on Biological Clocks Michael W. Young 1 Laboratory of Genetics The Rockefeller University 1230 York Avenue New York, New York 10021

Similar documents
Biological Clocks. Lu Chen, Ph.D. MCB, UC Berkeley. What is biological clock?

Biological Clocks. Lu Chen, Ph.D. MCB, UC Berkeley. Why Does Melatonin Now Outsell Vitamin C??

Clicker Question. The Need to Decompose. Mechanism and Reduction: Decomposing Circadian Clocks

Transcription Regulation And Gene Expression in Eukaryotes (Cycle G2 # )

CIRCADIAN SIGNALING NETWORKS

Advance in circadian rhythm genetics in mammals

The Nobel Assembly at Karolinska Institutet has today decided to award. the 2017 Nobel Prize in Physiology or Medicine. jointly to

Sleep-Wake Cycle I Brain Rhythms. Reading: BCP Chapter 19

Novel insights into photoreception, phototransduction and biological rhythms how might this impact on carcinogenesis?

Transcription Regulation And Gene Expression in Eukaryotes FS 2016 Graduate Course G2

LESSON 4.5 WORKBOOK How do circuits regulate their output?

Stochastic simulations

Time after time: inputs to and outputs from the mammalian circadian oscillators

Welcome to Bi !

Environmental stimulus perception and control of circadian clocks Nicolas Cermakian* and Paolo Sassone-Corsi

T. WU 1, Y. NI 1, F. ZHUGE 1, Z. FU 1. Introduction

Stochastic simulations

The Success of Decomposition

PHYSIOLOGY AND MAINTENANCE Vol. V - Biological Rhythms - Tarja Porkka-Heiskanen, Jarmo T. Laitinen

Neurons and Hormones 1. How do animals perform the right behaviors at the right time? In the right context?

Light and the circadian clock : effects on human health

Dark and light adaptation: a job that is accomplished mainly in the retina

Signaling components that drive circadian rhythms Garrick K Wang* and Amita Sehgal

Modeling Rhythms on Differents Levels: Cells, Tissues, and Organisms

Biological Rhythms. Today s lecture

Make sure you remember the Key Concepts

Molecular mechanism of cell-autonomous circadian gene. expression of Period2, a crucial regulator of the mammalian

MOLECULAR BASES OF CIRCADIAN RHYTHMS

Phase Shifts of Circadian Transcripts in Rat Suprachiasmatic Nucleus

SUPPLEMENTARY INFORMATION

What Is There Left to Learn about the Drosophila Clock?

Circadian photoreception in humans: More than meets the eye

Circadian Gene Expression in the Suprachiasmatic Nucleus

Circadian rhythm and Sleep. Radwan Banimustafa MD

Review. The Network of Time: Understanding the Molecular Circadian System. Till Roenneberg and Martha Merrow

Targeting of the attenuated diphtheria toxin (adta) into the melanopsin locus. a,

Light, body clocks and sleep - SE13

Lighting and Melatonin: How Lighting Impacts Melatonin Suppression and Regulation

Seeing Ultraviolet. Keeping Time with Neuropsin

From the reveller to the lark

Strange vision: ganglion cells as circadian photoreceptors

Sleep, Dreaming and Circadian Rhythms

Sleep Biology: Circadian Rhythms and Mother Nature

Utility of colored-light pupil response in patients with age-related macular degeneration

Entrainment of the circadian clock in humans: mechanism and implications for sleep disorders.

Methods in Enzymology. Volume 393. Circadian Rhythms. Michael W. Young

Circadian rhythms new functions for old clock genes?

3.4 Photoisomerization reactions

Scientific Background Discoveries of Molecular Mechanisms Controlling the Circadian Rhythm

Circadian Rhythms in Physiology and Behavior. The Persistence of Memory, Salvador Dali, 1931

CP116. Mixed-Color LED Lighting with Circadian Benefits

Peripheral Clocks: Keeping Up with the Master Clock

BioNSi: A Discrete Biological Network Simulator Tool

Supplemental Information. Melanopsin-Encoded Response Properties. of Intrinsically Photosensitive. Retinal Ganglion Cells

Metronomes that Regulate the Day

Beyond our eyes: the non-visual impact of light

Artificial organisms that sleep

Lecture 3 Vision 2 The Retina

Posttranslational Control of the Neurospora Circadian Clock

I A~ll C,,, i' 3, RE OR o TYPE AND "'OATE S COVEiRED" ANNUAL 15 Mar 92 TO 14 Mar 93

Strong Resetting of the Mammalian Clock by Constant Light Followed by Constant Darkness

