20
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      scite_
       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Blue light receptor CRY1 regulates HSFA1d nuclear localization to promote plant thermotolerance

      research-article
      Overview Bookmark

            SUMMARY

            Temperature increases as light intensity rises, but whether light signals can be directly linked to high temperature response in plants is unclear. Here, we find that light pre-treatment enables plants to survive better under high temperature, designated as light-induced thermotolerance (LIT). With short-term light treatment, plants induce light-signaling pathway genes and heat shock genes. Blue light photoreceptor cryptochrome 1 (CRY1) is required for LIT. We also find that CRY1 physically interacts with the heat shock transcription factor A1d (HsfA1d) and that HsfA1d is involved in thermotolerance under light treatment. Furthermore, CRY1 promotes HsfA1d nuclear localization through importin alpha 1 (IMPα1). Consistent with this, CRY1 shares more than half of the chromatin binding sites with HsfA1d. Mutation of CRY1 (cry1–304) diminishes a large number of HsfA1d binding sites that are shared with CRY1. We present a model where, by coupling light sensing to high-temperature stress, CRY1 confers thermotolerance in plants via HsfA1d.

            In brief

            Light pre-treatment endows plants with light-induced thermotolerance, enhancing their ability to survive in high temperatures. We show that blue light triggers CRY1 to activate HSFs in the nucleus. CRY1 and HsfA1d share common binding sites and regulate downstream gene expression, promoting survival of plants at elevated temperatures.

            Graphical Abstract

            Main article text

            INTRODUCTION

            Plants employ the heat shock response (HSR) as a means to acclimate to heat stress caused by elevated temperatures14 in response to heat shock. Plant cells elevate expression of molecular chaperones to ensure protein stability, correct folding, triage, and degradation, thereby restoring protein homeostasis (proteostasis).58 The pivotal controllers of chaperones are heat shock transcription factors (HSFs).912 In the presence of high-temperature stimuli, HSFs amass in the nucleus and initiate the upregulation of chaperones and other genes associated with the HSR.1315 Additionally, it has been observed that excessive light or high light intensity triggers the translocation of HSF gene family member HsfA1d into the nucleus, promoting an increase in heat shock protein (HSP) gene expression in Arabidopsis.16,17

            On Earth, fluctuations in sunlight intensity correspond closely with shifts in temperature. Over time, plants have evolved mechanisms to interlink light and temperature sensing, given that exposure to light is recognized to augment resistance to heat.18 Genetic investigations have indeed linked plant survival at elevated temperatures to chloroplast signaling.19 Analyses of diurnal gene expression have unveiled the regulation of chaperone protein genes by light/dark cycles in Arabidopsis thaliana.19,20 Moreover, light has been shown to be involved in the amplification of the activity of HSRs and unfolded protein responses in the endoplasmic reticulum, both of which are integral to the HSR.16,21,22 It has been documented that the red light receptor phytochrome B (phyB) collaborates with chloroplasts as a temperature sensor, harmonizing light and temperature signals. As the temperature rises, phyB activity is repressed, leading to a reduction in phyB nuclear body size. Furthermore, genes regulated by phyB are modulated in response to temperature changes.23,24 Additionally, the blue-light receptor CRY1 interacts with phytochrome-interacting factor 4 (PIF4) to suppress auxin biosynthesis at elevated temperatures, thereby curtailing hypocotyl elongation in a process termed thermomorphogenesis.25 Collectively, these findings underscore the close intertwining of light and heat signaling, with light receptors serving as pivotal connectors between these two pathways.

            However, it is noteworthy that the bulk of these studies have been conducted under conditions of moderate heat stress rather than in the face of severe or potentially lethal heat levels. Consequently, whether light plays a role in promoting adaptation to exceptionally high temperatures (i.e., thermotolerance) remains an enigma. Moreover, the precise light receptor governing plant thermotolerance and the mechanisms by which it exerts its influence remain elusive. In this study, we delve into the role of the blue-light receptor CRY1 in the induction of light-induced thermotolerance in plants. Through its capacity to interact with HsfA1d, CRY1 facilitates the nuclear import of HsfA1d, orchestrated by nuclear importin alpha 1 (IMPα1), thereby modulating the binding of HsfA1d to chromatin under light conditions. Our investigation dissects the intricate mechanisms that unite light signaling with the HSR.

            RESULTS

            Light-induced thermotolerance in plants

            Prior research has elucidated that the thermotolerance of plants under light conditions is regulated by chloroplast signaling.19 To discern the specific influence of chloroplasts and ascertain whether the light-signaling pathway independently triggers heat tolerance, we conducted an experiment using three sets of Arabidopsis plants. These plants were initially grown in darkness at 22°C for a duration of 4 days. Subsequently, one set remained in darkness at 22°C, another was subjected to white light exposure (50 μmol m−2 S−1) for 1 h at 22°C, and the third set was exposed to 34°C for 30 min in the dark (Figure S1A). Following this, all plants were exposed to a lethal temperature of 45°C for 2 h, followed by a recovery phase at 22°C under white light for 7 days. Intriguingly, our observations revealed that pre-exposure to light facilitated thermotolerance akin to pre-treatment at 34°C. In stark contrast, plants that were continuously grown in darkness succumbed to heat stress (Figure 1A). This enhancement of thermotolerance subsequent to light treatment was also evident in wheat and canola (Figure S1). We termed this phenomenon light-induced thermotolerance (LIT). We further sought to investigate whether the LIT phenomenon remains valid for plants cultivated under light conditions. To address this, we cultivated two sets of Arabidopsis plants in low-intensity light (50 μmol m−2 S−1) for 4 days. Subsequently, the light intensity was augmented to 75 μmol m−2 S−1 for a duration of 1 h for one set, while the other set was maintained at the initial light intensity prior to subjecting them to daytime heat shock. Our findings demonstrated that elevating light intensity for 1 h heightens the heat resistance of Arabidopsis plants (Figure S1D).

            Additionally, we sought to determine whether the LIT phenomenon is influenced by different photoperiods. We cultivated Arabidopsis seedlings under varying photoperiod conditions (day/night = 16 h/8 h, 12 h/12 h, or 8 h/16 h) for 4 days. Following this, the seedlings were pre-treated with 1 h of light exposure at midnight, followed by a heat shock at 45°C for 1 h. After a recovery period of 4 days, our results unveiled that longer daily light durations correlate with heightened thermotolerance in Arabidopsis seedlings. Notably, pretreatment with light exposure at midnight conferred a significant level of thermotolerance across all examined photoperiods (Figure S2A). Furthermore, extended daily light durations even augmented the thermotolerance of seedlings during the daytime (Figure S2B). These findings collectively suggest that a longer circadian rhythm day length contributes positively to the thermotolerance of Arabidopsis seedlings. Moreover, the LIT phenomenon demonstrated its applicability to seedlings cultivated under diverse photoperiod conditions.

            Light-induced expression pattern resembles heat shock genes

            In the realm of plants, the activation of the HSR in general and the specific transcriptional upregulation of HSP101 (encoding a disaggregate chaperone) have paramount significance for survival during instances of heat stress.26,27 Our investigation confirmed the increase in HSP101 mRNA expression following light exposure and heat treatment through semi-quantitative PCR (semi-qPCR) and qPCR analyses (Figure 1B).

            To ascertain the extent of transcriptional response commonality between light treatment and heat stress, we conducted mRNA deep sequencing (mRNA-seq) on dark-grown etiolated Col-0 seedlings subjected to either 1 h of white light exposure or a temperature of 34°C for 30 min. The transcriptome-wide analysis revealed substantial concordance between gene expression in response to light and heat (Figures 1C1E; Table S1). Remarkably, more than half of the genes induced by light exposure can also be induced by heat stress, culminating in a total of 582 genes commonly triggered by both stimuli (Figures 1C and 1D). The Gene Ontology (GO) terms were enriched, and the top 10 of each sector were listed (Figures 1E and S3). The results demonstrated that the 1,090 genes induced by light exposure were enriched in functions related to chloroplasts, photosystems, HSRs, light responses, and translation elongation factor activity (Figure S3A). The 2,518 genes induced by heat stress were primarily enriched in processes such as protein folding, chloroplast and photosystem structure and function, water transport, and translation elongation (Figure S3B). Notably, the 582 genes that responded to both light and heat exhibited enrichment in categories including protein folding, light response, translation elongation factor activity, hydrogen peroxide transmembrane transport, chlorophyll binding, and photosystem reaction (Figure 1E).

            Conversely, treatments of 34°C heat and light pre-exposure resulted in the decreased expression of 2,081 and 909 genes, respectively. Interestingly, among these downregulated genes, 286 were shared between the two conditions (Table S1). GO analysis highlighted that these co-downregulated genes were implicated in light intensity response, lipid and hormone responses, transcription and gene expression, cellular components of lipid droplets, nuclei, and plastids (Figures S3C and S3D; Table S1). Notably, among the common genes induced by light and heat, more than 22 were annotated as HSPs or HSP-like genes, underscoring the shared capacity of light and heat to induce the HSR. These findings unveil a mutually reinforcing relationship between exposure to light and heat stress.

            Blue light initiates HSR through CRY1

            Plants exhibit distinct responses to different wavelengths within the white light spectrum. To explore the feasibility of inducing LIT through specific spectral ranges, we pre-conditioned Col-0 seedlings with light spanning far-red-, red-, or blue-spectrum light before exposing them to 45°C. Notably, seedlings subjected to pre-exposure with far-red or red light failed to manifest thermotolerance, whereas those pre-exposed to blue light exhibited enhanced survival rates akin to those pre-exposed to white light (Figure 2A). Significantly, only blue light pre-exposure led to the activation of HSP101 gene expression (Figure 2C).

            Among light receptors, Cryptochrome 1 and 2 (CRY1 and CRY2) are attuned to blue light, while phytochrome A (phyA) and phyB are responsive to red and far-red light, respectively.2831 In an endeavor to pinpoint the specific light receptor responsible for thermotolerance, we evaluated canonical heat-induced thermotolerance and LIT in null mutant lines (phyA-211, phyB-9, cry1–304, and cry2–1). The survival rates of all tested lines, including Col-0 and the mutant lines, were enhanced upon pre-heat treatment at 34°C before exposure to severe heat stress (Figure S4). Notably, LIT was evident in all mutant lines except for cry1–304, which exhibited a marked impairment in acquiring LIT, resembling the survival rate of dark-grown Col-0 (negative control), where less than 10% of seedlings survived (Figure 2B). While cry2–1 demonstrated a deficit in LIT, the extent of its sensitivity was notably lower compared with the cry1 mutant (Figure 2B). Corresponding with the phenotypic findings, cry1–304 displayed diminished induction of HSP101 in response to white light (Figure 2D).

