Abstract
The circadian clock allows organisms to coordinate biochemical and physiological processes over one day. Changes in lighting conditions as they occur naturally over seasons or manmade by jet lag or shift work, advance or delay clock phase to synchronize physiology to the environment. Within the suprachiasmatic nucleus (SCN) of the hypothalamus, circadian timekeeping and resetting have been shown to depend on both membrane depolarization and intracellular second-messenger signaling. In both processes, voltage-gated calcium channels (VGCCs) mediate calcium influx resulting in the activation of intracellular signaling pathways that activate Period (Per) gene expression. However, the precise mechanism how these processes are gated in a concerted manner is unknown. Here we show that cycling-dependent kinase 5 (Cdk5) activity is modulated by light and gates phase shifts of the circadian clock. We found that knock-down of Cdk5 in the SCN of mice affects phase delays but not phase advances. This is associated with uncontrolled calcium influx into SCN neurons and an unregulated protein kinase A (PKA) – calcium calmodulin dependent kinase (CaMK) – cAMP response element-binding protein (CREB) signaling pathway. Accordingly, genes such as Per1 are not induced by light in the SCN of Cdk5 knock-down mice. Our experiments identified an important light modulated kinase that affects rapid clock phase adaptation. This finding indicates how light responsiveness and clock phase are coordinated to adapt activity onset to seasonal changes, jet-lag and shift work.
The circadian system coordinates biochemical and physiological functions in our body and synchronizes them to the environmental day-night cycle. Misalignment of the internal body clock with the external light-dark cycle, induced by shift work or jet lag, leads to inefficient regulation of body functions. As a consequence, circadian misalignment can lead to obesity, cancer, addictive behaviors, cardiovascular disease, and neurological disorders 1. Therefore, it is crucial to understand how the environment impacts the clock and how these entities interact.
In mammals, the master circadian clock is located in the ventral part of the hypothalamus just above the optic chiasm in the suprachiasmatic nuclei (SCN), which coordinate daily cycles of physiology and behavior 2. Molecular daily oscillations are generated at the cellular level by a cell-autonomous transcription-translation feedback loop (TTFL) involving a set of clock genes 3 and post-translational modifiers such as kinases 4, 5. Circuit-level interactions among SCN cells produce a coherent daily oscillation 6, which can be modulated by light signals to match the environmental light-dark cycle. Light is perceived by melanopsin containing photosensitive retinal ganglion cells (pRGCs) in the eye, and the signal produced in these cells travels via the retinohypothalamic tract (RHT) to the SCN 7. The release of glutamate at the RHT terminals stimulates AMPA/NMDA receptors, leading to Ca2+ influx into the SCN 8. Additionally, the activity of various kinases is changed, including DARPP-32 (dopamine and cAMP-regulated phosphoprotein of 32 kD), PKA (protein kinase A), and CaMK (Ca2+/calmodulin-dependent kinases). This cascade finally culminates in the phosphorylation of CREB (cyclic AMP response element binding protein) 9, 10, 11, 12, 13. This event promotes chromatin phosphorylation 14 and acetylation via the recruitment of CRTC1 (cAMP-regulated transcriptional co-activator 1) and the histone acetyltransferase CBP (CREB-binding protein), involving the clock protein PER2 15. As a consequence, immediate-early gene and clock gene expression is induced 16, 17, 18, causing a phase shift of the TTFL in oscillating cells of the SCN 6. This manifests at the behavioral level in a change of locomotor activity onset (phase shift) the day after the light pulse. The direction of the phase shift depends on the clock’s temporal state. Light perceived in the early night promotes phase delays. On the other hand, a light pulse late at night promotes phase advances, whereas light in the middle of the day does not alter the clock phase 19. For this so-called resetting of the circadian clock, the Per1 and Per2 genes appear to be important in the mouse. While Per1 is essential for phase advances, Per2 function is necessary for phase delays 20, 21.
Voltage-gated calcium channels (VGCCs) are classified into high voltage-activated channels (HVA), which include L-type and low voltage-activated (LVA) subtypes, also known as T-type channels 22. T-type VGCCs are involved in phase delays, whereas L-type VGCCs are related to phase advances 23, 24. CaV3.1, CaV3.2, and CaV3.3 belong to the T-type channel family, which is critically important for neuronal excitability 25. The activity of these T-type channels are regulated by various kinases, including PKA 26, PKC 27, and CDK5 (cyclin-dependent kinase 5) 28.
Cdk5 is a proline-directed serine/threonine kinase that forms a complex with its neural activators p35 or p39 29, 30 and cyclin I 31. The complex of Cdk5 and its activators controls various neuronal processes such as neurogenesis, neuronal migration, and synaptogenesis 32,33. In vivo and in vitro experiments show that Cdk5 kinase activity is low in the light phase and high in the dark phase 34, 35. It regulates the circadian clock in the SCN via phosphorylation of PER2 at serine 394 (Per2Brdm domain). Upon phosphorylation by Cdk5, PER2 is stabilized and enters the nucleus to participate in the regulation of the TTFL and CREB-related transcriptional events 15, 34. Since Cdk5 regulates the T-type channel CaV3.1 28and the circadian clock via PER2 phosphorylation 34, we analyzed a potential role of Cdk5 in the light-mediated clock resetting mechanism.
Results
Cdk5 knock-down in the SCN impairs light-induced phase delays
Light perceived in the dark period elicits changes in the clock phase 19. To test whether Cdk5 plays a role in this process, we knocked down Cdk5 in the SCN via stereotaxic application of adeno-associated viruses (AAVs). We injected an adenovirus expressing shRNA to silence Cdk5 (shCdk5) and, as a control, an adenovirus expressing a control shRNA (scr) into the SCN 34. Consistent with our previous observations 34, we found that silencing Cdk5 in the SCN reduced its expression in the SCN (Supplementary Fig. 1a) and the expression of PER2 (Supplementary Fig. 1b). Under constant darkness (DD) conditions this knock-down of Cdk5 shortened clock period in mice as assessed by wheel-running activity (Fig. 1a, b and Supplementary Fig. 1c). This period was not influenced by light pulses (Supplementary Fig. 1d). However, onset of activity was affected after releasing mice into constant darkness (DD). Light at zeitgeber time (ZT) 14 (where ZT0 is lights on and ZT12 is lights off) delayed the clock phase, whereas light at ZT22 advanced it in control (scr) animals, with light at ZT10 having no effect (Fig. 1a, c, Aschoff type II protocol). The animals with silenced Cdk5 in the SCN (shCdk5) behaved similarly to controls (scr), except for ZT14. Light did not elicit a phase delay at this time, suggesting that Cdk5 plays a role in the phase delay mechanism.
To corroborate our observations, we performed the same experiment in DD (Aschoff type I protocol). The shCdk5 animals displayed a shorter period compared to scr controls (Fig. 1d, e), consistent with previous observations 34. After determination of each animal’s clock period, we administered light pulses of 15 min. at circadian times (CT) 10, CT14, and CT22 for each animal (orange stars, Fig. 1d). Light at CT10 had no effect on both the shCdk5 and scr control mice (Fig. 1f). Light applied at CT14 promoted a phase delay in scr control mice. However, silencing of Cdk5 impaired the delay of the clock phase (Fig. 1d, f), which is consistent with the observation at ZT14 (Fig. 1a, c). Light at CT22 elicited normal phase advances in shCdk5 and scr controls (Fig. 1d, f), similar to the light pulse applied at ZT22 (Fig. 1a, c). From these experiments, we conclude that Cdk5 plays a role in delaying the clock phase in response to a light pulse in the early activity period of mice.
Cdk5 activity is modulated by light in the early night
Since we observed that Cdk5 plays a significant role in the phase shift of the circadian clock, we questioned whether the light signal at ZT14 could affect the levels of Cdk5 and its co-activator p35 in the SCN. To this end, we collected SCN samples at ZT14 in the dark or after a 15 min. light pulse, and we performed a western blot on total protein extracts. To control proper light induction, we measured the light-dependent phosphorylation of PKA (Fig. 2a, b) and CaMKII (Supplementary Fig. 2a, b). We confirmed that PKA and CaMKII phosphorylation levels were increased by light in the SCN (Fig. 2a, b, Supplementary Fig. 2a, b). Interestingly, we observed that light could also increase the p35 protein level, although the levels of Cdk5 were not affected (Fig. 2a, c). Because the levels of the Cdk5 co-activator p35 were increased by light, we wondered whether this event would affect the kinase activity of Cdk5/p35. We performed an in vitro kinase assay using the immunoprecipitated Cdk5 present in SCN tissue collected from mice not exposed to light or exposed to light at ZT14. We used the recombinant histone H1 as substrate in the presence of radioactive ATP 34. Surprisingly, our results indicated that Cdk5 kinase activity was decreased by light (Fig. 2d, e), suggesting that light may affect the interaction between Cdk5 and p35. Therefore, we performed a co-immunoprecipitation experiment using an antibody against Cdk5 as bait to challenge this hypothesis. Our results revealed that the SCN extract from mice that received a light pulse at ZT14 contained less pulled down p35 in a complex with Cdk5 (Fig. 2f). Taken together, the results support the hypothesis that light affects Cdk5 activity via interference with the formation of a complex of Cdk5 with its co-activator p35. Interestingly, the light pulse at ZT14 might affect more than just Cdk5/p35 protein: protein interactions and additional unknown proteins may be involved (Supplementary Fig. 2c).