A CLOCKWORK WEB: CIRCADIAN TIMING IN BRAIN AND PERIPHERY, IN HEALTH AND DISEASE

A PER/TIM/DBT Interval Timer for Drosophila s Circadian Clock

Neurobiology of Circadian Rhythms

The Ticking CLOCK of HSV-2 Pathology Rebecca J. Bayliss 1 and Vincent Piguet 1,2,3

Neuroscience - Problem Drill 13: The Eye and Visual Processing

Nonvisual effects of light. Prof. Grega Bizjak, PhD Laboratory of Lighting and Photometry Faculty of Electrical Engineering University of Ljubljana

Carlson (7e) PowerPoint Lecture Outline Chapter 6: Vision

Pediatrics Grand Rounds 16 July University of Texas Health Science Center at San Antonio

Multiple Photoreceptors Contribute to Nonimage-forming Visual Functions Predominantly through Melanopsincontaining

2. METHODS. 2.1 Apparatus

Altered Entrainment and Feedback Loop Function Effected by a Mutant Period Protein

The Drosophila melanogaster life cycle

CRYPTOCHROME: The Second Photoactive Pigment in the Eye and Its Role in Circadian Photoreception

Transcriptional Feedback Loop Regulation, Function, and Ontogeny in Drosophila

This document is a required reading assignment covering chapter 4 in your textbook.

Molecular Signals of Mammalian Circadian Clock

Vision Phototransduction of light By. Prof/Faten zakareia Physiology Dept College of Medicine King Saud University

Test of visual pathway function

The Circadian Clock: Regulating the Rhythm of Life

Simulation of Drosophila Circadian Oscillations, Mutations, and Light Responses by a Model with VRI, PDP-1, and CLK

Thesis of Ph.D. dissertation THE ROLE OF THE PHYTOCHROME B PHOTORECEPTOR IN THE REGULATION OF THE PLANT CIRCADIAN CLOCK AND CIRCADIAN RHYTHMS

Construction of the Visual Image

Supplementary Figure 1. Expression of the inducible tper2 is proportional to Dox/Tet concentration in Rosa-DTG/Per2 Per2-luc/wt MEFs.

The Role of Circadian Regulation in Cancer

HUMAN CENTRIC LIGHTING. System overview

The ins and outs of circadian timekeeping Steven A Brown* and Ueli Schibler

THE (HIDDEN ) BENEFITS OF DAYLIGHTING

Developmental regulation of melanopsincontaining retinal ganglion cells and its effects on circadian function

Biological rhythms. Types of biological rhythms

All mammalian cells investigated to date seem to possess

Circadian Phase Entrainment via Nonlinear Model Predictive Control

Cancer. October is National Breast Cancer Awareness Month

PROMOTER ANALYSIS OF MAMMALIAN CLOCK CONTROLLED GENES

Closing the circadian negative feedback loop: FRQ-dependent clearance of WC-1 from the nucleus

Lecture 8. Arousal & Sleep. Cogs17 * UCSD

Robert James Lucas. Professor of Neuroscience, Faculty of Life Sciences, University of

Problem Set 8 Key 1 of 8

Biological Bases of Behavior. 6: Vision

Introduction to Physiological Psychology

Transcription:

Neuron, Vol. 36, 1001 1005, December 19, 2002, Copyright 2002 by Cell Press Big Ben Rings in a Lesson on Biological Clocks Michael W. Young 1 Laboratory of Genetics The Rockefeller University 1230 York Avenue New York, New York 10021 Thomas Gage came to North America in 1754, serving as a British officer and custodian of the American colonies in the closing years of the French and Indian Wars. He settled in New York City, married a New Yorker, Margaret Kemble, and eventually was elevated to Com- mander-in-chief of the British forces. Gage lost his chance for a comfortable retirement in the colonies in April of 1775 when he was told to bring order to Massachusetts. It was General Gage who marched against the colonial militia at Lexington and Concord, touching off the American Revolution. Gage resigned his post in the Americas in October of 1775 and returned to England. There, he and his American wife moved into a newly built house on Portland Place, a broad street near the center of London. Portraits of Thomas and Margaret still frame the fireplace in one of the house s largest rooms, now part of a conference center and headquarters of the Novartis Foundation, a scientific charity originally formed by CIBA corporation in 1949. For the past 50 years, the Foundation has been or- ganizing small international conferences in biology and medicine in the Gage house. Eight three-day confer- ences are held each year on topics of high interest. These symposia are attended by 25 30 scientists who come more as discussants than as speakers. In fact, more than half of the printed schedule is devoted to specific or general discussions. This September, No- vartis was the site of a spirited examination of molecular clocks and their responses to light. work from two labs (Froehlich et al., 2002; He et al., 2002) points to a similar mechanism for the photoresponse in Neurospora. Two transcription factors, the White Collar proteins (WC-1 and WC-2), have long been implicated in the generalized transduction of signals from blue light. The proteins work together, binding DNA as a WC-1/ WC-2 complex. Both labs find that these complexes bind flavin (FAD), specifically through an interaction with WC-1. WC-1 and WC-2 are essential components of the fungal circadian clock. However, mutations that eliminate flavin binding do not abolish rhythmicity. These mutants are blind to the resetting effects of light, but can be synchronized by other environmental stimuli such as cycling temperature. Given the parallel mechanisms of fungal and fly clocks, it might be expected that a clock-resetting, flavin binding photoreceptor in mammals would be forthcom- ing. But here a stimulating debate rages on. Russell Foster reminded us that for years, action spectra have been used to argue that entrainment depends on an opsin in mammals. On this side, the argument leans heavily on two classes of photoresponses that persist in mice genetically deficient for all rods and cones of the retina: (1) undiminished photoentrainment of circadian rhythms and (2) light-stimulated contraction of the pupil (pupillary response). Both responses are most sensitive to light with a wavelength of 479 nm (Bellingham and Foster, 2002), squarely in a range expected for an opsinbased photopigment. Although the absorbance spectra are well resolved for several opsins, the Cryptochromes can be problematic, with variable spectra that are influ- enced by the presence of both flavin and pterin and by changes in spectral sensitivity of the chromophores that depend on their redox state (reviewed by Sancar, 2000; Foster and Helfrich-Forster, 2001). Adding fuel to the case for opsin, new presumptive photopigments have been discovered in this class. One of special interest, melanopsin, is expressed in the ganglion cells of the retina and would therefore be expected to survive ablation of the rods and cones. Foster reviewed several lines of evidence that melanopsin-expressing gan- glion cells form a new light-sensing tissue distinct from the image-forming visual system. First, at least some of these retinal cells project to the suprachiasmatic nu- cleus (SCN), the major site for circadian regulation of behavioral rhythms in mammals (Gooley et al., 2001; Hannibal et al., 2002). Second, electrophysiological assays show that retinal ganglion cells directly respond to light, even if removed from the retina, and that the action spectra of the responses peak at about 480 nm (Berson et al., 2002). Hence, they must autonomously produce a photoreceptor and are most sensitive to wavelengths previously implicated in circadian and pu- pillary responses. Finally, in addition to SCN projections, some melanopsin-containing ganglion cells project to Hunting for a Circadian Photoreceptor How does light reset our sleep/wake cycle? In Drosoph- ila, the path runs to Timeless (TIM), a transcription factor and essential component of the circadian clock, which is rapidly degraded in the presence of light. 24 hr waves of tim RNA and protein accumulation keep the fly clock running. When the fly is synchronized to its environment, the waves come at night. However, if light is extended into the early evening, as with a plane trip from New York to California, accumulation of new TIM proteins is delayed and the clock slows. In contrast, light delivered after midnight, after TIM has filled the nucleus, prematurely ends TIM s activity, shifting the molecular clock forward. For both advances and delays, TIM s elimination generally involves an unusual photoreceptor, Cryptochrome (CRY). This protein uses bound chromophores, flavin and pterin, to absorb blue light. CRY will physically associate with TIM, and light stimulates phosphorylation and ubiquitination of TIM in wild- areas of the brainstem mediating the pupillary response type, but not in CRY-deficient flies. (Hattar et al., 2002). As Jay Dunlap described for us at Novartis, recent So shouldn t this stream of successes wrap up the 1 Correspondence: young@rockefeller.edu search in mammals for an elusive circadian photoreceptor? Perhaps not. As Foster cautions, while the spectral

