied by a delay 
in Per phosphorylation and nuclear entry and disruption inthe nucleus by an 
advance in Per phosphorylation and disappearance. These results suggest 
amechanism for how a unidirectional environmental signal elicits a 
bidirectional clock response (Lee, 1996). Many circadian features of the 
Tim cycle resemble those of the Per cycle. However, Tim israpidly degraded 
in the early morning or in response to light, releasing Per from the 
complex. ThePer-Tim complex is a functional unit of the Drosophila 
circadian clock, and Tim degradation maybe the initial response of the 
clock to light (Zeng, 1996). Drosophila Clock protein (dClock) is a 
transcription factor that is required for the expression of the 
circadianclock genes period (per) and timeless (tim). dClock undergoes 
circadian fluctuations in abundance, is phosphorylated throughout a daily 
cycle, andinteracts with Per, Tim, and/or the Per-Tim complex during the 
night but not during most of the day. Both Per and Tim copurify with dClock 
in a time-of-day-specific manner: Per and Timare first detected at ZT12 
(beginning of the dark period), followed by increases in amounts that reach 
peak values at ZT23.9 (just before the lights go on). Between ZT16 (a third 
of the way through lights off) and ZT23.9, the amounts of all three 
proteins in immune complexes increase, even though the totallevels of Tim 
and Per in head extracts peak at ZT16 and ZT20, respectively. This suggests 
that during thenight dClock is present in limiting amounts compared to Per 
and Tim. Despite the higher levels ofimmunoprecipitated dClock between ZT4 
and ZT8 compared to values obtained between ZT12 and ZT16, very little, 
ifany, Per and Tim are detected. A likely explanation forthis is that 
between ZT4 and ZT8 the total levels of Per and especially those of Tim are 
at, or close to, trough values. Thus, the interaction of Per and Tim with 
dClock is mainly restricted to nighttime hours (Lee, 1998). Analysis of 
immune complexes derived from a period mutant clearly indicate that in the 
absence of Per, Tim canstill interact with dClock. Because Tim is 
apparently located exclusively in thecytoplasm in the absence of Per, this 
result could suggest that thenuclear localization of dClock also requires 
Per or a functional oscillator. Alternatively, low levels of Tim might 
beable to enter the nucleus in the absence of Per. In contrast, several 
attempts to visualize a specific interaction betweenPer and dClock in the 
absence of Tim were unsuccessful. There are atleast two nonmutually 
exclusive reasons that might account for thr inability to detect Per in 
dClock-containingimmune complexes prepared from tim mutant flies: (1) the 
levels of Per are very low in tim mutant flies and as such the amounts of 
Per that copurify with dClock are below thedetection limit, and (2) the 
interaction of Per with dClock requires Tim, possibly via formation of the 
Per-Timcomplex and/or a dependence for nuclear localization (Lee, 1998 and 
references).Attempts were made to measure the relative amounts of dClock 
that interact with Per and Tim as a function of time in anLD cycle. Head 
extracts were incubated with antibodies against either Per or Tim, and 
immune complexes probed fordClock, Per, and Tim. At ZT20 almost identical 
levels of dClock copurify with antibodies directedagainst either Per or 
Tim. Equivalent amounts of Per were also present in both immunepellets, but 
1.6-fold more Tim is immunoprecipitated with antibodies toTim, as compared 
to those directed against Per. These results are almost identicalwith a 
previous study showing that (1) in head extracts prepared from flies 
collected at ZT20, 80% of thetotal amount of Per is bound to Tim in a 1:1 
stoichiometric relationship, and (2) there is 1.5-1.8 times more Tim, as 
compared to Per. Thus, the current results suggest that at ZT20 the 
majority of the Per and Timproteins that interact with dClock are in the 
form of a heterodimeric Per-Tim complex. During the early day, only 
lowlevels of dClock are detected in immune complexes obtained using either 
antibodies to Per or Tim, in agreement with results using anti-dClock 
antibodies. Furthermore,it is mainly versions of Per and Tim that are 
essentially free of one another that interact with dClock during the 
earlyday (Lee, 1998). How might a trimeric complex containing Per, Tim, and 
dClock be assembled? Presumably the HLH domain ofdClock does not 
participate in mediating protein-protein interactions in this putative 
trimeric complex, because neitherPer nor Tim seems to have a similar 
dimerization region. The only other regions that have been shown to 
mediateprotein-protein interactions are the PAS domain found in Per and 
dClock and a not so well characterized region in Tim thatspans 400 amino 
acids and interacts with the PAS domain of Per. It is tempting tospeculate 
that one or both of these domains has the capacity to engage in at least 
trimeric formation. Although these studiesdo not address the nature of the 
trimeric interaction, they indicate that PAS-containing proteins are not 
limited to binary interactions (Lee, 1998). These results suggest that Per 
and Tim participate intranscriptional autoinhibition by physically 
interacting with dClock or a dClock-containing complex. Nevertheless, in 
the absence of Per or Tim, thelevels of dClock are constitutively low, 
indicating that Per and Tim also act as positive elements in the feedback 
loop by stimulating the production ofdClock. Although Per and Tim inhibit 
dClock activity, Per and Tim arerequired for the high-level production of 
dClock protein and mRNA. Thus, Per andTim appear to be the main "motor" of 
the Drosophila circadian oscillator, driving both positive and negative 
elements ofthe transcriptional-translational feedback loop. These 
observations suggest an explanation for the previously unexplainedfinding 
that the levels of Per mRNA in per mutant flies are approximately half as 
high as those obtained at peak times inwild-type flies. In contrast, 
mutations that abolish Neurospora FRQ activity result in high levels of 
frqRNA, suggesting that the frq-based circadian oscillator in Neurospora is 
based on a more simple negativetranscriptional feedback loop. How Per and 
Tim stimulate dClock expression is not clear.They may interact with other 
transcription factors and act as coactivators. Alternatively, they may 
block the function ofnegative factors leading to the stimulation of gene 
expression. In addition to regulating the transcriptional activity of 
thedClock-CYC complex, Per and Tim might also interact with other 
transcription factors that are not involved in thecircadian oscillator and 
as such molecularly couple the timekeeping mechanism to downstream effector 
pathways (Lee, 1998). The cyclic expression of the Period (Per) and 
Timeless (Tim)proteins is critical for the molecular circadian feedback 
loop inDrosophila. The entrainment by light of the circadian clock 
ismediated by a reduction in Tim levels. To elucidate the mechanism ofthis 
process, the sensitivity of Tim regulation by light was testedin an in 
vitro assay with inhibitors of candidate proteolyticpathways. The data 
suggest that Tim is degraded through aubiquitin-proteasome mechanism. In 
addition, in cultures fromthird-instar larvae, Tim degradation is blocked 
specifically byinhibitors of proteasome activity. Degradation appears to 
bepreceded by tyrosine phosphorylation. Finally, Tim is ubiquitinatedin 
response to light in cultured cells (Naidoo, 1999).An in vitro assay was 
developed to investigatethe nature of the Tim light response.Flies were 
entrained to a 12 hour light/12 hour dark cycle andprepared head extracts 
from flies collected at either ZT (zeitgebertime) 20 or immediately after a 
1-hour light pulse delivered atZT 19 (ZT0 = lights on; ZT12 = lights 
off).These extracts were incubated with Tim protein immunoprecipitatedfrom 
fly heads. After a 1-hour incubation at room temperature, Tim levels were 
assayed by protein immunoblots. Addition of the pulsedextract reduces the 
Tim signal. Unpulsedhead extract has no effect on the level of Tim, 
indicating that thereduction is light-specific. This light-induced 
reduction is alsoobserved in tim0 flies (which lack Tim protein) andis, in 
fact, routinely higher in these flies, which may suggest 
somedown-regulation by the clock in wild-type flies. Animmunoprecipitated 
Per substrate is not degraded by addition of thepulsed extract (Naidoo, 
1999).In order to determine the nature of the proteolytic activity, several 
general classes of protease inhibitors were assayed. Inhibitors ofserine 
proteases [phenylmethylsulfonyl fluoride (PMSF) and aprotinin]and aspartate 
proteases (pepstatin) are not very effective inblocking Tim degradation. 
