Skip to main content
Cellular and Molecular Gastroenterology and Hepatology logoLink to Cellular and Molecular Gastroenterology and Hepatology
. 2021 Sep 14;12(5):1847–1872.e0. doi: 10.1016/j.jcmgh.2021.08.001

The Circadian Clock Gene, Bmal1, Regulates Intestinal Stem Cell Signaling and Represses Tumor Initiation

Kyle Stokes 1, Malika Nunes 1, Chantelle Trombley 1, Danilo EF L Flôres 2, Gang Wu 2, Zainab Taleb 1, Abedalrhman Alkhateeb 3, Suhrid Banskota 4, Chris Harris 5, Oliver P Love 5, Waliul I Khan 4, Luis Rueda 3, John B Hogenesch 2, Phillip Karpowicz 1,
PMCID: PMC8591196  PMID: 34534703

Abstract

Background & Aims

Circadian rhythms are daily physiological oscillations driven by the circadian clock: a 24-hour transcriptional timekeeper that regulates hormones, inflammation, and metabolism. Circadian rhythms are known to be important for health, but whether their loss contributes to colorectal cancer is not known. We tested the nonredundant clock gene Bmal1 in intestinal homeostasis and tumorigenesis, using the Apcmin model of colorectal cancer.

Methods

Bmal1 mutant, epithelium-conditional Bmal1 mutant, and photoperiod (day/night cycle) disrupted mice bearing the Apcmin allele were assessed for tumorigenesis. Tumors and normal nontransformed tissue were characterized. Intestinal organoids were assessed for circadian transcription rhythms by RNA sequencing, and in vivo and organoid assays were used to test Bmal1-dependent proliferation and self-renewal.

Results

Loss of Bmal1 or circadian photoperiod increases tumor initiation. In the intestinal epithelium the clock regulates transcripts involved in regeneration and intestinal stem cell signaling. Tumors have no self-autonomous clock function and only weak clock function in vivo. Apcmin clock-disrupted tumors show high Yes-associated protein 1 (Hippo signaling) activity but show low Wnt (Wingless and Int-1) activity. Intestinal organoid assays show that loss of Bmal1 increases self-renewal in a Yes-associated protein 1–dependent manner.

Conclusions

Bmal1 regulates intestinal stem cell pathways, including Hippo signaling, and the loss of circadian rhythms potentiates tumor initiation. Transcript profiling: GEO accession number: GSE157357.

Keywords: Circadian Rhythms, Intestinal Stem Cells, Colorectal Cancer, Hippo Pathway

Abbreviations used in this paper: ANOVA, analysis of variance; BH, Benjamini-Hochberg; cKO, conditional knockout; GSEA, Gene Set Enrichment Analysis; ISC, intestinal stem cell; Lgr5+, Leucine-rich repeat-containing G-protein coupled receptor 5; LL, constant light cycle; PBS, phosphate-buffered saline; PHH3, phosphorylated histone H-3; Tnf, tumor necrosis factor; Wnt, Wingless and Int-1; Yap, Yes-associated protein 1

Graphical abstract

graphic file with name fx1.jpg


Summary

The loss of circadian rhythms in the intestine leads to aberrant regulation of stem cell signaling pathways and increased tumor initiation.

Cancer is a disease that commences with the initiation of tumors, and progresses when hyperproliferative tumor cells develop into increasingly malignant phenotypes.1 How the body’s physiology precisely contributes to tumor initiation vs progression is not clear. Circadian rhythms are daily changes in physiology driven by the circadian clock: a self-sustaining, 24-hour, transcriptional feedback mechanism comprising the transcription factors Clock/Bmal1 and their repressors Per1-3/Cry1-2.2 In addition to this core mechanism, a regulatory loop composed of Nr1d1-2 regulates the transcription of Bmal1 to confer robustness; post-transcriptional, translational, and epigenetic mechanisms also affect circadian clock function.2 Self-sustaining daily changes in the abundance and activity of the core clock components underlies 24-hour transcriptional rhythms that are present in nearly all cells of the body, where more than 40% of the genome is rhythmic.3 In doing so, the clock is thought to coordinate the timing of compatible and incompatible physiological processes throughout the body, thereby maximizing health and fitness. Daily circadian rhythms in hormones, inflammation, and metabolic processes are essential to health,4,5 and are the same physiological processes often dysregulated in tumor cells.1

Circadian disruption, an outcome of frequent activities such as shift work, increases cancer incidence,6, 7, 8, 9, 10 and therefore a link between circadian rhythms and cancer has been actively sought.11, 12, 13, 14 Cancers can harbor abnormal expression or mutations in circadian genes,15, 16, 17 but the circadian rhythmicity of cancers has not been established. For instance, breast cancer cells have been shown to have normal18 or abnormal circadian gene expression,19 raising the question of whether certain cancerous clones are clock-dead, or whether clock function is correlated with different stages of malignancy. Loss of circadian clock activity leads to cellular changes such as disruptions in metabolism,20,21 cell cycle,22,23 apoptosis,24,25 and the DNA damage response,26,27 cellular processes whose dysfunction are known to be protumorigenic. Animal models have shown that the loss of clock genes increases cancer,28, 29, 30, 31 although it is worth noting that questions have been raised about the strains of mice used in these studies,11,14,32 as well as discrepancies in tumorigenesis noted between different clock mutants.13,14 In addition to the circadian clock as a transcriptional regulator, environmental factors that cause circadian disruption are complex and involve dysfunction in the orchestration of body-wide processes.33 Thus, the effect of circadian rhythms on colorectal cancer are poorly understood. How does the complete loss of circadian rhythms in the body, vs loss at the cellular level, lead to tumor initiation or progression? This is an important question, relevant in the role of circadian health as a factor in colorectal cancer prevention and treatment.

To test the role of the clock in colorectal tumorigenesis, we used the Apcmin mouse strain, which models human colorectal cancer in a mechanistically conserved manner.34,35 This strain has only 1 functional copy of the gene Apc: intestinal epithelial cells in these mice can undergo spontaneous loss of heterozygosity of the remaining allele and subsequent tumor initiation events at random positions in the epithelium. Apc mutant tumor-initiating cells are reminiscent of proliferative intestinal stem cells (ISCs),36,37 which divide throughout life to produce differentiated intestinal cells and regenerate intestinal epithelia.38 This is because the Apc gene is a negative regulator of Wnt signaling, a pathway required for the maintenance and proliferation of Lgr5+ (Leucine-rich repeat-containing G-protein coupled receptor 5) ISCs. Inflammation is also a critical factor in regeneration,39 and potent driver of tumorigenesis in the intestine.40 Recent studies have shown that intestinal cells can assume a fetal-like state during regeneration,41,42 and inflammation can activate the Hippo tumor-suppressor pathway to promote regeneration.42, 43, 44, 45 Thus, both the Wnt and Hippo pathways are regenerative factors that are co-opted during tumorigenesis to drive tumor growth.

We previously showed that regeneration, in both Drosophila and mice, is regulated by the circadian clock.46, 47, 48 ISCs show 24-hour proliferation and cell signaling characteristic of circadian rhythmicity, and loss of clock function causes arrhythmic and dysfunctional regeneration. These same processes are regulated by the clock in epidermal stem cells, suggesting they are conserved in epithelia.49 We hypothesized that complete loss of circadian rhythms would increase intestinal tumorigenesis. Congenic Apcmin mice lacking Bmal1 have a 2-fold higher tumor initiation probability, which is recapitulated in Apcmin mice housed under constant light. In the intestinal epithelium, the circadian clock regulates a small number of rhythmic genes, including the Hippo signaling pathway transducer Tead4. However, Apcmin tumors show reduced circadian rhythms in vivo, and tumor cells have no capacity to generate cell-autonomous circadian rhythms in vitro, showing that loss of rhythms is linked to. tumorigenesis. Furthermore, clock-dead Apcmin + Bmal1 mutant tumor organoids down-regulate the Wnt pathway and up-regulate the Hippo pathway, and show increased Hippo-dependent self-renewal. Our results show that the circadian clock is a physiological repressor of early tumor-initiation events, and that the core clock gene Bmal1 participates in the remodeling of stem cell–regenerative signaling pathways in the epithelium.

Results

Loss of Bmal1 Increases Tumor Initiation in the Intestine

Bmal1-/- mutant mice are viable littermates from heterozygous crosses but show no circadian rhythms or clock activity.50 We crossed Apcmin mice on a C57BL/6 background with Bmal1-/- null mutant mice also on a C57BL/6 background, then offspring were backcrossed 7 generations to create a congenic strain. Littermates from congenic heterozygous Apcmin/+; Bmal1+/- crosses were examined at 3 months of age to avoid age-related phenotypes.51 Bmal1-deficient mice have low and arrhythmic serum levels of corticosterone, a known regulator of circadian physiology52 consistent with previous findings (Figure 1A).53 Large tumors (>1 mm) and small tumors (<1 mm) were quantified throughout the intestine. Control (Apc+/+; Bmal1+/+) mice had no tumors, and neither did the Bmal1-/- mice (Apc+/+; Bmal1-/-), indicating that the loss of Bmal1 alone does not drive tumor initiation (Figure 1B–D). However, Apcmin + Bmal1-/- mice (Apcmin/+; Bmal1-/-) develop 2 times more tumors compared with Apcmin controls (Apcmin/+; Bmal1+/+) (Figure 1B–D). The presence of many small tumors suggests that tumorigenesis is higher in the absence of Bmal1.

Figure 1.

Figure 1

Loss of Bmal1 increases tumor initiation in the intestine. (A) Apc+/+;Bmal1+/+ (wild-type controls, n = 17), Apc+/+;Bmal1-/- (Bmal1-/- mutants, n = 20), Apcmin;Bmal1+/+ (Apcmin controls, n = 17), and Apcmin;Bmal1-/- (Apcmin + Bmal1-/- mutants, n = 18) were tested. Blood serum corticosterone levels were higher and diurnal in control and Apcmin control genotypes, but invariant and low in both Bmal1-/- genotypes (ANOVA, f(6, 52) = 2.766; P = .0208). (B) Representative images showing tumors in Apcmin controls (left) and Apcmin + Bmal1-/- mutants (right). Scale bar: 500 μm. (C and D) Tumors larger than 1 mm in diameter were increased significantly in Apcmin + Bmal1-/- (ANOVA, f(3, 46) = 80.56; P < .0001), as well as the smaller (<1 mm) tumors (ANOVA, f(3, 47) = 57.41; P < .0001). (E and F) Cell proliferation (Ki67+) is more intense and higher in Apcmin + Bmal1-/- tumors (t test, P = .0338). Scale bar: 50 μm. (G and H) Mitotic cells (PHH3+) are increased in Apcmin + Bmal1-/- tumors (ANOVA, f(2, 390) = 5.936; P = .0029). Scale bar: 25 μm. (I) No significant difference in cell death (caspase 3 [Cas3]+) was observed in the tumors. (J) Apcmin + Bmal1-/- tumors show reduced tumor dysplasia (t test, P < .0001). (K) Tumor size in Apcmin + Bmal1-/- was not affected. (L and M) The number of tumors was equivalent in 1-month-old Apcmin controls compared with Apcmin + Bmal1-/- mice. (N and O) Proliferation marker (Ki67) was more intense in the tumors but the percentage of Ki67+ cells was equivalent. Scale bar: 50 μm. Histologic quantification shown for n = 10 crypts/regions per animal, and n ≥ 3 animals per condition. All error bars represent SEM. Zeitgeber Time (ZT) is indicated, where ZT 0–12 is the light phase and ZT 12–24 is the dark phase of a photoperiod cycle. DAPI, 4′,6-diamidino-2-phenylindole.

We measured proliferation in intestinal crypts and tumors using Ki67, a marker of cell-cycle activity, and phosphorylated-histone H3 (PHH3), a marker of mitosis. Apcmin + Bmal1-/- intestinal crypts and tumors show stronger expression levels of Ki67 and an increased number of Ki67+ cells (Figures 1E and F and 2A and B). PHH3 cell number is increased in Apcmin + Bmal1-/- crypts and tumors and shows daily variation (Figures 1G and H and 2E and F). This indicates that loss of Bmal1 increases proliferation in the Apcmin background. Apcmin + Bmal1-/- tissues also showed reduced cell death (Figures 1I and 2G), as marked by cleaved caspase-3. Histopathologic analysis showed that Bmal1 loss slightly reduces dysplasia (Figure 1J), but does not affect tumor size (Figure 1K). To test whether tumor growth is increased in an age-dependent fashion, we examined mice at earlier stages of tumor initiation. One-month-old mice lacking Bmal1 did not show an increase in tumor number yet (Figure 1L and M), but Ki67 expression levels and/or numbers were increased in tumors (Figure 1N and O) and in crypts (Figure 2C and D). Thus, the phenotype of Bmal1 loss of function is the generation of more tumors through increased proliferation. We conclude that in this manner Bmal1 negatively regulates tumorigenesis.

Figure 2.

Figure 2

Loss of Bmal1 alters tissue homeostasis. (A and B) Ki67+ expression was more intense and increased Ki67+ cells were present in Apcmin + Bmal1-/- crypts at 3 months of age (t test, P = .0012). Scale bar: 50 μm. (C and D) Earlier, at 1 month, these proliferation differences also were present, suggesting the tissue of Bmal1-/- mutants is primed for increased tumor initiation before tumors double in number (t test, P = .0166). Scale bar: 50 μm. (E and F) Slightly increased mitosis (PHH3+) was observed in Apcmin + Bmal1 mutant crypts at 3 months (ANOVA f(2, 218) = 3.386; P = .0352). Scale bar: 25 μm. (G) Reduced apoptosis (caspase 3 [Cas3]+) was observed in Apcmin + Bmal1-/- crypts (t test, P = .031). (H and I) β-cat positive (activated) nuclei were equal, suggesting that Wnt signaling was equally active in Apcmin control (left) and Apcmin + Bmal1 mutant (right) crypts. Scale bar: 50 μm. (J) Slightly fewer crypts were noted in Apcmin + Bmal1 mutant animals (t test, P = .0013). (K) Slightly reduced crypt depth was noted in the Apcmin + Bmal1 mutant (t test, P < .0001). (L) Slightly more cells also were found in the same (t test, P = .0123). (M and N) The number of secretory goblet cells (Alcian Blue) was higher in the Apcmin + Bmal1 mutant animals as well (t test, P < .0001). Scale bar: 100 μm. (O and P) Significantly more Paneth cells (lysozyme [Lyz]+) were present in Apcmin + Bmal1 mutant animals (t test, P = .0005). Scale bar: 25 μm. Histologic quantification is shown for n = 10 crypts/regions per animal, and n ≥ 3 animals per condition. All error bars represent SEM. β-Cat, β-catenin; DAPI, 4′,6-diamidino-2-phenylindole.