Neuron 1002 downregulate their own expression. In one of these loops, PER and TIM (or PER alone) physically associate with CLK/CYC heterodimers to suppress their transcrip- tion-promoting activity. In the remaining loop, high lev- els of VRI somehow suppress per, tim, and vri RNA levels (Blau and Young, 1999). The structure of VRI, a bzip protein, indicates that unlike PER and TIM, it should function directly on DNA. One more instance of cycling gene expression is seen in the fly clock. Clk RNA cycles with a phase opposite that of per, tim, and vri. As in the case of the VRI loop, Clk cycling has had its mysteries and its clues: mutations of per or tim lead to low, constitutive expression of Clk. This response is reversed, giving high constitutive Clk expression in per and/or tim mutants, if the CLK protein is also mutated. Somehow, CLK must repress its own activity while PER and TIM activate it (Glossop et al., 1999). Two new studies, one by Paul Hardin and colleagues and the other by Justin Blau s laboratory, may simplify this mechanism. As explained by Paul Hardin, VRI s regulation of per and tim is probably indirect VRI may act at the center of the CLK loop. In fact the role of VRI may be compared to that of a new clock protein, REV-ERB, recently found to control feedback within a circuit of the mammalian clock. In mammals, a circadian network composed of two feedback loops has been described (Shearman et al., 2000), and there are striking parallels between these and two of the loops mentioned above for the fly. In a first loop (Figure 1A), the mammalian PER proteins appear to have changed partners; they are now stabilized by heterodimerization with CRY rather than TIM, so that PER/CRY complexes accumulate in the nucleus and physically associate with CLOCK/BMAL1 to suppress Per and Cry transcription (Lee et al., 2001). CLOCK and BMAL1 are the orthologs of Drosophila s CLK and CYC, but in mammals it is Bmal1 rather than Clock whose expression cycles within a second feedback loop (Figure 1A). As in the fly, PER somehow positively regulates this Bmal1 expression (Lee et al., 2001). REV-ERB was introduced by Ueli Schibler. Schibler s laboratory has shown that, in mammals, the orphan nuclear receptor REV-ERB links the PER/CRY and BMAL1 feedback loops: CLOCK/BMAL1 appears to di- rectly activate transcription of Rev-erb, while REV- ERB protein binds to and represses the Bmal1 pro- moter (Figure 1A; Preitner et al., 2002). In this scheme, PER positively regulates Bmal1 expression because it reduces transcription of Rev-erb. Figure 1B gives one view of how such a clock might work. For simplicity, oscillations in the mouse liver are modeled, and only averaged expression profiles of Rev- erb, one Per (Per1), and one Cry (Cry2) are shown (Lee et al., 2001; Preitner et al., 2002). Each cycle would be composed of three timed regulatory steps. One that turns Per, Cry, and Rev-erb expression on, and two more that act sequentially to turn these genes off. REV- ERB would be the initial repressor of Per, Cry, and Rev- erb gene activity. PER/CRY complexes would step in later to keep the genes switched off after REV-ERB s work is done. If in the scheme of Figure 1B the beginning of a cycle is taken as dawn (ZT 0), high levels of CLOCK/ BMAL1 activity first set off a wave of Per, Cry, and Rev- sensitivity of the retinal ganglion cells matches that linked to circadian photoresponses, it has not yet been determined that this reflects absorbance and activity of melanopsin. More importantly, Steve Kay described new work indicating that mice genetically deficient for melanopsin still produce photo-entrainable circadian rhythms. There are deficiencies in such responses if short pulses of blue light are substituted for cycles of daylight. Nevertheless, another photoreceptor must be able to provide substantial function in mice. Lastly, as pointed out by Russell Van Gelder, whose interests have been running toward the Cryptochromes, these too are expressed in retinal ganglion cells, and there is compelling evidence against opsins as the only circa- dian photoreceptors. Retinol Binding Protein (RBP) is the only known trans- porter of retinol from liver stores to the retina where it is converted to retinal, the opsin chromophore. RBPdeficient mice (genetically null), maintained on a vitamin A-free diet, become visually blind, but without significant depreciation of their circadian photoresponse (Thomp- son et al., 2001). In contrast to these tests for opsin function, mice genetically deficient for both of their Cryptochrome genes and for their rods and cones show a strongly diminished response in two pertinent, al- though indirect, assays: c-fos induction in the SCN (previously linked to photo-entrainment of circadian rhythms; Selby et al., 2000), and the pupillary response, which Van Gelder now described. These indirect assays are required because the Crytochromes are also essen- tial for the production of circadian rhythms in mammals. In tests of the pupillary response, Russell Van Gelder and colleagues have compared the light sensitivity of mice missing their rods and cones (retinal degenerate, rd) to those mutant for rd and both Cryptochromes. A 20-fold reduction in the magnitude of the response was observed for rd; Cry1; Cry2 versus rd when mice were illuminated at a wavelength very close to that previously found to be most effective in producing pupil contraction in wild-type mice, and encompassing peak absorption of the opsins. While a precise measurement of the action spectra is still needed, simply interpreted, Cryptochrome or a Cryptochrome-dependent signaling path- way involving another photopigment should be activated by wavelengths of light usually associated with stimulation of opsins. Perhaps there will be a replay of the Drosophila story in which circadian clocks can react to light through a complex mix of opsin- and Cryptochrome-dependent steps. The Clockworks Most of what we know about the inner workings of circadian clocks in animals has come from genetic and biochemical studies in Drosophila (Panda et al., 2002b). Three genes in the fly, timeless, period (per), and vrille (vri), are synchronously transcribed with a circadian rhythm. Genetic studies have shown that per and tim are essential for circadian rhythmicity as is the normal cycling expression of vri. Their activities rise and fall together because all three genes are bound and transcribed by a common complex of transcription factors, Clock (CLK) and Cycle (CYC). The PER, TIM, and VRI proteins compose two distinct feedback loops that