            Moreover, the impairment of LIT was further substantiated through the assessment of another cry1 null mutant, cry1–349, whose phenotype aligned with the lack of LIT induction upon blue light exposure. Intriguingly, in cry1–349, it was the pre-heat treatment at 34°C rather than light treatment that triggered seedling thermotolerance (Figure S5). These findings collectively underscore the pivotal role of the blue light receptor CRY1 in initiating LIT.

            CRY1 interaction with HsfA1d

            Employing the yeast two-hybrid system, we identified that HsfA1d-NT (1–220 amino acids [aa]) interacts with the full-length CRY1, its N-terminal segment (1–490 aa), and the C-terminal fragment (491–681 aa) of CRY1. This suggests that HsfA1d exhibits the capability to interact with at least two distinct regions of CRY1 (Figure 3A). We corroborated the interaction between full-length CRY1 and HsfA1d through a split-luciferase (split-LUC) assay within the Nicotiana benthamiana transient expression system (Figure 3B). Further substantiating this interaction, we introduced a 35S:: HsfA1d-FLAG expression cassette into Arabidopsis (HsfA1d-FLAG/Col-0) and observed a marked enhancement in Arabidopsis survival rates following 45°C treatment when HsfA1d was overexpressed (Figure S6A). Through immunoprecipitation (IP) utilizing an anti-FLAG antibody against the FLAG-tagged protein in the HsfA1d-FLAG/Col-0 line, we confirmed the interaction between CRY1 and HsfA1d (Figure 3C). Moreover, the interaction between CRY1 and HsfA1d in the nucleus was established through bimolecular fluorescence complementation (BiFC) (Figure 3D). Collectively, these findings underscore the stable interaction between CRY1 and HsfA1d in Arabidopsis. Notably, HsfA1d belongs to the HsfA1 subfamily, which serves as a crucial master transcriptional regulator of the HSR in Arabidopsis,1,32 making it a compelling candidate for mediating LIT.

            HsfA1d mediates LIT

            The HSR is evolutionarily conserved among eukaryotes, and HSFs stand as the principal transcriptional regulators within this response. Arabidopsis possesses 21 HSF homologs, while yeast only contains a solitary HSF gene.9,10 Among these, the HsfA1 subfamily encompasses HsfA1a, HsfA1b, HsfA1d, and HsfA1e and emerges as the primary regulator of the induced HSR in Arabidopsis. Intriguingly, our investigation illuminated that pre-treatment with light failed to elevate the survival rate of the HsfA1 quadruple mutant (hsfa1sqk) when subjected to 45°C exposure. Moreover, expression of the marker gene HSP101 exhibited poor induction in response to either light or heat exposure (Figures 4A and 4B). Remarkably, the reintroduction of HsfA1d into hsfa1sqk (hsfa1a/b/e mutant) effectively reinstated light- and heat-induced thermotolerance, concurrently re-establishing the expression of HSP101 (Figures 4A and 4B). Consequently, the HsfA1 subfamily is deemed essential for LIT, with HsfA1d emerging as the pivotal mediator sufficient to trigger this phenomenon.

            Joint regulation of heat shock gene expression by HsfA1d and CRY1 in response to light

            To comprehensively understand the combined influence of CRY1 and HsfA1d on the transcriptional responses triggered by light and heat, we conducted RNA-seq analysis on dark-grown Col-0, cry1–304, and hsfa1sqk seedlings following exposure to white light for 1 h or 34°C heat for 30 min (Table S1). In the hsfa1sqk mutant, among the 154 annotated light-responsive genes (Figure 4C, denoted in blue), 12 genes (8%) exhibited differential expression (fold change > 2 or < 0.5, adjusted p < 0.05) relative to Col-0 in response to light and heat shock. Strikingly, for the 186 annotated heat-responsive genes (Figure 4C, highlighted in red), 52 genes (28%) displayed differential expression in the hsfa1sqk mutant compared with Col-0 in response to light and heat shock. In the case of the cry1–304 mutant, only 3 heat-responsive genes demonstrated reduced expression (fold change ≤ 0.5) compared with Col-0 under heat stress, while 63 genes exhibited reduced expression in response to light. Similarly, under heat stress, only 2 light-responsive genes exhibited reduced expression in cry1–304 relative to Col-0, in contrast to 32 genes exhibiting reduced expression in response to light (Figure 4C). These observations suggest that HsfA1 transcription factors are instrumental in triggering the transcriptional response to heat shock under light and heat conditions. However, CRY1’s involvement is specific to the light-induced HSR.

            Subsequently, to delineate the transcriptional signature associated with LIT, we identified the set of genes whose induction is dependent on the HsfA1 family and CRY1. In the hsfa1sqk strain, 74 genes exhibited a reduction in expression of >2-fold relative to Col-0 in response to heat, and 70 genes exhibited a similar reduction in response to light. Impressively, 54 of these genes showed reduced expression in response to light and heat stimuli (Figure 4D; Table S1). Among these 54 genes, 42 genes demonstrated a reduction of more than 2-fold in cry1–304 relative to Col-0 (Figures 4D and 4E). This final set of 42 genes predominantly encompassed HSP genes and HSF genes (Figure 4E). We designate these 42 genes as the set of genes co-regulated by CRY1 and the HsfA1 family in response to light, making them potential contributors to LIT.

            Shared target genes of CRY1 and HsfA1d

            Given the nuclear interaction between CRY1 and HsfA1d, coupled with the transcription factor role of HsfA1d, it is plausible that the CRY1-HsfA1d complex binds to chromatin, thereby influencing gene expression. To explore their potential co-occupancy on chromatin sites, we conducted chromatin IP sequencing (ChIP-seq) analysis on 5-day-old HsfA1d-FLAG/Col-0 and GFP-CRY133 plants, cultivated under a 16 h/8 h light/dark regimen (Table S2). The ChIP-seq data analysis unveiled that HsfA1d-FLAG and GFP-CRY1 were bound to 4,917 and 2,327 chromosomal regions, respectively, with a notable overlap of 1,483 sites (Figure 5A). Among the 42 genes exhibiting reduced expression under light and heat stimuli in the hsfa1sqk and cry1–304 mutants relative to Col-0, we identified 35 and 15 genes as targets of HsfA1d and CRY1, respectively, with a remarkable 12 genes being common to both (Table S1). This observation indicates that genes associated with the HSR are potentially regulated directly by HsfA1d and CRY1. Intriguingly, the co-occupying genes extend beyond core heat stress response genes like HSFB1 and HSFB2b. Among them RAV2, a gene linked to drought, salt, and oxidative stress, which are hallmarks of heat stress, also stands out. RAV2’s involvement in photo-protection against abiotic stress further suggests a possible connection between biotic stress and light signaling.34

            Considering the divergence in binding maps between HsfA1d-FLAG and GFP-CRY1 lines as deduced from the ChIP-seq data, we postulate that the transcriptional co-occupancy of genes may be altered between the cry1–304 and hsfa1sqk mutants. Of note, we observed the presence of bromodomain-containing 4 (BRD4), a gene belonging to the bromodomain and extraterminal family, among the co-occupying genes of HsfA1d and CRY1. Notably, BRD4 plays a role in regulating transcription.35 Further analysis revealed changed binding patterns of HSFB1, HSFB2b, RAV2, and BRD4 by CRY1 and HsfA1d based on the ChIP-seq data (Figure 5C). This observation was substantiated by ChIP-qPCR experiments, which confirmed significantly altered associations of CRY1 and HsfA1d with these four target genes between the HsfA1d-FLAG and GFP-CRY1 lines (Figure 5B). Taken together, these findings underscore the physical interaction of CRY1 and HsfA1d with common gene sequences, including those pivotal for thermotolerance.

            CRY1 facilitates nuclear localization of HsfA1d under blue light

            Having observed the nuclear interaction between HsfA1d and CRY1 in A. thaliana16 (Figure 3D), we proceeded to investigate whether the nuclear localization of both proteins is influenced by light exposure. We initially examined the nuclear localization of CRY1 in response to light using 35S::GFP-CRY1 (GFP-CRY1) transgenic Arabidopsis seedlings33 (Figure S6B). Employing nuclear and cytoplasmic protein fractionation followed by western blotting, we observed increased nuclear accumulation of GFP-CRY1 after exposure to light (Figures 6A, left, and S7A). To delve further, we conducted a fluorescence-based analysis of nuclear accumulation of GFP-CRY1 through fluorescence microscopy and bleach recover after blue light treatment and dark (DK) conditions. The study revealed that blue light not only enhanced the nuclear accumulation of GFP-CRY1 in terms of quantity but also expedited the recovery rate after photobleaching (Figures S8A and S8B). Specifically, under DK conditions, the fluorescence intensity (FI) of nuclear GFP-CRY1 was only restored to approximately 66% of its initial value after bleaching, whereas with blue light exposure, FI restoration reached around 90%. Furthermore, confocal microscopy demonstrated a notable increase in global GFP FI in the hypocotyls of Arabidopsis seedlings subjected to light, underscoring the overall elevation in CRY1 accumulation (Figure S8C). In essence, blue light was found to augment the accumulation and nuclearization of CRY1 in Arabidopsis.

            Conversely, we also noted that heat stress at 34°C triggered the nuclearization of HsfA1d (Figure S7D). To explore whether light has a similar effect on the nuclear localization of HsfA1d, we tracked localization of HsfA1d in HsfA1d-FLAG/Col-0 lines (Figure S6C). Employing fractionation and Western blot analysis, we demonstrated that light exposure also enhanced the nuclear accumulation of HsfA1d (Figures 6A, right, and S7B). Consequently, light exposure exhibited the ability to bolster nuclear localization of CRY1 and HsfA1d.

            In the context of Arabidopsis, nuclear localization of the far-red receptor phyA is reliant on the interaction between far-red elongated hypocotyl 1 (FHY1) and IMPα1.36 Intriguingly, our investigations unveiled interactions between CRY1 and HsfA1d, respectively, with IMPα1 in the yeast two-hybrid system (Figure 6B), and these interactions were further corroborated in the split-LUC assay (Figure 6C). Notably, the presence of CRY1 was found to enhance the interaction between HsfA1d and IMPα1 (Figure 6D). Importantly, the effects of blue light on the interaction of CRY1-HsfA1d and CRY1-IMPα1 were examined through the split-LUC assay, revealing significant intensification of the interaction in response to blue light treatment (Figure S10). These findings led us to speculate that, in the presence of blue light, CRY1 may facilitate nuclear localization of HsfA1d through a physical interaction, potentially aided by IMPα1.