A recent study proposed that the circadian clock regulates the Wnt pathway ligand, Wnt3A, in the Paneth cells of intestinal organoids.54 Because dysfunction of Wnt is a driver of tumorigenesis, we tested if increased Wnt signaling disruption precedes the increase in Apcmin + Bmal1-/- tumor initiation. β-catenin, the transducer of Wnt signaling, was unaltered (Figure 2H and I). Apcmin + Bmal1-/- untransformed crypts showed reductions in the number (Figure 2J) and size (Figure 2K) of crypts, but an increase in the overall number of crypt cells present (Figure 2L). These changes did not resemble the heightened proliferation that would be expected from Wnt increased activity. However, we found an increase in the production of secretory cell types, including Paneth cells (lysozyme-positive) and goblet cells (Alcian Blue) in the Apcmin + Bmal1-/- intestine (Figure 2M–P). These data are consistent with previous reports showing Hippo pathway effects on secretory cell differentiation.43,55

The Normal Epithelium Shows Self-Sustaining Circadian Clock Function, but Tumors Do Not

To test how Bmal1 functions as a transcriptional regulator in the epithelial crypt, we derived organoids from congenic wild-type control mice (Apc+/+; Bmal1+/+) vs Bmal1-/- mice (Apc+/+; Bmal1-/-). Twelve hours after passaging, organoids were synchronized using a single pulse of the glucocorticoid dexamethasone,46 and then fractions were collected every 2 hours from 24 to 48 hours after synchronization to avoid immediate early response gene effects.56 RNA was sequenced, and transcriptomes were analyzed by MetaCycle57 (Figure 3A). Comparing wild-type vs Bmal1-/- organoid circadian expression showed that only 41 genes are expressed rhythmically in a Bmal1-dependent manner (base expression, >0.5; relative amplitude, >0.05; BH (Benjamini-Hochberg), q <0.1) (Figure 3B, Supplementary Table 1). These include well-known circadian clock components such as Bmal1 itself, Nr1d1-2, Nr1f3, Per3, Cry1-2, and Tef (Figure 3C, Supplementary Table 1). Additional rhythmic targets, previously implicated in ISC biology, are notable including the markers: Anxa2, 7, 858; differentiation regulators: Bmp3, Cav2, EphB336; growth regulators: Nras1; DNA repair component: Parp358; and regeneration regulators: Kit and Tead455,59 (Figure 3C, Supplementary Table 1). No cell-cycle regulators were detected. A second organoid assay, performed over 2 days to test a subset of the top rhythmic genes, showed that circadian clock genes showed consistent self-sustaining 24-hour rhythms (Figure 3D). However, the Hippo pathway component Tead4 showed time-dependent variation that had less consistent 24-hour rhythmicity. These results suggest that Bmal1 regulates clock transcription in the normal intestinal epithelium, but that its targets are limited to a small number of genes that include several known ISC regulators.

Figure 3.

Figure 3

Analysis of circadian transcription in the crypt epithelium. (A) Organoids derived from the ileum of control and Bmal1 mutant mice were passaged, then synchronized using dexamethasone. RNA was isolated at 2-hour intervals from 24 to 48 hours in free-running conditions. RNA sequencing data were analyzed for circadian rhythmicity using MetaCycle’s meta2D function (n = 2 organoid lines/genotype). (B) Heat maps showing median-normalized gene expression (Exp./Med.) ordered by MetaCycle phase to depict all significant circadian genes found in controls (41 genes). The same genes were arrhythmic in Bmal1-/- mutants. (C) Graphs showing cycling genes from the RNA sequencing: Nr1d1 (q = 0.00039), Cry1 (q = 0.00039), Bmp3 (q = 0.057), Ephb3 (q = 0.0084), Kit (q = 0.067), and Tead4 (q = 0.0098). Mutants do not show circadian rhythms in any genes (q > 0.1). (D) Two-day quantitative polymerase chain reaction (qPCR) was used to validate the results of the RNA sequencing. Pmpca was used as the endogenous control, the gene that was determined by RNA sequencing to be stable across 24 hours in all genotypes. All error bars represent SEM. Dex, Dexamethasone; RPKM, Reads Per Kilobase of transcript, per Million mapped reads.

We performed the same experiment using Apcmin control organoids (Apcmin/+; Bmal1+/+) and Apcmin + Bmal1-/- organoids (Apcmin/+; Bmal1-/-) derived from microdissected tumors (Figure 4A). Apcmin organoids express 238 rhythmic genes and Apcmin + Bmal1-/- express 263, using the same criteria as described earlier (Figure 4B, Supplementary Table 1). The circadian clock was dysfunctional in both genotypes. Even control tumors with wild-type Bmal1 do not have self-sustaining circadian clock transcription, indeed these down-regulate most clock components (Supplementary Table 2). Only 4 rhythmic genes overlapped between genotypes, showing that the 24-hour variation in gene expression is unique (Figure 4C). The top genes in Apcmin (Golga1, Dolk, Rnf128) or Apcmin + Bmal1-/- (Mlxipl, Msmo1, Fam76b) were tested over 2 days, and although some showed variation over 24-hour organoid culture (Figure 4D), these were not self-sustaining circadian rhythms over 48 hours, suggesting stochastic expression, with the exception of Rnf128 and Msmo1, which have 24-hour rhythms (Figure 4E). We conclude that tumor organoids show 24-hour variation in gene expression, independent of Bmal1, but have no autonomous, self-sustaining, rhythmic, transcriptional clock mechanism present.

Figure 4.

Figure 4

Analysis of circadian transcription in the tumor epithelium. (A) Tumor organoids were derived from Apcmin and Apcmin + Bmal1-/- mice, and then RNA was analyzed by sequencing as shown in Figure 2 (n = 2 organoid lines/genotype). (B) Heat maps median-normalized gene expression (Exp./Med.) ordered by MetaCycle phase show all rhythmic genes. (C) Venn diagram showing that there is nearly no overlap between rhythmic transcripts identified from wild-type and mutant samples. (D) Graphs of the RNA sequencing of the top 3 genes from each genotype: Golga1 (q = 0.015), Dolk (q = 0.015), Rnf128 (q = 0.015), Mlxipl (q = 0.0053), Msmo1 (q = 0.0053), and Fam76b (q = 0.0055). (E) Validation test using 2-day quantitative polymerase chain reaction (qPCR) indicated that only Rnf128 (ANOVA, f(12,24) = 1.378; P = .2427) and Msmo1 (ANOVA f(12, 26) = 4.613; P = .0005) show self-sustaining rhythms. All error bars represent SEM. Dex, Dexamethasone; RPKM, Reads Per Kilobase of transcript, per Million mapped reads.

Epithelial Self-Renewal Is Regulated by Bmal1

We next compared changes in gene expression between genotypes over the full 24-hour period. There were 111 genes changed, either up-regulated or down-regulated, when comparing Bmal1+/+ controls and Bmal1-/- mutant organoids (Supplementary Table 2). Bmal1-/- organoids showed 67 genes increased in expression (Figure 5A, Supplementary Table 2), which included inflammatory markers such as Reg3a, Reg3b, and Cntf, indicative of a stress/inflammatory state.60 Compared with nontransformed wild-type controls, Apcmin tumor organoids up-regulate genes involved in proliferation, Wnt signaling, and inflammation, but down-regulate genes involved in metabolism and clock activity (Supplementary Tables 2 and 3). This is consistent with the role of Apc as a Wnt pathway repressor. Apcmin + Bmal1-/- organoids up-regulate or down-regulate 2398 genes when compared with Apcmin organoids (Supplementary Table 2). Of note, Wnt target genes and apoptosis genes were down-regulated in the absence of Bmal1, but Hippo target genes, as well as many genes involved in proliferation, were up-regulated (Figure 5B and C, Supplementary Tables 2 and 3). To confirm the increase in Hippo signaling, we performed Gene Set Enrichment Analysis (GSEA)61 using a gene set containing genes previously found to be regulated by the Hippo pathway transducers Yap and Taz in the intestinal epithelium.43 Yes-associated protein 1 (Yap)–up-regulated genes are enriched in Apcmin; Bmal1-/- organoids (Figure 5D), consistent with the role of the Hippo pathway in promoting cell survival.

Figure 5.

Figure 5

Epithelial self-renewal is regulated by Bmal1. (A) Heat map showing 24-hour fold change in relative mean expression of Bmal1-/- organoids compared with control organoids. Circadian clock genes were down-regulated, and inflammation markers were up-regulated in Bmal1 mutant organoids compared with controls. (B) Heat map showing 24-hour fold change in relative mean expression of Apcmin + Bmal1-/- organoids compared with Apcmin control organoids. Wnt pathway targets were down-regulated, whereas Hippo pathway targets were up-regulated. Cell proliferation genes also showed an increase in Apcmin + Bmal1-/-. (C) GO (Gene Ontology) analysis of pathways down-regulated by Apcmin + Bmal1 mutant tumor organoids, relative to Apcmin controls. Wnt signaling was decreased, whereas cell proliferation was increased in Bmal1 mutants, together with changes in transcriptional regulation. A Benjamini cut-off value of 0.05 was used for GO analysis. (D) Apcmin + Bmal1-/- have up-regulated Yap signaling compared with Apcmin controls (enrichment score, 0.44; normalized enrichment score, 2.083), nominal P value = .000, False Discovery Rate (FDR) q value = 0.000, Family Wise Error Rate (FWER) P value = .000). (E) After derivation, individual organoids from each genotype were dissociated into single cells and plated separately to assess the number of subclones, and the number of crypt buds per subclone. (F) Representative images of organoid subclones. Scale bar: 100 μm. (G) Individual Bmal1-/- organoids developed more subclones (t test, P = .0004), indicating increased self-renewal ability. (H) Bmal1-/- subclones had more crypt buds, indicating further increased self-renewal of ISCs (t test, P < . 0075). (I) Schematic shows clonal analysis of Apcmin organoids. (J) Representative images of Apcmin and Apcmin + Bmal1-/- organoid subclones. Scale bar: 100 μm. (K) Apcmin + Bmal1-/- tumor organoids produce more subclones indicating increased self-renewal (t test, P = .0444). (L) Apcmin control and Apcmin + Bmal1-/- mutant organoids were treated with Yap inhibitor; dimethyl sulfoxide (DMSO) is the vehicle control. Apcmin + Bmal1-/- organoids showed increased expression of the Yap target gene Ereg, which was reduced by Yap inhibitor (ANOVA, f(3,10) = 19.55; P = .002). (M) Organoids were pretreated with Yap inhibitor before subcloning. Apcmin + Bmal1-/- organoids showed increased self-renewal, which was disrupted by pretreatment of Yap inhibitor (ANOVA, f(3, 24) = 7.087; P = .00144). Data show n = 10 clones from 2 to 3 lines for quantifications. All error bars represent SEM. Inflam, Inflammation; qPCR, quantitative polymerase chain reaction; Red-Ox, Reduction-Oxidation; Rel Mean Expr, Relative Mean Expression; Sig, Signalling; Yap-i, Yap-inhibitor.

A tumor-initiating cell’s ability to generate a tumor is thought to be related to self-renewal, a property of adult tissue stem cells.62 Both crypt progenitors and/or ISCs contribute to tumor initiation,37,63 and self-renewal can be tested in organoid culture, where single Lgr5+ ISCs64 or Lgr5- intestinal precursors can generate organoids.65 We developed an organoid assay to measure self-renewal directly, by directly subcloning 1 single organoid dissociated into single cells and measuring secondary clone number and crypt formation in the resulting subclones (Figure 5E). This showed self-renewal at the level of a single organoid, spawned from 1 stem cell, rather than from a population whose secondary organoid numbers are an average that obscures individual organoid self-renewal. Based on our results, we predicted that Bmal1-/- epithelial cells, which have higher tumor initiation (Figure 1B–D), would have increased self-renewal. Indeed, nontransformed Bmal1-/- organoids produced twice as many secondary organoids from a single organoid (Figure 5F and G), and these secondary organoids had many more crypts present, further indicating increased self-renewal (Figure 5H). Transformed Apcmin + Bmal1-/- organoids also showed higher self-renewal ability (Figure 5I–K), suggesting that both nontransformed and transformed Bmal1-/- epithelial cells have an organoid initiation advantage. To test the role of the Hippo pathway, we treated Apcmin + Bmal1-/- organoids with small-molecule inhibitors of Yap and its binding partner Tead.55,66 Yap inhibition reduces the expression of Ereg, a critical Yap/Taz target gene that regulates ISC growth43 (Figure 5L), but does not affect the expression of the Wnt pathway target Axin2 (not shown). Treatment of Apcmin; Bmal1-/- organoids with the Yap inhibitor abolished their increased self-renewal (Figure 5M), and treatment with a Tead4-specific inhibitor showed a similar effect (not shown). Overall, these results suggest that Bmal1 suppresses a Hippo-dependent, self-renewal pathway in the intestinal epithelium, and in its absence the self-renewal of ISCs or tumor-initiating clones is increased.