1003 Figure 1. Model for Sequentially Acting Feedback Loops in the Mammalian Clock (A) The network depicted would depend on early transcriptional regulation of Bmal1 gene expression by the orphan nuclear receptor REV-ERB (R) (Preitner et al., 2002), and later posttranscriptional regulation of BMAL1 (BML) protein activity by PER (P) and CRY (C). (B) In this model, the REV-ERB and PER/ CRY feedback loops collaborate to restrict BMAL1 function to a few hours each day. In the morning (ZT 0 6), CLOCK/BMAL1 activity induces a wave of Per, Cry, and Rev-erb transcription. Although accumulation of PER and CRY proteins is delayed until after dark (Lee et al., 2001), REV-ERB protein levels climb throughout much of the day (Preitner et al., 2002). REV-ERB directly inhibits Bmal1 expression and therefore depresses the pool of BMAL1 protein by early afternoon (ZT 6 10). Per, Cry, and Rev-erb transcription stall at that time due to their dependence on BMAL1. Falling Rev-erb transcription and elimination of REV-ERB protein allows reinitiation of Bmal1 transcription. However, in the absence of REV-ERB, long-lived complexes of nuclear PER and CRY form a second stage of repression that inactivates accumulating BMAL1 proteins overnight. BMAL1 activity will reemerge as PER and CRY degrade shortly before dawn. This starts the next molecular cycle. ZT 0 is dawn and ZT 12 is sunset in a 12 hr/12 hr light/dark cycle. Profiles of RNA and proteins represented are from mouse liver (see text). erb transcription (step 1). This wave hits its peak in the afternoon (ZT 8) because rising levels of REV-ERB shut down transcription of Bmal1, exhausting the pool of CLOCK/BMAL1 complexes (Preitner et al., 2002). Since BMAL1 is needed to maintain the pool of REV-ERB, by sunset (ZT 12) levels of both BMAL1 and REV-ERB are low. Transcription of Per and Cry also declines during this interval due to loss of BMAL1 (completing step 2). Because the PER and CRY proteins are stabilized by heterodimerization, these proteins persist when Per and Cry RNAs are gone. Nuclear PER/CRY complexes be- come most abundant around midnight (ZT 18) (Lee et al., 2001). Although absence of REV-ERB now permits a wave of Bmal1 transcription, the activity of new CLOCK/ BMAL1 complexes should be suppressed by physical interaction with nuclear PER/CRY (step 3). This prolongs suppression of Per, Cry, and Rev-erb transcription until PER and CRY are degraded just before dawn, allowing high levels of CLOCK/BMAL1 activity to emerge and start the cycle again. Day-Length Measurements and Seasonal Timing It has long been known that organisms use their circadian clocks to measure day length. This is particularly evident in plants where flowering time and abscission are set to the changing seasons. Steve Kay explained how his laboratory has begun to pick apart the molecular signals tying such photoperiodic measurements to the clock. Several years ago Kay isolated a mutation, toc1-1, that caused short-period (21 hr) circadian rhythms in Arabidopsis. This mutant also showed a defect in flowering; buds appeared in advance of the expected season, and flowering was induced when plants were held in relatively short-day cycles (e.g., 8 hr light/16 hr dark). Yet, this early flowering did not appear if the environ- mental cycle matched the period of the mutant (7 hr light/14 hr dark). That suggested that photoperiodic measurement was altered in these plants because of their circadian defect. Because it had recently been shown that another gene in this system, CONSTANS (CO), accelerates flowering in long days and is expressed with a circadian rhythm (Suarez-Lopez et al., 2001), Kay and his colleagues won- dered how the pattern of CONSTANS cycling might change in toc1-1 plants. They found that in short-day cycles (8 hr light/16 hr dark), wild-type plants accumulated most of their CO RNA at night, but in toc1-1 plants this RNA accumulated early, with most of the rise oc- curring before nightfall (Yanovsky and Kay, 2002). Most CO accumulation occurred at night in both wild-type and toc1-1 plants if they were maintained in 21 hr cycles with comparably short days (7 hr light/14 hr dark), and neither flowered. CONSTANS encodes a transcription factor, and its effects on flowering are probably due to activation of a target gene, FLOWERING LOCUS T (FT). In short-day cycles (8 hr light/16 hr dark), this gene is induced at nightfall in toc1-1 mutants, but not in wild-type plants. Therefore, it appears that the daytime production of CO in the mutants is able to switch FT on (Yanovsky and Kay, 2002). Could the important signal for FT expression come from superposition of light and CO? Kay s group found that mutations of either the photoreceptor cryp- tochrome 2 or phytochrome A blunted levels of FT ex- pression without affecting profiles of CO accumulation. On the other hand, mutation of CO also suppressed light-induced production of FT. Light and CO must coop- erate to raise FT levels and trigger flowering (Yanovsky and Kay, 2002). Since CO is regulated by a circadian timer, this constitutes a molecular system for measuring daylength.