However,degradation is inhibited by the proteasomal 
inhibitorsacetyl-leu-leu-norleucinal (ALLN), cbz-leu-leu-norvalinal(ZL2NVaH 
or MG115) and cbz-leu-leu-leucinal(ZL3H or MG132). These peptidealdehydes 
strongly inhibit the chymotryptic activity of theeukaryotic 26S proteasome. 
Timdegradation is also blocked by bestatin, a metalloproteaseinhibitor, and 
by leupeptin, which inhibits cysteine proteases andhas some effects on 
other proteolytic systems, including theproteasome. The precise mechanism 
of actionin this case is not known. Consistent with a role for the 
proteasome,depletion of ubiquitin from the extract blocks Timdegradation 
(Naidoo, 1999). Although the in vitroassay indicates a mechanism for Tim's 
response to light, itsusefulness is limited by its variability.To verify 
the findings of this assay, an in vivosystem was developed. Thus, a primary 
culture assay was used to test the effect of two proteasomal inhibitors 
(lactacystinand MG115) on the Tim light response. Lactacystin, a 
microbialmetabolite, is the most specific one known; a naturally occuring 
inhibitorof the proteasome. It spontaneouslyhydrolyzes into 
clastolactacystin B lactone, which is the activespecies that reacts with 
the proteasome, inhibiting its chymotrypticand tryptic peptidase-like 
activity. MG115 is a potent synthetic peptide aldehydeinhibitor. For the 
assay, the centralnervous system (CNS) of third-instar larvae was dissected 
and maintained in culture medium for1 hour. Some samples were exposed to a 
pulse of light for20 min and were fixed at the end of the hour. Dark 
controlsamples were also incubated for an hour in the dark. Tim expression 
was then examined in thelateral neurons (clock cells), which were located 
by costaining withan antibody to pigment-dispersing hormone. Strong Tim 
staining is seen in lateral neurons ofunpulsed tissue, but little to no Tim 
in CNS tissue that has receiveda light pulse. The effect of inhibitors was 
tested by adding them to theculture medium at the start of the incubation. 
Tissue treated withlactacystin and MG115 before the light pulse revealed 
robust Timstaining in the lateral neurons. The strong inhibition by MG115 
isconsistent with a report that this is a much more effective inhibitorof 
proteolysis in intact cells than it is of in vitro hydrolysis 
ofmacromolecular substrates. The 100%block by lactacystin may reflect 
variable permeability or instabilityof the lactone metabolite (Naidoo, 
1999). Proteasomes are multicatalytic, multisubunit proteolytic 
complexeswith highly conserved structures; they play a key role in avariety 
of cellular processes, including the cell cycle,transcriptional regulation, 
removal of abnormal proteins from thecell, antigen presentation, and even 
in theturnover of a mammalian circadian-regulated protein. The Tim response 
to light is blockedspecifically, in two different assays, by several 
inhibitors of theproteasome; this is important, given that lactacystin, 
which wasthought to affect only the proteasome, has been shown to also act 
on a second multisubunit enzyme. Because the newly identified enzyme 
isinsensitive to ALLN, it cannot account for the Tim response. For the 
ubiquitin-proteasome system, prolineglutamate serine threonine (PEST) 
regions sometimes serve as putativedegradation/phosphorylation signals in 
the target molecule. The Tim protein sequence reveals the presenceof seven 
PEST regions concentrated near the NH2 and COOHtermini (Naidoo, 1999). Most 
cellular proteins that are degraded by theproteasome are ubiquitinated and 
then targeted to the proteasome. To determine whether Tim is 
ubiquitinated,which would also demonstrate that it is a direct target of 
theproteasome, a cell culture system was used. Tim and ahemagglutinin 
(HA)-tagged ubiquitin octamer were expressed under heat shock control in 
Drosophila S2 cells. After a30-min heat shock, cells were either maintained 
in the dark ortreated with light for 2 hours, after which the cells were 
lysedand immunoprecipitates of Tim were probed with an antibody to HA. Tim 
was found to be ubiquitinated in response to light. The effect is specific 
for Tim, because Peris not ubiquitinated with or without light treatment. 