Epithelial Bmal1 Contributes to Increased Tumor Initiation

To what extent does the epithelium play a role in the increased tumor initiation of Bmal1-/- mutants in vivo? To address this question, we crossed Apcmin mice with Villin-Cre67 and Bmal1flox/flox mice68 to delete exon 8—the DNA transactivation domain—thereby conditionally abolishing Bmal1 and thus circadian transcriptional activity only in the epithelial cells in the Apcmin background (Figure 6A). Three-month-old Apcmin conditional knockout (cKO) control mice (Apcmin/+; Bmal1flox/flox) and Apcmin + Bmal1 cKO mice (Apcmin/+; Vilcre/+; Bmal1flox/flox) were tested, showing that the Apcmin + Bmal1 cKO mice had an equal number of large tumors, but 2-fold higher small tumors (Figure 6B and C). This shows that loss of Bmal1 in the epithelium partially recapitulates the full mutant phenotype.

Figure 6.

Figure 6

Epithelial Bmal1 contributes to increased tumor initiation. (A) Bmal1 was expressed in the intestine of controls, but was absent in the intestine of Bmal1 cKO (Apcmin; VilCre/+; Bmal1fl/fl). In the liver, both controls and cKO expressed Bmal1, indicating the specific deletion of Bmal1 from the intestine (f(3,8) = 11.48; P = .0026; n = 3 per genotype). (B and C) Apcmin + Bmal1 ckO (n = 7) have an equivalent number of tumors greater than 1 mm, but significantly more tumors smaller than 1 mm, compared with Apcmin cKO controls (n = 8) (t test, P = .027). (D) Tnf is higher in Apcmin +_Bmal1-/- mutants compared with controls and both of the cKO genotypes (ANOVA, f(3, 16) = 8.42; P = .0014; n = 4–6 mice per genotype). (E and F) Tumor tissue from Apcmin + Bmal1-/- tumors show an increased number of infiltrating CD45+ blood cells than the other genotypes (ANOVA, f(3, 65) = 6.824; P = .005). Histologic quantification shown for n = 10 regions per animal, and n ≥ 3 animals per condition. Scale bar: 10 μm. All error bars represent SEM. DAPI, 4′,6-diamidino-2-phenylindole; qPCR, quantitative polymerase chain reaction; Zeitgeber Time.

Are there other factors that drive tumorigenesis in the Bmal1-/- mutant? The circadian clock regulates many different rhythms in physiology, including daily cycles of metabolism and the endocrine and immune systems.4,12 Inflammation is an important driving factor in colorectal tumorigenesis,39 and the circadian clock also recently was shown to regulate inflammation in the intestine.69,70 We therefore asked whether Apcmin + Bmal1-/- mice have increased inflammation, which together with the epithelium could contribute to tumor initiation. Blood was analyzed from the full-body KO and cKO mice, and tested for proinflammatory cytokines. Analysis of a panel of cytokines showed that Apcmin + Bmal1-/- mice have increased levels of Tumor Necrosis Factor (Tnf) (Figure 6D), a clock-regulated proinflammatory cytokine.46 However, Apcmin + Bmal1 cKO mice did not show this increase in Tnf. To test whether local inflammation surrounding the epithelium is higher, CD45+ blood cell infiltration into tumors was quantified. cKO controls, Apcmin + Bmal1 cKO, and Apcmin + Bmal1+/+ tumors, have equivalent lower numbers of CD45+ cells, while Apcmin + Bmal1-/- tumors have significantly higher CD45+ numbers, indicating a proinflammatory state (Figure 6E and F). We conclude that global circadian disruption in Apcmin + Bmal1-/- mice drives a protumorigenic inflammatory phenotype that is not present in the Apcmin + Bmal1 cKO.

Yap Activity Is Increased in Bmal1 Mutant Tumors

The Hippo signaling transducer, Yap, is required for tumorigenesis in the Apcmin colorectal cancer model.43 We therefore probed Apcmin controls and Apcmin + Bmal1-/- with an antibody marking Yap to determine if Hippo signaling is perturbed when Bmal1 is absent. Apcmin + Bmal1-/- epithelial tumors have higher levels of Yap, and the signal shows an active/nuclear localization compared with Apcmin controls (Figure 7A and B). Protein was extracted from the tumors of Apcmin control and Apcmin + Bmal1-/- mice and analyzed by Western blot to further test Hippo activity. Both total Yap and activated Yap (nonphosphorylated S127) are higher in Apcmin + Bmal1-/- tumors (Figure 7C). This shows that Yap expression and activity is higher in Bmal1 mutant tumors consistent with the Yap signature of Apcmin + Bmal1-/- tumor organoids (Figure 5D).

Figure 7.

Figure 7

Yap activity is increased in Bmal1 mutant tumors. (A and B) Representative images showing increased Yap+ nuclear signal in the cells of Apcmin + Bmal1-/- tumors (arrowheads show the cell nucleus in inset image ). Yap-activated nuclei are present in higher numbers (t test, P = .0186). Scale bar: 50 μm. (C) Western blots showing increased levels of total and activated Yap in Apcmin; Bmal1-/- mutant tumors. (D and E) Representative images showing increased Yap+ nuclear signal in the cells of Apcmin + Bmal1 cKO that are present in higher numbers (t test, P = .0009). Scale bar: 50 μm. (F and G) Control tumors show higher expression of Lgr5 RNA (blue) while Apcmin + Bmal1 cKO tumors show higher Tead4 RNA expression (red). The area of Tead4+ cells is significantly higher in Apcmin + Bmal1 cKO (ANOVA f(3, 36) = 9.241; P = .0001). Scale bar: 50 μm. (H and I) Apcmin + Bmal1 cKO tumors show higher levels of the Yap target gene Ereg (blue), but lower levels of the Wnt target gene, Axin2 (red), relative to controls. The area of Ereg+ cells is significantly higher (ANOVA f(3, 36) = 9.422; P < .0001). Histologic quantification shown for n = 5–10 regions per animal, and n ≥ 3 animals per condition. Scale bar: 100 μm. All error bars represent SEM.

To address whether this Yap increase is owing to epithelial Bmal1 function, Apcmin cKO control mice and Apcmin + Bmal1 cKO mice were tested for markers of the Wnt and Hippo pathways. Apcmin + Bmal1 cKO mice have increased levels of Yap localized in the nucleus (Figure 7D and E) similar to those in Apcmin + Bmal1-/- mutants. Expression of the Hippo pathway component Tead455 also was increased in Apcmin + Bmal1 cKO tumors (Figure 7F and G). We further probed for the Wnt target, Axin2, and the Yap target, Ereg, in the Apcmin + Bmal1 cKO tumors. The Bmal1 cKO tumors have lower Wnt but higher Hippo pathway activity (Figure 7H and I). The results of the Axin2/Ereg analysis mirror those of the Lgr5/Tead4 expression almost exactly, both indicating that the Bmal1 tumors have higher Hippo signaling at the expense of Wnt. Of note, Wnt markers (Lgr5 and Axin2) and Hippo markers (Tead4 and Ereg) are enriched in separate cells, suggesting these reflect changes in the tumor cell population. Together, these data show Bmal1 cKO tumors have lower Wnt but higher Hippo activity, consistent with the Bmal1-dependent transcriptional program in Apcmin + Bmal1-/- organoids (Figure 4B–D). We conclude that Bmal1 negatively regulates the Hippo pathway in the tumor epithelium and, in its absence, these display higher Hippo activity.

In Vivo Tumors Have Weak Circadian Rhythms

Because tumor organoids do not show rhythmic circadian clock transcription in vitro (Figure 4), we asked whether circadian clock disruption is a general feature of intestinal tumors. We re-examined Apcmin control mice, housed under normal photoperiod, to determine whether heterogeneity in clock activity or Wnt/Hippo signaling is present in the different tumors initiated. Individual tumors from a cohort of mice over a 24-hour timeline were microdissected (n = 30 mice, 5 per time point), and compared with adjacent nontransformed tissue for gene expression (n = 180 tumor/tissue pairs, n = 30 per time point). Apcmin tumors showed lower expression of the clock genes Nr1d1, Bmal1, and Per2 than the surrounding nontransformed tissue, and at the population level (average) clearly showed reduced circadian rhythms in vivo (Figure 8A–F). This expression was heterogenous but nearly all individual tumors showed lower clock gene expression than the surrounding nontransformed tissue. We further tested circadian clock activity in the epithelium by staining for the clock component, Nr1d1,2 in Apcmin controls and Apcmin + Bmal1-/- mutants. Nr1d1 showed daily changes in its levels and nuclear localization in the normal crypt epithelium of Apcmin control animals (Figure 8G and H). Apcmin + Bmal1-/- crypts did not show these daily fluctuations but high Nr1d1 levels at all times examined. In tumors present in the same animals tested, Nr1d1 levels fluctuated in tumors in Apcmin controls at the same time as nontransformed tissue, and Apcmin + Bmal1-/- had high levels at all times (Figure 8I and J). Nr1d1 protein in tumors thus showed diurnal rhythms such as the surrounding tissue, corroborating the RNA analysis. Overall, we conclude that weak diurnal rhythms in clock components persist in tumors in vivo.

Figure 8.

Figure 8

In vivo tumors have weak circadian rhythms. (A and B) Quantitative reverse-transcription polymerase chain reaction (RT-qPCR) graphs show transcript levels obtained from Apcmin mice housed under a light dark cycle. Individual tumors were compared directly with their adjacent nontransformed tissue taken from the ileum of the small intestine. Six time points over 24 hours were tested, from 5 mice per time point (n = 30 tumors per time point). Upper graphs shows individual tumor expression, and lower graphs show the average of that time point. Nr1d1 expression in individual tumors is lower but rhythmic (2-way ANOVA, f(5, 168) = 12.01; P < .0001). (C and D) Bmal1 (2-way ANOVA, f(5, 168) = 6.49; P < .0001) also is lower but rhythmic. (E and F) Per2 shows lower expression in tumors relative to tissue as well (f(1, 168) = 25.50; P < .0001). (G and H) Nr1d1 antibody in nontransformed intestinal crypts tested at 3 time points over 24 hours (Zeitgeber Time [ZT]4, 12, 20). White outlines shows the crypt border. Apcmin controls have diurnal changes in Nr1d1 in crypt cells, while Apcmin + Bmal1-/- nontransformed crypts show arrhythmic levels (2-way ANOVA, f(2, 116) = 351.9; P < .0001). (I and J) Nr1d1 staining in tumors from the same animals. Apcmin control tumors show diurnal changes, while Apcmin + Bmal1-/- tumors are again arrhythmic (2-way ANOVA, f(2, 27) = 127.4; P < .0001), n = 3 mice per time point. Scale bar: 25 μm. Histologic quantification shown for n = 10 crypts/regions and n ≥ 3 animals per condition. All error bars represent SEM. DAPI, 4′,6-diamidino-2-phenylindole.

A circadian regulation of the Wnt pathway was reported previously in organoids,54 hence we examined Wnt signaling in these same samples. In vivo, the intestine showed daily changes in the Wnt pathway targets Axin2, Lgr5, Ccnd1, and cMyc (Figure 9A–H). The Hippo pathway genes Ereg and Tead4 similarly showed diurnal changes (Figure 9I–L). Although tumors showed heterogeneity in the expression of Wnt/Hippo target genes, at the population level daily changes were evident, with tumors generally showing increased levels of these cancer and proliferation-related pathways. To further analyze these data, we tested the Pearson correlations of clock genes to Wnt genes in all of these samples. These showed that the normal correlations between the timing of clock and Wnt activity were altered in tumors (Figure 9M and N). For instance, the negative correlation between Bmal1 and Per2 broke down, whereas correlations between Nr1d1 and Axin2, cMyc, or Ccnd1 increased. Wnt pathway genes were highest toward the end of the light phase (Zeitgeber Time 8–12), and these rhythms were exacerbated in tumors despite their lower clock activity. These data indicate that individual tumors are heterogenous but all of them had reduced diurnal patterns of circadian clock components that did not correlate to reductions in protumorigenic signaling.

Figure 9.

Figure 9

Analysis of circadian clock, Wnt and hippo genes in the intestine. Quantitative reverse-transcription polymerase chain reaction (RT-qPCR) graphs from same samples as shown in Figure 8. The Wnt target genes were as follows: (A and B) Lgr5 (2-way ANOVA, f(5, 168) = 2.50; P = .033), (C and D) Axin2 (2-way ANOVA, f(5, 168) = 9.24; P = .0027), (E and F) Ccnd1 1-way ANOVA, f(1, 168) = 8.947; P = .0032), and (G and H) cMyc (2-way ANOVA, f1, 168) = 71.98; P < .0001), and were higher in tumors but also had diurnal changes similar to the surrounding tissue. (I and J) The Hippo component Tead4 is diurnally rhythmic in the intestine and its tumors (2-way ANOVA, f(11, 168) = 5.125; P < .0001). (K and L) The Hippo target Ereg, is rhythmic in tumors but with a different rhythm than surrounding tissue (2-way ANOVA f(5, 168) = 2.746; P = .02). (A and B) Pearson correlation showing the relationship between clock genes and Wnt pathway targets in nontransformed intestine (tissue) vs transformed tumors (tumor). Tissue normally shows a negative correlation (dark blue) between Bmal1 and Nr1d1, or Per2, and weaker correlation between Per2 and Nr1d1 and Wnt target genes (Axin2, cMyc, and Ccdn1). Tumors show loss of negative clock correlations, indicating weakened circadian transcription (where positive and negative regulators are expected to be antiphasic and thus negatively correlated). Increased positive correlation (dark red) between clock targets Per2 and Nr1d1 and Wnt target genes was observed in tumors. This indicates clock function was weakened, but daily rhythms in Wnt signaling were present in tumors. Error bars represent SEM.