Neuron 1004 Connecting the Clock to Animal Behavior basic structure of nighttime activity and daytime rest is and Physiology maintained (Stokkan et al., 2001; Damiola et al., 2000). How do molecular oscillators get their work done? Opin- Complete independence, though, is rare and work deions have changed as autonomous molecular clocks scribed by Paolo Sassone-Corsi has shown that even have been mapped to tissues both inside and outside a broken cellular clock can sometimes be fixed by the nervous system. Since oscillating transcription fac- placing it in a neighborhood of properly running clocks. tors compose circadian clocks, timed signals could be Fibroblasts taken from mice that are genetically defiproduced by coupling these factors, more or less di- cient for mper1 function are rhythmic in culture with rectly, to genes encoding specific effectors of behavior 20 hr cycles. Nevertheless, the same cells assume a and physiology. Individual clock-containing cells would phase expected for wild-type peripheral tissues and differentially interpret a common set of oscillating near 24 hr periodicity if transplanted into wild-type mice signals. (Pando et al., 2002). Where do factors synchronizing Over the past few months, a flood of microarray work peripheral clocks come from? Do some emerge from in Arabidopsis, Drosophila, and mammals has rein- oscillating gene action in peripheral, nonneural tissues? forced this view. In all of these systems, it is now esti- Do some peripheral clocks send signals to the brain, mated that at least 5% 10% of the gene content is bypassing the SCN to regulate feeding and other speexpressed with a circadian rhythm. In Arabidopsis and cialized behaviors? There are intriguing puzzles to solve in Drosophila, these impressive figures are derived from here. looking at complex RNA pools from mixed cell types (e.g., whole plants or the Drosophila head; reviewed by Circadian Rhythms, Cell Cycles, Growth Control Ceriani et al., 2002). Hundreds of genes cycle and their There is a long history of experimental work seeking a phases of expression cover most times of day and night. connection between circadian clocks and the cell cycle. In this way, the clock enforces a sequence of gene For some eukaryotic, unicellular systems, the evidence expression. With each hour, new genes are added while is quite good photocycles that entrain circadian others are taken offline. Mammalian arrays have made rhythms reset 24 hr rhythms of cell division that persist it clear that startling differences in these programs occur for many days in constant darkness or constant light between cell types. The liver, heart, and suprachias- (reviewed by Edmunds, 1988). Are there comparable matic nucleus (SCN) each support cycling expression interactions in multicellular organisms, even mammals? of several hundred genes. Yet fewer than 10% of the Cheng Chi Lee produced a list of novel phenotypes genes are common in any pairwise comparison of the in Per 2 mutant mice that indicate action of PER as a tissues (Storch et al., 2002; Panda et al., 2002a). tumor suppressor, as a regulator of cell growth, and as Some of these oscillating, tissue-specific gene proda component of the DNA damage response (Fu et al., ucts can have profound effects on behavior. Microarray 2002). First, there is a highly significant difference in studies in mammals included the SCN because this rethe frequency of spontaneous and irradiation-induced gion of the hypothalamus is a command center for locotumors and hyperplasia in Per2 mice. In fact, all Per2 motor activity rhythms. In rodents, transplanted SCN mice develop an otherwise rarely encountered, salivary will restore rhythms following surgical ablation, even gland hyperplasia by the age of 8 months. Per2 mice when encapsulated so that only small diffusible molewere also found to be unusually resistant to irradiationcules contact the surrounding brain. Now two oscillating induced apoptosis. This deficiency appears to be linked genes, one encoding TGF (Kramer et al., 2001) and to reduced induction of p53 and associated cytochrome another prokineticin 2 (PK2) (Cheng et al., 2002), appear c regulation. Even more surprising, Lee s laboratory reto form SCN-borne signals mediating rhythmic behavior. Both are normally expressed at low levels at night when ports that genes composing the clock are induced by mice (TGF ) and rats (PK2) are active. That suggested irradiation in wild-type but not Per2 mice, suggesting that absence of these factors permits locomotor activity. a possible role for the clock itself in the apoptotic re- Consistent with this, continuous infusion of either factor sponse. to sites near the SCN blocks most locomotor activity. Timed expression of several genes central to cell cycle As pointed out by Chuck Weitz in his description of work control is altered in Per2 mice. For example, both cyclin with TGF, this is an effect downstream of the cellular A and cyclin D1 mrnas accumulate in hepatocytes from clocks of the SCN. By measuring the phase of the reapamplitude of both rhythms appear to be substantially wild-type mice with a circadian rhythm. The phase and pearing behavioral rhythm when infusion stops, it is seen that activity resumes with a new phase that depends altered in Per2 mice (Fu et al., 2002). Since cyclin D1 is on the number of circadian ( 24.5 hr) cycles passed a target for c-myc, the latter gene s regulation was also since infusion began (Kramer et al., 2001). Simply interthat examined; c-myc too oscillates with a circadian rhythm preted, the SCN dynamically modifies levels of gross is significantly altered in Per2 mutants. Lee sug- motor behavior over the course of a day through the gested that, in particular, strong upward swings in c-myc clock-dependent control of certain gene activity. expression may underlie the salivary gland hyperplasia Distributing clocks to tissues with different, some- his laboratory sees in Per2 mice. times conflicting, functions can produce surprising relenging Testing a focused proposal of this sort will be chal- sponses. For example, in rodents, restricting food to an because circadian clocks command such broad hour or two in the middle of the day resets the phase programs of timed gene expression. Recall that in the of molecular clocks in the liver, but not the SCN. Conseexpression mouse, 24 hr clocks not only determine the sequential quently, animals show new behaviors associated with of hundreds of genes, but the list of clock- rhythmic bursts of daytime anticipation of food, but the controlled targets is tissue specific, amplifying the com-