Cdk5 impacts the CREB signaling pathway via calcium/calmodulin-dependent kinases (CaMK)
Deletion of a cAMP-responsive element (CRE) in the Per1 promoter blunted light-induced Per1 expression in the SCN at night 36. Because nocturnal light induces phosphorylation of CRE binding protein (CREB) and phosphorylated CREB (p-CREB) can bind to CREs 37, 38, 39, we investigated whether Cdk5 is involved in the pathway evoking the CREB phosphorylation at serine-133 (pSer-133), a site known to be involved in phase delays, and Per1 induction 11. Therefore, we performed immunohistochemical analysis using an antibody detecting phosphate on CREB at serine 133 (p-CREB-S133) (Fig. 3a, Supplementary Fig. 3b, control Supplementary Fig. 3c). In the SCN of control animals (scr), we observed p-CREB-S133 in nuclei of neurons after the light was delivered at ZT14 but not in controls (Fig. 3a, arrowheads). In contrast, p-CREB-S133 was already detected in nuclei before the light pulse in shCdk5 animals (Fig. 3a, arrowheads), indicating that Cdk5 plays a role in gaiting the phosphorylation of CREB.
The CREB/CRE transcriptional pathway has been shown to be activated by calcium/calmodulin-dependent kinase II (CaMKII) and mitogen-activated protein kinase (MAPK) 40, 41, 42. Pharmacological inhibition of CaMKII but not of MAPK affected light-induced phase delays in hamsters 43. Therefore, we tested whether phosphorylated CaMKII (p-CaMKII) is affected by the knock-down of Cdk5 in the SCN of mice. We observed that p-CaMKII presence (alpha isoform) in the cytoplasm of SCN cells increased after light at ZT14 compared to no light in control animals (Fig. 3b, left panels). In shCdk5 SCN, however, p-CaMKII was already present before the light pulse in significantly higher levels than controls (Fig. 3b, control Supplementary Fig. 3d). This result indicates that Cdk5 is gating the phosphorylation of CaMKII alpha.
CaMKII has been shown to shuttle Ca2+/calmodulin (Ca2+/CAM) to the nucleus to trigger CREB phosphorylation and gene expression 44. Therefore, we investigated whether CAM localization was influenced by a light pulse and whether Cdk5 plays a role in this process. We observed that in control animals, CAM was distributed evenly in the cytoplasm of cells in SCN tissue before a light pulse. However, after the light pulse, it was localized around the nuclei (Fig. 3c). Interestingly, in the SCN of shCdk5 animals, CAM was already localized around the nuclei before the light administration and remained there after the light pulse, suggesting that Cdk5 is gating CAM localization in the cell.
Once delivered to the nucleus, Ca2+/CAM triggers a highly cooperative activation of the nuclear CaMK cascade, including CaMKIV, to rapidly phosphorylate CREB for the transcription of target genes 44, 45. Therefore, we tested whether a light pulse affected the phosphorylation of CaMKIV and whether this was influenced by Cdk5. In control animals, we detected p-CaMKIV to be strongly present in the SCN after but not before a light pulse (Fig. 3d, control Supplementary Fig. 3e). In shCdk5 SCN, p-CaMKIV was always detectable independent of the light pulse (Fig. 3d). This indicated that Cdk5 was gating phosphorylation of CaMKIV.
Calcium entry is regulated by channels, such as T-type VGCC, which are involved in phase delays 23. Previous reports show that Cdk5 directly or indirectly can phosphorylate Cav3.1 in vitro 28. Thus, we looked at the influence of light and Cdk5 on the T-type channel Cav3.1 using immunohistochemical staining. We observed that the level of Cav3.1 protein was significantly increased on the surface of SCN cells after the light pulse (Fig. 3e, blue bars). This suggests that light inhibits internalization and degradation of this channel. Interestingly, in the Cdk5 depleted SCN cells, Cav3.1 staining was already high on the cell surface before the light signal (Fig. 3e, red bars). We observed no difference in the Cav3.1 signal between SCN samples obtained from shCdk5 mice before and after the light pulse (Fig. 3e, red bars), suggesting that Cdk5 may be directly or indirectly involved in the regulation of Cav3.1 localization. This is consistent with previously described effects of Cdk5 on the cellular localization of other receptors such as the D2 and TRPV1 receptors 46, 47.
Cdk5 modulates neuronal activity in response to light at ZT14
Neuronal activity in response to light at ZT14 requires calcium influx. At night neuronal cell membranes are hyperpolarized, creating a Ca2+ gradient. A light stimulus at night promotes membrane depolarization and VGCC activation, which evokes a Ca2+ influx into SCN neurons ultimately changing the phase of the circadian clock 48, 49. Our results shown in figure 3 indicate that Cdk5 regulates the gating between light and the CaMKII pathway, which relies on Ca2+ availability. Thus, we tested whether Cdk5 regulated the light-mediated Ca2+ influx into SCN neurons. To this end, we employed in vivo calcium imaging to assess changes in calcium levels in the SCN in freely moving mice after 15 minutes of a light pulse given at ZT14. First, we injected an adeno-associated virus (AAV) expressing the shCdk5 sequence into the SCN to silence Cdk5. This AAV is co-expressing the calcium indicator GCaMP7 under the neuron-specific synapsin 1 promoter. As a control, we injected an AAV carrying a NSshRNA (scrambled sequence) instead of shCdk5 (see Materials and Methods). Consistent with our previous results, the construct expressing shCdk5 in the SCN produced a shortened free-running period in mice (Supplementary Fig. 4a-c). Animals injected with AAV were implanted with a chronic optical fiber placed above the SCN to allow for longitudinal imaging of GCaMP7 signals using fiber photometry. After habituation, ΔF/F0 (or the ratio of change in GCaMP7 fluorescence to the baseline fluorescence, see methods) was recorded before and after light pulse delivery at ZT14 in both groups of mice (Fig. 4a).
We observed an increase of calcium activity in control mice (scramble) during 15 min. of the light pulse at ZT14, which was also sustained for over 15 min. after the light pulse (Fig. 4b, black trace). In contrast, the ΔF/F0 in shCdk5 mice during and after the 15 min. light pulse was significantly lower compared to the control animals (Fig. 4b, red trace). Interestingly, we could observe random, but regular calcium transients, unrelated to the light stimulus during and after the light pulse (Fig. 4b, at -1, 2-3, 9, 12, and 14 min during the light pulse and 5, 9 and 12 min after the light pulse). A Ca2+ transient was observed right before the light was given at ZT14 (Fig. 4b), which showed the same magnitude as those observed during and after the light stimulus.
This activity was significantly decreased in shCdk5 mice during the last five minutes of the light pulse as compared to the baseline levels (see Methods; Fig. 4c; Supplementary Fig. 4d). Interestingly, 5 min. and 15 min. after the light pulse, the calcium mediated activity remained significantly lower in shCdk5 animals compared to controls (Fig. 4c and Supplementary Fig. 4d). Finally, mice were sacrificed, and the GFP signal was assessed by immunostaining to verify virus expression in the SCN (Fig. 4d). The outlined circle in red indicates where the fibers were located. Taken together, these results indicate that Cdk5 modulates Ca2+ mediated neuronal signaling.
Cdk5 regulates the DARPP32-PKA axis
The cAMP-activated Protein Kinase A (PKA) signaling pathway, leading to phosphorylation of CREB, plays a pivotal role in regulating phase delays in photic resetting 50, 51. Since the PKA signaling pathway can be induced in vivo 10, 52 and in vitro 53, we wondered whether Cdk5 could play a role in PKA- mediated CREB phosphorylation. To this end, we employed Förster resonance energy transfer (FRET) a widely used method to investigate molecular interactions between proteins such as CREB: CBP in living cells 15, 54.
We transfected control (wt) and Cdk5 knock-out (Cdk5 KO) NIH 3T3 cell lines 34 with ICAP (an indicator of CREB activation due to phosphorylation) and stimulated the cells with forskolin in presence of Ca2+. Phosphorylation of the CREB domain in the reporter decreased the FRET signal after 30 min. while non phosphorylation increased it. We observed that the FRET signal in wt cells strongly decreased within 30 min. after the stimulus compared to baseline (Fig. 5a, blue trace). In contrast, the FRET signal in Cdk5 KO cells rose towards baseline after an initial decline in response to forskolin (Fig. 5a, red trace). This indicated that Cdk5 is involved in the phosphorylation of CREB. Of note is that the forskolin solvent DMSO can’t stimulate CREB phosphorylation on its own (Supplementary Fig. 5a).