            To test this hypothesis, we conducted a cross between HsfA1d-FLAG/Col-0 and the cry1–304 mutant, yielding HsfA1d-FLAG/cry1–304 (Figure S6D). Subsequent analysis involving fractionation of nuclear and cytoplasmic proteins, followed by western blotting, revealed that light-induced nuclear localization of HsfA1d was enhanced in HsfA1d-FLAG/Col-0 but remained unaltered in HsfA1d-FLAG/cry1–304 (Figures 6E and S7C). Hence, our results conclusively demonstrate that CRY1 actively promotes light-dependent nuclear localization of HsfA1d.

            Decreased binding loci and affinity of HsfA1d in cry1 mutants

            To delve deeper into the impact of CRY1 on chromatin binding of HsfA1d, we conducted ChIP-seq of HsfA1d in HsfA1d-FLAG/cry1–304 mutants under the same growth conditions as previously detailed (Table S2). The result unveiled a stark reduction in the number of HsfA1d binding sites in the cry1–304 background (721) compared with the Col-0 background (4,917), with 502 sites overlapping between the two (Figures 5A and 7A). Of significance, there were 190 overlapping binding sites between GFP-CRY1 and HsfA1d-FLAG in HsfA1d-FLAG/cry1–304 (GFP-CRY1 vs. HsfA1d-FLAG/cry1–304), exhibiting a notable decrease in contrast to the 1,483 overlapping sites in HsfA1d-FLAG/Col-0 (Figures 5A and 7B). These findings strongly suggest that, in cry1–304 mutants, about the majority (approximately 89%; Table S2) of shared binding loci between CRY1 and HsfA1d are lost.

            Further assessment of CRY1’s impact on HsfA1d′s chromatin affinity involved counting binding peaks and generating a heatmap of HsfA1d binding to transcription start sites (TSSs) in HsfA1d-FLAG/Col-0 and HsfA1d-FLAG/cry1–304 mutant backgrounds. The heatmap clearly portrays diminished HsfA1d binding signals in the HsfA1d-FLAG/cry1–304 background, characterized by a dramatic reduction in binding sites at TSSs (Figures 7C and 7D). Moreover, IGV plots (Itegrative Genomics Viewer) illustrated that the binding peak of HsfA1d-FLAG targets, particularly BRD4, in the cry1–304 background were significantly diminished, as validated by ChIP-qPCR (Figures 7E and 7F). qRT-PCR results supported this observation, showing that, in terms of control and overexpression levels of HsfA1d, the transcription levels of HSFB1, HSFB2b, and BRD4 in the cry1–304 background were markedly decreased in comparison with the Col-0 background (Figures S9A and S9B). This indicates that the absence of CRY1 might exert an influence on the binding site and binding affinity.

            The complex formed by CRY1 and HsfA1d governed the expression of a specific gene set, including heat response genes, which contribute to a plant’s survival under high temperature stress (Figures 4C4E). In line with this, the overexpression of GFP-CRY1 in Col-0 led to an enhanced survival rate after exposure to harsh temperature treatment (Figure 7G), and the introduction of MYC-CRY1 complemented the heat tolerance phenotype observed in the cry1–304 line (Figure S9C).

            DISCUSSION

            This study uncovers a novel interplay between light signaling and the HSR, shedding light on the direct correlation between light exposure and the HSR. While previous studies have shown that high light/high irradiation triggers stress responses separately,16,17,37,38 transcriptome-wide analysis disclosed a significant overlap between light-induced genes and heat-induced genes, particularly in terms of functional categories such as the light-harvesting complex and HSPs (Figures 1C1E and S3), indicating coordinated signaling that enables plants to pre-adapt to rising temperatures via LIT.

            The investigation demonstrated that LIT is prompted by blue light, specifically through the action of the blue light receptor CRY1, rather than red or far-red light (Figure 2), unlike the PIF-dependent activities previously linked to CRY1, which revolve around photomorphogenesis at elevated temperatures, not thermotolerance.25,39 CRY1 physically interacts with HsfA1d in the nucleus, and plants that lose HsfA1s failed to trigger LIT (Figure 3D). It is speculated that the interaction between CRY1 and HsfA1d in the nucleus enables light-dependent nuclear localization of HsfA1d to initiate the light-dependent HSR and promotes LIT. Co-occupancy of these two factors at promoters implies their involvement in transcriptional regulation. CRY1 and HsfA1d share many common genomic binding sites, including heat stress genes such as HsfB1, HsfB2b, and HSPs (Figure 5). Importantly, CRY1 is responsible for the light-induced heat stress response but not high-temperature-induced heat stress. Nevertheless, HsfA1d is involved in light- and heat-induced heat-stress responses (Figure 4A). It indicates that CRY1 perceives light signals and transduces them to downstream genes through its interaction with HsfA1d. Thus, both CRY1and HsfA1d are required for LIT.

            HsfA1s, including HsfA1a, HsfA1b, and HsfA1e, make up a major transcriptional family involved in acquired heat tolerance.40 This family triggers a transcriptional cascade composed of multiple transcription factors under high-temperature conditions, which is important for the activation of the HSR transcriptional regulatory network. While HsfA1d is pivotal for LIT, the roles of other members of the HsfA1 subfamily (HsfA1a, HsfA1b, and HsfA1e) remain to be explored because they often exhibit redundant functions. Indeed, we verified the interaction of CRY1 and HsfA1a/b/e (Figures S11AS11C). As we know, there are two cryptochromes (CRY1 and CRY2) responsible for regulating various blue light responses in Arabidopsis.41 Moreover, considering the similarity in sequence and function between CRY1 and CRY2, it is plausible that CRY2 could also contribute to LIT, which was verified by a heat resistance test of the cry2–1 mutant line (Figure 2B), although it is less sensitive regarding heat tolerance than cry1–304, which may result from the variation of their roles in blue light signaling. Further, like CRY1, CRY2 also physically interacts with HsfA1d and IMPα1 (Figure S11D and S11E). The mechanism behind CRY2-mediated heat tolerance and the biological significance of the interaction between CRY2 and HsfA1d or IMPα1 deserves further exploration.

            CRY1 and HsfA1d respond to light exposure by translocating into the nucleus, allowing them to transmit the light signal into the nucleus for the rewiring of the heat stress response (Figures 6A, S7A, and S7B). We found that IMPα1, which regulated far-red light receptor phyA nuclear translocation, also interacted with CRY1 and HsfA1d (Figures 6B and 6C). Moreover, the co-expressed CRY1 enhanced the interaction between HsfA1d and IMPα1 (Figure 6D). This may result in nuclear transport of HsfA1d by CRY1 when exposed to light. Consistent with this, HsfA1d failed to accumulate in the nucleus when exposed to light in cry1 mutations (Figures 6E and S7C). Notably, cry1 mutations reduced the number of occupied sites and the binding affinity of HsfA1d, and most of the lost sites previously bound by HsfA1d were co-occupied by CRY1 (Figures 7A7D). Paradoxically, although the binding of HsfA1d to HsfB1 and HsfB2b remained unchanged, the expression of these two genes was decreased. This implies that CRY1 might also play a role in transcriptional activation in addition to affecting the nuclear localization of HsfA1d, possibly because of its transcription activation domain. Despite these complex interactions, overexpression of CRY1 enhanced the survival rate of DK-grown seedlings under high-temperature stress after light pre-treatment, while cry1 mutations reduced that (Figures 2B and 7G). The intricate interplay between CRY1, HsfA1d, and their complex potentially alters promoter binding and gene expression regulation to modulate plant thermotolerance. This study proposes a model in which blue-spectrum light triggers CRY1 to activate HSFs, inducing chaperone genes, and the increased chaperone expression promotes survival at elevated temperatures (Figures 7H and 7I).

            The heat-shock response is activated with input from the CRY1 system when high-energy light is present. Similarly, the red/far-red light sensor phyB, which is also a thermosensor in plants,42,43 responds to lower-energy light and primes the HSR.44 Additionally, phyB is responsible for sensing the high proportion of far-red light in autumn to induce cold-responsive genes to prepare for winter frosts.45 Conversely, we propose that CRY1 is responsible for sensing the increasing blue light intensity during spring and summer to increase thermotolerance. On shorter timescales, these systems may operate on diurnal heating and cooling cycles to exert circadian control over proteostasis. From a practical standpoint, the crosstalk between CRY1 and HsfA1d endows plants with stress tolerance and could be a useful tool for modifying crop plants to adapt to high-temperature stress under global climate warming.

            Limitations of the study

            Our study identifies a molecular pathway underlying the phenomenon of blue LIT in plants. Light exposure enhances the translocation of CRY1 to the nucleus and facilitates nuclear accumulation of HsfA1d through physical interaction, which requires the cooperation of IMPα1, but it is not clear how the CRY1-HsfA1d interaction affects the HsfA1d-IMPα1 module; it is possible that additional factors are involved in CRY1-HsfA1d nuclear import. In the nucleus, CRY1 and HsfA1d co-occupy genomic binding sites and activate transcription of heat-related genes to confer LIT. However, HsfA1d’s binding on some genes, like HsfB1 and HsfB2b, could still be observed in cry1–304 while their transcription level was reduced. This suggests that CRY1 may regulate transcription of HsfB1 and HsfB2b independent of HsfA1d or that other transcription factors are involved in regulating the expression of these genes. Furthermore, we found that CRY1-HsfA1d physically interact with each other, but the mechanism of how light affects the association of CRY1-HsfA1d is lacking. More regulators associated with the CRY1-HsfA1d complex need to be identified to fully elucidate the underlying mechanisms. Finally, in this study we analyzed plant seedlings grown in controlled conditions with DK-to-light transfer, which is not a natural condition of plant growth. Additional regulation models need to be developed for natural day-light cycle growth conditions.

            STAR★METHODS

            RESOURCE AVAILABILITY
            Lead contact

            Further information and requests for reagents and source data should be directed to and will be fulfilled by the lead contact, Xu Zheng (zhengxu@123456henau.edu.cn).

            Materials availability

            All plant materials and plasmids generated in this paper will be shared by the lead contact upon request. This study did not generate new unique reagents.

            Data and code availability

            RNA-seq and ChIP-seq data generated in this study have been deposited in the Sequence Read Archive (SAR) of the National Center for Biotechnology Information (SAR: https://www.ncbi.nlm.nih.gov/sra/PRJNA842487), and are publicly available as of the date of publication.