1005 plexity of the temporal code. Presumably, a Per2 mutant response ripples through the entire program of rhythmic expression in each tissue. Surely connections between individual clock-controlled genes and elements of the Per2 phenotype will remain speculative for awhile. These difficulties aside, the mouse Per2 study does remind us that a dominant mutation of this gene has been associated with FASPS, a familial sleep disorder in humans (Toh et al., 2001). It would be fascinating to study the range of phenotypic effects associated with this Per2 mutation in transgenic mice. (2002a). Coordinated transcription of key pathways in the mouse by the circadian clock. Cell 109, 307 320. Panda, S., Hogenesch, J.B., and Kay, S.A. (2002b). Circadian rhythms from flies to human. Nature 417, 329 335. Pando, M.P., Morse, D., Cermakian, N., and Sassone-Corsi, P. (2002). Phenotypic rescue of a peripheral clock genetic defect via SCN hierarchical dominance. Cell 110, 107 117. Preitner, N., Damiola, F., Lopez-Molina, L., Zakany, J., Duboule, D., Albrecht, U., and Schibler, U. (2002). The orphan nuclear receptor REV-ERB alpha controls circadian transcription within the positive limb of the mammalian circadian oscillator. Cell 110, 251 260. Sancar, A. (2000). Cryptochrome: the second photoactive pigment in the eye and its role in circadian photoreception. Annu. Rev. Bio- References chem. 69, 31 67. Selby, C.P., Thompson, C., Schmitz, T.M., Van Gelder, R.N., and Bellingham, J., and Foster, R.G. (2002). Opsins and mammalian Sancar, A. (2000). Functional redundancy of cryptochromes and photoentrainment. Cell Tissue Res. 309, 57 71. classical photoreceptors for nonvisual ocular photoreception in Berson, D.M., Dunn, F.A., and Takao, M. (2002). Phototransduction mice. Proc. Natl. Acad. Sci. USA 97, 14697 14702. by retinal ganglion cells that set the circadian clock. Science 295, Shearman, L.P., Sriram, S., Weaver, D.R., Maywood, E.S., Chaves, 1070 1073. I., Zheng, B., Kume, K., Lee, C.C., van der Horst, G.T., Hastings, Blau, J., and Young, M.W. (1999). Cycling vrille expression is re- M.H., and Reppert, S.M. (2000). Interacting molecular loops in the quired for a functional Drosophila clock. Cell 99, 661 671. mammalian circadian clock. Science 288, 1013 1019. Ceriani, M.F., Hogenesch, J.B., Yanovsky, M., Panda, S., Straume, Stokkan, K.A., Yamazaki, S., Tei, H., Sakaki, Y., and Menaker, M. M., and Kay, S.A. (2002). Genome-wide expression analysis in Dro- (2001). Entrainment of the circadian clock in the liver by feeding. sophila reveals genes controlling circadian behavior. J. Neurosci. Science 291, 490 493. 