Previous studies have described that Ca2+-mediated CREB transcription of target genes requires PKA activity 42. However, to date it is not clear whether there is a parallel (synergistic) relationship between PKA and Ca2+ signaling pathways or whether they are sequentially depending on each other (Fig 5b, cartoon model). To address this question, we performed the following FRET experiment. NIH 3T3 cells were stimulated with forskolin in either the presence of Ca2+ with EGTA (Ca2+ chelator) (Fig. 5b, orange line), without EGTA (Fig. 5b, blue line) or completely depleted of Ca2+ (Fig. 5b, salmon colored line). We observed that under normal conditions the FRET signal goes down, comparable to the signal seen in figure 5a, indicating a higher Ser-133 KID phosphorylation compared to the baseline (Fig 5b, blue signal). When we added EGTA (removing Ca2+), the FRET signal increased to the baseline level after the forskolin treatment (Fig 5b, orange signal). The cells depleted of Ca2+ were also not responsive to the forskolin stimulus since the FRET signal moved towards the baseline level within 30 min. (Fig. 5b, salmon colored signal). Together, our results indicate that CREB phosphorylation is modulated by Cdk5 via Ca2+ signaling as suggested in figure 3. Interestingly, PKA appeared not to directly phosphorylate CREB, because CREB did not pull-down p-PKA in an immunoprecipitation experiment. In contrast p-CaMKIV did interact with CREB (Supplementary Fig. 5b, c) suggesting that CREB is most likely phosphorylated by CaMKIV, which is probably indirectly regulated by PKA activity.
Next, we aimed to investigate what the possible pathway could be through which PKA regulates CaMKIV. Previous studies have shown that Cdk5 regulates PKA activity via DARPP32 55 (Fig. 5c). Therefore, we asked whether DARPP32 phosphorylation was light-dependent and whether Cdk5 would modulate this process. We sacrificed mice either receiving a light pulse at ZT14 or no light. Cryo-sections containing the SCN were stained with an antibody recognizing phosphorylated Thr-75 (pThr-75) of DARPP32. We observed that DARPP32 is highly phosphorylated at ZT14 with the light signal reducing the phosphorylation levels in the cytoplasm and nuclei significantly (Fig. 5d, blue bars; Supplementary Fig. 5d, e). In contrast, silencing of Cdk5 led to a dramatic decrease of the pThr-75 signal in the cytoplasm and nuclei of SCN cells at ZT14 and light did not have an effect (Fig. 5d, red bars, Supplementary Fig. 5e). These observations are consistent with the view that Cdk5 phosphorylates DARPP32 and that light inhibits this process probably through an unknown mechanism inactivating Cdk5.
Non phosphorylated DARPP32 promotes PKA activity, which is characterized by phosphorylation at Thr-197 in the catalytic site of PKA 56, 57. Therefore, we asked whether decreased levels of p-DARPP32 after the light stimulus at ZT14 could inversely correlate with the phosphorylation state of PKA. Thus, we performed immunostaining on coronal brain sections containing the SCN using an antibody recognizing the phosphorylated Thr-197 of PKA. We observed that PKA phosphorylation significantly increased after the light pulse in the SCN tissue obtained from control (scr) mice (Fig. 5e, right panel, blue bars). However, in SCN from shCdk5 mice, the phosphorylation level was already elevated before the light pulse compared to scr control (Fig. 5e, left panels, top micrographs), and it was also sustained after the light pulse (Fig. 5e, left panels, bottom micrographs, right panel, red bars). Our results indicate that Cdk5 gates PKA phosphorylation induced by the light pulse at ZT14. Many observations indicate that active PKA can stimulate the Ca2+ influx through Cav3 T-type voltage gated channels, including Cav3.1 27, 58. The molecular mechanism normally requires physical interaction between the channel and PKA, followed by phosphorylation which influences the gating properties 59. Therefore, we performed a co-immunostaining in the same SCN sections collected before (Fig. 3) to detect both Cav3.1 and phospho-PKA (the active form). We observed that the colocalization between Cav3.1 and phospho-PKA dramatically increased after the light pulse in the SCN tissue of control (scr) mice (Fig. 5f, scr left panels yellow color, and blue bars in the right panel). Interestingly, the colocalization level of the two proteins was already high in the shCdk5 SCN tissue before the light pulse, compared to controls (Fig. 5f scramble vs. shCdk5, left panel, top micrographs). The colocalization level between Cav3.1 and phospho-PKA in the shCdk5 tissues was not influenced by the light pulse (Fig. 5f, right panel, red bars). Altogether our results suggest that Cdk5 gates the PKA-Cav3.1 interaction in response to the light signal at ZT14 in an indirect way via DARPP32.
Cdk5 affects light-induced gene expression
Light perceived in the dark period leads not only to phase shifts but is also paralleled by the induction of immediate early genes and certain clock genes in the SCN 16, 17, 18, 60, 61. This process involves the PKA – CaMK – CREB signaling pathway (reviewed in 62). Therefore, we investigated whether Cdk5 is involved in the signal transduction process to induce immediate early genes and clock genes in the SCN in response to light. To that extent, we performed a time-course profile of light-induced genes and immediate early genes. We collected SCN from mice that received a nocturnal light pulse at ZT14 at different time points after the stimulus over 2 hours (Fig. 6). In agreement with previous studies, Per1 and Dec1 mRNA expression was induced by light peaking at 1h after the stimulus. Conversely, Per2 and Dec2 mRNA expression was not affected by the light pulse at ZT14 (Fig. 6a-d, blue bars) 18, 61, 63. Knock-down of Cdk5 abolished this light-driven Per1 and Dec1 gene induction (Fig. 6a, c, red bars), indicating an involvement of Cdk5 in the light-driven activation process of these clock genes. As previously reported, expression of the clock gene Bmal1 was not light-inducible 34, 64 and not affected by shCdk5 (Fig. 6e). The injection of the control scr and shCdk5 constructs was successful, as demonstrated by the expression of eGFP mRNA in the analyzed SCN (Fig. 6f).
Interestingly, the knock-down of Cdk5 did not affect light-mediated induction of cFos expression, which peaked at 0.5h after the light pulse (Fig. 6g). In contrast, Egr1, another immediate early gene, which is involved in synaptic plasticity, learning, and memory 65, was light-inducible in control but not in shCdk5 animals (Fig. 6h). This suggests that the immediate early gene cFos is regulated by a different mechanism compared to Egr1 and the clock genes Per1 and Dec1 in response to a light stimulus at ZT14.
Vasoactive intestinal polypeptide (VIP) was described to play a role in phase-shifting the SCN clock 66. Furthermore, the light-induced expression of clock genes is localized in VIP-positive cells in the SCN that are essential for clock resetting 67. Therefore, we tested whether Vip gene expression is affected by shCdk5. We observed that a light pulse did not induce Vip expression significantly in the SCN, nor did shCdk5 affect its general expression (Supplementary Fig. 6a). This suggests that Cdk5 does not regulate Vip expression and modulate phase shifts via VIP.
Salt inducible kinase 1 (Sik1) is involved in the regulation of the magnitude and duration of phase shifts by acting as a suppressor of the effects of light on the clock 68. Therefore, we tested how a light pulse affected Sik1 expression in the SCN and whether Cdk5 might play a role in its regulation. We observed that Sik1 was significantly induced by a light pulse in the SCN of control mice after 0.5 h. However, the knock-down of Cdk5 abolished this induction (Fig. 6i). This suggests that Cdk5 modulates Sik1 expression to regulate the magnitude of the behavioral response to light.
The light-inducible small G-protein Gem limits the circadian clock phase-shift magnitude by inhibiting voltage-dependent calcium channels 69. We tested whether a light pulse affected Gem expression in the SCN and whether this was involving Cdk5. We observed that Gem was significantly induced by light 1 hour after light administration (Fig. 6j, blue bars). Interestingly, knock-down of Cdk5 abolished this induction (Fig. 6i, red bars), but Gem levels seemed to be slightly but not significantly elevated already before light administration (Fig. 6i, time point 0). This indicates that Cdk5 influences light induced Gem expression and that Cdk5 may also affect basal Gem expression before the light pulse. Interestingly, similar results as for light induced gene expression in shCdk5 SCN were observed in SCN of Per2Brdm1 mutant mice (Supplementary Fig. 6b, c).
Phase shifts of the circadian clock can also be studied in cell cultures using forskolin instead of light as a stimulus 53. In accordance with our in vivo experiments (Fig. 6), expression of Per1 but not Per2 mRNA was induced in synchronized NIH 3T3 fibroblast cells after forskolin treatment (Supplementary Fig. 6d, e, blue bars). Comparable to the experiments in the SCN, Per1 induction was abolished in Cdk5 knock-out cells (Supplementary Fig. 6d). In contrast, cFos mRNA induction was not affected in Cdk5 knock-out cells (Supplementary Fig. 6f), which is consistent with our observations in the SCN (Fig. 6g).