Extended light treatment also degrades Tim inthese cells, and this 
degradation is inhibited by the proteasomeinhibitor MG115. Although these 
data implicate a ubiquitin-proteasomal mechanism,they do not preclude a 
role for other proteolyic systems (Naidoo, 1999). To investigatea possible 
role for phosphorylation in the degradation of Tim, the effect of several 
kinase inhibitors in the in vivoprimary culture assay were examined. The 
tyrosine kinase inhibitor genisteinblocks the degradation of Tim in the 
lateral neurons after a pulseof light, whereas the serine-threonine 
inhibitors staurosporin andcalphostin C and the MEK inhibitor PD98059 do 
not. These results suggest that tyrosine kinaseactivity precedes 
degradation of Tim. The concentrations of genisteinthat were effective in 
this assay suggest ac-src-like kinase activity, althoughthe concentration 
dependence must be interpreted with caution,because it could be a measure 
of permeability or drug stability (Naidoo, 1999).To determine whether the 
tyrosine phosphorylation occurs on Timitself, protein immunoblots of Tim 
immunoprecipitates were probed withan antibody to phosphotyrosine. After 20 
min of light treatmentat ZT19, Tim could be detected with the antibody 
tophosphotyrosine. Tim in the 'dark'samples is sometimes detected with this 
antibody but notconsistently, which suggests that tyrosine phosphorylation 
of Tim isincreased by light. The mobility of the Tim band in the 
light-treatedsample is also reduced, presumably because of phosphorylation 
(Naidoo, 1999). Together, these data indicate that the Tim response to 
lightinvolves tyrosine phosphorylation and ubiquitination, followed 
byproteasomal degradation. What then is the role of the proteasome pathway 
infree-running behavioral rhythms? Are the mechanisms that degrade Timin 
response to light the same as those that degrade it in constantdarkness? If 
this is the case, light may serve only to further activate aprocess that is 
already under way. It is proposed that cyclic turnover ofTim under 
free-running conditions is mediated by phosphorylation,which targets it for 
degradation, perhaps by the proteasome. Tim isprogressively phosphorylated 
throughout the night, and maximallyphosphorylated forms are found just 
before the rapid decline ofprotein levels. From this point on, untilthe 
middle of the day, Tim levels remain low because of the lowlevels of RNA. 
As the repression of transcription is released, mostlikely because of the 
decrease in Per levels, RNA accumulates andprotein also starts to 
accumulate, albeit slowly, because it is stillsubject to phosphorylation 
and degradation. When the rate of Timsynthesis exceeds the rate of 
phosphorylation/degradation, higherlevels of protein are observed, but as 
the phosphorylation programcontinues and RNA levels are reduced (because of 
negative feedback),levels of the protein drop off. Light could enhance Tim 
degradationby increasing Tim phosphorylation and/or by increasing 
proteolytic activityin some manner. This model would predict that the 
presenceof light accelerates the falling phase of the protein and delays 
therising phase, both through the same mechanism (Naidoo, 
1999).Phosphorylation is an important feature of pacemaker organization in 
Drosophila. Genetic and biochemical evidence suggestsinvolvement of the 
casein kinase I homolog doubletime (dbt) in the Drosophila circadian 
pacemaker. Two novel dbt mutants have been characterized. Both cause a 
lengthening of behavioral period and profoundly alter period (per) and 
timeless (tim)transcript and protein profiles. The Per profile shows a 
major difference from the wild-type program only during the morninghours, 
consistent with a prominent role for Dbt during the Per monomer degradation 
phase. The transcript profiles are delayed,but there is little effect on 
the protein accumulation profiles, resulting in the elimination of the 
characteristic lag between the mRNA and protein profiles. These resultsand 
others indicate that light and post-transcriptional regulation play major 
roles in defining the temporal properties of the protein curves and suggest 
that this lag isunnecessary for the feedback regulation of per and tim 
protein on per and tim transcription (Suri, 2000). Bothmutations, when 
presented in the context of the highly similar yeastcasein kinase I HRR25, 
severely reduce kinase activity on peptidesubstrates. The long-period 
phenotypes are likely caused byinsufficient Dbt activity, so it takes 
longer to reach some requiredlevel of Per phosphorylation. It is also 
assumed that both mutants areexpressed at a level similar to that of 
wild-type Dbt (Suri, 2000).Both dbth andDbtg/+ have 29 hr periods and 
aresimilar in all other respects, suggesting that the phenotypes are 
notidiosyncratic features of the mutations but reflect the role of Dbt 
inthe pacemaker. Although the mutant flies entrain to imposed 24 
hrphotoperiods, the LD locomotor activity patterns indicate that there isno 
anticipation of the morning or evening light/dark transitions, andthe 
evening activity peak is delayed by several hours into the night.The 
altered LD patterns are probably a consequence of the longerperiods. 
Indeed, flies that carry pers as well asdbth have a period of 22.5 hr 
andmanifest robust anticipation of both morning and evening transitions 
aswell as an advanced evening activity peak. Both dbt mutantLD profiles 
resemble that of the 29 hr periodperl mutant strain, consistent withthis 
altered period notion (Suri, 2000).The molecular features of the 
perlcircadian program are difficult to compare with those of 
wild-typeflies, because the mutant rhythms are weak and of low amplitude as 
wellas long period even under 12 hr LD entraining conditions. In contrast, 
Per and Tim cyclingin the long-period dbt mutants is robust. Protein levels 
arecomparable with those in wild-type flies during the night, and levelsin 
the two mutant strains appear even higher than wild-type levelsduring the 
daytime. Previous work suggests a role forDbt-catalyzed phosphorylation in 
targeting Per for degradation: thisprobably reflects slower protein 
turnover during the morning in thedbt mutants. The Tim phosphorylation 
pattern in the mutantsdid not show any noticeable difference from the 
wild-type pattern.These observations suggest that the modest mutant effects 
on the Tim profiles are indirect, perhaps through a primary effect of the 
dbt mutants on Per (Suri, 2000).Per phosphorylation is still readily 
observable in both mutant lines.In fact, there is a hint that Per is even 
hyperphosporylated in thesestrains. Although this might reflect 
phosphorylation events that nevertake place in a wild-type background, less 
active Dbt mutants might beexpected to depress the magnitude as well as the 
kinetics of thetemporal phosphorylation program. This suggests that Per 
might not be adirect Dbt substrate in vivo but is only 
influencedindirectly, through intermediates that are direct Dbt targets. 