Circadian Rhythm Loss Potentiates Tumor Initiation

Circadian rhythms in mice are entrained by photoperiod, and the behavioral and physiological rhythms that ensue consolidate circadian clock function in tissues throughout the body.71 Our results thus far show the effects of Bmal1 loss in tumor initiation; however, it is not clear if this is a feature of disrupted circadian rhythms or noncircadian Bmal1 functions. To address this, we exposed Apcmin mice to constant light levels (LLs) that lead to circadian disruption72 and quantified tumor incidence at 3 months of age. Wild-type mice did not develop tumors in constant light, but Apcmin mice showed a 2-fold increase in both small (<1 mm) and large (>1 mm) tumor initiation in constant light compared with those under normal photoperiod (light dark cycle) (Figure 10A–C), essentially recapitulating the effects observed in the Apcmin + Bmal1 mutant (Figure 1C and D). This confirms that disruption of circadian rhythms itself was able to increase tumor initiation in the Apcmin background. We further tested Hippo signaling and cell proliferation in the LL circadian-disrupted tumors to see whether the same Bmal1-dependent increases were present. Apcmin mice exposed to constant light showed an increase in Yap-positive tumor cells, with concentrated staining in the nuclei (Figure 10D and E) identical to both the full body and conditional Bmal1 knockout mice (Figure 7A, B, D, and E). Ki67 was increased in the crypts (Figure 10F and G) and tumors of Apcmin LL mice (Figure 10H and I), similar to the Bmal1-/- mutant (Figure 1E and F). These data show that environmental disruption to circadian entrainment through increased light exposure leads to a Yap-active tumor phenotype that resembles the Bmal1 loss of function phenotype.

Figure 10.

Figure 10

Circadian rhythm loss potentiates tumor initiation. Control mice and Apcmin mice were tested on a normal 12-hour light/12-hour dark photoperiod (light dark cycle [LD]) vs constant light (LL, 700 Lux) (n = 5–6 per genotype). (A) Representative images of tumors found in LD and LL conditions. Scale bar: 200 μm. (B and C) Apcmin mice in LL develop significantly more large tumors greater than 1 mm in size (ANOVA, f(3, 20) = 20.58; P < .0001), and small tumors of less than 1 mm (ANOVA, f(3, 20) = 28.71; P < .0001) than LD mice, indicating that constant light recapitulates the Bmal1 mutant phenotype. No tumors were observed in control mice, under both LD and LL conditions, without the Apcmin allele present. Error bars represent SEM. (D and E) Representative images of total Yap-stained tumors in Apcmin (LD) and Apcmin (LL) show increased Yap+ nuclear signal in the cells of Apcmin + Bmal1-/-, which are present in higher numbers (t test, P = .0023). Scale bar: 50 μm. (F and G) Cell proliferation (Ki67+) is more intense and increased in Apcmin + Bmal1-/- crypts (t test P = .0341). Scale bar: 50 μm. (H and I) Proliferation also is increased in LL tumors (t test P = .0447). Histologic quantification shown for n = 10 crypts and n ≥ 3 animals per condition. Scale bar: 50 μm. All List of Antibodies Used error bars represent SEM.

Discussion

The effect of circadian rhythms on tumorigenesis is a long-standing question. Studies have shown that shift workers have a higher cancer incidence,6,7,9,10 suggesting that circadian rhythm disruption is a risk factor. However, the epidemiologic data are not yet conclusive,14 and studies using animal models to test how clock genes relate to cancer also have been equivocal.13 Early reports have indicated that lymphoma was increased in Per2 mutant mice,28 and this same strain recently was shown to have increased liver and lung tumors.30,31 Indeed, Per2 mutant mice have increased intestinal tumorigenesis when crossed to the Apcmin model.29 However, Per2 is redundant with its homologs Per1 and Per3, and the circadian clock is active in the Per2 mutant, which has circadian rhythms present.2,73 Hence, it is not clear if the increased tumorigenesis resulting from Per2 loss is owing to its role as a circadian clock gene, or to its nonclock functions.

Using the Apc model of colorectal cancer we show that tumor initiation is regulated negatively by the circadian clock. Loss of the nonredundant clock gene, Bmal1, in a congenic Apcmin mouse strain, and circadian disruption in the Apcmin strain effected by constant light, both increased tumor number (Figures 1B–D and 10A–C). Conditional deletion of Bmal1 in the intestine also increased tumor number (Figure 6B and C), showing that Bmal1 and/or the epithelial clock specifically played a role in suppressing tumor initiation. These experiments confirmed that circadian rhythm disruption in the epithelium is a causal factor in tumorigenesis. Both Bmal1-/- organoids and Apcmin + Bmal1-/- organoids have increased self-renewal (Figure 5E–K), consistent with the idea that these have an enhanced ability to initiate tumors. Although proliferation was increased, we did not observe that Bmal1 loss increased tumor size (Figure 1K), suggesting that the suppressive effects of Bmal1 may be more complex than simply driving general tumor growth. These data provide insight into the precise stages of tumorigenesis, in which the circadian clock exerts its effects in this tissue.

Bmal1 loss decreases tumorigenesis in the skin,49 as does its binding partner Clock,74 whereas its negative regulator Per2 has the opposite effect in the skin and lung.30,49 A recent study showed that the clock component Nr1d1 is a tumor suppressor, whose activation in animal models dramatically reduced cancer.75 Further studies are needed to determine whether other clock genes have distinct effects on Apcmin tumor initiation. Our results add to a growing body of literature showing that clock gene disruption leads to cancer in a tissue-specific manner.

Recent studies have shown that tumors in the blood and brain both show and require circadian clock gene function.76,77 How is this possible? Is each tumorigenic event different, so that some tumors require clock function while others do not? Using circadian wild-type Apcmin mice, we found that in nearly all intestinal tumors, clock gene rhythms were present but at lower amplitude than the surrounding tissue in vivo (Figure 8A–F). Organoids derived from these tumors do not show self-autonomous clock gene transcription rhythms (Figure 4, Supplementary Table 1), showing that these cells lack a circadian oscillator. In vivo, circadian rhythmicity throughout the body is driven by hormones, responses to feeding, or other daily environmental changes experienced by the individual.52,78 The timing of food availability has been shown to improve circadian clock expression rhythms in tumor cells,79,80 and can even rescue Bmal1 loss of function in the liver.81 This explains how Apcmin tumor cells mimic the host circadian program in vivo, aligning their expression of circadian clock genes with the normal physiological rhythms that surround them. This disrupted clock function in tumors is consistent with reports that cMyc, increased in Apcmin tumors (Figure 9G and H), inhibits circadian clock transcription.20,82,83 Indeed, we noted the presence of a cMyc transcriptional signature in Apcmin + Bmal1-/- organoids compared with Apcmin control (Supplementary Table 5).

Despite their core clock deficiency, Apcmin tumor organoids showed 24-hour variation in hundreds of transcripts in vitro, and Apcmin + Bmal1-/- tumor organoids showed 24-hour variation in another set of transcripts (Supplementary Table 1). We cannot rule out that some of these 24-hour variations in organoids without a functional clock are false positives caused by genome-wide testing of thousands of genes; nevertheless, the ability of Apcmin + Bmal1-/- organoids to show rhythms is a curious phenomenon, previously found to occur in both transcription and metabolism.84, 85, 86 Further experiments are required to classify the nature of these self-sustained oscillations in the absence of the core transcriptional oscillator, and their possible biological implications in organoid culture. In contrast to the tumor epithelium, the nontransformed epithelium shows robust circadian clock activity87,88 (Figures 8 and 9). Using organoid cultures, we detected 41 self-sustaining rhythmic genes dependent on Bmal146 (Figure 3). Other studies have detected many more rhythmic genes in the intestine.89, 90, 91, 92, 93 Our RNA sequencing approach differed from these studies in several important respects: (1) testing only the cells of the intestinal epithelium, enriched for crypt precursors present in the first 2 days of organoid cultures; (2) using the Bmal1-/- mutant to distinguish putative core clock targets from rhythmic but non–Bmal1-regulated genes; (3) testing circadian gene expression under free-running in vitro conditions to exclude environmental responses (such as photoperiod or feeding) from self-sustaining circadian rhythmicity; and (4) performing high-resolution transcriptomic analysis, which would exclude stochastic noncircadian changes in daily gene expression. We have attempted to follow the guidelines proposed for optimal detection of circadian transcription,56 and it is apparent that epithelial crypt cells have relatively few rhythmic circadian gene targets. Among these are genes known to be involved in ISC biology, suggesting the circadian clock does indeed regulate aspects of tissue renewal of interest for future study. An important caveat to our analysis is that the gene expression of differentiating cells may obscure circadian expression and therefore our analysis should not be considered exhaustive. Additional tests of specific differentiated cell types in organoids, or longer (ie, 48-hour) timelines, would be informative.

Although our study has focused on the epithelium, other aspects of physiology that are dysregulated in Bmal1-/- mice51 also may contribute to tumor initiation. The circadian clock is known to regulate many aspects of inflammation,94 and we report that dysfunction in immune system activity may be an additional factor (Figure 6D–F), when the clock is disrupted throughout the body. It has been shown that glucocorticoid rhythms in the body suppress tumor growth.95 Our analysis, showing lower glucocorticoid levels in Bmal1-/- null genotypes (Figure 1A), also supports the additional contribution of disrupted endocrine homeostasis to tumorigenesis in Apcmin + Bmal1-/- mice. Both inflammatory and metabolic contributions to tumor growth during circadian disruption will be important to test to distinguish the effects of these contributing factors.96

This study reports circadian clock regulation of the Hippo pathway. Apcmin + Bmal1-/- organoids show higher Yap/Tead target genes (Figure 5B and D); Apcmin + Bmal1-/- mutant, Apcmin Bmal1 intestinal cKO, and Apcmin circadian-disrupted (LL) tumors show more Yap activity, and higher Tead4 and Ereg expression (Figures 7 and 10). These changes are associated with concomitant lower levels of Wnt target genes in both organoids and tumors. Wnt pathway activation is the primary cause of Apcmin-induced tumorigenesis,36 but the Hippo pathway is implicated in Apcmin tumorigenesis as well.43 Studies have shown that these pathways may be mutually antagonistic, and consistent with this notion we note that Wnt and Hippo target-expressing cells generally are separate (Figure 7F–I). We therefore hypothesize that Bmal1 negatively regulates the Hippo signaling pathway in the tumor epithelium, and Wnt pathway regulation emerges as a secondary effect. Intestinal regeneration has been proposed to involve the reprogramming of a fetal-like state with high Yap activity.42,43,97 Crypt cell populations readily dedifferentiate during regeneration,38 where Yap activity is precisely controlled before the system returns to a primarily Wnt-driven, self-renewal state.65 An endogenous timekeeper such as the circadian clock would be an ideal system to control the timing of regenerative pathway responses, thereby dictating the time when Wnt and Hippo signaling pathways are engaged during stress and regeneration. This is analogous to the role of the circadian clock in regulating signaling pathways in epidermal stem cells.49 Understanding the circadian regulation of ISC signaling will help develop cancer-preventing measures, as well as providing insight into the diagnosis and treatment of colorectal cancers in individuals subject to shift work or other forms of circadian disruption.

Methods

Animal Housing and Breeding

Mice were housed on a 12-hour light/12-hour dark cycle with ad libitum food in a barrier facility. Apcmin mice (#002020; Jackson Labs, Bar Harbor, ME) were crossed with Bmal1+/- null mutant mice (#009100; Jackson Labs). Progeny were back-crossed for 7 generations to generate a genetically pure strain before experiments began. Bmal1flox/flox mice (#007668; Jackson Labs) were crossed with VillinCre/+ mice (#021504; Jackson Labs), or Apcmin mice, then the 2 strains were crossed to obtain the Apcmin/+; Bmal1flox/flox and Apcmin/+; VilCre/+; Bmal1flox/flox for the cKO experiments. Genotyping was performed according to protocols available at Jackson Labs (www.jax.org). All animals were tested for tumor initiation at 1 or 3 months of age. Mice maintained under constant light (LL) were housed in these conditions from 21 days of age to 3 months of age. Light levels were approximately 700 Lux, measured at the back of each cage, and all objects were removed so that light fully penetrated throughout each cage. Mice were maintained under Canadian Council for Animal Care guidelines (University of Windsor: AUPP 17-21).

Tissue Collection

All animals were killed using CO2, followed by cervical dislocation at the times indicated in the figure panels. Intestines then were removed and flushed with ice-cold, phosphate-buffered saline to remove feces. For histologic analysis and tumor counting, the entire length of the small intestine was fixed in 4% paraformaldehyde (PFA) + phosphate-buffered saline (PBS) (Electron Microscopy Sciences, Hatfield, PA) for 2 hours at room temperature. The intestine was subdivided into 4 regions of equal length longitudinally: the proximal intestine closest to the stomach containing duodenum and jejunum, the proximal–medial region containing the jejunum, the distal–medial region containing the jejunum and ileum, and the distal region containing the ileum. In Figure 1 and Supplementary Figure 1, the 2 medial regions are combined. Tumors were counted visually and tumors greater than or less than 1 mm were scored by comparing size with a 1-mm diameter glass capillary tube. Tissue was frozen in Tissue-Tek OCT (Optimal Cutting Temperature) Compound (#62550-12; Electron Microscopy Sciences). Tissue used for RNA-Scope (ACD, Newark, CA) analysis was fixed in 4% PFA + PBS for 24 hours at 4ºC. Tissue used for gene expression analysis was placed in RNAlater buffer (Qiagen, Germantown, MD) at 4ºC overnight (O/N), then stored at -80ºC. Blood samples were collected in heparinized tubes, centrifuged to remove blood cells at room temperature, and then stored at -80ºC until analysis. Tnf and interleukin-6 blood cytokine measurements were performed by Eve Technologies (Calgary, Alberta, Canada). Corticosterone levels were determined by enzyme-linked immunosorbent assay (ADI-900-097; Enzo Life Sciences, Farmingdale, NY) in duplicate according to kit instructions at a 1:80 dilution with 1% steroid displacement buffer. Samples were assayed in 2 groups, with intra-assay variation of 7.3% and 7.7%, and interassay variation of a pooled mouse control was 9.5% and 9.3%.