Collectively, our expression data provide evidence that Cdk5 regulates light and forskolin-mediated expression of genes critical for the regulation of phase delays of the circadian clock. Immediate early genes, such as Egr1, are regulated in a similar manner, whereas others, such as cFos, are regulated by another mechanism not involving Cdk5.
Discussion
In this study, we investigated the role of Cdk5 in rapid phase shifts of the circadian clock. We found that Cdk5 activity is regulated by light and that Cdk5 is necessary for phase delays but not phase advances. We identified Cdk5 to play a major role in the modulation of Ca2+ levels and gating of the PKA-CaMK-CREB signaling pathway and coordinate it with the presence of PER2 in the nucleus of SCN cells.
In a previous study, we identified the protein kinase Cdk5 to regulate the phosphorylation and nuclear localization of the clock protein PER2 34. Because PER2 and protein kinases are involved in the photic signaling mechanism of clock phase adaptation 15, 20, 62, 70, 71, we tested the involvement of Cdk5 in this process. The phenotype of Cdk5 knock-down (shCdk5) in the SCN of mice resembled the phenotypes observed in Per2 mutant (Per2Brdm1) and neuronal Per2 knock-out (nPer2 ko) mice. ShCdk5, as well as Per2Brdm1and nPer2 ko animals, showed strongly reduced phase delays in response to a short light pulse given at ZT14 (Fig. 1a, c) 20 or CT14 (Fig. 1d, f) 21, 72. These mouse lines displayed a shortened period consistent with previous observations (Fig. 1b, e) 34, 73. Our results indicate that Cdk5 is not only involved in the regulation of the circadian clock mechanism via nuclear localization of PER2 but also plays an important role in the molecular mechanism that leads to a delay of clock phase in response to a light pulse in the early dark phase or early subjective night.
Since Cdk5 mediates the effects of light at the behavioral (Fig. 1) level, we tested the influence of light on Cdk5 protein accumulation and kinase activity in the SCN at ZT14 (Fig. 2). We observed no change in the protein accumulation of Cdk5. On the other hand, Cdk5 kinase activity was reduced in the SCN after a light pulse at ZT14 (Fig. 2d, e) which was surprising in the context of increased p35 levels (Fig. 2a, c) and augmented PKA phosphorylation (Fig. 2a, b). However, this observation is in line with what we previously reported, where we demonstrated that Cdk5 kinase activity was low during the light phase and higher during the dark phase 34. It appeared, however, that p35 was not interacting with Cdk5 after light at ZT14 (Fig. 2f). Additional interactions of Cdk5 with unknown proteins may also be lost (Supplementary Fig. 2c). These observations suggest that Cdk5 was most likely modified in response to light leading to loss of interaction with p35 and other proteins. Ser159 of Cdk5 mediates the specificity of the Cdk5-p35 interaction 74, and therefore, phosphorylation of this site by an unknown kinase may mediate the loss of Cdk5 activity. Several additional phosphorylation sites in Cdk5 have been identified, of which phosphorylation of S47 renders Cdk5 inactive 75. Which one of the phosphorylation sites in Cdk5 is modulated by light and what additional interactors may be involved in this process remains to be established.
Light in the early portion of the dark phase elicits phase delays, which involve T-type calcium channels, PKA-signaling, and Ca2+ signaling, ending in the phosphorylation of CREB (reviewed in 62). We observed that in shCdk5 mice, CREB was already phosphorylated in the absence of light, although the total protein amount did not change (Fig. 3a, Supplementary Fig. 3a, b). Similarly, CaMKII and CaMKIV were shown to be phosphorylated and, therefore, activated only after the light pulse in control animals (Fig. 3b, d). Conversely, these kinases were highly phosphorylated in a light-independent manner in the SCN of shCdk5 animals (Fig. 3b, d) indicating that Cdk5 had a suppressive function on the phosphorylation of CaMKII and CaMKIV.
A stimulus can promote calmodulin (CAM) involving CaMKII gamma to translocate from calcium channels to the nucleus to promote CaMKIV phosphorylation and activation 44. Unexpectedly, we observed that a light stimulus can have a similar but distinct effect on CAM in SCN cells (Fig. 3c). CaMKII alpha was phosphorylated after a light pulse at ZT14, which lead to perinuclear localization of CAM in control mice, while this localization pattern was already observed in shCdk5 animals independently of the light stimulus (Fig. 3c). In control mice that received no light CAM showed a diffuse expression pattern similar to the T-type calcium channel Cav3.1 at ZT14.
Interestingly, this light driven localization pattern was echoed by the change in cellular distribution of the T-type calcium channel Cav3.1 known as internalization/externalization. Again, the presence of Cdk5 suppressed the localization of this channel to the cell membrane in the absence of light, with light allowing localization to the cell membrane (Fig. 3e). This observation is reminiscent of investigations described previously in which Cdk5 appeared to play an important role in channel translocation 76, 77 as well as in receptor translocation 46, 47. Thus, our findings are in accordance with the view that Cdk5 plays a crucial role in light stimulus driven cell dynamics.
Calcium plays an important role in circadian and phase-shifting biology 48. Circadian calcium fluxes in the cytosol of SCN neurons have been demonstrated in vitro 78 and they change rapidly as a response to light perceived by the retina 49. We performed in vivo live imaging to detect Ca2+ levels in the SCN using fiber photometry with protein-based Ca2+ indicators such as GCaMP 79. With this approach, we observed that calcium fluxes in the SCN of control mice increased during and after a light pulse, but this change was significantly dampened in shCdk5 animals (Fig. 4b, c). Interestingly, although the Ca2+ influx was generally reduced in the SCN of shCdk5 mice, we observed random Ca2+ activity, which were independent of any light stimulus. These transients were observed also at the beginning of ZT14, before the light pulse (Fig. 4b, c). These results may indicate the presence of a calcium leak reminiscent of the already active phosphorylation cascade observed in the shCdk5 SCN in the absence of light (Fig. 3). We do not know, however, whether internal calcium stores involving ryanodine receptors 80 are altered by Cdk5 as well and how this would contribute to the observed phenotypes.
The PKA signaling pathway is involved in the resetting of the circadian phase (reviewed in 50). Interference with PKA activation in the early subjective night led to reduced phase delay responses as observed in vitro in the SCN 52. Here we find that Cdk5 plays an inhibitory role in PKA phosphorylation and activation. The FRET approach shows that in cells the lack of Cdk5 makes cells unresponsive to forskolin (Fig. 5a), an agent known to mitigate phase shifts in cells via PKA 53. Interestingly, PKA appears to influence phase shifts and CREB phosphorylation indirectly via a Ca2+ dependent mechanism (Fig. 5b) with phosphorylated CaMKIV being the kinase that phosphorylates CREB (Supplementary Fig. 5b, c). This observation is in contrast with previous studies that suggested a direct phosphorylation of CREB by PKA 10, 52. However, p-PKA is mostly located in the cytoplasm (Fig. 5e) while p-CaMKIV is in the nuclei (Fig. 3d). Furthermore, our experiments indicate that CREB did not interact with p-PKA but did with p-CaMKIV (suppl Fig. 5b, c), supporting the notion that PKA regulates CREB phosphorylation indirectly via CaMKIV in the SCN.
Because we observed that PKA was already phosphorylated in the dark when Cdk5 was silenced (Fig.5e), we asked how Cdk5 could negatively regulate PKA phosphorylation. A previous study described that Cdk5 can phosphorylate DARPP32 to suppresses PKA activity 81. Furthermore, Darpp-32 KO mice show attenuated phase delays 12 resembling shCdk5 mice (Fig. 1). In accordance with these studies, we found an inverse correlation between p-DARPP32 (Fig. 5d) and p-PKA (Fig. 5e), implying that Cdk5 indirectly inhibits PKA activity via DARPP32.
Our results imply that PKA action on CREB might be mediated via T-type calcium channels such as Cav 3.1 (Fig. 5f). This assumption is reasonable because PKA can phosphorylate Cav 3.1 channels and increase electrical conductivity, which leads to a higher influx of Ca2+ 26. To that extent, our results indicate that a higher co-localization of p-PKA with Cav 3.1 is associated with an activation of the CaMKinase pathway and CREB phosphorylation.