Forexample, Dbt may phosphorylate and activate a direct Per kinase or 
aspecific protease. In this context, Per has not yet been shown to be 
adirect Dbt substrate. It is also possible that Dbt is a 
functionallyrelevant but minor Per kinase. In this case, the bulk of the 
Permobility shift on SDS-PAGE is a consequence of other kinases. BecausePer 
persists for several hours longer in the mutants than in wild-typeflies, 
the other kinases would continue to function and give rise toeven more 
highly phosphorylated species than are usually observed.These would be an 
indirect consequence of weak dbt activityand delayed degradation. A final 
possibility is that the enhanced anddelayed Per phosphorylation simply 
reflects some misregulation of Dbt activity (Suri, 2000). Careful analysis 
of the Per and Tim protein profiles in the long-perioddbt mutants suggests 
that Dbt acts in the late night andmorning phase of the molecular cycle: 
the mutants leave the earlyevening protein profile almost unaltered. This 
indicates thatdbt probably targets nuclear, monomeric Per. It hasalso been 
suggested that Dbt acts in the early night to destabilize cytoplasmic Per, 
thus delaying nuclear entry and repression. The dbt mutants reportedhere do 
not significantly change this early night, presumptivecytoplasmic phase of 
accumulation. It is possible that Dbt prefers freePer over Per complexed to 
Tim. If free Per is a better substrate, thenDbt mutants should show a 
greater effect in the late night and earlymorning, after a large fraction 
of Tim has disappeared. Alternatively,Dbt might influence only marginally 
the Per accumulation phase for someother reason. But dbt mutant larvae 
accumulate high levelsof hypophosphorylated Per, which suggests that Dbt is 
the major Perkinase and strongly influences Per accumulation as well as 
degradation. There is evidence, however,that much of this Per accumulation 
occurs in cells and tissues where Per is not normally detectable, making 
the connection with the normalPer-Tim cycle uncertain (Suri, 2000).To 
assess the effect of the dbt mutants on transcription, per and tim mRNA 
cycling was assayed in wild-type anddbt mutant flies. Both mutant profiles 
are delayed by 4-5hr. This is presumably because of the delayed 
disappearance of Per aswell as Tim, which has been suggested to repress per 
andtim transcription. This relationship is very similar to thatpreviously 
reported for the perSmutant strain; in this case, the clock proteins 
disappear more quickly,leading to an advance in the RNA profiles. The 
perSeffect is more pronounced on Per than on Tim, consistent with thenotion 
that monomeric Per might be the major transcriptional repressor. In any 
case, comparable results in the threemutants indicate a solid relationship 
between the timing of the declinein protein levels and the timing of the 
subsequent increase inper and tim transcription (Suri, 2000).Based on these 
observations, a possible model for Dbt function in the Drosophila pacemaker 
is presented. In the cytoplasm, normal destabilization of Per delays 
substantialbuildup of Per-Tim complexes and the consequent nuclear 
transport of the dimeric Per-Tim complex. In the nucleus, Per 
destabilizationrelieves repression. In Dbt mutants, Per degradation is much 
slower.This prolongs repression and delays the per andtim mRNA upswing in 
the next cycle (Suri, 2000). There is an impressive relationship between 
the per andtim RNA profiles in comparison to the evening locomotoractivity 
peak. In all cases, these RNA and locomotor activity begin to increase 
atapproximately the same time, i.e., around ZT7 in the middle of 
thedaytime. Mutants or physiological manipulations that affect the timing 
of the RNA profiles affect the timing of the evening activity peak 
inparallel. This fits withthe emerging view, from mammalian as well as 
Drosophila work, that cycling transcription plays an important role 
incircadian output as well as within the central pacemaker oscillator. A 
further implication of theserelationships is that the protein oscillations 
from one day affectbehavior as well as the RNA profiles on the next one: 
the morningdecline and eventual disappearance of Per and Tim terminate a 
proteincycle from the previous day, which then causes the subsequent 
increasesin both RNA levels and locomotor activity (Suri, 2000). In 
contrast, the delayed Per and Tim disappearance in the mutants haslittle if 
any effect on the subsequent protein accumulation phase(ZT13-ZT20) under 
these standard LD conditions; it is hardly affected, and both proteins peak 
at approximately the same time asthey do in the wild-type flies 
(ZT19-ZT21). Because of the delayed RNArise in the mutants, the per and tim 
RNAaccumulation profiles almost coincide with those of the proteins,between 
ZT15 and ZT21. This indicates that the timing of the RNA riseis 
insufficient to time the protein rise. The increase in proteinlevels may 
reflect protein half-life regulation, which is uncoupledfrom the underlying 
mRNA levels, at least under some circumstances (Suri, 2000). The 
coincidence of the protein and RNA curves also raises doubts aboutthe 
importance of the 4-6 hr lag between these two accumulationprofiles. The 
data presented in this study indicate that the lag is dispensable forrobust 
behavioral and molecular oscillations. This is especiallyrelevant for the 
RNA fluctuations. Despite evidence that at leastper mRNA fluctuations may 
not be necessary for coreoscillator function, theynormally correlate with 
other molecular and behavioral circadianfluctuations. Moreover, there are 
substantial data indicating that Perand Tim feedback regulate these 
transcriptional oscillations. There is also considerable experimental 
evidence as well as theoretical models, to suggest that thenormal 4-6 hr 
lag between the RNA and protein curves is essential forgenerating these 
robust, high-amplitude transcriptional oscillations. The general view 
isthat the protein accumulation delay gives enough time for transcriptionto 
increase substantially, before protein levels have increasedsufficiently to 
inhibit transcription. The presence ofrobust transcriptional oscillations 
without the delayed proteinaccumulation makes this scheme less likely. It 
redirects focus towardsome post-transcriptional delay (e.g., the timing of 
nuclear entry ofthe Per-Tim dimer), which is predicted to be functional and 
important fortranscriptional feedback regulation. It is important to note 
that theseconclusions are based on biochemical experiments with 
whole-headextracts. It is still possible that the mRNA-protein lag may 
beimportant in the specific pacemaker neurons of Drosophila (Suri, 
2000).All of these experiments were performed under LD conditions. When 
thelight comes on at ZT24, it causes a rapid decline in Tim levels. In 
DDconditions, therefore, Tim levels are much higher in the earlysubjective 
day, as expected. But a major, unanticipated difference wasthat the Per and 
Tim profiles in the dbt mutant flies areprofoundly delayed in DD, as 
evidenced by the late appearance offaster-migrating species. This occurs 
without a comparable change inthe RNA profiles, giving rise to a 
quasi-normal lag between RNA andprotein. The light-mediated advance of the 
protein curves and theabsence of a comparable light reset of the RNA 
profile reinforce theindependent regulation of the accumulation phase of 
the clockRNAs and proteins: only the RNA profiles are influenced bythe 
declining phase of the protein cycle of the previous day, whereasonly the 
protein profiles appear to be reset by the light entrainmentstimulus. The 
data are therefore consistent with a post-translationalroute of light 
entrainment, perhaps mediated by some aspect of thenormal light effect on 
Tim. This presumably contributes tothe daily advance of the dbt mutant 
clock under LDconditions, which counteracts the 5 hr period-lengthening 
effect thatwould take place under DD conditions (Suri, 2000). Further 
understanding of the role of Dbt in the clock will requireexperiments that 
directly address Dbt function and regulation. Forexample, it is possible 
that temporal regulation of Dbt activity makesa major contribution to the 
temporal phosphorylation profile and moregenerally to the normal timing of 
the circadian program. Additionally,the extent to which Dbt modifies other 
pacemaker proteins is not clear.It is possible that these other putative 
Dbt substrates may also beintimately connected to the pacemaker mechanism. 