Staining and Western Blot

Tissue was washed 3× in PBS and placed in 30% sucrose in PBS O/N. Tissue then was equilibrated in OCT for 1 hour and frozen at -80ºC. Tissue was sectioned at 10 μm using a Leica CM 1950 Cryostat (Leica Biosystems, Concord, Ontario). H&E staining and Alcian Blue staining was performed as per previous protocol.46 Immunohistochemistry was performed as previously described.46 Tumor dysplasia was blindly evaluated using a previously published scoring system: the number of adenomas and crypt architecture was assessed in the distal, medial, and proximal sites in the tissue, and the alteration in mucosa architecture, epithelial changes, hyperplasia, crypt abscesses, and crypt loss were scored from 0 to 4 (most severe) score.98,99 3,3′-Diaminobenzidine tetra hydrochloride staining was performed using horseradish peroxidase/3,3′-diaminobenzidine tetra hydrochloride (ABC) Detection IHC Kit as per the manufacturer’s protocol (ab64261; Abcam). RNA in situ probe hybridization and detection was performed using the RNAScope method according to the manufacturer’s protocol (ACD, Newark, CA). Antibodies used are shown in Table 1.

Table 1.

List of Antibodies Used

Antibody Dilution Assay Supplier Product number
Phosphorylated-histone H3 (Ser10) 1:1000 IF Upstate/Millipore (Oakville, Ontario) 06-570
Cleaved caspase 3 1:200 IF Cell Signaling Tech (Whitby, Ontario) 9661S
Lysozyme 1:200 IF Agilent/Dako (Santa Clara, CA) A0099
Nr1d1 1:250 IF Abnova (Taipei City, Taiwan) H00009572 M02
Yap (D841X) XP (R) 1:100
1:1000
DAB
WB
Cell Signaling Tech 140745
β-catenin 1:250 DAB Abcam (Toronto, Ontario) Ab32572
Ki67 1:100 DAB Abcam Ab16667
CD45 1:200 IF Abcam ab10558
Nonphosphorylated YAP (Ser127) (E6U8Z) rabbit mAb 1:1000 WB Cell Signaling Tech 29495S
Tubulin 1:10,000 WB Sigma T5168
DK α-rabbit-HRP 1:10,000 WB Jackson Immunoresearch (West Baltimore Pike, PA) 711-035-152
DK α-mouse-HRP 1:10,000 WB Jackson Immunoresearch 715-035-150

DAB, 3,3′-diaminobenzidine tetra hydrochloride; DK, Donkey; HRP, horseradish peroxidase; IF, Immuno-Fluoresence; mAb, Monoclonal Antibody; WB, Western Blot; Yap, Yes-associated protein 1.

Tumors were microdissected, cut into small pieces, and then lysed in RIPA buffer containing Pierce Protease and Phosphatase Inhibitor (Pierce, Waltham, MA). Electrophoresis was performed on Mini-PROTEAN TGX Stain-free gels (Bio-Rad, Hercules, CA) and Western blot on polyvinylidene difluoride (Thermo Scientific, Waltham, MA). Western blot were blocked for 30 minutes with 5% milk in PBS- and 0.1% Tween 20. Primary antibodies were incubated overnight at 4ºC. Secondary antibodies were incubated for 1 hour at room temperature. Horseradish peroxidase was developed using Super Signal Femto (Thermo Scientific) and imaged on a Bio-Rad gel Imager. All samples were collected at Zeitgeber Time 8. The antibodies used are shown in Table 1.

Microscopy

Samples were imaged using a slide scanner (Axio Scan.Z1; Zeiss, Toronto, Canada). The tumor area was quantified using Zen Blue Software (Zeiss). Organoid cloning images were obtained with a 5× objective on an AXIO Vert.A1 microscope (Zeiss). Images were processed using Adobe (San Jose, CA) Photoshop CS5. Antibody-positive cells were quantified as the total number of labeled cells or nuclei/total number of 4′,6-diamidino-2-phenylindole–positive nuclei. Ten crypts were sampled per animal for each intestinal region (proximal, medial, and distal) for quantification of PHH3, lysozyme, cleaved caspase-3, Ki67, Nr1d1, Yap, Tead4, Lgr5, Ereg, and Axin2. Regions were averaged together for results shown if no significant differences were observed.

RNA Purification

Tissue samples were homogenized using a bullet blender (Next Advance, Troy, NY), and RNA was isolated using the RNEasy mini Purification Kit (Qiagen). Complementary DNA was produced using the iScript RT Supermix (Bio-Rad) according to the manufacturer’s protocol.

Quantitative Polymerase Chain Reaction

Transcript levels were quantified using iTaq Universal SYBR Green Supermix (Bio-Rad) on a Viia7 real-time polymerase chain reaction machine (Thermo Fisher Scientific). Primer sequences were obtained from PrimerBank (https://pga.mgh.harvard.edu/primerbank/) and validated at r2 ∼ 0.98, with an efficiency of approximately 80–110. The primers used are listed in Table 2.

Table 2.

List of Quantitative Polymerase Chain Reaction Primers Used

Axin2 TGACTCTCCTTCCAGATCCCA TGCCCACACTAGGCTGACA
Bmal1 TGACCCTCATGGAAGGTTAGAA GGACATTGCATTGCATGTTGG
Ccnd1 GCGTACCCTGACACCAATCTC CTCCTCTTCGCACTTCTGCTC
cMyc ATGCCCCTCAACGTGAACTTC GTCGCAGATGAAATAGGGCTG
Cry1 CACTGGTTCCGAAAGGGACTC CTGAAGCAAAAATCGCCACCT
Dolk1 CAGTGTGGGACCGATACTCCT CCAAGCAAAGGCATGACCA
Ereg CTGCCTCTTGGGTCTTGACG GCGGTACAGTTATCCTCGGATTC
Fam76b GAATGTCGGATTGCACATCCT GGTCTGAGGTGCTCCATATTTC
Gapdh AGGTCGGTGTGAACGGATTTG TGTAGACCATGTAGTTGAGGTCA
Golga1 AAGAATTGGATGCACGAACCA AGCCGCTCGTTTAAGTCAGAT
Lgr5 CCTACTCGAAGACTTACCCAGT GCATTGGGGTGAATGATAGCA
Msmo1 AAACAAAAGTGTTGGCGTGTTC AAGCATTCTTAAAGGGCTCCTG
Mlixpl AGATGGAGAACCGACGTATCA ACTGAGCGTGCTGACAAGTC
Pmpca CTGCACACAGACGGTTCAG GTCCAGAGTGGTAACTTTGGTTT
Nr1d1 TGAACGCAGGAGGTGTGATTG GAGGACTGGAAGCTATTCTCAGA
Rnf128 ATTCAAAGAGGCATCCAAGTCAC TGCATTTCGTAATCTTCGAGCAG

Organoid Experiments

Intestinal organoids were derived from mice as previously reported.46 For the 24-hour RNA sequencing, organoid biological replicates were collected every 2 hours (n = 2 derived lines of each of the 4 genotypes). For the 48-hour analyses, organoid biological replicates (n = 3 derived lines of each of the 4 genotypes) were collected every 4 hours. RNA sequencing experiments were performed by splitting organoids, allowing a 12-hour recovery, and then 1-hour treatment with 0.1 μm dexamethasone (Sigma, Oakville, Ontario) in base media containing epidermal growth factor. Media then was replaced with fresh organoid culture media. Samples were collected from 24 hours after dexamethasone treatment, every 2 hours, and immediately lysed in RLT (Qiagen) + 1% β-mecaptoethanol (M6250; Sigma Aldrich) and flash frozen in liquid nitrogen. For organoid cloning assays, 5 large organoids were selected from 1 animal of each genotype and dissociated for 10 minutes using 100 μL TrypLE Express (12605010; Thermo Fisher) + 1:1000 Y-27632 (72304; Stem Cell Tech, Vancouver, British Columbia). Enzymatic dissociation was quenched with 100-μL base organoid media containing epidermal growth factor + 1:1000 Y-27632. Single cells then were plated in ENR (EGF, Noggin, R-Spondin) / EN (EGF, Noggin) media + Y-27632 for 1 day. The following day, Y-27632 was removed and replaced with fresh ENR/EN media. Fast-growing Apcmin tumor organoids were quantified 5 days later, whereas slower-growing nontransformed organoids were quantified 15 days later. In all experiments, organoid media was changed every 2–5 days, depending on need. Yap chemical inhibition was performed using 3 mmol/L Verteporfin (AOB5623; Aobious, Gloucester, MA), Tead inhibition using 3 mmol/L TED-347 (AOB17707; Aobious), and 1:750 dimethyl sulfoxide vehicle control (D2650; Sigma) was applied to organoid media.

RNA Sequencing and Gene Expression Analysis

A total of 1 μg RNA per sample was prepared at the Donnelly Sequencing Centre (http://ccbr.utoronto.ca/donnelly-sequencing-centre) for RNA integrity assessment and sequencing. Libraries were prepared using 96 NEB–NEBNext Ultra II Directional RNA Library Prep Kit including quality control and DNase treatment (#E7765; New England Biolabs, Ipswich, MA). Sequencing was performed on a 1 Flowcell, Illumina NovaSeq6000 (Illumina), S2; 100 cycles; v1 chemistry read 1, 101 bp; index read 1, 8 bp; and index read 2, 8 bp. Raw transcriptome sequencing reads were aligned against GRCm38 and Ensembl release 96 transcript annotations using STAR v2.5.100 RSEM v1.3.0101 was used for transcript quantification. Libraries were excluded from further analyses in the case of low-quality alignment (Supplementary Table 4). Transcripts per million values for all remaining libraries in each genotype were merged into 1 expression table file. Quantile normalization was performed for each expression table using the preprocessCore package. Rhythmic analysis and differential expression analysis were performed as follows. First, genes with null expression in at least 1 sample within a table were excluded from that table. Rhythmicity for the remaining transcripts then was evaluated with MetaCycle,57 and we considered all transcripts with a base expression greater than 0.5, relative amplitude greater than 0.05, and a BH q value less than 0.1 as rhythmic. The number of rhythmic transcripts was as follows: 41 in control organoids, 0 in Bmal1 mutant, 238 in Apcmin control, and 263 in Apcmin + Bmal1 mutant (Supplementary Table 1). We next evaluated gene-by-gene (n = 17,543) the differences in overall expression across 24 hours (all time points) between genotypes/expression tables, using analysis of variance (ANOVA), followed by the post hoc Tukey test. For each gene, pairwise expression fold changes between genotypes were calculated as the ratio between the average 24-hour expression in the first genotype and the average in the second genotype. With a Tukey q value less than 0.05 and |log2(fold change)| greater than 1.5, the number of differentially expressed genes was as follows: 111 between control and Bmal1 mutant, 6405 between control and Apcmin control, and 2398 between Apcmin control and Apcmin + Bmal1 mutant (Supplementary Table 2). GSEA was conducted in the GSEA 4.1.0 software downloaded from https://www.gsea-msigdb.org/gsea/index.jsp. Genes selected to be input into GSEA had to be expressed in two thirds of the samples (Supplementary Table 5). The gene set (size = 425) for GSEA analysis was generated using genes found to be up-regulated 2-fold or greater in43 RNA sequencing data set.

Statistics

Statistical analysis was performed using Prism 8 (GraphPad, San Diego, CA). An unpaired t test, 1-way ANOVA, or 2-way ANOVA were used with a post-test (Table 3). RNA sequencing data analyses were performed in R. MetaCycle was applied using JTK and LS algorithms and the Fisher method for P value integration. For comparisons between genotypes, the P values from ANOVA and the post hoc Tukey tests were corrected with the BH method. In these tests, genes that were absent in any 1 genotype were assigned the lowest expression value found in the expression tables. Pearson heatmaps were plotted using the Python 3.6–Seaborn library based on the Pearson correlation between the participant genes. Cubic spline interpolation was used to model the growth of a gene throughout time points (Figures 8 and 9). The splines are natural where the first derivative ∂1 and the second derivative ∂2 both equal 0 at each time point. Implementation of the cubic spline interpolation was performed using Matlab 2013a.

Table 3.