            This paper does not report original code.

            All plant materials and plasmids generated in this paper will be shared by the lead contact upon request.

            EXPERIMENTAL MODEL AND SUBJECT DETAILS

            For cultivation of Triticun aestiun (Chinese spring), Brassica napus (Zhongshuang 11), and Arabidopsis seeds were surface sterilized for 15 min with 1% sodium hypochlorite. After 3 days germination, seedlings were transferred to soil and kept growing in the dark or 16/8 h light/dark regime (white light, WL) (50 μmol m−2S−1) under 22°C for 4 days, followed by treatment, and recovery at 22°C under WL for 4 days (Figure S1A).

            All strains were in Col-0 background. phyA-211,46,47 phyB-9,47,48 cry1–304,49,50 cry1–349,51 and cry2–150,52 were null mutants to the corresponding genes. All light receptor mutant seeds were kindly provided by Dr Chentao Lin. HSFA1a/b/day/e quadruple mutation (hsfa1sqk) and HsfA1a/b/e were kindly provided by Dr Hsiang-chin Liu.1 Transgenic line over expressing GFP-CRY1 and MYC-CRY1 (MYC-CRY1/cry1–304) were kindly provided by Chentao Lin33 and Hongquan Yang,53 respectively.

            The full length CDS of HsfA1d (AT1G32330, 1458bp) was amplified with gene specific primers (Table S3), and cDNAs reverse-transcribed from mRNA of Arabidopsis (Col-0) as template. The PCR product was cloned into the site XhoI-XbaI of binary vector pEarlyGate-100. The HsfA1d gene was under the control of the cauliflower mosaic virus 35S promoter. The construct was verified by sequencing and transformed into Agrobacterium tumefaciens strain GV3101 for following transformation of Arabidopsis. Arabidopsis plants were transformed with flower dip method described by Clough and Bent.54

            METHOD DETAILS
            RT-PCR

            RNA was extracted from plants by TransZol regent (Cat No. ET101, TransGen Biotech, Beijing, China). For RT-PCR, the mRNA was converted to cDNA by PrimeScript RT reagent Kit with gDNA Eraser (Perfect Real Time) (Taraka, Code No: RR047Q). Quantitative RT-PCR analyses for expression of target genes were performed according to the manual of the SYBR supermix (BIO-RAD). Three replicates were performed for each sample, with ACTIN2 as the reference gene. The qPCR was performed on QuantStudio 6. The semi-quantitative RT-PCR was performed using the same primers for HSP101, using 18S rRNA as a reference. Primers were listed in Table S3.

            Nuclear Fractionation

            Nuclear fractionation was performed as described previously.55 Briefly, 1.5g fresh seedlings was homogenized in 3 mL Honda buffer (HB: 2.5% Ficoll 400, 5% dextran T40, 0.4 M Sucrose, 25 mM Tris-HCl, pH 7.5, 10 mM MgCl2, 1 mM DTT, 1 mM PMSF, and 1×complete protease inhibitor mixture) and filtered through a 62 μm pore nylon mesh, then added Triton X-100 (with final concentration of 0.5%) followed by incubating on ice for 15 min and centrifuged (1500g for 5 min). The supernatant was centrifuged (12000 g, 10 min) and saved as nuclei free fraction and the pellet was gently resuspended in 1 mL HB (with 0.1% Triton X-100). Centrifuged the mixture (1500 g, 5 min) and resuspended the pellet in 1 mL HB and centrifuged once more (100 g for 1 min). Then, the supernatant was transferred to a new 1.5 mL eppendorf tube and centrifuged (2000 g, 5 min). The resulting pellet was resuspended in 300 μL buffer G (GB: 1.7 M Sucrose, 10mM Tris-HCl, pH 8.0, 0.15% Triton X-100, 2 mM MgCl2, 5 mM DTT, and 1 ×protease inhibitor mixture) and was added on the top of 300 μL GB in a new 1.5 mL eppendorf tube. Finally, the preparation of the sample was centrifuged (16,000 g, 60 min) and the resulting pellet was resuspended in 100 μL HB with 100 μL SDS loading buffer. The prepared samples were blotted with anti-Flag (Sigma, F1804) and Anti-CRY1 (kindly provided by Dr Chentao Lin) antibody for HsfA1d-Flag and GFP-CRY1 with Histone H3 and Actin (ACT) as loading control. Anti-Histone H3 (Phyto AB, PHY033S) and anti-ACTIN2 (Ambart, 26F7) antibodies were used to detect nuclear and cytosolic markers, respectively.

            Yeast Two-Hybrid Analysis

            The different combinations of bait plasmid BD-HsfA1d (1–220aa), BD-IMPα1, and prey plasmids AD-CRY1NT (1–490aa), AD-AD-CRY1CT (491–681aa) and AD-CRY1 were co-transformed into yeast strain Y2H. The empty GAL4 BD bait vector (pGBKT7) and GAL4 AD prey vector (pGADT7) combination was used as a negative control. For the construction of bait and prey plasmid, the coding sequences of HsfA1d, CRY1, CRY1 (1–490aa), CRY1 (491–681aa), IMPα1, were amplified from Arabidopsis cDNA. The deletion series of HsfA1d was generated from HsfA1d by inverse PCR. Yeast two-hybrid analysis was performed using the Matchmaker Y2H system (Clontech, Mountain View, CA, USA).

            RNA sequencing

            Total RNA was extracted using Trizol reagent following manufacturer’s instructions (Invitrogen, CA, USA). Sequencing libraries were generated from the purified mRNA using the VAHTS Universal V6 RNA-seq Library Kit for MGI (Vazyme, Nanjing, China) following the manufacturer’s recommendations with unique index codes. Sequencing was performed on MGI-SEQ 2000 platform by Frasergen Bioinformatics Co., Ltd. (Wuhan, China).

            Low quality reads, such as reads with adaptor sequences, reads with >5% N, or >20% bases with quality <Q20 (percentage of sequences with sequencing error rates <1%), were removed using perl script. The clean reads were mapped to the Arabidopsis genome (Tair 10) using HISAT2.56

            Differentially expression genes between sample groups was evaluated by DESeq2.57 The false discovery rate (FDR) was used to identify the threshold of P-value in multiple tests in order to compute the significance of the differences. Here, only genes with |log2(−FoldChange)| ≥1 and FDR significance score (padj) < 0.01 were used for subsequent analysis.

            Chromatin immunoprecipitation and data analysis

            ChIP was performed on 5-day old seedlings grown in the 16h/8h light/dark regime, then were transplanted or not transplanted to 34°C for 30min. Cross-linking was performed with 40 mL of 1% formaldehyde in a vacuum for 30 min followed by quenching with 125 mM glycine for 10 min. After rinsing seedlings with water, nuclei isolation buffer was added to grind the treated sample on ice. We filtered out the slag with a 200-purpose nylon mesh, and finally squeezed the filtrate into 5 mL EP tubes by hand (one glove for each sample) and place on ice for 15 min. After centrifugation at 2000 × g 4°C for 5 min, we discarded the supernatant. Tissues were ground in 500μL ChIP buffer (50 mM HEPES pH 7.5, 140 mM NaCl, 1 mM EDTA, 1% Triton x–100, 0.1% DOC) and sonicated on ice 10 times using a probe sonicator (75W 25% Duty Factor 12000s). 1 mL was transferred to a 1.5 mL tube and spun to remove tissues debris. Input was set aside, and a serial IP was performed. First, 25 μL of anti-FLAG magnetic beads (50% slurry, Sigma) were added the mixture was incubated for overnight at 4°C on a rotator. Beads were separated with a magnet and the supernatant was removed. Beads were washed twice with 1 mL ChIP buffer (5 min incubations at 4°C between each wash), washed again with LiCl buffer (1 mL), and finally wash once with TE buffer (1mL). Beads were separated with a magnet and eluate was transferred to a fresh tube. Bound material was eluted with 100 μL TE +1% SDS by incubating at 65°C for 2h. Beads were separated with a magnet and eluate was transferred to a fresh tube and Add 10 μl 5M NaCl solution to the liquid incubated 3–4 h at 65°C to reverse crosslinks. Protein was degraded by adding 5μL 20 mg/mL proteinase K in 243 μL TE (supplemented 2 μL GlycoBlue) and incubating at 37°C for 2h. DNA fragments were separated from protein by adding 500 μL phenol/chloroform/isoamyl alcohol (25:24:1), and the aqueous layer was added to a fresh tube. 55 μL of 4M LiCl was added along with 1 mL of 100% EtOH, and DNA was precipitated at −20°C overnight. DNA was pelleted by spinning for 30 min at 4°C and resuspended in 50 μL TE.

            The PE library with an approximately 300 bp insert size was constructed according to the Illumina library preparation protocol (Il-lumina Inc., San Diego, CA, USA). The library quantification and size were measured using Qubit 3.0 Fluorometer (Life Technologies, Carlsbad, CA, USA) and Bioanalyzer 2100system (Agilent Technologies, CA, USA). Subsequently, sequencing was performed on the Illumina NovaSeq6000 platform.

            In ChIP-Seq analysis pipeline. The interference information, containing adapter sequence, low-quality bases and undetected bases (indicated by N) were filtered using Trimmomatic58 (v0.38, with parameters: LEADING:3, TRAILING:3, SLIDINGWINDOW:4:15, MINLEN:8) and quality was assessed using FastQC59 in paired-end raw data. After obtaining clean data through quality control, the Bowtie2 were used to map clean reads to the reference genome60 (v2.3.5, with parameters: dovetail, no-unal, very sensitive local, no mixed, no discordant), and the low-quality mapping was further screen out. PCR redundancy and organelle alignment by Samtools61 (v1.9, with default parameters). The retained valid pairs were used for subsequent analysis.

            MACS262 (v2.1.1.20160309) software was used to perform Peak Calling (with parameters: -f BAMPE -B –SPMR –keep-dup all). After that, the R scripts with Deeptools software63 were performed to illustrate the signal enrichment analysis of peak regions, gene body regions and the regions from transcription start site (TSS) with default parameters. ChIPseeker64 was used to obtain gene annotations on peak regions at the same time.