Addressing theseissues would provide a much deeper understanding of the 
role ofphosphorylation in the pacemaker (Suri, 2000).Tissue-specific 
overexpression of the glycogen synthase kinase-3 (GSK-3) ortholog shaggy 
(sgg) shortens the period of the Drosophila circadian locomotor activity 
cycle. The short period phenotype has been attributed to premature nuclear 
translocation of the Period/Timeless heterodimer. Reducing Sgg/GSK-3 
activity lengthens period, demonstrating an intrinsic role for the kinase 
in circadian rhythmicity. Lowered sgg activity decreases Timeless 
phosphorylation, and GSK-3ß specifically phosphorylates Timeless in vitro. 
Overexpression of sgg in vivo converts hypophosphorylated Timeless to a 
hyperphosphorylated protein whose electrophoretic mobility, and light and 
phosphatase sensitivity, are indistinguishable from the rhythmically 
produced hyperphosphorylated Timeless of wild-type flies. These results 
indicate a role for Sgg/GSK-3 in Timeless phosphorylation and in the 
regulated nuclear translocation of the Period/Timeless heterodimer 
(Martinek, 2001).Two independent lines of evidence suggest that sgg 
regulates the period of molecular cycling primarily through effects on 
nuclear translocation of the Per/Tim heterodimer: (1) the transition point 
between delays and advances of the phase response curve, an indicator for 
nuclear entry of Per/Tim complexes, is advanced by 3 hr in flies 
overexpressing sgg; (2) nuclear Per is detected 2 hr earlier in the lateral 
neurons of larvae overexpressing sgg than in wild-type LNs (Martinek, 
2001).sgg-induced shifts in the timing of nuclear translocation are likely 
to reflect changes in Tim phosphorylation that are in turn connected to 
altered levels of Per and Tim. Because Per and Tim are overproduced when 
sgg activity is low, it is suggested that sgg-dependent Tim phosphorylation 
accelerates Per/Tim heterodimerization or directly promotes nuclear 
translocation of Per/Tim complexes in wild-type flies. In this view, 
decreased Tim phosphorylation in sgg mutants would tend to retard nuclear 
transfer, and so require higher concentrations of the Per and Tim proteins 
at times of nuclear entry (Martinek, 2001).\n\nTim can be directly 
phosphorylated by GSK-3ß in vitro. Such experiments suggest a mechanism 
involving direct interaction of Sgg/GSK-3 and Tim in vivo, but do not 
exclude indirect regulation of Tim phosphorylation by this enzyme in the 
fly. Nor do these results rule out the involvement of additional protein 
kinases. For example, a tyrosine-linked phosphorylation of Tim has been 
implicated in the degradation of Tim by the proteasome. Because Sgg would 
not be expected to promote tyrosine phosphorylation, this kinase should not 
regulate all aspects of Tim function (Martinek, 2001).Sgg/GSK-3 is well 
known for its central role in Wingless/Wnt signaling. Surprisingly, recent 
work has indicated that the vertebrate ortholog of Double-time, casein 
kinase Iepsilon, may also participate in this developmental pathway. For 
example, in Xenopus, inhibition of casein kinase Iepsilon produces 
developmental abnormalities closely corresponding to a loss of Wnt 
function. Casein kinase Iepsilon stabilizes ß-catenin and binds and 
phosphorylates Dishevelled, both established components of the Wnt signal 
transduction pathway. It is remarkable that two kinases that function 
together to provide specific developmental regulation may both act as 
controlling elements in a patently unrelated behavioral process. This could 
reflect an underlying synergism between Sgg/GSK-3 and casein kinase 
1epsilon. Certainly the activities of both kinases must be integrated at 
some level for coherent transduction of Wnt signals. Because Dbt and Sgg 
appear to produce opposing effects on Per/Tim nuclear transfer, with Dbt 
retarding transfer and Sgg accelerating the process, the relative 
activities of these kinases could establish an important focus for 
stabilizing the period of Drosophila's circadian rhythms. For example, a 
control point composed of offsetting kinase activities might contribute to 
such homeostatic mechanisms as temperature compensation of the clock. In 
preliminary work, the effects on circadian rhythmicity of two other 
elements of the wg signal transduction pathway were examined. A 
temperature-sensitive allele of wg fails to show any effect on rhythmic 
locomotor activity, and a heat shock-dishevelled-rescued dsh mutant 
produces no circadian abnormalities. Thus, sgg's participation in the 
circadian oscillator may be unrelated to its function in wg signaling 
(Martinek, 2001).The clock gene double-time (dbt) encodes an ortholog of 
casein kinase Iepsilon that promotes phosphorylation and turnover of the 
Period protein. Whereas the period, timeless, and Clock genes of Drosophila 
each contribute cycling mRNA and protein to a circadian clock, dbt RNA and 
Dbt protein are constitutively expressed. Robust circadian changes in Dbt 
subcellular localization are nevertheless observed in clock-containing 
cells of the fly head. These localization rhythms accompany formation of 
protein complexes that include Per, Tim, and Dbt, and reflect periodic 
redistribution between the nucleus and the cytoplasm. Nuclear 
phosphorylation of Per is strongly enhanced when Tim is removed from 
Per/Tim/Ddt complexes. The varying associations of Per, Ddt and Tim appear 
to determine the onset and duration of nuclear Per function within the 
Drosophila clock (Kloss, 2001).Dbt RNA levels are constant throughout the 
day. In this respect, the same is true for Dbt protein levels, since there 
was no detectable circadian oscillation of Dbt accumulation in timed head 
extracts. Furthermore, a variety of mutations disrupting the circadian 
clock and molecular oscillations have no effect on the level of Dbt 
protein. Thus, production of Dbt protein is not under the control of clock 
genes. In contrast, the subcellular localization of Dbt in the lateral 
neurons and photoreceptor cells changes over the course of a daily cycle. 