Details of Statistical Tests

Figure 1
 A Two-way ANOVA f(6, 52) = 2.766; P = .0208 J Unpaired t test P < .0001
 C One-way ANOVA
Tukey post-test
f(3, 46) = 80.56; P < .0001
P < .0001
K Unpaired t test P = .7113
 D One-way ANOVA
Tukey post-test
f(3, 47) = 57.41; P < .0001;
P < .0001
L Unpaired t test P = .5447
 F Unpaired t test P = .0338 M Unpaired t test P = .4881
 H One-way ANOVA
Tukey post-test
f(2, 390) = 5.936; P = .0029;
ZT12 P = .0030
O Unpaired t test P = .3793
 I Unpaired t test P = .1036
Figure 2
 B Unpaired t test P = .0012 J Unpaired t test P = .0013
 D Unpaired t test P = .0166 K Unpaired t test P < .0001
 F Two-way ANOVA
Tukey post-test
f(2, 218) = 3.386; P = .0352
ZT4 P = .0246
L Unpaired t test P = .0123
 G Unpaired t test P = .031 N Unpaired t test P = .0005
 I Unpaired t test P = .1409 P Unpaired t test P < .0001
Panel Test Value
Figure 3
 C MetaCycle2d Nr1d1 (P = 4.9-08; q = 0.00039), Cry1 (P = 3.8-08; q = 0.00039), Bmp3 (P = .00012; q = 0.057), Ephb3 (P = 8.0-06; q = 0.0084), Kit (P = .00016; q = 0.067), Tead4 (P = 1.09-05; q = 0.0098)
 D Two-way ANOVA Nr1d1 (f(12, 37) = 5.993; P < .0001), Cry1 (f(1, 37) = 413.1; P < .0001), Tead4 (f(12, 37) = 15.36; P < .0001)
Figure 4
 D MetaCycle2d Golga (P = 1.20-06; q = 0.015),
Dolk (P = 2.83-06; q = 0.015), RNF128 (P = 5.81-06; q = 0.015), Mlxipl (P = 4.26-07; q = 0.0053), Msmo1 (P = 1.04-06; q = 0.0053), Fam76b (P = 1.44-06; q = 0.0055)
 E Two-way ANOVA Golga (f(12,26) = 8.393; P < .0001),
Dolk (f(12,26) = 3.557; P = .0033) RNF128 (f12, 25) = 3.284; P = .0058), Mlxipl (f(12,15) = 1.220; P = .3623), Msmo1(f12, 13) = 2.051; P = .1067), Fam76b (f(12, 26) = 6.062; P < .0001)
Panel Test Value Panel Test Value
Figure 5
 A Gene-wise 1-way ANOVA (4 genotypes) with BH correction
Gene-wise Tukey post-test (2 genotypes) with BH correction
f(3,84) > 2.75; P < .047; q < 0.05; P < .00357; q < 0.05 H Unpaired t test P = .0075
 B Gene-wise 1-way ANOVA (4 genotypes) with BH correction
Gene-wise Tukey post-test (2 genotypes) with BH correction
f(3,84) > 2.75; P < .047; q < 0.05; P < .02815; q < 0.05 K Unpaired t test P = .0444
 C GO analysis: DAVID 6.8 P < .0002; Benjamini < 0.05
Gene set > 9
L One-way ANOVA
Tukey post-test
Apcmin; Bmal1-/-DMSO vs Yap-i
f(3, 10) = 19.55; P = .0002; P = .0068
 D ES
NES
Nominal P value
FDR q-value
FWER P value
ES, 0.44; NES, 2.083; P = .000; q =0.000; P = .000 M. One-way ANOVA
Tukey post-test:
DMSO vs DMSO
Apcmin; Bmal1-/-DMSO vs YAP
f(2,43) = 4.69; P = .0144; P = .0272; P = .0044
 G Unpaired t test P = .0004
Figure 6
 A One-way ANOVA
Tukey post-test
f(3,8) = 11.48; P = .0026; P = .0029 D One-way ANOVA
Tukey post-test
f(3, 16) = 8.420; P = .0014; P = .0217
 B Unpaired t test 0.9172 F One-way ANOVA
Tukey post-test
f(3, 65) = 6.824; P = .005; P = .0182
 C Unpaired t test P = .027
Figure 7
 B Unpaired t test P = .0186 G One-way ANOVA
Sidak post-test
f(3, 36) = 9.241; P = .0001
P = .0096
 E Unpaired t test P = .0009 I One-way ANOVA
Sidak post-test
f(3, 36) = 9.422; P < .0001
P = .0064
 D Unpaired t test P = .027
Figure 8
 A Two-way ANOVA f(5, 168) = 12.01; P < .0001 H Two-way ANOVA
Tukey post-test
f(2, 116) = 351.9; P < .0001
ZT4 P < .0001
ZT12 P < .0001
 C Two-way ANOVA f(5, 168) = 6.49; P < .0001 J Mixed-effect analysis
Sidak post-test
f(2, 27) = 127.4; P < .0001
ZT12 < 0.0001
 E Two-way ANOVA f(1, 168) = 25.50; P < .0001
Panel Test Value
Figure 9
 A Two-way ANOVA f(5, 168) = 2.50; P = .033
 C Two-way ANOVA f(5, 168) = 9.24; P = .0027
 E Two-way ANOVA f(1, 168) = 8.947; P = .0032
 G Two-way ANOVA f(1, 168) = 71.98; P < .0001
 I Two-way ANOVA f(1, 168) = 41.51; P < .0001
 K Two-way ANOVA F(5, 168) = 2.746; P = .02
Panel Test Value Panel Test Value
Figure 10
 B One-way ANOVA
Tukey post-test
f(3, 20) = 20.58; P < .0001
P = .0027
G Unpaired t test P = .0341
 C One-way ANOVA
Tukey post-test
f(3, 20) = 28.71; P < .0001
P = .0014
I Unpaired t test P = .0447
 E Unpaired t test P = .0023

ANOVA, analysis of variance; DMSO, dimethyl sulfoxide; ES, enrichment score; FDR, False Discovery Rate; FWER, Family-Wise Error Rate; NES, normalized enrichment score; Yap-i, Yap Inhibitor; ZT, Zeitgeber Time.

Acknowledgments

The authors would like to thank members of the Karpowicz Laboratory for their support, Sharon Lavigne for logistic support, and David Weaver for helpful comments on the manuscript.

CRediT Authorship Contributions

Kyle Stokes, MSc (Formal analysis: Equal; Investigation: Lead; Writing – original draft: Supporting)

Malika Nunes, MSc (Investigation: Supporting)

Chantelle Trombley, MSc (Investigation: Supporting)

Danilo E. F. L. Flôres, PhD (Data curation: Lead; Formal analysis: Supporting)

Gang Wu, PhD (Data curation: Supporting; Formal analysis: Lead)

Zainab Taleb, MSc (Investigation: Supporting)

Abedalrhman Alkhateeb, PhD (Data curation: Supporting; Formal analysis: Supporting)

Suhrid Banskota, PhD (Investigation: Supporting)

Chris Harris, MSc (Investigation: Supporting)

Oliver P. Love, PhD (Investigation: Supporting; Supervision: Supporting)

Waliul I. Khan, PhD (Investigation: Supporting; Supervision: Supporting)

Luis Rueda, PhD (Formal analysis: Supporting; Supervision: Supporting)

John B. Hogenesch, PhD (Data curation: Supporting; Formal analysis: Supporting; Supervision: Supporting)

Phillip Karpowicz, PhD (Conceptualization: Lead; Investigation: Lead; Supervision: Lead)

Footnotes

Conflicts of interest The authors disclose no conflicts.

Funding Funded by the Natural Sciences and Engineering Research Council of Canada Alexander Graham Bell Scholarship (K.S.); Sao Paulo Research Foundation grant FAPESP 2019/04451-3 (D.F.); Mitacs Accelerate Postdoctoral Fellowship (A.A.); Canadian Institutes of Health Research grant PJT-156262 and the Natural Sciences and Engineering Research Council of Canada grant RGPIN-2019-06739 (W.I.K.); Natural Science and Engineering Research Council and the Canada Research Chairs program (O.P.L.); Natural Sciences and Engineering Research Council of Canada grant RGPIN-2019-04696 (L.R.); National Institute of Neurological Disorders and Stroke grant 5R01NS054794-13, National Heart, Lung, and Blood Institute grant 5R01HL138551-02, and the National Cancer Institute grant 1R01CA227485-01A1 (J.B.H.); and the Canadian Institutes of Health Research grant PJT-388014, Canada Foundation for Innovation/Ontario Research Fund, and the Ontario Institute for Regenerative Medicine, and Crohn’s and Colitis Canada (P.K.). The contents of this article are solely the responsibility of the authors and may not represent the official views of the sponsoring agencies.

Supplementary Material

Supplementary Table S1
mmc1.xlsx (310.7KB, xlsx)
Supplementary Table S2
mmc2.xlsx (2.2MB, xlsx)
Supplementary Table S3
mmc3.xlsx (40.5KB, xlsx)
Supplementary Table S4
mmc4.xlsx (17.3KB, xlsx)
Supplementary Table S5
mmc5.xlsx (94.6KB, xlsx)