            ChIP-qPCR assay

            ChIP assays were performed as described above in chromatin immunoprecipitation part. Then, we performed real-time PCR to examine the relative enrichment of certain DNA fragments over the wild type (Col-0) by Roche LC480 with primers listed in Table S3 The 2−ΔΔCT values were calculated to analyze the relative gene expression.65

            Co-Immunoprecipitation and western blotting

            For in vivo Co-IP assays from plants, the samples were lysed in 600 μL plant IP buffer (50 mM Tris, pH 7.5, 150 mM NaCl, 1 mM EDTA, 10 mM NaF, 25 mM b-glycerophosphate, 2 mM sodium orthovanadate, 10% glycerol, 0.1% Tween 20, 1 mM DTT) and 1×Complete Protease Inhibitor Cocktail (Roche). Then 500 μL of extract was incubated with anti-FLAG beads (SIGMA) for 4 h at 4°C. The beads were washed 4 times with 500 μL lysis buffer. The eluate was subjected to western blotting using anti-CRY1 and anti-FLAG antibodies.

            The nuclear fraction, or the IP beads were boiled in SDS protein loading buffer at 90°C for 10 min 15 μL of each sample was loaded into SDS-PAGE gels. The gels were run at 30 mA for 1.5 h, and blotted to PVDF membrane at 225 mA for 1.5 h. After 1 h blocking in Li-Cor blocking buffer, the membrane was incubated with anti-FLAG primary antibody (SIGMA) for 1 h, anti-CRY1 (kindly provided as a gift by Chentao Lin). The fluorescent signal scanned with the Li-Cor/Odyssey system.

            Luciferase reporter system

            Tim-nLUC (nLUC) and Tim-cLUC (cLUC) vector system were used for the split-LUC assay. The coding sequences of CRY1, IMPα1, HsfA1a/b/e were amplified from Arabidopsis cDNA and inserted into the site Kpn I-Sal I of Tim-nLUC or Tim-nLUC. These vectors, including nLUC and cLUC, were introduced into Agrobacterium tumefaciens strain GV3101 separately. The different combinations of Agrobacterium tumefaciens strains with nLUC or cLUC were infiltrated into leaves of N. benthamiana. For the combination of IMPα1-nLUC and HsfA1d-cLUC, a GV3101 strain harboring pCAMBIA2300:CaMV35S::GFP (2300-GFP) or pCAMBIA2300:CaMV35-S::CRY1 (2300-CRY1) was co-infiltrated to elucidate effects of CRY1 to nuclear localization of HsfA1d.

            Agroinfiltration of N. benthamiana was performed following the method of Shi et al.66 After 2 days of infiltration, the infiltrated leaves were sprayed with 1 mM D-luciferin (Sigma, 115144–35-9), and kept in the dark for 5 min. The luminescence signal was visualized and recorded using Night SHADE LB 985 system (Berthold, Germany) with a 30 s exposure time, 4 × 4 binning, slow readout, and high gain. Each combination of constructs was repeated for three times with highly similar results.

            Bimolecular fluorescence complementation assay

            For interaction analysis of CRY1 and HsfA1d, a bimolecular fluorescence complementation (BiFC) experiment was executed in leaf mesophyll protoplast of Arabidopsis. HsfA1d, CRY1 and HY5 genes were amplified from Arabidopsis cDNA and HsfA1d and CRY1 were inserted into the site Xho I-Hind III of PNYFP and PCYFP, respectively, to get PNYFP-CaMV35S::CRY1-nYFP (CRY1-nYFP) and PCYFP-CaMV35S::HsfA1d-cYFP (HsfA1d-cYFP). HY5 was cloned to the site Xba I-BamH I of pUC-CaMV35S:mCherry to get pUC-CaMV35S::HY5-mCherry (HY5-mCherry). HY5-mCherry was co-introduced into Arabidopsis protoplast with CRY1-nYFP and HsfA1d-cYFP as a nuclear maker. Preparation and transfection of Arabidopsis protoplast please refer to Wu. et al.67 Fluorescence signal were captured with Zeiss LSM710 confocal microscope. Wavelengths for excitation and emission were 488 nm and 525 nm for YFP, and 580 nm and 610 nm for mCherry.

            QUANTIFICATION AND STATISTICAL ANALYSIS

            GraphPad Prism 8 was used for all statistical analyses. Three independent parallel experiments were performed with similar results. Values were Means ± SD. p < 0.05 was considered to be statistically significant. Unless otherwise specified, the student t-test was used to determine the significance of the difference. All the Arabidopsis lines used in this study were Col-0 background, except as otherwise indicated.

            Supplementary Material

            1
            2
            3
            4

            ACKNOWLEDGMENTS

            We thank Chentao Lin for the light receptor mutant seeds and the anti-CRY1 antibody and Hsiang-chin Liu for Arabidopsis heat shock factor mutant seeds. We are grateful to the Jing-Ke Weng and Mary Gering labs for providing and maintaining the greenhouse and X. Wang, B. Christ, and Valentina Carballo for helping us set up plant gene transformation. We thank O. Corradin, S. Rouskin, S. Lourido, and H. Lodish and their lab members for valuable discussions and Z. Feder, M. Truttmann, Daowen Wang, and Hongquan Yang for helpful comments and suggestions and critical reading of the manuscript. We are grateful to Thomas Volkert and the Whitehead Genome Technology Core and N. Azubuine and T. Yangchen for a constant supply of plates and media. This work was supported by the National Nature Science Foundation of China (no. 30900840 to X.Z.), the Opening Foundation of Co-construction State Key Laboratory of Wheat and Maize Crop Science (no. SKL2022KF02 to J.G.), the Startup Grant of Henan Agricultural University (no. 30601733 to X.Z.), and the NIH Early Independence Award of the USA (DP5 OD017941 to D.P.).

            Footnotes

            DECLARATION OF INTERESTS

            The authors declare no competing interests.

            SUPPLEMENTAL INFORMATION

            Supplemental information can be found online at https://doi.org/10.1016/j.celrep.2023.113117.

            REFERENCES

            1. LiuHC, and CharngYY (2013). Common and distinct functions of Arabidopsis class A1 and A2 heat shock factors in diverse abiotic stress responses and development. Plant Physiol. 163, 276–290. [Cross Ref] .[PubMed]

            2. LindquistS (1986). The heat-shock response. Annu. Rev. Biochem. 55, 1151–1191.[PubMed]

            3. RichterK, HaslbeckM, and BuchnerJ (2010). The heat shock response: life on the verge of death. Mol. Cell 40, 253–266. [Cross Ref] .[PubMed]

            4. ZhuJ-K (2016). Abiotic stress signaling and responses in plants. Cell 167, 313–324. [Cross Ref] .[PubMed]

            5. DobsonCM (2003). Protein folding and misfolding. Nature 426, 884–890.[PubMed]

            6. LabbadiaJ, and MorimotoRI (2015). The biology of proteostasis in aging and disease. Annu. Rev. Biochem. 84, 435–464.[PubMed]

            7. JacobP, HirtH, and BendahmaneA (2017). The heat-shock protein/chaperone network and multiple stress resistance. Plant Biotechnol. J. 15, 405–414.[PubMed]

            8. TiwariLD, KhungarL, and GroverA (2020). AtHsc70–1 negatively regulates the basal heat tolerance in Arabidopsis thaliana through affecting the activity of HsfAs and Hsp101. Plant J. 103, 2069–2083. [Cross Ref] .[PubMed]

            9. NoverL, BhartiK, DöringP, MishraSK, GanguliA, and ScharfKD (2001). Arabidopsis and the heat stress transcription factor world: how many heat stress transcription factors do we need? Cell Stress Chaperones 6, 177–189. [Cross Ref] .[PubMed]

            10. AnckarJ, and SistonenL (2011). Regulation of HSF1 function in the heat stress response: implications in aging and disease. Annu. Rev. Biochem. 80, 1089–1115.[PubMed]

            11. ScharfK-D, BerberichT, EbersbergerI, and NoverL (2012). The plant heat stress transcription factor (Hsf) family: structure, function and evolution. Biochim. Biophys. Acta 1819, 104–119.[PubMed]

            12. KastEJ, and KaplinskyN (2022). HSFA1d regulates the kinetics of heat-induced HSP17.6 expression in Arabidopsis. MicroPubl. Biol. 2022. [Cross Ref].

            13. ScharfKD, BerberichT, EbersbergerI, and NoverL (2012). The plant heat stress transcription factor (Hsf) family: structure, function and evolution. Biochim. Biophys. Acta 1819, 104–119. [Cross Ref] .[PubMed]

            14. MorimotoRI, KlineMP, BimstonDN, and CottoJJ (1997). The heat-shock response: regulation and function of heat-shock proteins and molecular chaperones. Essays Biochem. 32, 17–29.[PubMed]

            15. GarbuzDG (2017). Regulation of heat shock gene expression in response to stress. Mol. Biol. 51, 352–367. [Cross Ref].

            16. JungH-S, CrispPA, EstavilloGM, ColeB, HongF, MocklerTC, PogsonBJ, and ChoryJ (2013). Subset of heat-shock transcription factors required for the early response of Arabidopsis to excess light. Proc. Natl. Acad. Sci. USA 110, 14474–14479.[PubMed]

            17. NishizawaA, YabutaY, YoshidaE, MarutaT, YoshimuraK, and ShigeokaS (2006). Arabidopsis heat shock transcription factor A2 as a key regulator in response to several types of environmental stress. Plant J. 48, 535–547.[PubMed]

            18. LaudeHH (1939). Diurnal cycle of heat resistance in plants. Science 89, 556–557.

            19. DickinsonPJ, KumarM, MartinhoC, YooSJ, LanH, ArtavanisG, CharoensawanV, SchöttlerMA, BockR, JaegerKE, and WiggePA (2018). Chloroplast signaling gates thermotolerance in Arabidopsis. Cell Rep. 22, 1657–1665.[PubMed]

            20. DashS, Van HemertJ, HongL, WiseRP, and DickersonJA (2012). PLEXdb: gene expression resources for plants and plant pathogens. Nucleic Acids Res. 40, D1194–D1201.[PubMed]

            21. LuDP, and ChristopherDA (2008). Light enhances the unfolded protein response as measured by BiP2 gene expression and the secretory GFP-2SC marker in Arabidopsis. Physiol. Plantarum 134, 360–368.