Dbt is consistently detected in the nucleus. However, at the end of the day 
and in the early part of the night, a substantial increase is found in 
cytoplasmic Dbt, coincident with the cytoplasmic accumulation of Per 
proteins and Per/Tim complexes. Furthermore, when Per/Tim complexes 
translocate to the nucleus at ZT18, and early during the day when Per 
remains in the nucleus in absence of Tim, a substantial nuclear 
accumulation of Dbt is observed. These changes in subcellular location of 
Dbt appear to be influenced exclusively by the locus of Per accumulation 
(in the presence or absence of Tim). Tim protein has little or no effect on 
the localization of Dbt because Dbt is always detected in the nucleus in 
per01 flies, which lack Per and have a substantial amount of Tim in the 
cytoplasm. Consequently, there is circadian regulation of Dbt proteins, in 
the form of a changing subcellular distribution. The fact that the movement 
of Per and Tim from the cytoplasm to the nucleus predicts the distribution 
of Dbt implies a close correspondence between maximum levels of Per/Tim 
complex and cytoplasmic levels of Dbt. Such a relationship could indicate 
that Tim associates with cytoplasmic Per once the latter protein has 
effected cytoplasmic localization of most cellular Dbt (Kloss, 
2001).Because Dbt preferentially accumulates in nuclei in the absence of 
Per, cytoplasmic Per proteins must affect this default localization at 
certain times of day in wild-type flies. Although the half-life of Dbt has 
not been determined, Dbt RNA and proteins are constantly synthesized. 
Therefore, the subcellular fate of newly translated Dbt may simply depend 
on whether cytoplasmic Per is available to associate with Dbt and retard 
its nuclear translocation. Alternatively, accumulation of Dbt may involve 
mechanisms promoting both nuclear import and export, with the predominant 
localization of Dbt governed by the presence or absence of cytoplasmic Per. 
Regardless of the specific mechanism, since Dbt has also been implicated in 
vital developmental and cellular functions that are not mediated through 
Per, an important product of any device generating cycling subcellular 
localization of this kinase could be temporal regulation of its access to 
alternative substrates (Kloss, 2001).Dbt has been shown to be a component 
of the cytoplasmic activity that destabilizes Per. Evidence was also found 
that Dbt influences the stability of nuclear Per proteins. However, it has 
been unclear whether Dbt acts in both subcellular compartments, or whether 
nuclear stability of Per is affected by a Dbt-dependent phosphorylation in 
the cytoplasm, with delayed effects once Per translocates into the nucleus. 
This study shows that Dbt proteins are found both in the cytoplasm and in 
the nucleus. Coupled with the finding that Per proteins are always found 
associated with Dbt, this suggests that Dbt is required both in the nucleus 
and in the cytoplasm for Per phosphorylations (Kloss, 2001).The 
simultaneous changes in subcellular localization of Per, Tim, and Dbt make 
it likely that direct physical associations among these proteins cause the 
cycling Dbt localizations. Per and Dbt proteins can associate in vitro and 
in cultured cells. Per/Dbt complexes can be recovered at all times during 
the day from head extracts, regardless of whether the majority of these 
proteins are localized in the cytoplasm or in the nucleus. Thus, Per 
proteins are associated with Dbt proteins in vivo when Per is in a Per/Tim 
complex and when Per proteins are free from Tim (Kloss, 2001).Conversely, 
while Dbt binds to Per and Per/Tim complexes, no evidence has been found 
that Tim protein, free from Per, associates with Dbt in vivo. This finding 
is in line with the conclusion that Dbt's effects on the circadian clock 
are primarily mediated through Per (Kloss, 2001).Extensive efforts have 
failed to obtain a functional assay for bacterially produced, recombinant 
Dbt in vitro. The putative kinase domains of Dbt and its mammalian ortholog 
CKIepsilon are very closely related (86% aa identity), so it was surprising 
to find that recombinant, mammalian CKIepsilon readily phosphorylates 
Drosophila Per and human Per in vitro. These observations suggest that Dbt 
function might be tightly regulated in the fly. It has been established 
that truncation of mammalian CKIepsilon substantially increases its 
activity in vitro, and truncated forms of the enzyme were used in the above 
mentioned Per and hPer assays. Although a corresponding truncation of Dbt 
failed to generate activity, such studies of mammalian CKIepsilon also 
indicate more complex regulation for this kinase in vivo (Kloss, 
2001).Without direct kinetic measurements of the activity of Dbt at 
different times of day, it cannot be determine whether Dbt function is 
under circadian control. However, it can be asked whether Per 
phosphorylation in vivo is (1) dependent upon the presence of Dbt and (2) 
influenced by Tim. In timUL flies entrained to LD 12:12, where Per remains 
complexed with TimUL for a prolonged interval in the nucleus, Per remains 
hypophosphorylated during the dark phase. Because wild-type flies begin to 
phosphorylate their Per proteins during the dark phase of such LD cycles, 
the results with timUL suggest that Tim influences the timing of 
light-independent Per phosphorylation (Kloss, 2001).Light-triggered removal 
of TimUL protein is correlated with a rapid and progressive increase in the 
level of Per phosphorylation. Because a similar, cytoplasmic association of 
Per and Dbt in tim01 flies results in cytoplasmic Per degradation, and such 
Per degradation requires Dbt, the most parsimonious explanation of these 
results should be that nuclear association of Per with TimUL protects Per 
from phosphorylation and, secondarily, from turnover. It has been shown 
that light eliminates Tim, but will not promote Per phosphorylation in a 
hypomorphic mutant of Dbt (dbtP). Thus, Per phosphorylation appears to be 
influenced by the formation of Per/Tim complexes, and only when Per is free 
from Tim is it subject to phosphorylation by a Dbt-dependent mechanism. 
While this view is favored, it is also possible that light directly 
activates elements of a Dbt-dependent mechanism to promote some Per 
phosphorylations, or that additional factors associate with Per (or Dbt) 
after Tim is removed by light. Such factors would then be essential for 
Dbt-regulated phosphorylation of Per (Kloss, 2001).The following is a model 
for the accumulation, phosphorylation, and degradation of Per: 
Dbt-dependent phosphorylation of Per in the cytoplasm is thought to delay 
the accumulation of Per proteins until lights off. Increasing Tim levels 
result in stable Per/Tim/Dbt complexes containing hypophosphorylated Per. 
These complexes are translocated to nuclei, where continued physical 
association of Tim with Per prolongs the cycle. Subsequently, the formation 
of Per free from Tim allows the clock to advance by Dbt-dependent 
phosphorylation of nuclear Per. This phosphorylation could be indirectly 
controlled by Dbt. The cycle restarts after degradation of phosphorylated 
nuclear Per proteins. According to this model, Dbt would have opposing 
effects on the cycle at different times of day and in different subcellular 
compartments. This regulation would determine the onset and duration of 
Per's activity in the nucleus, and should therefore be required to 
establish rhythmicity and set the period of Drosophila's circadian clock 
(Kloss, 2001).The biological clock synchronizes the organism with the 
environment, responding to changes in light and temperature. Drosophila 
Cryptochrome (Cry), a putative circadian photoreceptor, interacts with the 
clock protein Timeless (Tim) in a light-dependent manner. Although Tim 
dimerizes with Period (Per), no association between Cry and Per has 
previously been revealed, and aspects of the light dependence of the 
Tim/Cry interaction are still unclear. Behavioral analysis of double 
mutants of per and cry suggest a genetic interaction between the two loci. 