References

  • 1.Hanahan D., Weinberg R.A. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
  • 2.Takahashi J.S. Transcriptional architecture of the mammalian circadian clock. Nat Rev Genet. 2017;18:164–179. doi: 10.1038/nrg.2016.150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Zhang R., Lahens N.F., Ballance H.I., Hughes M.E., Hogenesch J.B. A circadian gene expression atlas in mammals: implications for biology and medicine. Proc Natl Acad Sci U S A. 2014;111:16219–16224. doi: 10.1073/pnas.1408886111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Bass J., Lazar M.A. Circadian time signatures of fitness and disease. Science. 2016;354:994–999. doi: 10.1126/science.aah4965. [DOI] [PubMed] [Google Scholar]
  • 5.Janich P., Meng Q.J., Benitah S.A. Circadian control of tissue homeostasis and adult stem cells. Curr Opin Cell Biol. 2014;31C:8–15. doi: 10.1016/j.ceb.2014.06.010. [DOI] [PubMed] [Google Scholar]
  • 6.Schernhammer E.S., Laden F., Speizer F.E., Willett W.C., Hunter D.J., Kawachi I., Colditz G.A. Rotating night shifts and risk of breast cancer in women participating in the nurses' health study. J Natl Cancer Inst. 2001;93:1563–1568. doi: 10.1093/jnci/93.20.1563. [DOI] [PubMed] [Google Scholar]
  • 7.Schernhammer E.S., Laden F., Speizer F.E., Willett W.C., Hunter D.J., Kawachi I., Fuchs C.S., Colditz G.A. Night-shift work and risk of colorectal cancer in the nurses' health study. J Natl Cancer Inst. 2003;95:825–828. doi: 10.1093/jnci/95.11.825. [DOI] [PubMed] [Google Scholar]
  • 8.Flynn-Evans E.E., Mucci L., Stevens R.G., Lockley S.W. Shiftwork and prostate-specific antigen in the National Health and Nutrition Examination Survey. J Natl Cancer Inst. 2013;105:1292–1297. doi: 10.1093/jnci/djt169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Wegrzyn L.R., Tamimi R.M., Rosner B.A., Brown S.B., Stevens R.G., Eliassen A.H., Laden F., Willett W.C., Hankinson S.E., Schernhammer E.S. Rotating night-shift work and the risk of breast cancer in the Nurses' Health Studies. Am J Epidemiol. 2017;186:532–540. doi: 10.1093/aje/kwx140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Um K., Park C.S., Yoo C., Ahn Y.S., Kim M., Jeong K.S. Risk factors including night shift work of colorectal polyp. Ann Occup Environ Med. 2020;32:e26. doi: 10.35371/aoem.2020.32.e26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Sancar A., Lindsey-Boltz L.A., Gaddameedhi S., Selby C.P., Ye R., Chiou Y.Y., Kemp M.G., Hu J., Lee J.H., Ozturk N. Circadian clock, cancer, and chemotherapy. Biochemistry. 2015;54:110–123. doi: 10.1021/bi5007354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Masri S., Sassone-Corsi P. The emerging link between cancer, metabolism, and circadian rhythms. Nat Med. 2018;24:1795–1803. doi: 10.1038/s41591-018-0271-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Yu E.A., Weaver D.R. Disrupting the circadian clock: gene-specific effects on aging, cancer, and other phenotypes. Aging (Albany NY) 2011;3:479–493. doi: 10.18632/aging.100323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Sancar A., Van Gelder R.N. Clocks, cancer, and chronochemotherapy. Science. 2021;371 doi: 10.1126/science.abb0738. [DOI] [PubMed] [Google Scholar]
  • 15.Alhopuro P., Bjorklund M., Sammalkorpi H., Turunen M., Tuupanen S., Bistrom M., Niittymaki I., Lehtonen H.J., Kivioja T., Launonen V., Saharinen J., Nousiainen K., Hautaniemi S., Nuorva K., Mecklin J.P., Jarvinen H., Orntoft T., Arango D., Lehtonen R., Karhu A., Taipale J., Aaltonen L.A. Mutations in the circadian gene CLOCK in colorectal cancer. Mol Cancer Res. 2010;8:952–960. doi: 10.1158/1541-7786.MCR-10-0086. [DOI] [PubMed] [Google Scholar]
  • 16.Momma T., Okayama H., Saitou M., Sugeno H., Yoshimoto N., Takebayashi Y., Ohki S., Takenoshita S. Expression of circadian clock genes in human colorectal adenoma and carcinoma. Oncol Lett. 2017;14:5319–5325. doi: 10.3892/ol.2017.6876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Wang X., Yan D., Teng M., Fan J., Zhou C., Li D., Qiu G., Sun X., Li T., Xing T., Tang H., Peng X., Peng Z. Reduced expression of PER3 is associated with incidence and development of colon cancer. Ann Surg Oncol. 2012;19:3081–3088. doi: 10.1245/s10434-012-2279-5. [DOI] [PubMed] [Google Scholar]
  • 18.Lellupitiyage Don S.S., Lin H.H., Furtado J.J., Qraitem M., Taylor S.R., Farkas M.E. Circadian oscillations persist in low malignancy breast cancer cells. Cell Cycle. 2019;18:2447–2453. doi: 10.1080/15384101.2019.1648957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Broadberry E., McConnell J., Williams J., Yang N., Zindy E., Leek A., Waddington R., Joseph L., Howe M., Meng Q.J., Streuli C.H. Disrupted circadian clocks and altered tissue mechanics in primary human breast tumours. Breast Cancer Res. 2018;20:125. doi: 10.1186/s13058-018-1053-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Altman B.J., Hsieh A.L., Sengupta A., Krishnanaiah S.Y., Stine Z.E., Walton Z.E., Gouw A.M., Venkataraman A., Li B., Goraksha-Hicks P., Diskin S.J., Bellovin D.I., Simon M.C., Rathmell J.C., Lazar M.A., Maris J.M., Felsher D.W., Hogenesch J.B., Weljie A.M., Dang C.V. MYC disrupts the circadian clock and metabolism in cancer cells. Cell Metab. 2015;22:1009–1019. doi: 10.1016/j.cmet.2015.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Marcheva B., Ramsey K.M., Buhr E.D., Kobayashi Y., Su H., Ko C.H., Ivanova G., Omura C., Mo S., Vitaterna M.H., Lopez J.P., Philipson L.H., Bradfield C.A., Crosby S.D., JeBailey L., Wang X., Takahashi J.S., Bass J. Disruption of the clock components CLOCK and BMAL1 leads to hypoinsulinaemia and diabetes. Nature. 2010;466:627–631. doi: 10.1038/nature09253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Kowalska E., Ripperger J.A., Hoegger D.C., Bruegger P., Buch T., Birchler T., Mueller A., Albrecht U., Contaldo C., Brown S.A. NONO couples the circadian clock to the cell cycle. Proc Natl Acad Sci U S A. 2013;110:1592–1599. doi: 10.1073/pnas.1213317110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Plikus M.V., Vollmers C., de la Cruz D., Chaix A., Ramos R., Panda S., Chuong C.M. Local circadian clock gates cell cycle progression of transient amplifying cells during regenerative hair cycling. Proc Natl Acad Sci U S A. 2013;110:E2106–E2115. doi: 10.1073/pnas.1215935110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Lee J.H., Sancar A. Circadian clock disruption improves the efficacy of chemotherapy through p73-mediated apoptosis. Proc Natl Acad Sci U S A. 2011;108:10668–10672. doi: 10.1073/pnas.1106284108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Lee J.H., Sancar A. Regulation of apoptosis by the circadian clock through NF-kappaB signaling. Proc Natl Acad Sci U S A. 2011;108:12036–12041. doi: 10.1073/pnas.1108125108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Gaddameedhi S., Selby C.P., Kaufmann W.K., Smart R.C., Sancar A. Control of skin cancer by the circadian rhythm. Proc Natl Acad Sci U S A. 2011;108:18790–18795. doi: 10.1073/pnas.1115249108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Gery S., Komatsu N., Baldjyan L., Yu A., Koo D., Koeffler H.P. The circadian gene per1 plays an important role in cell growth and DNA damage control in human cancer cells. Mol Cell. 2006;22:375–382. doi: 10.1016/j.molcel.2006.03.038. [DOI] [PubMed] [Google Scholar]
  • 28.Fu L., Pelicano H., Liu J., Huang P., Lee C. The circadian gene Period2 plays an important role in tumor suppression and DNA damage response in vivo. Cell. 2002;111:41–50. doi: 10.1016/s0092-8674(02)00961-3. [DOI] [PubMed] [Google Scholar]
  • 29.Wood P.A., Yang X., Taber A., Oh E.Y., Ansell C., Ayers S.E., Al-Assaad Z., Carnevale K., Berger F.G., Pena M.M., Hrushesky W.J. Period 2 mutation accelerates ApcMin/+ tumorigenesis. Mol Cancer Res. 2008;6:1786–1793. doi: 10.1158/1541-7786.MCR-08-0196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Papagiannakopoulos T., Bauer M.R., Davidson S.M., Heimann M., Subbaraj L., Bhutkar A., Bartlebaugh J., Vander Heiden M.G., Jacks T. Circadian rhythm disruption promotes lung tumorigenesis. Cell Metab. 2016;24:324–331. doi: 10.1016/j.cmet.2016.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Mteyrek A., Filipski E., Guettier C., Okyar A., Levi F. Clock gene Per2 as a controller of liver carcinogenesis. Oncotarget. 2016;7:85832–85847. doi: 10.18632/oncotarget.11037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Antoch M.P., Toshkov I., Kuropatwinski K.K., Jackson M. Deficiency in PER proteins has no effect on the rate of spontaneous and radiation-induced carcinogenesis. Cell Cycle. 2013;12:3673–3680. doi: 10.4161/cc.26614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Roenneberg T., Merrow M. The circadian clock and human health. Curr Biol. 2016;26:R432–R443. doi: 10.1016/j.cub.2016.04.011. [DOI] [PubMed] [Google Scholar]
  • 34.Moser A.R., Pitot H.C., Dove W.F. A dominant mutation that predisposes to multiple intestinal neoplasia in the mouse. Science. 1990;247:322–324. doi: 10.1126/science.2296722. [DOI] [PubMed] [Google Scholar]
  • 35.Su L.K., Kinzler K.W., Vogelstein B., Preisinger A.C., Moser A.R., Luongo C., Gould K.A., Dove W.F. Multiple intestinal neoplasia caused by a mutation in the murine homolog of the APC gene. Science. 1992;256:668–670. doi: 10.1126/science.1350108. [DOI] [PubMed] [Google Scholar]
  • 36.Medema J.P., Vermeulen L. Microenvironmental regulation of stem cells in intestinal homeostasis and cancer. Nature. 2011;474:318–326. doi: 10.1038/nature10212. [DOI] [PubMed] [Google Scholar]
  • 37.Barker N., Ridgway R.A., van Es J.H., van de Wetering M., Begthel H., van den Born M., Danenberg E., Clarke A.R., Sansom O.J., Clevers H. Crypt stem cells as the cells-of-origin of intestinal cancer. Nature. 2009;457:608–611. doi: 10.1038/nature07602. [DOI] [PubMed] [Google Scholar]
  • 38.Gehart H., Clevers H. Tales from the crypt: new insights into intestinal stem cells. Nat Rev Gastroenterol Hepatol. 2019;16:19–34. doi: 10.1038/s41575-018-0081-y. [DOI] [PubMed] [Google Scholar]
  • 39.Karin M., Clevers H. Reparative inflammation takes charge of tissue regeneration. Nature. 2016;529:307–315. doi: 10.1038/nature17039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Grivennikov S.I., Greten F.R., Karin M. Immunity, inflammation, and cancer. Cell. 2010;140:883–899. doi: 10.1016/j.cell.2010.01.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Wang Y., Chiang I.L., Ohara T.E., Fujii S., Cheng J., Muegge B.D., Ver Heul A., Han N.D., Lu Q., Xiong S., Chen F., Lai C.W., Janova H., Wu R., Whitehurst C.E., VanDussen K.L., Liu T.C., Gordon J.I., Sibley L.D., Stappenbeck T.S. Long-term culture captures injury-repair cycles of colonic stem cells. Cell. 2019;179:1144–1159 e15. doi: 10.1016/j.cell.2019.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Yui S., Azzolin L., Maimets M., Pedersen M.T., Fordham R.P., Hansen S.L., Larsen H.L., Guiu J., Alves M.R.P., Rundsten C.F., Johansen J.V., Li Y., Madsen C.D., Nakamura T., Watanabe M., Nielsen O.H., Schweiger P.J., Piccolo S., Jensen K.B. YAP/TAZ-dependent reprogramming of colonic epithelium links ECM remodeling to tissue regeneration. Cell Stem Cell. 2018;22:35–49 e7. doi: 10.1016/j.stem.2017.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Gregorieff A., Liu Y., Inanlou M.R., Khomchuk Y., Wrana J.L. Yap-dependent reprogramming of Lgr5(+) stem cells drives intestinal regeneration and cancer. Nature. 2015;526:715–718. doi: 10.1038/nature15382. [DOI] [PubMed] [Google Scholar]
  • 44.Taniguchi K., Wu L.W., Grivennikov S.I., de Jong P.R., Lian I., Yu F.X., Wang K., Ho S.B., Boland B.S., Chang J.T., Sandborn W.J., Hardiman G., Raz E., Maehara Y., Yoshimura A., Zucman-Rossi J., Guan K.L., Karin M. A gp130-Src-YAP module links inflammation to epithelial regeneration. Nature. 2015;519:57–62. doi: 10.1038/nature14228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Romera-Hernandez M., Aparicio-Domingo P., Papazian N., Karrich J.J., Cornelissen F., Hoogenboezem R.M., Samsom J.N., Cupedo T. Yap1-driven intestinal repair is controlled by group 3 innate lymphoid cells. Cell Rep. 2020;30:37–45 e3. doi: 10.1016/j.celrep.2019.11.115. [DOI] [PubMed] [Google Scholar]
  • 46.Stokes K., Cooke A., Chang H., Weaver D.R., Breault D.T., Karpowicz P. The circadian clock gene BMAL1 coordinates intestinal regeneration. Cell Mol Gastroenterol Hepatol. 2017;4:95–114. doi: 10.1016/j.jcmgh.2017.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Parasram K., Bernardon N., Hammoud M., Chang H., He L., Perrimon N., Karpowicz P. Intestinal stem cells exhibit conditional circadian clock function. Stem Cell Rep. 2018;11:1287–1301. doi: 10.1016/j.stemcr.2018.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Karpowicz P., Zhang Y., Hogenesch J.B., Emery P., Perrimon N. The circadian clock gates the intestinal stem cell regenerative state. Cell Rep. 2013;3:996–1004. doi: 10.1016/j.celrep.2013.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Janich P., Pascual G., Merlos-Suarez A., Batlle E., Ripperger J., Albrecht U., Cheng H.Y., Obrietan K., Di Croce L., Benitah S.A. The circadian molecular clock creates epidermal stem cell heterogeneity. Nature. 2011;480:209–214. doi: 10.1038/nature10649. [DOI] [PubMed] [Google Scholar]
  • 50.Bunger M.K., Wilsbacher L.D., Moran S.M., Clendenin C., Radcliffe L.A., Hogenesch J.B., Simon M.C., Takahashi J.S., Bradfield C.A. Mop3 is an essential component of the master circadian pacemaker in mammals. Cell. 2000;103:1009–1017. doi: 10.1016/s0092-8674(00)00205-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Kondratov R.V., Kondratova A.A., Gorbacheva V.Y., Vykhovanets O.V., Antoch M.P. Early aging and age-related pathologies in mice deficient in BMAL1, the core component of the circadian clock. Genes Dev. 2006;20:1868–1873. doi: 10.1101/gad.1432206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Balsalobre A., Brown S.A., Marcacci L., Tronche F., Kellendonk C., Reichardt H.M., Schutz G., Schibler U. Resetting of circadian time in peripheral tissues by glucocorticoid signaling. Science. 2000;289:2344–2347. doi: 10.1126/science.289.5488.2344. [DOI] [PubMed] [Google Scholar]
  • 53.Leliavski A., Shostak A., Husse J., Oster H. Impaired glucocorticoid production and response to stress in Arntl-deficient male mice. Endocrinology. 2014;155:133–142. doi: 10.1210/en.2013-1531. [DOI] [PubMed] [Google Scholar]
  • 54.Matsu-Ura T., Dovzhenok A., Aihara E., Rood J., Le H., Ren Y., Rosselot A.E., Zhang T., Lee C., Obrietan K., Montrose M.H., Lim S., Moore S.R., Hong C.I. Intercellular coupling of the cell cycle and circadian clock in adult stem cell culture. Mol Cell. 2016;64:900–912. doi: 10.1016/j.molcel.2016.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Imajo M., Ebisuya M., Nishida E. Dual role of YAP and TAZ in renewal of the intestinal epithelium. Nat Cell Biol. 2015;17:7–19. doi: 10.1038/ncb3084. [DOI] [PubMed] [Google Scholar]