            22. NawkarGM, KangCH, MaibamP, ParkJH, JungYJ, ChaeHB, ChiYH, JungIJ, KimWY, YunD-J, and LeeSY (2017). HY5, a positive regulator of light signaling, negatively controls the unfolded protein response in Arabidopsis. Proc. Natl. Acad. Sci. USA 114, 2084–2089.[PubMed]

            23. LegrisM, KloseC, BurgieES, RojasCCR, NemeM, HiltbrunnerA, WiggePA, SchäferE, VierstraRD, and CasalJJ (2016). Phytochrome B integrates light and temperature signals in Arabidopsis. Science 354, 897–900. [Cross Ref] .[PubMed]

            24. JungJH, DomijanM, KloseC, BiswasS, EzerD, GaoM, KhattakAK, BoxMS, CharoensawanV, CortijoS, (2016). Phytochromes function as thermosensors in Arabidopsis. Science 354, 886–889. [Cross Ref] .[PubMed]

            25. MaD, LiX, GuoY, ChuJ, FangS, YanC, NoelJP, and LiuH (2016). Cryptochrome 1 interacts with PIF4 to regulate high temperature-mediated hypocotyl elongation in response to blue light. Proc. Natl. Acad. Sci. USA 113, 224–229. [Cross Ref] .[PubMed]

            26. MerretR, CarpentierMC, FavoryJJ, PicartC, DescombinJ, Bousquet-AntonelliC, TillardP, LejayL, DeragonJM, and CharngYY (2017). Heat Shock Protein HSP101 Affects the Release of Ribosomal Protein mRNAs for Recovery after Heat Shock. Plant Physiol. 174, 1216–1225. [Cross Ref] .[PubMed]

            27. McLoughlinF, KimM, MarshallRS, VierstraRD, and VierlingE (2019). HSP101 Interacts with the Proteasome and Promotes the Clearance of Ubiquitylated Protein Aggregates. Plant Physiol. 180, 1829–1847. [Cross Ref] .[PubMed]

            28. NagataniA, ReedJW, and ChoryJ (1993). Isolation and initial characterization of Arabidopsis mutants that are deficient in phytochrome A. Plant Physiol. 102, 269–277.[PubMed]

            29. ReedJW, NagataniA, ElichTD, FaganM, and ChoryJ (1994). Phytochrome A and phytochrome B have overlapping but distinct functions in Arabidopsis development. Plant Physiol. 104, 1139–1149.[PubMed]

            30. MocklerTC, GuoH, YangH, DuongH, and LinC (1999). Antagonistic actions of Arabidopsis cryptochromes and phytochrome B in the regulation of floral induction. Development 126, 2073–2082.[PubMed]

            31. GuoH, YangH, MocklerTC, and LinC (1998). Regulation of flowering time by Arabidopsis photoreceptors. Science 279, 1360–1363. [Cross Ref] .[PubMed]

            32. Nishizawa-YokoiA, NosakaR, HayashiH, TainakaH, MarutaT, TamoiM, IkedaM, Ohme-TakagiM, YoshimuraK, YabutaY, and ShigeokaS (2011). HsfA1d and HsfA1e involved in the transcriptional regulation of HsfA2 function as key regulators for the Hsf signaling network in response to environmental stress. Plant Cell Physiol. 52, 933–945.[PubMed]

            33. GaoJ, WangX, ZhangM, BianM, DengW, ZuoZ, YangZ, ZhongD, and LinC (2015). Trp triad-dependent rapid photoreduction is not required for the function of Arabidopsis CRY1. Proc. Natl. Acad. Sci. USA 112, 9135–9140. [Cross Ref] .[PubMed]

            34. FuM, KangHK, SonSH, KimSK, and NamKH (2014). A subset of Arabidopsis RAV transcription factors modulates drought and salt stress responses independent of ABA. Plant Cell Physiol. 55, 1892–1904. [Cross Ref] .[PubMed]

            35. DevaiahBN, LewisBA, ChermanN, HewittMC, AlbrechtBK, RobeyPG, OzatoK, SimsRJ3rd, and SingerDS (2012). BRD4 is an atypical kinase that phosphorylates serine2 of the RNA polymerase II carboxy-terminal domain. Proc. Natl. Acad. Sci. USA 109, 6927–6932. [Cross Ref] .[PubMed]

            36. HelizonH, Rösler-DaltonJ, GaschP, von HorstenS, EssenLO, and ZeidlerM (2018). Arabidopsis phytochrome A nuclear translocation is mediated by a far-red elongated hypocotyl 1-importin complex. Plant J. 96, 1255–1268. [Cross Ref] .[PubMed]

            37. KleineT, KindgrenP, BenedictC, HendricksonL, and StrandÅ (2007). Genome-wide gene expression analysis reveals a critical role for CRYPTOCHROME1 in the response of Arabidopsis to high irradiance. Plant Physiol. 144, 1391–1406.[PubMed]

            38. van RooijenR, HarbinsonJ, and AartsMGM (2018). Photosynthetic response to increased irradiance correlates to variation in transcriptional response of lipid-remodeling and heat-shock genes. Plant direct 2, e00069.[PubMed]

            39. PedmaleUV, HuangS.-s.C, ZanderM., ColeBJ., HetzelJ., LjungK., ReisPAB., SrideviP., NitoK., NeryJR., (2016). Cryptochromes interact directly with PIFs to control plant growth in limiting blue light. Cell 164, 233–245.[PubMed]

            40. LIUHC, LIAOHT, and CHARNGYY (2011). The role of class A1 heat shock factors (HSFA1s) in response to heat and other stresses in Arabidopsis. Plant Cell Environ. 34, 738–751.[PubMed]

            41. BaeG, and ChoiG (2008). Decoding of light signals by plant phytochromes and their interacting proteins. Annu. Rev. Plant Biol. 59, 281–311. [Cross Ref] .[PubMed]

            42. LegrisM, KloseC, BurgieES, RojasCCR, NemeM, HiltbrunnerA, WiggePA, SchäferE, VierstraRD, and CasalJJ (2016). Phytochrome B integrates light and temperature signals in Arabidopsis. Science 354, 897–900.[PubMed]

            43. JungJ-H, DomijanM, KloseC, BiswasS, EzerD, GaoM, KhattakAK, BoxMS, CharoensawanV, CortijoS, (2016). Phytochromes function as thermosensors in Arabidopsis. Science 354, 886–889.[PubMed]

            44. KarayekovE, SellaroR, LegrisM, YanovskyMJ, and CasalJJ (2013). Heat shock–induced fluctuations in clock and light signaling enhance phytochrome B–mediated arabidopsis deetiolation. Plant Cell 25, 2892–2906.[PubMed]

            45. FranklinKA, and WhitelamGC (2007). Light-quality regulation of freezing tolerance in Arabidopsis thaliana. Nat. Genet. 39, 1410–1413.[PubMed]

            46. RattanapisitK, ChoMH, and BhooSH (2016). Lysine 206 in Arabidopsis phytochrome A is the major site for ubiquitin-dependent protein degradation. J. Biochem. 159, 161–169. [Cross Ref] .[PubMed]

            47. SuL, HouP, SongM, ZhengX, GuoL, XiaoY, YanL, LiW, and YangJ (2015). Synergistic and Antagonistic Action of Phytochrome (Phy) A and PhyB during Seedling De-Etiolation in Arabidopsis thaliana. Int. J. Mol. Sci. 16, 12199–12212. [Cross Ref] .[PubMed]

            48. CassonSA, and HetheringtonAM (2014). phytochrome B Is required for light-mediated systemic control of stomatal development. Curr. Biol. 24, 1216–1221. [Cross Ref] .[PubMed]

            49. YangY, LiY, LiX, GuoX, XiaoX, TangD, and LiuX (2008). Comparative proteomics analysis of light responses in cryptochrome1–304 and Columbia wild-type 4 of Arabidopsis thaliana. Acta Biochim. Biophys. Sin. 40, 27–37. [Cross Ref] .[PubMed]

            50. StoelzleS, KagawaT, WadaM, HedrichR, and DietrichP (2003). Blue light activates calcium-permeable channels in Arabidopsis mesophyll cells via the phototropin signaling pathway. Proc. Natl. Acad. Sci. USA 100, 1456–1461. [Cross Ref] .[PubMed]

            51. ShalitinD, YuX, MaymonM, MocklerT, and LinC (2003). Blue light–dependent in vivo and in vitro phosphorylation of Arabidopsis cryptochrome 1. Plant Cell 15, 2421–2429.[PubMed]

            52. NakagawaM, and KomedaY (2004). Flowering of Arabidopsis cop1 mutants in darkness. Plant Cell Physiol. 45, 398–406. [Cross Ref] .[PubMed]

            53. SangY, LiQH, RubioV, ZhangYC, MaoJ, DengXW, and YangHQ (2005). N-terminal domain-mediated homodimerization is required for photoreceptor activity of Arabidopsis CRYPTOCHROME 1. Plant Cell 17, 1569–1584. [Cross Ref] .[PubMed]

            54. CloughSJ, and BentAF (1998). Floral dip: a simplified method for Agrobacterium-mediated transformation of Arabidopsis thaliana. Plant J. 16, 735–743.[PubMed]

            55. ZhengX, WuS, ZhaiH, ZhouP, SongM, SuL, XiY, LiZ, CaiY, MengF, (2013). Arabidopsis phytochrome B promotes SPA1 nuclear accumulation to repress photomorphogenesis under far-red light. Plant Cell 25, 115–133.[PubMed]

            56. KimD, PaggiJM, ParkC, BennettC, and SalzbergSL (2019). Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat. Biotechnol. 37, 907–915.[PubMed]

            57. LoveMI, HuberW, and AndersS (2014). Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550.[PubMed]

            58. BolgerAM, LohseM, and UsadelB (2014). Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120.[PubMed]

            59. AndrewsS (2010). FastQC: a quality control tool for high throughput sequence data. Babraham Bioinformatics (Babraham Institute).

            60. LangmeadB, and SalzbergSL (2012). Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359.[PubMed]

            61. LiH, HandsakerB, WysokerA, FennellT, RuanJ, HomerN, MarthG, AbecasisG, and DurbinR; 1000 Genome Project Data Processing Subgroup (2009). The sequence alignment/map format and SAMtools. Bioinformatics 25, 2078–2079.