Light induced phosphorylation of CREB leads to induction of immediate early genes and clock genes (reviewed in 62). Accordingly, we observed that the clock genes Per1 and Dec1 but not Per2, Dec2, and Bmal1 were induced in the SCN by light at ZT14 (Fig. 6a-e, blue bars) consistent with previous findings 15, 18, 61, 63. The light induction of Per1 and Dec1 was abolished in shCdk5 animals (Fig. 6a, c, red bars) as well as in Per2Brdm1mutant mice (Supplementary Fig. 6b, c), suggesting involvement of Cdk5 and Per2 in induction of these genes. In contrast, light induction of the immediate early gene cFos was neither affected in shCdk5 nor Per2Brdm1SCN (Fig. 6g, Supplementary Fig. 6f), resembling the normal cFos induction in Per2 KO animals 15. This indicates that the light signaling mechanism for cFos induction is different from the one mediating induction of Per1 and Dec1. Interestingly, however, the light-inducible genes Sik1 and Gem, which are involved in limiting the effects of light on the clock 68, 69 were not light-inducible in shCdk5 animals (Fig. 6i, j) supporting the view that the factors that drive (Per1, Dec1) or limit (Sik1, Gem) the effects of light on the clock are regulated by the same mechanism. Interestingly, neither lack of Per1, Dec1 20, 82 nor Sik1 or Gem 68, 69 alone abolish phase delays. Of note is that lack of cFos or Egr1 did not affect phase delays either 83, 84. Furthermore, the neuropeptide vasoactive intestinal peptide (VIP), which is important in circadian light responses 67, was not inducible by a light pulse at ZT14 in control as well as shCdk5 animals (Supplementary Fig. 6a), indicating that Cdk5 acts upstream of VIP signaling. Overall, our data suggest that Cdk5 not only regulates the light sensitive PKA – CaMK-CREB signaling pathway but ultimately also affects gene expression. The combination of lack of induction of many genes in the Cdk5-regulated pathway is responsible for the manifestation of rapid behavioral phase delays.
Based on this and our previous studies, we propose the following molecular model for light-mediated phase delays (Fig. 7). The model is divided into two parts. One part describes the state before the light pulse, and the second part the mechanism after the light pulse. The state before the light pulse (ZT12-14) is depicted in the gray area in Figure 7. As reported previously, Cdk5 is active right after dark onset 34, depicted as the active Cdk5/p35 complex (blue). This has two consequences: 1) PER2 (red) is phosphorylated and translocates to the nucleus 34, and 2) DARPP32 is phosphorylated and thus inhibiting PKA activity 81. Hence, before the light pulse at ZT14, the nucleus is supplied with PER2, which appears to be necessary for light-mediated behavioral phase delays 20, 21, 72. In parallel, the signaling pathway necessary to phosphorylate CREB is turned off. This state can then be dramatically changed when light is applied at ZT14 evoking glutamate and PACAP release at the synapses between the RHT and the SCN. Interaction between p35 and Cdk5 is abolished (Fig. 2) thereby inactivating Cdk5 and stopping phosphorylation of PER2 and DARPP32. Since significant amounts of PER2 are already in the nucleus this probably has no consequences on nuclear PER2 function. However, DARPP32 is not phosphorylated anymore and the block on PKA is released. At the same time, PKA becomes phosphorylated due to PACAP and cAMP signaling, leading to activation of Cav3.1 by PKA (Fig. 5f, 26). This leads to CaMKII and CaMKIV phosphorylation and, ultimately, to the phosphorylation of CREB in the nucleus (Fig. 3, 45). Phospho-CREB builds up a complex with CRTC1/CBP and PER2 15 to activate gene expression of the Per1, Dec1, Sik1, and Gem genes. In the activation complex the amount of PER2 present in the nucleus may at least in part affect the magnitude of the phase delay, which is depending on the time the light pulse is given. In conclusion, Cdk5 activity is gating both processes, the pre-light condition as well as the post-light condition, leading to a concerted activation of a set of light-responsive genes that impinge on behavioral phase delays in response to nocturnal light exposure.
Methods
Animals and housing
All mice were housed with chow food (3432PX, Kliba-Nafag) and water ad libitum in transparent plastic cages (267 mm long, ×207 mm wide, ×140 mm high; Techniplast Makrolon type 2 1264C001) with a stainless-steel wire lid (Techniplast 1264C116), kept in soundproof ventilated chambers at constant temperature (22 ± 2 °C) and humidity (40 – 50%). All mice were entrained to a 12-h light–dark cycle (LD cycle), and the time of day was expressed as Zeitgeber time (ZT; ZT0 lights on, ZT12 lights off). Four-month-old 129/C57BL6 mixed males were used for the experiments. Housing and experimental procedures were performed per the guidelines of the Schweizer Tierschutzgesetz and the declaration of Helsinki. The state veterinarians of the Canton of Fribourg and Bern approved the protocol (license numbers: 2021-19-FR; BE45/18; BE21/22).
Locomotor activity monitoring
Locomotor activity parameters were analyzed by monitoring wheel-running activity, as described in 85, and calculated using the ClockLab software (Actimetrics). To analyze free-running rhythms, animals were entrained to LD 12:12 and released into constant darkness (DD). The internal period length was determined from a regression line drawn through the activity onsets of ten days of stable rhythmicity under constant conditions, calculated using the respective inbuilt functions of the ClockLab software (Acquisition Version 3.208, Analysis Version 6.0.36). For better visualization of daily rhythms, locomotor activity records were double-plotted, which means that each day’s activity is plotted twice, to the right and below that of the previous day. For the analysis of light-induced resetting, we used an Aschoff type II and I protocol 85. For type II, mice maintained in LD 12:12 were subjected to a 15 min. light pulse (LP, 500 lux) at ZT10 (no phase shift), 14 (phase delay), and 22 (phase advance). Subsequently, they were released into DD for ten days, and phase shift was measured. For type I, mice maintained in DD were subjected to a 15 min. light pulse at Circadian Time (CT) 10, 14, or 22. A circadian hour equals 1/24 of the endogenous period (τ), calculated as follows: circadian hour = tau/24 hours. To convert ZT hours to CT hours, we multiply the circadian hour by the actual hours needed to apply the light pulse at a specific time [i.e., CT14= (tau/24 hours) *14]. The phase shift was determined by fitting a regression line through the activity onsets of at least 7 days under LD conditions before the light pulse and a second line through the activity onsets of at least 7 days under DD after the light pulse. The first two days after the administration of the light pulse were not considered for the calculation of the phase shift. The distance between the two regression lines determined the phase shift. Before starting any new protocol, mice were allowed to stabilize their circadian oscillator for 10 days. The corresponding figure legends indicate the number of animals used in the behavioral studies.
Light pulse and tissue isolation
Light pulse (LP., 500 lux) was given at ZT14, and mice were sacrificed at appropriate indicated times. Brains were collected, and SCN tissue was isolated for western blot or RT-qPCR use. For immunofluorescence experiments, mice were perfused with 4% PFA and cryoprotected in 30% sucrose. Tissue isolation at ZT14 without a light pulse was used as light-induction negative control.
RNA extraction and cDNA synthesis
Total RNA was extracted from confluent 6 cm petri dishes or frozen SCN tissue using the Microspin RNA II kit (Machery & Nagel, Düren, Germany) according to the manufacturer’s instructions. 0.5 μg of total RNA was converted to single-strand cDNA in a total volume of 10 μL using the SuperScript IV VILO kit (Thermo Fisher Scientific, Waltham MA, USA) according to the manufacturer’s instructions. The samples were diluted to 200 μL with pure water. 5 μL of each sample were mixed with 7.5 μL of KAPA probe fast universal real-time PCR master mix (Merck, Darmstadt, Germany) and 2.5 μL of the indicated primer/probe combinations. For the subsequent real-time PCR, a Rotorgene 6000 machine was used (Qiagen, Hilden, Germany) and analyzed with the propriety software.