22, 9305 9319. Storch, K.F., Lipan, O., Leykin, I., Viswanathan, N., Davis, F.C., Wong, Cheng, M.Y., Bullock, C.M., Li, C., Lee, A.G., Bermak, J.C., Belluzzi, W.H., and Weitz, C.J. (2002). Extensive and divergent circadian gene J., Weaver, D.R., Leslie, F.M., and Zhou, Q.Y. (2002). Prokineticin 2 expression in liver and heart. Nature 417, 78 83. transmits the behavioural circadian rhythm of the suprachiasmatic Suarez-Lopez, P., Wheatley, K., Robson, F., Onouchi, H., Valverde, nucleus. Nature 417, 405 410. F., and Coupland, G. (2001). CONSTANS mediates between the Damiola, F., Le Minh, N., Preitner, N., Kornmann, B., Fleury-Olela, circadian clock and the control of flowering in Arabidopsis. Nature F., and Schibler, U. (2000). Restricted feeding uncouples circadian 410, 1116 1120. oscillators in peripheral tissues from the central pacemaker in the Thompson, C.L., Blaner, W.S., Van Gelder, R.N., Lai, K., Quadro, L., suprachiasmatic nucleus. Genes Dev. 14, 2950 2961. Colantuoni, V., Gottesman, M.E., and Sancar, A. (2001). Preservation Edmunds, L.N., Jr. (1988). Cellular and Molecular Bases of Biological of light signaling to the suprachiasmatic nucleus in vitamin A-defi- Clocks (New York: Springer-Verlag). cient mice. Proc. Natl. Acad. Sci. USA 98, 11708 11713. Foster, R.G., and Helfrich-Forster, C. (2001). The regulation of circa- Toh, K.L., Jones, C.R., He, Y., Eide, E.J., Hinz, W.A., Virshup, D.M., dian clocks by light in fruitflies and mice. Philos. Trans. R. Soc. Ptacek, L.J., and Fu, Y.H. (2001). An hper2 phosphorylation site Lond. B Biol. Sci. 356, 1779 1789. mutation in familial advanced sleep phase syndrome. Science 291, Froehlich, A.C., Liu, Y., Loros, J.J., and Dunlap, J.C. (2002). White 1040 1043. Collar-1, a circadian blue light photoreceptor, binding to the fre- Yanovsky, M.J., and Kay, S.A. (2002). Molecular basis of seasonal quency promoter. Science 297, 815 819. time measurement in Arabidopsis. Nature 419, 308 312. Fu, L., Pelicano, H., Liu, J., Huang, P., and Lee, C. (2002). The circadian gene Period2 plays an important role in tumor suppression and DNA damage response in vivo. Cell 111, 41 50. Glossop, N.R., Lyons, L.C., and Hardin, P.E. (1999). Interlocked feedback loops within the Drosophila circadian oscillator. Science 286, 766 768. Gooley, J.J., Lu, J., Chou, T.C., Scammell, T.E., and Saper, C.B. (2001). Melanopsin in cells of origin of the retinohypothalamic tract. Nat. Neurosci. 4, 1165. Hannibal, J., Hindersson, P., Nevo, E., and Fahrenkrug, J. (2002). The circadian photopigment melanopsin is expressed in the blind subterranean mole rat, Spalax. Neuroreport. 13, 1411 1414. Hattar, S., Liao, H.W., Takao, M., Berson, D.M., and Yau, K.W. (2002). Melanopsin-containing retinal ganglion cells: architecture, projections, and intrinsic photosensitivity. Science 295, 1065 1070. He, Q., Cheng, P., Yang, Y., Wang, L., Gardner, K.H., and Liu, Y. (2002). White collar-1, a DNA binding transcription factor and a light sensor. Science 297, 840 843. Kramer, A., Yang, F.C., Snodgrass, P., Li, X., Scammell, T.E., Davis, F.C., and Weitz, C.J. (2001). Regulation of daily locomotor activity and sleep by hypothalamic EGF receptor signaling. Science 294, 2511 2515. Lee, C., Etchegaray, J.P., Cagampang, F.R., Loudon, A.S., and Reppert, S.M. (2001). Posttranslational mechanisms regulate the mammalian circadian clock. Cell 107, 855 867. Panda, S., Antoch, M.P., Miller, B.H., Su, A.I., Schook, A.B., Straume, M., Schultz, P.G., Kay, S.A., Takahashi, J.S., and Hogenesch, J.B.