            62. FengJ, LiuT, QinB, ZhangY, and LiuXS (2012). Identifying ChIP-seq enrichment using MACS. Nat. Protoc. 7, 1728–1740.[PubMed]

            63. RamírezF, RyanDP, GrüningB, BhardwajV, KilpertF, RichterAS, HeyneS, DündarF, and MankeT (2016). deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160–W165.[PubMed]

            64. YuG, WangL-G, and HeQ-Y (2015). ChIPseeker: an R/Bioconductor package for ChIP peak annotation, comparison and visualization. Bioinformatics 31, 2382–2383.[PubMed]

            65. LivakKJ, and SchmittgenTD (2001). Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25, 402–408. [Cross Ref] .[PubMed]

            66. ShiY, LeeLY, and GelvinSB (2014). Is VIP1 important for Agrobacterium-mediated transformation? Plant J. 79, 848–860.[PubMed]

            67. WuF-H, ShenS-C, LeeL-Y, LeeS-H, ChanM-T, and LinC-S (2009). Tape-Arabidopsis Sandwich - a simpler Arabidopsis protoplast isolation method. Plant Methods 5, 16. [Cross Ref] .[PubMed]

            Floating objects

            Figure 1.
            Light promotes thermotolerance and induces the HSR in A. thaliana

            (A) Survival rate of A. thaliana under lethal heat shock pre-treated by moderate heat shock or white light. A. thaliana (Col-0) seedlings were grown in the DK for 4 days and then transferred to white light (50 μmol m−2 S−1) for 1 h or heat shocked at 34°C for 30 min or kept in the DK prior to 2 h of exposure to 45°C. The survival rate of seedlings was quantified after 4 days of recovery (22°C, 16 h white light/8 h DK) (Figure S1A). The histogram at the bottom shows the rate of survival after the corresponding treatment. Means ± SD, n ≥ 50; n represents number of Arabidopsis plants. *p < 0.05, **p < 0.01 vs. DK.

            (B) HSP101 gene expression level was monitored over a time course of light or heat exposure. Samples were harvested 6, 12, 15, and 30 min after 34°C heat shock or after 15, 30, and 60 min of white light exposure. Gene expression was checked by semi-quantitative RT-PCR normalized with 18S rRNA expression, and qRT-PCR was normalized by ACTIN2.

            (C and D) RNA-seq of poly(A) + mRNA was performed on DK-grown plants with or without heat shock for 30 min at 34°C or white light exposure for 1 h. Levels of gene transcription were quantified as the |log2(fold change)| ≥1 and a false discovery rate (FDR) significance score p-adjusted value (padj) of less than 0.05 of the light- and heat-treated samples compared with the DK-grown control. The Venn diagram (C) and heatmap display the differently expressed genes (padj < 0.05) by light and heat shock at 34°C (D).

            (E) analysis of significantly expressed genes induced by light and heat shock at 34°C.

            Figure 2.
            Blue light-induced thermotolerance of Arabidopsis through CRY1

            (A) Survival rate of A. thaliana under lethal heat shock pre-treated with different colors of light. Col-0 seedlings were grown in the DK for 4 days and transferred to far-red (2.5 μmol m−2 S−1), red (30 μmol m−2 S−1), and blue (20 μmol m−2 S−1) light for 1 h before treatment at 45°C for 2 h. Following 4 days of recovery, plants were photographed, and survival statistics were quantified. Three biological replicates were implemented for statistics. Means ± SD, n ≥ 50; n represents number of Arabidopsis plants. *p < 0.05, **p < 0.01 vs. far red.

            (B) Survival rate of the wild-type (WT) and different light receptor mutants of A. thaliana under lethal heat shock. Col-0 and light receptor mutants (phyA-211, phyB-9, cry1–304, and cry2–1) seedlings were grown in the DK for 4 days and transferred to white light for 1 h before 2 h at 45°C. Following 4 days of recovery, plants were photographed, and the survival rate was quantified. Three biological replicates were implemented for statistics. Means ± SD, n ≥ 40; n represents number of Arabidopsis plants. *p < 0.05, **p < 0.01 vs. Col-0.

            (C) HSP101 expression level in Col-0 seedlings following exposure to far-red, red, and blue light for 1 h. qRT-PCR was performed as in Figure 1B. Means ± SD, n = 3 (replicates). *p < 0.05, **p < 0.01 vs. DK.

            (D). HSP101 expression level in Col-0 and light receptor mutants in the DK and following 1 h of white light exposure. Gene expression was quantified by qRT-PCR normalized by ACTIN2. Significant differences in log2 fold change of HSP101 expression between Col-0 and light receptor mutants were analyzed. Means ± SD, n = 3 (replicates). *p < 0.05, **p < 0.01 vs. Col-0.

            Figure 3.
            CRY1 physically interacts with HsfA1d

            (A) Yeast two hybrid one-on-one interaction assay. This assay showed interactions between pGADT7 (AD), AD-CRY1, AD-CRY1NT (1–490 amino acids [aa]), AD-CRY1CT (491–681 aa), and pGBKT7 (BD) (top) or BD-HsfA1d (1–220 aa) (bottom).

            (B) Split-LUC assay of HsfA1d-cLUC (carbonyl terminal of luciferase) and CRY1-nLUC (amino terminal of luciferase) in N. benthamiana.

            (C) Co-immunoprecipitation (coIP) between HsfA1d-FLAG and CRY1 in Arabidopsis. Protein was isolated from HsfA1d-FLAG/Col-0 and Col-0 seedlings. IP was performed with beads conjugated with a FLAG tag, and the eluate was detected with an anti-FLAG antibody and CRY1 antibody.

            (D) Bimolecular fluorescence complementation (BiFC) assay of HsfA1d-cYFP (carbonyl terminal of yellow fluorescent protein) and CRY1-nYFP (amino terminal of yellow fluorescent protein) in the nucleus of A. thaliana leaf epidermic protoplasts. Constructs encoding HsfA1d-cYFP and CRY1-nYFP (top) or nYFP (bottom) were used to transfect protoplasts. A construct encoding HY5-mCherry was co-transformed to mark the nucleus. Transformed protoplasts were visualized by confocal microscope. Scale bars, 10 μm.

            Figure 4.
            HsfA1 and CRY1 genes are required for activating the heat shock transcriptional response upon light exposure

            (A) Pre-exposure to white light promoted survival of Arabidopsis seedlings in lethal heat shock through HsfA1d. Col-0, hsfa1a/b/d/e quadruple mutants (hsfa1sqk), and hsfa1a/b/e triple mutants (hsfa1sqk/A1d) were cultured in DK for 4 days, followed by 1-h white light exposure and then 2-h heat shock at 45°C. After 4 days of recovery, plants were photographed.

            (B) qRT-PCR analysis of HSP101 expression levels. Expression of HSP101 in Col-0, hsfa1sqk, and hsfa1sqk/A1d was determined by qRT-PCR. All of these materials were cultured in DK room and then randomly divided into three groups: DK, kept in DK; light, 1-h white light exposure; HS (heat shock), 30 min at 34°C. Expression levels were normalized by ACTIN2. Means ± SD, n = 3 (replicates). *p < 0.05, **p < 0.01 vs. the same treatment in Col-0.

            (C) Correlation of differentially expressed genes (DEGs) between mild heat shock (34°C) and light-induced genes and RNA-seq of poly(A) + mRNA of DK-grown Col-0, hsfa1sqk, and cry1–304 plants and those treated by 30-min heat shock (34°C) or 1-h white light. Gene expression changes were quantified as a fold change greater than 2 or less than 0.5 (adjusted p < 0.05) with respect to Col-0. Red dots, 34°C treatment-induced DEGs analyzed by Gene Ontology (GO); blue dots, light exposure-induced DEGs analyzed by GO.

            (D) Venn diagram showing the number of DEGs with >2-fold reduced expression in each category of (C).

            (E) The fold change of the 42 genes with reduced expression in cry1–304 and hsfa1sqk mutants with respect to Col-0 under heat treatment and light exposure.

            Figure 5.
            HsfA1d and CRY1 co-occupy chromatin regions

            (A) Venn diagram showing the number of shared and distinct bound chromatin sites by HsfA1d-FLAG (blue) and GFP-CRY1 (green) in the Col-0 background of A. thaliana growing in the 16 h/8 h light/DK regimen.

            (B) ChIP-qPCR analysis verified that HsfA1d-FLAG (blue) and GFP-CRY1 (green) associate with HsfB1, HsfB2b, RAV2, and BRD4 genes. Col-0 plants were used as a negative control. Amplification of HsfB1, HsfB2b, RAV2, and BRD4 genes was normalized by ACTIN2 with amplification of TA3 as a negative control. Means ± SD, n = 3 (replicates). *p < 0.05, **p < 0.01 vs. WT.

            (C) HsfA1d and CRY1 ChIP-seq displayed binding peaks on HsfB1, HsfB2b, RAV2, and BRD4 genes. The input controls are shown in black.

            Figure 6.
            CRY1 promotes the nuclear localization of HsfA1d through physical interaction with IMPα1

            (A) Light-induced nuclear accumulation of GFP-CRY1 and HsfA1d-FLAG. HsfA1d-FLAG/Col-0 and GFP-CRY1 seedlings were cultured in DK for 4 days and then exposed to white light for 0 min, 30 min, or 60 min. DK- and light-treated seedlings were sampled, and nuclear fractions were isolated and blotted with anti-FLAG and anti-GFP antibodies. Nuclear fractions were verified by histone H3 but not ACTIN2, while cytoplasm fractions were verified by ACTIN2 but not histone H3. Relative signal intensity of GFP-CRY1 and HsfA1d-FLAG was measured by ImageJ, and relative values of treated samples to corresponding untreated controls were calculated. Three independent parallel experiments were performed (for the other two replicates, see Figure S7).

            (B) Yeast two-hybrid assay showing that HsfA1d and CRY1 interacted with IMPα1.

            (C) CRY1and HsfA1d interact with IMPα1, as seen by split-LUC assay in N. benthamiana.

            (D) CRY1 promoted the nuclear translocation of HsfA1d, indicated by enhanced LUC signal in the presence of overexpressed CRY1 in the split-LUC assay (top). LUC signals of different combinations of constructs were quantitated (bottom). Means ± SD, n = 3 (replicates). *p < 0.05, **p < 0.01 vs. IMPα1-nLUC + HsfA1d-cLUC + 2,300-GFP.

            (E) The null mutant of the cry1–304 gene in A. thaliana attenuated nuclear translocation of HsfA1d. HsfA1d-FLAG/Col-0 and HsfA1d-FLAG/cry1–304 were cultured, light treated, and blotted as in (A). Three independent parallel experiments were performed (for the other two replicates, see Figure S7).

            Figure 7.
            CRY1 increased the quantity of chromatin regions co-occupied by HsfA1d and enhanced the heat resistance of A. thaliana

            (A and B) Venn diagram showing the number of shared and distinct binding sites of HsfA1d-FLAG in the Col-0 (blue) and cry1–304 (green) background (A) and those of GFP-CRY1 (blue) and HsfA1d-FLAG/cry1–304 (green) (B). All seedlings were grown in the 16 h/8 h light/DK regimen for 5 days and sampled for ChIP-seq.