To investigate whether this is reflected in a physical interaction, a 
yeast-two-hybrid system was employed that revealed a dimerization between 
Per and Cry. This is further supported by a coimmunoprecipitation assay in 
tissue culture cells. The light-dependent nuclear interactions of Per and 
Tim with Cry require the C terminus of Cry and may involve a trans-acting 
repressor. Thus, as in mammals, Drosophila Cry interacts with Per, and, as 
in plants, the C terminus of Cry is involved in mediating light responses 
(Rosato, 2001). The genetic interaction between per and cry prompted an 
investigatation of the possibility of a physical interaction between Per 
and Cry using a yeast-two-hybrid system. A full-length Cry protein, 
directly fused to LexA (bait), was challenged with Per(233-685) as prey. 
This fragment includes the major protein/protein interaction domains 
described for Per. A fragment of Tim(377-915) that is known to bind to Per 
and contains the relevant regions for Per/Tim dimerization as prey was also 
tested. No interactions were observed between LexA-Cry and both 
Per(233-685) and Tim(377-915) fragments in the dark. Cry has been shown to 
interact with full-length Tim, but not Per, under constant light. In light, 
LexA-Cry binds strongly to Per(233-685), but not to Tim(377-915). LexA-Cry 
was also challenged with full-length Per and Tim, both in darkness and 
light. No interactions were observed in the dark. Under constant light, 
only full-length Tim showed evidence of dimerization with LexA-Cry. Three 
conclusions are drawn from these results: Per and Tim interactions with 
LexA-Cry are light dependent; the N and/or the C terminus of Tim are 
required for the association with LexA-Cry, and there is an inconsistency 
between the results obtained from full-length Per and the fragment 
Per(233-685). In regard to the latter, the well-established Per/Tim 
interaction was retested using LexA-Tim bait with Per and Per(233-685) 
preys in darkness and light. No interactions were observed using 
full-length Per. Subsequent Western blot analysis has revealed that, in 
this system, full-length Per is poorly expressed, thereby explaining the 
lack of interactions in yeast with this construct. Nevertheless, a strong 
interaction between LexA-Cry and Per(233-685) could be demonstrated. This 
discrepancy between the current results and contradictory published results 
must reside in the different yeast-two-hybrid systems employed. Evidence 
was also found for a Tim-independent Cry/Per complex using 
coimmunoprecipitation (Rosato, 2001).Cryptochromes are believed to interact 
with a signaling factor after light exposure, and evidence has been found 
in plants for a role of the C-terminal domain in signaling. Since the 
coimmunoprecipitation result supports the view that the interaction between 
LexA-Cry and Per(233-685) in yeast reflects a meaningful association 
between Per and Cry, the power of yeast genetics was exploited to test the 
regulatory role of the C terminus of Drosophila Cry. Twenty residues were 
deleted from the Cry C terminus to create CryDelta and it was challenged 
with Per(233-685) and full-length Tim in darkness and light. An interaction 
was evident in both conditions, with no obvious difference between them. It 
has been suggested that LexA-Cryb is unable to interact with Tim in yeast 
cells because it may have lost its photoresponsiveness. Both LexA-Cryb and 
LexA-CrybDelta, which are strongly expressed in yeast, are nevertheless 
unable to interact with Per(233-685) or with Tim. Given the light 
independence of CryDelta, it is suggested that the D[410]N substitution in 
Cryb probably confers a gross structural defect to LexA-Cryb, rather than 
simply affecting its photoreceptor ability (Rosato, 2001).To further map 
the interaction between Cry and Per, LexA-CryDelta was challenged with 
several overlapping Per fragments. It was confirmed that LexA-Tim (377-915) 
interacts with the PAS A domain (Per[233-390]) and Per(233-685). 
LexA-CryDelta does not associate with Per(233-390), nor with the PAS A + B 
region (Per[233-485]), but interacts with the downstream C domain 
(Per[524-685], which includes the perS site. From these results, it is 
speculated that Tim and Cry may interact with different regions of the Per 
protein and, since Cry associates with region(s) of Tim external to the 
(377-915) fragment, it is hypothesized that Per, Tim, and Cry can be found 
in the same complex (Rosato, 2001). LexA-Cry requires light in order to 
interact with Per(233-685) and Tim. However, it cannot be ruled out a 
priori that it is the temperature increase, caused by the continuous light 
exposure, rather than light per se, that triggers Cry's interactions. 
LexA-Cry was therefore challenged with Per(233-685) and Tim at 37C in the 
dark, but no interactions were observed. Furthermore, since LexA-CryDelta 
does not require light, this variant was used to investigate the effect of 
temperature on Cry interactions (Rosato, 2001). Yeast patches were grown on 
X-gal plates at 30C and 37C in parallel. It was noted that at 37C, the 
LexA-CryDelta interaction with Per(233-685) is considerably weakened, 
whereas the control LexA-Tim(377-915)/Per(233-685) dimerization does not 
show any substantial temperature differences. The same temperature 
dependence is also observed when LexA-CryDelta is challenged with Tim and 
Per (524-685) (Rosato, 2001).Oscillations of the period (per) and timeless 
(tim) gene products are an integral part of the feedback loop that 
underlies circadianbehavioral rhythms in Drosophila melanogaster. Resetting 
this loop in response to light requires the putative circadian 
photoreceptorCryptochrome (Cry). The early events in photic resetting were 
dissected by determining the mechanisms underlying the Cry response tolight 
and by investigating the relationship between Cry and the light-induced 
ubiquitination of the Tim protein. In response to light, Cry is degraded by 
the proteasome through a mechanism that requires electron transport. 
Various Cry mutant proteins are not degraded, and this suggests that an 
intramolecular conversion is required for this light response. 