qPCR primers
Per1:
FW: GGC ATG ATG CTG CTG ACC ACG
RV: ACT GGG GCC ACC TCC AGT TC
TM: FAM-TGG CCC TCC CTC ACC TTA GCC TGT TCC T-BHQ1
Per2:
FW: TCC ACA GCT ACA CCA CCC CTT A
RV: TTT CTC CTC CAT GCA CTC CTG A
TM: FAM-CCG CTG CAC ACA CTC CAG GGC G-BHQ1
Dec1
FW: TGC AGA CAG GAG CGC ACA GT
RV: GCT TTGGGC AGG CAG GTA GGA
TM: FAM-TGG TTG CGC GCT GGG GAT CCG T-BHQ1
Dec2
FW: ACA GAA TGG GGA GCG CTC TCT GAA
RV: TGA AAC CCC GAG TGG AAC GCA
TM: FAM-CGC CGG TCC AGG CCG ACT TGG A-BHQ1
Bmal1
FW: GCA ATG CAA TGT CCA GGA AG
RV: GCT TCT GTG TAT GGG TTG GT
TM: FAM-ACC GTG CTA AGG ATG GCT GTT CAG CA-BHQ1
eGFP
FW: CAT CTG CAC CAC CGG CAA GC
RV: GGT CGG GGT AGC GGC TGA A
TM: FAM-TGC CCG TGC CCT GGC CCA CC-BHQ1
cFos
FW: GCC GGG GAC AGC CTT TCC TA
RV: TCT GCG CAA AAG TCC TGT GTG TTG A
TM: FAM-CCA GCC GAC TCC TTC TCC AGC ATG GGC-BHQ1
Egr1
FW: CGG CAG CAG CGC CTT CAA T
RV: GGA CTC TGT GGT CAG GTG CTC AT
TM: FAM-CCT CAA GGG GAG CCG AGC GAA CAA CCC-BHQ1
Sik1
FW: GGC TGC ACG ACC AGC AAT CG
RV: GGC GGT AGA AGA GTG GTG CTG TA
TM: FAM-TCC TGC ACC AGC AGA GGC TGC TCC AG-BHQ1
Gem
FW: TGG GAA AAT AAG GGG GAG AA
RV: AGC TTG CAC GGT CTG TGA TA
TM: FAM-CCA CTG CAT GCA GGT CGG GGA TGC C-BHQ1
Vip
FW: AGC AGA ACT TCA GCA CCC TAG ACA
RV: TCG GTG CCT CCT TGG CTG TT
TM: FAM-AGC CGG AAA GGC AGC CCT GCC T-BHQ1
Tprkb (normalisation probe for Tprkb)
FW: GGC TGG CAT CAG ACC CAC AGA
RV: GGG CCC GTA GAG TCG GGA AA
TM: FAM-CCT GCG TCT GCC CTC TGA GGG CTG-BHQ1
Atp5h (normalisation probe for Atp5h)
FW: TGC CCT GAA GAT TCC TGT GCC T
RV: ACT CAG CAC AGC TCT TCA CAT CCT
TM: FAM-TCT CCT CCT GGT CCA CCA GGG CTG TGT-BHQ1
Sirt2 (normalisation probe for Sirt2)
FW: CAG GCC AGA CGG ACC CCT TC
RV: AGG CCA CGT CCC TGT AAG CC
TM: FAM-TGA TGG GCC TGG GAG GTG GCA TGG A-BHQ1
Nono (normalisation probe for Nono)
FW: TCT TTT CTC GGG ACG GTG GAG
RV: GTC TGC CTC GCA GTC CTC ACT
TM: FAM-CGT GCA GCG TCG CCC ATA CTC CGA GC-BHQ1
Immunofluorescence
SCN cryosections (40 µM) were placed in a 24-well plate, washed three times with 1x TBS (0.1 M Tris/0.15 M NaCl) and 2x SSC (0.3 M NaCl/0.03 M tri-Na-citrate pH 7). Antigen retrieval was performed with 2xSSC heating to 85°C for 30 min. Then, sections were washed twice in 2x SSC and three times in 1x TBS pH 7.5 before blocking them for 1.5 hours in 10% fetal bovine serum (Gibco)/0.1% Triton X-100/1x TBS at RT. If the recipient species for some raised antibody was the mouse, we performed a Mouse on Mouse (MOM; Ab269452) blocking (2h) before 10% FBS to block endogenous mouse immunoglobulins in a mouse tissue section. After the blocking, the primary antibodies (Table 1), diluted in 1% FBS/0.1% Triton X-100/1x TBS, were added to the sections and incubated overnight at 4°C. The next day, sections were washed with 1x TBS and incubated with the appropriate fluorescent secondary antibodies diluted 1:500 in 1% FBS/0.1% Triton X-100/1x TBS for 3 hours at RT. (Alexa Fluor 488-AffiniPure Donkey Anti-Rabbit IgG (H+L) no. 711–545–152, Lot: 132876, Alexa Fluor647-AffiniPure Donkey Anti-Mouse IgG (H+L) no. 715–605–150, Lot: 131725, Alexa Fluor647-AffiniPure Donkey Anti-Rabbit IgG (H+L) no. 711–602–152, Lot: 136317 and all from Jackson Immuno Research). Tissue sections were stained with DAPI (1:5000 in PBS; Roche) for 15 min. Finally, the tissue sections were rewashed twice in 1x TBS and mounted on glass microscope slides. Fluorescent images were taken using a confocal microscope (Leica TCS SP5), and pictures were taken with a magnification of 63x with or without indicated additional zoom. Images were processed with the Leica Application Suite Advanced Fluorescence 2.7.3.9723. Immunostained sections were quantified using ImageJ version 1.49. Statistical analysis was performed on three animals per treatment.
Adeno Associated Virus (AAV) production and stereotaxic injections
Experiments were performed as previously described 34. Stereotaxic injections were performed on 4- to 5-month-old mice under isoflurane anesthesia using a stereotaxic apparatus (Stoelting). The brain was exposed by craniotomy, and the Bregma was used as a reference point for all coordinates. AAVs were injected bilaterally into the SCN (Bregma: anterior-posterior (AP) − 0.40 mm; medial-lateral (ML) ±0.00 mm; dorsal-ventral (DV) – 5.7 mm, angle + /- 3°) using a hydraulic manipulator (Narishige: MO-10 one-axis oil hydraulic micromanipulator, http://products.narishige-group.com/group1/MO-10/electro/english.html) at a rate of 40 nL/min through a pulled glass pipette (Drummond, 10 µl glass micropipette; Cat number: 5-000-1001-X10). The pipette was first raised 0.1 mm to allow the spread of the AAVs and later withdrawn 5 min after the end of the injection. After surgery, mice were allowed to recover for 2 weeks and entrained to LD 12:12 before behavior and molecular investigations.
The injected viruses were:
SsAAV-9/2-hSyn1-chI[mouse(shCdk5)]-EGFP-WPRE-SV40p(A)
ssAAV-9/2-hSyn1-chI[1x(shNS)]-EGFP-WPRE-SV40p(A)
Protein extraction from SCN tissue
The protocol was a modified version of what was published before 15. Isolated SCNs obtained from 4 different mice were pooled according to the indicated condition (either dark or 15 min after the light pulse). The pooled tissues were frozen in liquid N2 and resuspended in a brain-specific lysis buffer (50 mM Tris-HCl pH 7.4, 150 mM NaCl, 0.25% SDS, 0.25% sodium deoxycholate, 1 mM EDTA). They were homogenized using a pellet pestle, kept on ice for 30 min and vortexed for 30 s, followed by N2 freezing. Frozen samples were left to melt on ice. The samples were sonicated (10 s, 15% amplitude) and centrifuged for 20 min at 16,000 g at 4 °C. The supernatant was collected in new tubes, and the pellet was discarded.
Immunoprecipitation
The protocol was described before 34. The protein extract was diluted with the appropriate lysis buffer in a final volume of 250 µL and immunoprecipitated using the indicated antibody (ratio 1:50). The reaction was kept at 4°C overnight on a rotary shaker. The day after, samples were captured with 50 µL of 50% (w/v) protein-A agarose beads (Roche), and the reaction was kept at 4°C for 3 hr on a rotary shaker. Before use, beads were washed thrice with the appropriate protein buffer and resuspended in the same buffer (50% w/v). The beads were collected by centrifugation and washed three times with NP-40 buffer (100 mM Tris-HCl pH7.5, 150 mM NaCl, 2 mM EDTA, 0.1% NP-40). After the final wash, beads were resuspended in 2% SDS 10%, glycerol, 63 mM Trish-HCL pH 6.8, and proteins were eluted for 15 min at RT. Laemmli buffer was finally added, and samples were boiled for 5 min at 95° C and loaded onto 10% SDS-PAGE gels.
Western blot
Circa 40 µg o of protein was loaded onto 10% SDS-PAGE gel and run at 100 Volt for two hours. Protein migration was followed by a semidry transfer (40 mA, 1 h 30 s) using Hybond® ECL™ nitrocellulose. We subsequently performed red ponceau staining (0.1% of Ponceau S dye and 5%) on the membrane to confirm the successful transfer. The list of antibodies used in the paper is shown in Table 2. The membrane was washed with TBS 1x/Tween 0.1% and blocked with TBS 1x/BSA 5%/Tween 0.1% for 1 h. After washing, the membrane was blotted with the appropriate primary antibodies overnight. The day after, membranes were washed three times with TBS 1x/Tween 0.1%, followed by secondary antibody immunoblotting for 1 h at room temperature. The densitometric signal was digitally acquired with the Azure Biosystem.
In vitro kinase assay using immunoprecipitated Cdk5 from SCN
The protocol is the same as before 34. CDK5 was immunoprecipitated (4°C overnight with 2x Protein A agarose (Sigma-Aldrich)) from SCN samples at ZT14 in the dark or after a light pulse (LP., circa 500 lux) of 15min. After immunoprecipitation, samples were diluted in a washing buffer and split into two halves. One-half of the IP was used for an in vitro kinase assay. Briefly, 1 µg of histone H1 (Sigma-Aldrich) was added to the immunoprecipitated CDK5. Assays were carried out in reaction buffer (30 mM HEPES, pH 7.2, 10 mM MgCl2, and 1 mM DTT) containing [γ-32P] ATP (10 Ci) at 30°C for 1 hour and then terminated by adding SDS sample buffer and boiling for 5 min. Samples were subjected to 15% SDS-PAGE, stained by Coomassie Brilliant Blue, and dried, and then phosphorylated histone H1 was detected by autoradiography. The other half of the IP was used for Western blotting to determine the total CDK5 immunoprecipitated from the SCN samples. The following formula was used to quantify the kinase activity at each time point: ([32P] H1/total H1)/CDK5 IP protein.