  • 56.Hughes M.E., Abruzzi K.C., Allada R., Anafi R., Arpat A.B., Asher G., Baldi P., de Bekker C., Bell-Pedersen D., Blau J., Brown S., Ceriani M.F., Chen Z., Chiu J.C., Cox J., Crowell A.M., DeBruyne J.P., Dijk D.J., DiTacchio L., Doyle F.J., Duffield G.E., Dunlap J.C., Eckel-Mahan K., Esser K.A., FitzGerald G.A., Forger D.B., Francey L.J., Fu Y.H., Gachon F., Gatfield D., de Goede P., Golden S.S., Green C., Harer J., Harmer S., Haspel J., Hastings M.H., Herzel H., Herzog E.D., Hoffmann C., Hong C., Hughey J.J., Hurley J.M., de la Iglesia H.O., Johnson C., Kay S.A., Koike N., Kornacker K., Kramer A., Lamia K., Leise T., Lewis S.A., Li J., Li X., Liu A.C., Loros J.J., Martino T.A., Menet J.S., Merrow M., Millar A.J., Mockler T., Naef F., Nagoshi E., Nitabach M.N., Olmedo M., Nusinow D.A., Ptáček L.J., Rand D., Reddy A.B., Robles M.S., Roenneberg T., Rosbash M., Ruben M.D., Rund S.S.C., Sancar A., Sassone-Corsi P., Sehgal A., Sherrill-Mix S., Skene D.J., Storch K.F., Takahashi J.S., Ueda H.R., Wang H., Weitz C., Westermark P.O., Wijnen H., Xu Y., Wu G., Yoo S.H., Young M., Zhang E.E., Zielinski T., Hogenesch J.B. Guidelines for genome-scale analysis of biological rhythms. J Biol Rhythms. 2017;32:380–393. doi: 10.1177/0748730417728663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Wu G., Anafi R.C., Hughes M.E., Kornacker K., Hogenesch J.B. MetaCycle: an integrated R package to evaluate periodicity in large scale data. Bioinformatics. 2016;32:3351–3353. doi: 10.1093/bioinformatics/btw405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.De Vos M., Schreiber V., Dantzer F. The diverse roles and clinical relevance of PARPs in DNA damage repair: current state of the art. Biochem Pharmacol. 2012;84:137–146. doi: 10.1016/j.bcp.2012.03.018. [DOI] [PubMed] [Google Scholar]
  • 59.Schmitt M., Schewe M., Sacchetti A., Feijtel D., van de Geer W.S., Teeuwssen M., Sleddens H.F., Joosten R., van Royen M.E., van de Werken H.J.G., van Es J., Clevers H., Fodde R. Paneth cells respond to inflammation and contribute to tissue regeneration by acquiring stem-like features through SCF/c-Kit signaling. Cell Rep. 2018;24:2312–2328 e7. doi: 10.1016/j.celrep.2018.07.085. [DOI] [PubMed] [Google Scholar]
  • 60.Moor A.E., Harnik Y., Ben-Moshe S., Massasa E.E., Rozenberg M., Eilam R., Bahar Halpern K., Itzkovitz S. Spatial reconstruction of single enterocytes uncovers broad zonation along the intestinal villus axis. Cell. 2018;175:1156–1167 e15. doi: 10.1016/j.cell.2018.08.063. [DOI] [PubMed] [Google Scholar]
  • 61.Subramanian A., Tamayo P., Mootha V.K., Mukherjee S., Ebert B.L., Gillette M.A., Paulovich A., Pomeroy S.L., Golub T.R., Lander E.S., Mesirov J.P. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A. 2005;102:15545–15550. doi: 10.1073/pnas.0506580102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Kreso A., Dick J.E. Evolution of the cancer stem cell model. Cell Stem Cell. 2014;14:275–291. doi: 10.1016/j.stem.2014.02.006. [DOI] [PubMed] [Google Scholar]
  • 63.Schwitalla S., Fingerle A.A., Cammareri P., Nebelsiek T., Goktuna S.I., Ziegler P.K., Canli O., Heijmans J., Huels D.J., Moreaux G., Rupec R.A., Gerhard M., Schmid R., Barker N., Clevers H., Lang R., Neumann J., Kirchner T., Taketo M.M., van den Brink G.R., Sansom O.J., Arkan M.C., Greten F.R. Intestinal tumorigenesis initiated by dedifferentiation and acquisition of stem-cell-like properties. Cell. 2013;152:25–38. doi: 10.1016/j.cell.2012.12.012. [DOI] [PubMed] [Google Scholar]
  • 64.Sato T., Vries R.G., Snippert H.J., van de Wetering M., Barker N., Stange D.E., van Es J.H., Abo A., Kujala P., Peters P.J., Clevers H. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459:262–265. doi: 10.1038/nature07935. [DOI] [PubMed] [Google Scholar]
  • 65.Serra D., Mayr U., Boni A., Lukonin I., Rempfler M., Challet Meylan L., Stadler M.B., Strnad P., Papasaikas P., Vischi D., Waldt A., Roma G., Liberali P. Self-organization and symmetry breaking in intestinal organoid development. Nature. 2019;569:66–72. doi: 10.1038/s41586-019-1146-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Bum-Erdene K., Zhou D., Gonzalez-Gutierrez G., Ghozayel M.K., Si Y., Xu D., Shannon H.E., Bailey B.J., Corson T.W., Pollok K.E., Wells C.D., Meroueh S.O. Small-molecule covalent modification of conserved cysteine leads to allosteric inhibition of the TEAD⋅Yap protein-protein interaction. Cell Chem Biol. 2019;26:378–389.e13. doi: 10.1016/j.chembiol.2018.11.010. [DOI] [PubMed] [Google Scholar]
  • 67.Madison B.B., Dunbar L., Qiao X.T., Braunstein K., Braunstein E., Gumucio D.L. Cis elements of the villin gene control expression in restricted domains of the vertical (crypt) and horizontal (duodenum, cecum) axes of the intestine. J Biol Chem. 2002;277:33275–33283. doi: 10.1074/jbc.M204935200. [DOI] [PubMed] [Google Scholar]
  • 68.Storch K.F., Paz C., Signorovitch J., Raviola E., Pawlyk B., Li T., Weitz C.J. Intrinsic circadian clock of the mammalian retina: importance for retinal processing of visual information. Cell. 2007;130:730–741. doi: 10.1016/j.cell.2007.06.045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Wang S., Lin Y., Yuan X., Li F., Guo L., Wu B. REV-ERBalpha integrates colon clock with experimental colitis through regulation of NF-kappaB/NLRP3 axis. Nat Commun. 2018;9:4246. doi: 10.1038/s41467-018-06568-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Pagel R., Bar F., Schroder T., Sunderhauf A., Kunstner A., Ibrahim S.M., Autenrieth S.E., Kalies K., Konig P., Tsang A.H., Bettenworth D., Divanovic S., Lehnert H., Fellermann K., Oster H., Derer S., Sina C. Circadian rhythm disruption impairs tissue homeostasis and exacerbates chronic inflammation in the intestine. FASEB J. 2017;31:4707–4719. doi: 10.1096/fj.201700141RR. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Panda S. Circadian physiology of metabolism. Science. 2016;354:1008–1015. doi: 10.1126/science.aah4967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Lucassen E.A., Coomans C.P., van Putten M., de Kreij S.R., van Genugten J.H., Sutorius R.P., de Rooij K.E., van der Velde M., Verhoeve S.L., Smit J.W., Lowik C.W., Smits H.H., Guigas B., Aartsma-Rus A.M., Meijer J.H. Environmental 24-hr cycles are essential for health. Curr Biol. 2016;26:1843–1853. doi: 10.1016/j.cub.2016.05.038. [DOI] [PubMed] [Google Scholar]
  • 73.Pendergast J.S., Friday R.C., Yamazaki S. Distinct functions of Period2 and Period3 in the mouse circadian system revealed by in vitro analysis. PLoS One. 2010;5 doi: 10.1371/journal.pone.0008552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Hashikawa K.I., Katamune C., Kusunose N., Matsunaga N., Koyanagi S., Ohdo S. Dysfunction of the circadian transcriptional factor CLOCK in mice resists chemical carcinogen-induced tumorigenesis. Sci Rep. 2017;7:9995. doi: 10.1038/s41598-017-10599-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Sulli G., Rommel A., Wang X., Kolar M.J., Puca F., Saghatelian A., Plikus M.V., Verma I.M., Panda S. Pharmacological activation of REV-ERBs is lethal in cancer and oncogene-induced senescence. Nature. 2018;553:351–355. doi: 10.1038/nature25170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Puram R.V., Kowalczyk M.S., de Boer C.G., Schneider R.K., Miller P.G., McConkey M., Tothova Z., Tejero H., Heckl D., Jaras M., Chen M.C., Li H., Tamayo A., Cowley G.S., Rozenblatt-Rosen O., Al-Shahrour F., Regev A., Ebert B.L. Core circadian clock genes regulate leukemia stem cells in AML. Cell. 2016;165:303–316. doi: 10.1016/j.cell.2016.03.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Dong Z., Zhang G., Qu M., Gimple R.C., Wu Q., Qiu Z., Prager B.C., Wang X., Kim L.J.Y., Morton A.R., Dixit D., Zhou W., Huang H., Li B., Zhu Z., Bao S., Mack S.C., Chavez L., Kay S.A., Rich J.N. Targeting glioblastoma stem cells through disruption of the circadian clock. Cancer Discov. 2019;9:1556–1573. doi: 10.1158/2159-8290.CD-19-0215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Brown S.A., Zumbrunn G., Fleury-Olela F., Preitner N., Schibler U. Rhythms of mammalian body temperature can sustain peripheral circadian clocks. Curr Biol. 2002;12:1574–1583. doi: 10.1016/s0960-9822(02)01145-4. [DOI] [PubMed] [Google Scholar]
  • 79.Das M., Ellies L.G., Kumar D., Sauceda C., Oberg A., Gross E., Mandt T., Newton I.G., Kaur M., Sears D.D., Webster N.J.G. Time-restricted feeding normalizes hyperinsulinemia to inhibit breast cancer in obese postmenopausal mouse models. Nat Commun. 2021;12:565. doi: 10.1038/s41467-020-20743-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Filipski E., Levi F. Circadian disruption in experimental cancer processes. Integr Cancer Ther. 2009;8:298–302. doi: 10.1177/1534735409352085. [DOI] [PubMed] [Google Scholar]
  • 81.Chaix A., Lin T., Le H.D., Chang M.W., Panda S. Time-restricted feeding prevents obesity and metabolic syndrome in mice lacking a circadian clock. Cell Metab. 2018;29:303–319.e4. doi: 10.1016/j.cmet.2018.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Repouskou A., Prombona A. c-MYC targets the central oscillator gene Per1 and is regulated by the circadian clock at the post-transcriptional level. Biochim Biophys Acta. 2016;1859:541–552. doi: 10.1016/j.bbagrm.2016.02.001. [DOI] [PubMed] [Google Scholar]
  • 83.Shostak A., Ruppert B., Ha N., Bruns P., Toprak U.H., Project I.M.-S., Eils R., Schlesner M., Diernfellner A., Brunner M. MYC/MIZ1-dependent gene repression inversely coordinates the circadian clock with cell cycle and proliferation. Nat Commun. 2016;7:11807. doi: 10.1038/ncomms11807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Ko C.H., Yamada Y.R., Welsh D.K., Buhr E.D., Liu A.C., Zhang E.E., Ralph M.R., Kay S.A., Forger D.B., Takahashi J.S. Emergence of noise-induced oscillations in the central circadian pacemaker. PLoS Biol. 2010;8 doi: 10.1371/journal.pbio.1000513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Ray S., Valekunja U.K., Stangherlin A., Howell S.A., Snijders A.P., Damodaran G., Reddy A.B. Circadian rhythms in the absence of the clock gene Bmal1. Science. 2020;367:800–806. doi: 10.1126/science.aaw7365. [DOI] [PubMed] [Google Scholar]
  • 86.Krishnaiah S.Y., Wu G., Altman B.J., Growe J., Rhoades S.D., Coldren F., Venkataraman A., Olarerin-George A.O., Francey L.J., Mukherjee S., Girish S., Selby C.P., Cal S., Er U., Sianati B., Sengupta A., Anafi R.C., Kavakli I.H., Sancar A., Baur J.A., Dang C.V., Hogenesch J.B., Weljie A.M. Clock regulation of metabolites reveals coupling between transcription and metabolism. Cell Metab. 2017;25:961–974 e4. doi: 10.1016/j.cmet.2017.03.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Sladek M., Rybova M., Jindrakova Z., Zemanova Z., Polidarova L., Mrnka L., O'Neill J., Pacha J., Sumova A. Insight into the circadian clock within rat colonic epithelial cells. Gastroenterology. 2007;133:1240–1249. doi: 10.1053/j.gastro.2007.05.053. [DOI] [PubMed] [Google Scholar]
  • 88.Hoogerwerf W.A., Hellmich H.L., Cornelissen G., Halberg F., Shahinian V.B., Bostwick J., Savidge T.C., Cassone V.M. Clock gene expression in the murine gastrointestinal tract: endogenous rhythmicity and effects of a feeding regimen. Gastroenterology. 2007;133:1250–1260. doi: 10.1053/j.gastro.2007.07.009. [DOI] [PubMed] [Google Scholar]
  • 89.Hoogerwerf W.A., Sinha M., Conesa A., Luxon B.A., Shahinian V.B., Cornelissen G., Halberg F., Bostwick J., Timm J., Cassone V.M. Transcriptional profiling of mRNA expression in the mouse distal colon. Gastroenterology. 2008;135:2019–2029. doi: 10.1053/j.gastro.2008.08.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Thaiss C.A., Levy M., Korem T., Dohnalova L., Shapiro H., Jaitin D.A., David E., Winter D.R., Gury-BenAri M., Tatirovsky E., Tuganbaev T., Federici S., Zmora N., Zeevi D., Dori-Bachash M., Pevsner-Fischer M., Kartvelishvily E., Brandis A., Harmelin A., Shibolet O., Halpern Z., Honda K., Amit I., Segal E., Elinav E. Microbiota diurnal rhythmicity programs host transcriptome oscillations. Cell. 2016;167:1495–1510 e12. doi: 10.1016/j.cell.2016.11.003. [DOI] [PubMed] [Google Scholar]
  • 91.Weger B.D., Gobet C., Yeung J., Martin E., Jimenez S., Betrisey B., Foata F., Berger B., Balvay A., Foussier A., Charpagne A., Boizet-Bonhoure B., Chou C.J., Naef F., Gachon F. The mouse microbiome is required for sex-specific diurnal rhythms of gene expression and metabolism. Cell Metab. 2019;29:362–382 e8. doi: 10.1016/j.cmet.2018.09.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Kuang Z., Wang Y., Li Y., Ye C., Ruhn K.A., Behrendt C.L., Olson E.N., Hooper L.V. The intestinal microbiota programs diurnal rhythms in host metabolism through histone deacetylase 3. Science. 2019;365:1428–1434. doi: 10.1126/science.aaw3134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Liang X., Bushman F.D., FitzGerald G.A. Rhythmicity of the intestinal microbiota is regulated by gender and the host circadian clock. Proc Natl Acad Sci U S A. 2015;112:10479–10484. doi: 10.1073/pnas.1501305112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Man K., Loudon A., Chawla A. Immunity around the clock. Science. 2016;354:999–1003. doi: 10.1126/science.aah4966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Kiessling S., Beaulieu-Laroche L., Blum I.D., Landgraf D., Welsh D.K., Storch K.F., Labrecque N., Cermakian N. Enhancing circadian clock function in cancer cells inhibits tumor growth. BMC Biol. 2017;15:13. doi: 10.1186/s12915-017-0349-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Bishehsari F., Voigt R.M., Keshavarzian A. Circadian rhythms and the gut microbiota: from the metabolic syndrome to cancer. Nat Rev Endocrinol. 2020;16:731–739. doi: 10.1038/s41574-020-00427-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Ayyaz A., Kumar S., Sangiorgi B., Ghoshal B., Gosio J., Ouladan S., Fink M., Barutcu S., Trcka D., Shen J., Chan K., Wrana J.L., Gregorieff A. Single-cell transcriptomes of the regenerating intestine reveal a revival stem cell. Nature. 2019;569:121–125. doi: 10.1038/s41586-019-1154-y. [DOI] [PubMed] [Google Scholar]
  • 98.Raufman J.P., Shant J., Xie G., Cheng K., Gao X.M., Shiu B., Shah N., Drachenberg C.B., Heath J., Wess J., Khurana S. Muscarinic receptor subtype-3 gene ablation and scopolamine butylbromide treatment attenuate small intestinal neoplasia in Apcmin/+ mice. Carcinogenesis. 2011;32:1396–1402. doi: 10.1093/carcin/bgr118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Erben U., Loddenkemper C., Doerfel K., Spieckermann S., Haller D., Heimesaat M.M., Zeitz M., Siegmund B., Kuhl A.A. A guide to histomorphological evaluation of intestinal inflammation in mouse models. Int J Clin Exp Pathol. 2014;7:4557–4576. [PMC free article] [PubMed] [Google Scholar]
  • 100.Dobin A., Davis C.A., Schlesinger F., Drenkow J., Zaleski C., Jha S., Batut P., Chaisson M., Gingeras T.R. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21. doi: 10.1093/bioinformatics/bts635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Li Bo. Dewey Colin RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics. 2011;4(12):323. doi: 10.1186/1471-2105-12-323. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Table S1
mmc1.xlsx (310.7KB, xlsx)
Supplementary Table S2
mmc2.xlsx (2.2MB, xlsx)
Supplementary Table S3
mmc3.xlsx (40.5KB, xlsx)
Supplementary Table S4
mmc4.xlsx (17.3KB, xlsx)
Supplementary Table S5
mmc5.xlsx (94.6KB, xlsx)

Articles from Cellular and Molecular Gastroenterology and Hepatology are provided here courtesy of Elsevier

RESOURCES