Light-induced Tim ubiquitination precedes Cry degradation and is increased 
when electron transport is blocked. Thus, inhibition of electron transport 
may 'lock' Cry in an active state by preventing signaling required either 
to degrade Cry or to convert it to an inactive form. High levels of Cry 
block Tim ubiquitination, suggesting a mechanism by which light-driven 
changes in Cry could control Tim ubiquitination (Lin, 2001). The presence 
of endogenous Cry in S2 cells supports the idea that the light-dependent 
Tim ubiquitination is mediated by Cry. To determine if the Tim response to 
light requires Cry, Tim levels were assayed in light-pulsed and unpulsed 
cryb flies. While wild-type flies show the characteristic decrease in Tim 
levels with light treatment, this response is lacking in cryb flies (Lin, 
2001). The S2 cell system was used to determine the relationship between 
light-induced Cry degradation and Tim ubiquitination and degradation. One 
possibility considered was that Cry is required for Tim stability. In this 
model, light-induced Cry degradation would lead to Tim degradation, perhaps 
by exposing relevant sites on Tim to phosphorylation and ubiquitination 
events. Although Tim and Cry do not bind each other in the dark in the 
yeast two-hybrid system, they can be coimmunoprecipitated from S2 cells, 
suggesting that they are present in the same complex. Thus, removal of Cry 
in response to light could affect Tim processing. Alternatively, light 
exposure may lead to some conformational and/or redox changes in Cry that 
trigger downstream events, including Tim ubiquitination and Cry 
degradation. To distinguish between these two possibilities, the time 
course of Tim ubiquitination and that of Cry were examined for degradation 
in S2 cells. An increase in Tim ubiquitination is detected within 5 min of 
light exposure, while Cry levels in the same extracts remain unchanged up 
to the end of a 30-min light pulse. Thus, overall degradation of Cry does 
not appear to be required for Tim ubiquitination. The possibility that Cry 
is removed from a complex with Tim cannot be excluded: it is more likely 
that in response to light, Cry transmits a signal that leads to Tim 
ubiquitination (Lin, 2001). No degradation of Tim in S2 cells could be 
detected in response to light. This may be due, in part, to the HA tag on 
the ubiquitin, which could interfere with proteasomal digestion. However, 
other researchers have also noted that Tim is not turned over upon light 
exposure in S2 cells. Extended incubation (up to 6 h post-Tim induction) of 
transfected cells results in Tim degradation in both dark- and 
light-treated cells (Lin, 2001). Tim ubiquitination was examined in the 
presence of electron transport inhibitor DPI. Tim ubiquitination is 
increased by DPI, although Cry degradation is blocked, which is consistent 
with the idea that Tim ubiquitination does not require degradation of Cry. 
In fact, the increased Tim ubiquitination is most likely due to the 
accumulation of activated Cry, effected through a block either in 
degradation or in the reconversion of Cry to an inactive form (Lin, 2001). 
Light-induced Tim ubiquitination in S2 cells is thought to be mediated by 
endogenous Cry. To test the effects of increasing Cry levels on 
light-induced Tim ubiquitination, S2 cells were cotransfected with hs-tim, 
hs-Ub, and different concentrations of either pIZ-cry or hs-cry and Tim 
ubiquitination was assayed 2 h after light exposure (Lin, 2001). High 
concentrations of both hs-cry and pIZ-cry decrease Tim ubiquitination. 
However, ubiquitination of Tim is enhanced when hs-cry is transfected. 
pIZ-cry does not increase Tim ubiquitination when transfected at low 
concentrations, most likely because this plasmid yields higher levels of 
Cry expression. Taken together these observations indicate that small 
increases in Cry promote Tim ubiquitination after 2 h of light exposure but 
that high levels attenuate it. However, even in the presence of high levels 
of Cry, Tim ubiquitination increases during the first 10 to 15 min of light 
treatment. The block at later time points in Cry-overexpressing cells is 
indicative of a deficit in the maintenance of Tim ubiquitination, which may 
be due to enhanced deactivation of Cry (Lin, 2001). The data on the 
differential effects of low and high Cry concentrations are supported by 
results of Cry overexpression in transgenic flies. Flies that overexpress 
Cry under control of the tim promoter show enhanced resetting, while those 
that express Cry under the actin 5c promoter show a reduction of 
light-induced phase delays. The difference in the phenotypes of these two 
overexpression strains may lie in the level of overexpression. To determine 
whether the reduced resetting in the actin 5c line correlates with reduced 
Tim degradation in response to light, flies carrying a UAS-cry construct 
were crossed to others carrying an actin 5c promoter-GAL4 transgene and the 
resulting progeny was assayed for Tim expression. Tim expression was 
examined at different times of day by Western blotting of adult fly head 
extracts. In Cry overexpression flies Tim levels are considerably higher 
than wild-type levels at time points early in the day but equivalent to 
wild-type levels at all other time points. Thus, the effect is specific for 
the early part of the day, when Tim is normally turned over in response to 
light (Lin, 2001). Degradation of Cry by light invokes analogies with the 
plant photoreceptors, phytochrome (PHY) and Cry, both of which are degraded 
in response to light. Thus, it may be a common mechanism to control levels 
of the photoreceptor and thereby the strength of the photic response. 
Moreover, as noted here for Cry, PHY is known to be degraded by the 
proteasome (Lin, 2001). The role of the proteasome in degradation of both 
Cry and Tim also underscores similarities with the cell cycle. The cell 
cycle is characterized by cycling proteins that undergo phosphorylation and 
subsequent degradation, in many cases by the proteasome. Both Per and Tim 
are cyclically phosphorylated and phosphorylation plays a role in turnover 
of both proteins. For Tim, light-induced degradation is effected through an 
increase in phosphorylation and ubiquitination. Thus, as for the cell 
cycle, multiple proteins in the circadian cycle are turned over by the 
ubiquitin-proteasome pathway. However, Per turnover may utilize a different 
pathway since ubiquitination of Per has not been observed (Lin, 2001). Cry 
has been shown to block Per and Tim autoregulation of their own RNA 
synthesis in a light-dependent manner in S2 cells. Since Tim degradation is 
not detectable in S2 cells, it has been suggested that the inhibition of 
Tim activity by Cry, rather than its degradation, is the primary response 
to light. This block in Per-Tim activity may be the immediate response of 
the clock to light. Presumably this block persists as long as the photic 
signals are present and Cry is not degraded. However, a phase change of 
several hours, which can be produced with a pulse of Biological Overview 
Evolutionary Homologs Protein Interactions Developmental Biology Effects of 
Mutation ReferencesHome page: The Interactive Fly 1997 Thomas B. Brody, 
Ph.D.\nThe Interactive Fly resides on the\nSociety for Developmental 
Biology's Web server.\n\nDarkness and Flame: The Dark Side activation code 
generator\nDownload Zip https://t.co/1LrmoURDf1\n\n\n eebf2c3492\n

-- 
You received this message because you are subscribed to the Google Groups 
"Clementine Music Player" group.
To unsubscribe from this group and stop receiving emails from it, send an email 
to clementine-player+unsubscr...@googlegroups.com.
To view this discussion on the web visit 
https://groups.google.com/d/msgid/clementine-player/4893dc4d-6d62-4a82-ba00-2d1acf183b52n%40googlegroups.com.

Reply via email to