Cell Culture
NIH3T3 wt and CRISPR/Cas9 Cdk5 ko 34 mouse fibroblast cells (ATCCRCRL-1658) were maintained in Dulbecco’s modified Eagle’s medium (DMEM), containing 10% fetal calf serum (FCS) and 100 U/mL penicillin-streptomycin at 37°C in a humidified atmosphere containing 5% CO2. For any experiment, cells were synchronized with forskolin (100 µM).
Surgical procedure for fiber photometry recordings
Animals previously infected either with AAV9-hSyn1-chI[1x(shNS)]-jGCaMP7b-WPRE-SV40p(A) (scramble) or AAV9-hSyn1-chI[mouse(shCdk5)]- jGCaMP7b -WPRE-SV40p(A) (shCDK5), were injected with Metacam (Meloxicam, 5mg/kg s.c.) for analgesia, then anesthetized with 1.5 –2 % isoflurane/O2 mix. Mice were placed in a Kopf digital stereotactic frame. Their body temperature was kept constant at 37 °C via a feedback-coupled heating device (Panlab/Harvard Apparatus), and their eyes were covered with ointment (Bepanthen Augen- und Nasensalbe, Bayer). After the skin incision (formerly prepared aseptically), the skull bone was cleaned with saline to remove the remaining tissue. A small craniotomy to target the SCN was drilled into the skull (Micro-Drill from Harvard Apparatus with burrs of 0.7 mm diameter from Fine Science Tools), and the dura was carefully removed. An optical fiber implant (400 μm, 0.5 NA Core Multimode Optical Fiber, FT400ERT, inserted into ceramic ferrules, 2.5 mm OD; 440 μm ID, Thorlabs) was slowly implanted above the SCN to allow for imaging of GCaMP7b signals (AP: +0.40; ML: ±0.0; DV: -5.3; angle +/- 4°). One stainless steel screw was inserted into the skull over the cerebellum for stability purposes. The implant was then secured to the skull with dental cement (Paladur, Kulzer). After surgical procedures, mice were allowed to recover for one week and finally tethered with an optical patch cord.
Fiber photometry experimental design
GCaMP7b was excited with a blue LED (Doric, LED driver, assembled with 470 nm) at 480 Hz, and emission was sampled at 2’000 Hz with a photodetector (Doric, DFD_FPA_FC) through a fluorescence MiniCube (Doric, ilFMC6_IE(400-410)_E1(460-490)_F1(500-540)_E2(555-570)_F2(580-680)_S) and digitized with a national instruments USB-6002 DAQ device. Fiber photometry recordings were acquired using custom-written scripts in LabVIEW on a computer. All the recordings were started about 15 min before ZT14 to stabilize the fluorescent signal. For every trial at ZT14 a constant light pulse of 10000 Lux (Daylight Lamp) was manually turned on for 15 min (± 20 seconds), and the recording was stopped 15 minutes after the light was switched off. To allow mice to restore their circadian time to the 12-h light–dark cycle, the intertrial interval was of minimum 10 days. In order to align the light pulse to the photometry recording, a patch cord was connected to the light source and the photometry system
In vivo calcium imaging, data processing and analysis
The data were subdivided into control (scramble) and experimental (shCDK5) groups. The fluorescent signal was demodulated in the frequency band of 470 – 490 Hz at 10 Hz acquisition rate. Due to GCaMP7b variable photobleaching (i.e., the loss of fluorescence intensity as a function of light exposure), we filtered the demodulated signal using a 3 order Savitzky-Golay filter (every 100 s), and detrended it using a hug-line. We then calculated the ΔF/F0 as follows:
Finally, the ΔF/F0 was cut to the light pulse as follow: 5 minutes before light pulse, light pulse period and 15 minutes after the light. To exclude differences in the duration of the light pulse (± 20 seconds), the period analyzed was of 14 minutes. All data processing was performed using custom-written Matlab scripts.
Live FRET imaging
The protocol was performed as before 15. The following plasmid was used for the project: ICAP-NLS Vector carrying 15. Transfected NIH3T3 cells were starved for 4 h with 0.5% FBS DMEM. Subsequently, cells were washed twice with 1×HBTS without CaCl2 and MgCl2 (25 mM HEPES, 119 mM NaCl, 6gr/L Glucose, 5 mM KCl) and resuspended in the same buffer. NIH3T3 cells were imaged using an inverted epifluorescence microscope (Leica DMI6000B) with an HCX PL Fluotar 5x/0.15 CORR dry objective, a Leica DFC360FX CCD camera (1.4 M pixels, 20 fps), and EL6000 Light Source, and equipped with fast filter wheels for FRET imaging. Excitation filters for CFP and FRET: 427 nm (BP 427/10). Emission filters for CFP: 472 (BP 472/30) and FRET: 542 nm (BP 542/27). Dichroic mirror: RCY 440/520. One frame every 20 sec was acquired for at least 90 cycles (0.05 Hz frequency), and the recording lasted at least 30 min. The baseline response in the presence of HBTS was recorded for 2 min and 40s. At minute 3:00, 100 µM Forskolin, 2 mM CaCl2, and 2 mM MgCl2 were added to the cells. The time-lapse recordings were analyzed using LAS X software (Leica). Two regions of interest (ROI) were randomly selected for each cell, and 50 cells per plate were chosen randomly. A first ROI delimiting the background and a second ROI including the cell nucleus of NIH3T3 cell expressing NLS KIDKIX were used per cell. The ROI background values were subtracted from the ROI cell values for each channel. For baseline normalization, the FRET ratio R was expressed as a ΔR/R, where ΔR is R–R0, and R0 is the average of R over the last 120 s prior stimulus.
Statistical analysis
Statistical analysis of all experiments was performed using GraphPad Prism6 software. Depending on the data type, either an unpaired t-test or one- or two-way ANOVA with Bonferroni or Tukey’s post-hoc test was performed. Values considered significantly different are highlighted. [p<0.05 (*), p<0.01 (**), or p<0.001 (***)].
Data were compared via two-way repeated-measures ANOVA with post hoc Bonferroni’s corrections for multiple comparisons. Data distribution was assumed to be normal, but this was not formally tested. All data are displayed as mean ± standard error of the mean (SEM). No power calculations were performed to determine sample sizes, but similarly sized cohorts were used in previous studies. The experimenters were not blind to the conditions when acquiring or analyzing the data.
Sample numbers are indicated in the corresponding figure legends, and test details are only reported for significant results. Figures were prepared in Adobe Illustrator 2022.
Acknowledgements
Technical assistance by Antoinette Hayoz and Maude Marmy is acknowledged. This work was supported by the Swiss National Science Foundation (SNF) 310030_219880/1 to UA, 310030_197607 to DAG, 310030_219438/1 to ZY, the Inselspital University Hospital Bern, the European Research Council CoG-725850 to AA and the States of Berne and Fribourg.
Competing interests
We declare no conflict of interest.