            (C and D) ChIP-seq analysis of HsfA1d-FLAG-occupied transcription start sites (TSSs) in the Col-0 and cry1–304 backgrounds. The profiles (C) and heatmap (D) indicate that loss of function of CRY1 attenuated occupation of HsfA1d on TSSs.

            (E) ChIP-seq peaks of HsfA1d-FLAG on genes (HsfB1, HsfB2b, and BRD4) in the Col-0 (blue) and cry1–304 (green) backgrounds. Function loss of CRY1 affected HsfA1d occupation of the HsfB1 and BRD4 genes. The input controls are shown in black.

            (F) ChIP-qPCR analysis of HsfA1d-FLAG-occupied genes (HsfB1, HsfB2b, and BRD4) in the cry1–304 background. ChIP-qPCR was performed and analyzed as in Figure 5C. Means ± SD, n = 3. *p < 0.05, **p < 0.01 vs. WT.

            (G) Overexpression of GFP-CRY1 enhanced the thermotolerance of A. thaliana. Col-0 and GFP-CRY1 seedlings were grown in the 16 h/8 h light/DK regimen for 4 days, treated with 45°C for 2 h, and then photographed after 4 days of recovery. Means ± SD, n = 3 (replicates). *p < 0.05, **p < 0.01 vs. Col-0.

            (H) Cartoon model of light-induced transcriptional activation by CRY1 and HsfA1 transcription factors. HsfA1 is shown as a red trimer and CRY1 as a cyan oval. In the nucleus, the red, purple, and cyan genes were under control of HsfA1 only, HsfA1 and CRY1, or CRY1 only, respectively.

            (I) Model for upstream signaling that supported light-induced thermotolerance (LIT).

            KEY RESOURCES TABLE
            REAGENT or RESOURCESOURCEIDENTIFIER

            Antibodies

            anti-FlagSigmaCat# F1804; RRID:AB_262044
            anti-GFPAbwaysCat# AB0008; RRID:AB_2943672
            anti-CRY1Gifted by Chentao Lin, Fujian Agriculture and Forestry UniversityN/A
            anti-Histone H3Phyto ABCat# PHY033A; RRID:AB_2943673
            anti-ActinAbmartCat# M20009H; RRID:AB_2936239

            Bacterial and virus strains

            agrobacterium GV3101 strainProvided by Jianping Yang, Henan Agricultural UniversityN/A
            yeast Y2H strainWeidi, 2ND LabCat#YC1002

            Chemicals, peptides, and recombinant proteins

            TransZolTransGen BiotechCat#ET101
            PrimeScript RT reagent KitTarakaCat#RR047Q
            SYBR supermixBIO-RADCat#1708880B10
            cOmplete Protease Inhibitor CocktailRocheCat#05892970001
            HEPESSigmaCat#H3375
            IPTGInvitrogenCat#15529019
            PBSSolarbioCat#P1020
            SDSSolarbioCat#S8010
            GlucoseSolarbioCat#G8150
            TrisSolarbioCat#T8060
            GlycineSolarbioCat#G8200
            YPD agarSigmaCat#1500
            Beetle Luciferin, Potassium SaltPromegaCat#E1601
            agaroseinvitrogenCat#75510019-100
            X-α-galSolarbioCat#B8050
            Poly (ethylene glycol) 4000SigmaCat#89510
            DMSOSolarbioCat#D8371
            NP-40MilliporeCat#492018
            SD/-Ade/-His/-Leu/-Trp with agarCoolaberCat#PM2112
            lithium acetateSigmaCat#920320
            NaClSolarbioCat#S8210
            anti-FLAG M2 beadsSigmaCat#M8823
            anti-GFP beadsDia-AnCat#MIP0057S

            Critical commercial assays

            TransZolTransGen BiotechCat#ET101
            PrimeScript RT reagent KitTarakaCat#RR047Q
            SYBR supermixBIO-RADCat#1708880B10
            cOmplete Protease Inhibitor CocktailRocheCat#05892970001

            Deposited data

            transcriptome and ChIP-seq raw dataThis study https://www.ncbi.nlm.nih.gov/sra/PRJNA842487

            Experimental models: Organisms/strains

            Arabidopsis, mutant, phyA-211Gifted by Chentao Lin, Fujian Agriculture and Forestry UniversityN/A
            Arabidopsiss, mutant, phyB-9Gifted by Chentao Lin, Fujian Agriculture and Forestry UniversityN/A
            Arabidopsis, mutant, cry1-304Gifted by Chentao Lin, Fujian Agriculture and Forestry UniversityN/A
            Arabidopsis, mutant, cry1-349Gifted by Chentao Lin, Fujian Agriculture and Forestry UniversityN/A
            Arabidopsis, mutant, cry2-1Gifted by Chentao Lin, Fujian Agriculture and Forestry UniversityN/A
            Arabidopsis, GFP-CRY1Gifted by Chentao Lin, Fujian Agriculture and Forestry UniversityN/A
            Arabidopsis, mutant, hsfA1sQKGifted by Hsiang-chin Liu, National Taiwan UniversityN/A
            Arabidopsis, mutants, HsfA1a/b/eGifted by Hsiang-chin Liu, National Taiwan UniversityN/A
            Arabidopsis, MYC-CRY1/cry1-304Gifted by Hongquan Yang, Shanghai Normal UniversityN/A
            Arabidopsis, HsfA1d-FlagThis studyN/A
            Col-0, DKThis studyN/A
            Col-0, WLThis studyN/A
            HsfA1 d-Flag, DK-WLThis studyN/A
            HsfA1 d-Flag, DK-34°CThis studyN/A
            HsfA1 d-Flag, DKThis studyN/A
            HsfA1 d-Flag, WLThis studyN/A
            hsfA1sQK, DKThis studyN/A
            cry1-304, DKThis studyN/A
            Col-0, DK-WLThis studyN/A
            hsfA1sQK, DK-WLThis studyN/A
            cry1-304, DK-WLThis studyN/A
            Col-0, DK-34°CThis studyN/A
            hsfA1sQK, DK-34°CThis studyN/A
            cry1-304, DK-34°CThis studyN/A
            HsfA1 d-Flag/Col_0This studyN/A
            HsfA1 d-Flag/cry1-304This studyN/A
            GFP-CRY1This studyN/A

            Oligonucleotides

            Primer for vector instruction (See Table S3)This studyN/A

            Recombinant DNA

            BD-HsfA1d (1-220aa) vectorThis studyN/A
            BD-IMPα1 vectorThis studyN/A
            AD-CRY1NT (1-490aa) vectorThis studyN/A
            AD-AD-CRY1CT (491-681aa) vectorThis studyN/A
            AD-CRY1 vectorThis studyN/A
            CRY1-nLUC vectorThis studyN/A
            IMPα1-nLUC vectorThis studyN/A
            HsfA1 d-cLUC vectorThis studyN/A
            CRY1-cLUC vectorThis studyN/A
            CRY2-cLUC vectorThis studyN/A
            HsfA1 a-nLUC vectorThis studyN/A
            HsfA1 b-nLUC vectorThis studyN/A
            HsfA1d-nLUC vectorThis studyN/A
            HsfA1-nLUC vectorThis studyN/A
            CRY1-nYFP vectorThis studyN/A
            HsfA1d-cYFP vectorThis studyN/A
            2300-CRY1 vectorThis studyN/A
            HY5-mCherry vectorThis studyN/A

            Software and algorithms

            RNA-seq mappingHISAT2 http://www.psc.edu/user-resources/software/hisat2
            evaluating DEGsDESeq2N/A
            data filteringTrimmomatic v0.38 http://www.usadellab.org/cms/index.php?page=trimmomatic
            data quality assessingFastQC https://www.bioinformatics.babraham.ac.uk/projects/download.html#fastqc
            ChIP-seq mappingBowtie2 (v2.3.5) https://sourceforge.net/projects/bowtie-bio/
            ChIP-seq alignmentSamtools (V1.9) https://github.com/samtools/samtools/releases/download/1.9/samtools-1.9.tar.bz2
            ChIP-seq Peak CallingMACS2 (v2.1.1.20160309) https://pypi.python.org/pypi/MACS2
            illustrating the signal enrichment analysisDeeptoolsN/A
            gene annotationsChIPseekerN/A
            Highlights
            • Blue light activates the plant heat shock response via CRY1-mediated activation of HsfA1d

            • HsfA1d and CRY1 regulate the expression of heat shock genes in response to light

            • CRY1 promotes nuclear localization of HsfA1d under blue light through physical interaction

            • CRY1 shares binding sites of target genes with HsfA1d and affects binding affinity of HsfA1d

            Author and article information

            Journal
            101573691
            39703
            Cell Rep
            Cell Rep
            Cell reports
            2211-1247
            10 October 2023
            26 September 2023
            12 September 2023
            23 October 2023
            : 42
            : 9
            : 113117
            Affiliations
            [1 ]College of Agronomy, Henan Agricultural University, Zhengzhou 450046, China
            [2 ]Whitehead Institute for Biomedical Research, Cambridge, MA, USA
            [3 ]Department of Molecular Genetics and Cell Biology and Center for Physics of Evolving Systems, University of Chicago, Chicago, IL, USA
            Author notes
            [4]

            These authors contributed equally

            [5]

            Lead contact

            AUTHOR CONTRIBUTIONS

            Conceptualization: X.Z., D.P., and Y.S.; Methodology and performing of the majority of experiments: J.G., R.Z., L.Z., and J.W.; Co-immunoprecipitation, all ChIP assays, and ChIP-qPCR: J.G.; Nuclear fractionation: R.Z. (with the help of L.S.); Yeast two-hybrid analyses and western blotting: L.Z. (with the help of M.J.); Bioinformatics analyses: S.L. (with the assistance from X.Z. and J.G.); Phenotyping of plants: P.Z.; Original draft writing: X.Z., D.P., and Y.S.; Review and editing: X.Z., Y.S., J.G., and L.Z.; Funding acquisition: X.Z., J.G., and D.P.; Resources: J.Y., G.K., and Y.J.; Supervision, Y.S., Y. J., D.P. and X.Z.

            Article
            NIHMS1933914
            10.1016/j.celrep.2023.113117
            10591714
            37703177
            10597a35-3ce1-4b6f-9b90-58b813d55c3d

            This is an open access article under the CC BY-NC-ND license ( http://creativecommons.org/licenses/by-nc-nd/4.0/).

            History
            Categories
            Article

            Cell biology

            Comments

            Comment on this article