References
- 1.The Circadian Clock and Human HealthCurr Biol 26:R432–443
- 2.The Mammalian Circadian Timing System and the Suprachiasmatic Nucleus as Its PacemakerBiology (Basel) 8
- 3.Transcriptional architecture of the mammalian circadian clockNat Rev Genet 18:164–179
- 4.The intricate dance of post-translational modifications in the rhythm of lifeNat Struct Mol Biol 23:1053–1060
- 5.Orchestration of Circadian Timing by Macromolecular Protein AssembliesJ Mol Biol 432:3426–3448
- 6.Membrane Currents, Gene Expression, and Circadian ClocksCold Spring Harb Perspect Biol 9
- 7.Light as a central modulator of circadian rhythms, sleep and affectNat Rev Neurosci 15:443–454
- 8.Resetting the biological clock: mediation of nocturnal circadian shifts by glutamate and NOScience 266:1713–1717
- 9.Light and circadian rhythmicity regulate MAP kinase activation in the suprachiasmatic nucleiNat Neurosci 1:693–700
- 10.Regulation of CREB phosphorylation in the suprachiasmatic nucleus by light and a circadian clockScience 260:238–241
- 11.Phosphorylation of CREB Ser142 regulates light-induced phase shifts of the circadian clockNeuron 34:245–253
- 12.DARPP-32 involvement in the photic pathway of the circadian systemJ Neurosci 26:9434–9438
- 13.The Phosphorylation of CREB at Serine 133 Is a Key Event for Circadian Clock Timing and Entrainment in the Suprachiasmatic NucleusJ Biol Rhythms 33:497–514
- 14.Light induces chromatin modification in cells of the mammalian circadian clockNat Neurosci 3:1241–1247
- 15.PER2 mediates CREB-dependent light induction of the clock gene Per1Sci Rep 11
- 16.Light pulses that shift rhythms induce gene expression in the suprachiasmatic nucleusScience 248:1237–1240
- 17.A differential response of two putative mammalian circadian regulators, mper1 and mper2, to lightCell 91:1055–1064
- 18.Light-induced resetting of a mammalian circadian clock is associated with rapid induction of the mPer1 transcriptCell 91:1043–1053
- 19.A functional analysis of circadian pacemakers in nocturnal rodents II. The variability of phase response curvesJ Comp Physiol 106:253–266
- 20.MPer1 and mper2 are essential for normal resetting of the circadian clockJ Biol Rhythms 16:100–104
- 21.Phase responses to light pulses in mice lacking functional per or cry genesJ Biol Rhythms 19:518–529
- 22.Voltage-gated calcium channelsCold Spring Harb Perspect Biol 3
- 23.Voltage-gated calcium channels play crucial roles in the glutamate-induced phase shifts of the rat suprachiasmatic circadian clockEur J Neurosci 21:1215–1222
- 24.A specific role for the REV-ERBalpha-controlled L-Type Voltage-Gated Calcium Channel CaV1.2 in resetting the circadian clock in the late nightJ Biol Rhythms 29:288–298
- 25.Molecular physiology of low-voltage-activated t-type calcium channelsPhysiol Rev 83:117–161
- 26.Augmentation of Cav3.2 T-type calcium channel activity by cAMP-dependent protein kinase AJ Pharmacol Exp Ther 318:230–237
- 27.Temperature-dependent modulation of CaV3 T-type calcium channels by protein kinases C and A in mammalian cellsJ Biol Chem 282:32710–32718
- 28.Regulation of neuronal cav3.1 channels by cyclin-dependent kinase 5 (Cdk5)PLoS One 10
- 29.An isoform of the neuronal cyclin-dependent kinase 5 (Cdk5) activatorJ Biol Chem 270:26897–26903
- 30.p35 is a neural-specific regulatory subunit of cyclin-dependent kinase 5Nature 371:419–423
- 31.Cyclin I activates Cdk5 and regulates expression of Bcl-2 and Bcl-XL in postmitotic mouse cellsJ Clin Invest 119:3089–3101
- 32.Making a neuron: Cdk5 in embryonic and adult neurogenesisTrends Neurosci 32:575–582
- 33.Cdk5 regulates multiple cellular events in neural development, function and diseaseDev Growth Differ 56:335–348
- 34.Cyclin-dependent kinase 5 (CDK5) regulates the circadian clockElife 8
- 35.Physical Interaction between Cyclin-Dependent Kinase 5 (CDK5) and Clock Factors Affects the Circadian Rhythmicity in Peripheral OscillatorsClocks Sleep 4:185–201
- 36.cAMP response element induces Per1 in vivoBiochem Biophys Res Commun 531:515–521
- 37.Circadian and light-induced transcription of clock gene Per1 depends on histone acetylation and deacetylationMol Cell Biol 24:6278–6287
- 38.Bimodal regulation of mPeriod promoters by CREB-dependent signaling and CLOCK/BMAL1 activityProc Natl Acad Sci U S A 99:7728–7733
- 39.Ca2+/cAMP response element-binding protein (CREB)-dependent activation of Per1 is required for light-induced signaling in the suprachiasmatic nucleus circadian clockJ Biol Chem 278:718–723
- 40.CREB: a Ca(2+)-regulated transcription factor phosphorylated by calmodulin-dependent kinasesScience 252:1427–1430
- 41.Coupling of the RAS-MAPK pathway to gene activation by RSK2, a growth factor-regulated CREB kinaseScience 273:959–963
- 42.Cross talk between ERK and PKA is required for Ca2+ stimulation of CREB-dependent transcription and ERK nuclear translocationNeuron 21:869–883
- 43.Involvement of calcium-calmodulin protein kinase but not mitogen-activated protein kinase in light-induced phase delays and Per gene expression in the suprachiasmatic nucleus of the hamsterJ Neurochem 77:618–627
- 44.gammaCaMKII shuttles Ca(2)(+)/CaM to the nucleus to trigger CREB phosphorylation and gene expressionCell 159:281–294
- 45.Calcium/calmodulin-dependent protein kinase types II and IV differentially regulate CREB-dependent gene expressionMol Cell Biol 14:6107–6116
- 46.CDK5 inhibits the clathrin-dependent internalization of TRPV1 by phosphorylating the clathrin adaptor protein AP2mu2Sci Signal 12
- 47.Cdk5 phosphorylates dopamine D2 receptor and attenuates downstream signalingPLoS One 8
- 48.NMDA-evoked calcium transients and currents in the suprachiasmatic nucleus: gating by the circadian systemEur J Neurosci 13:1420–1428
- 49.Calcium response to retinohypothalamic tract synaptic transmission in suprachiasmatic nucleus neuronsJ Neurosci 27:11748–11757
- 50.Signaling in the suprachiasmatic nucleus: selectively responsive and integrativeCell Tissue Res 309:99–107
- 51.Phase delays to light and gastrin-releasing peptide require the protein kinase A pathwayNeurosci Lett 559:24–29
- 52.Differential cAMP gating of glutamatergic signaling regulates long-term state changes in the suprachiasmatic circadian clockJ Neurosci 20:7830–7837
- 53.Forskolin induces circadian gene expression of rPer1, rPer2 and dbp in mammalian rat-1 fibroblastsFEBS Lett 465:79–82
- 54.Imaging CREB activation in living cellsJ Biol Chem 285:23285–23295
- 55.DARPP-32: an integrator of neurotransmissionAnnu Rev Pharmacol Toxicol 44:269–296
- 56.Identification of phosphorylation sites in the recombinant catalytic subunit of cAMP-dependent protein kinaseJ Biol Chem 268:18626–18632
- 57.Physiological phosphorylation of protein kinase A at Thr-197 is by a protein kinase A kinaseMol Cell Biol 18:1416–1423
- 58.Protein kinase A regulation of T-type Ca2+ channels in rat cerebral arterial smooth muscleJ Cell Sci 126:2944–2954
- 59.Neuronal Cav3 channelopathies: recent progress and perspectivesPflugers Arch 472:831–844
- 60.Photic and circadian regulation of c-fos gene expression in the hamster suprachiasmatic nucleusNeuron 5:127–134
- 61.Dec1 and Dec2 are regulators of the mammalian molecular clockNature 419:841–844
- 62.Physiology of circadian entrainmentPhysiol Rev 90:1063–1102
- 63.Light affects behavioral despair involving the clock gene Period 1PLoS Genet 17
- 64.Light does not degrade the constitutively expressed BMAL1 protein in the mouse suprachiasmatic nucleusEur J Neurosci 18:125–133
- 65.The neuroplasticity-associated arc gene is a direct transcriptional target of early growth response (Egr) transcription factorsMol Cell Biol 25:10286–10300
- 66.Vasoactive intestinal polypeptide (VIP) phase-shifts the rat suprachiasmatic nucleus clock in vitroEur J Neurosci 13:839–843
- 67.SCN VIP Neurons Are Essential for Normal Light-Mediated Resetting of the Circadian SystemJ Neurosci 38:7986–7995
- 68.The CRTC1-SIK1 pathway regulates entrainment of the circadian clockCell 154:1100–1111
- 69.A light-induced small G-protein gem limits the circadian clock phase-shift magnitude by inhibiting voltage-dependent calcium channelsCell Rep 39
- 70.Protein Kinases in the Photic Signaling of the Mammalian Circadian ClockYale J Biol Med 92:241–250
- 71.Photic Entrainment of the Circadian SystemInt J Mol Sci 23
- 72.Liver-derived ketone bodies are necessary for food anticipationNat Commun 7
- 73.The mPer2 gene encodes a functional component of the mammalian circadian clockNature 400:169–173
- 74.Structure and regulation of the CDK5-p25(nck5a) complexMol Cell 8:657–669
- 75.Identification and characterization of a novel phosphoregulatory site on cyclin-dependent kinase 5Biochem Biophys Res Commun 504:753–758
- 76.The Drosophila TRPL ion channel shares a Rab-dependent translocation pathway with rhodopsinEur J Cell Biol 90:620–630
- 77.TRP Channels in VisionNeurobiology of TRP Channels
- 78.A Gq-Ca2+ axis controls circuit-level encoding of circadian time in the suprachiasmatic nucleusNeuron 78:714–728
- 79.Fast and sensitive GCaMP calcium indicators for imaging neural populationsNature 615:884–891
- 80.A neuronal ryanodine receptor mediates light-induced phase delays of the circadian clockNature 394:381–384
- 81.Phosphorylation of DARPP-32 by Cdk5 modulates dopamine signalling in neuronsNature 402:669–671
- 82.Disturbed clockwork resetting in Sharp-1 and Sharp-2 single and double mutant micePLoS One 3
- 83.The circadian system of c-fos deficient miceJ Comp Physiol A 178:563–570
- 84.Mice Lacking EGR1 Have Impaired Clock Gene (BMAL1) Oscillation, Locomotor Activity, and Body TemperatureJ Mol Neurosci 64:9–19
- 85.A guideline for analyzing circadian wheel-running behavior in rodents under different lighting conditionsBiol Proced Online 7:101–116
Article and author information
Author information
Version history
- Preprint posted:
- Sent for peer review:
- Reviewed Preprint version 1:
Copyright
© 2024, Brenna et al.
This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.
Metrics
- views
- 288
- downloads
- 11
- citations
- 0
Views, downloads and citations are aggregated across all versions of this paper published by eLife.