Skip to main content
Erschienen in: Longevity & Healthspan 1/2014

Open Access 01.12.2014 | Research

Transcriptional regulation of Caenorhabditis elegansFOXO/DAF-16 modulates lifespan

verfasst von: Ankita Bansal, Eun-Soo Kwon, Darryl Conte Jr, Haibo Liu, Michael J Gilchrist, Lesley T MacNeil, Heidi A Tissenbaum

Erschienen in: Longevity & Healthspan | Ausgabe 1/2014

Abstract

Background

Insulin/IGF-1 signaling plays a central role in longevity across phylogeny. In C. elegans, the forkhead box O (FOXO) transcription factor, DAF-16, is the primary target of insulin/IGF-1 signaling, and multiple isoforms of DAF-16 (a, b, and d/f) modulate lifespan, metabolism, dauer formation, and stress resistance. Thus far, across phylogeny modulation of mammalian FOXOs and DAF-16 have focused on post-translational regulation with little focus on transcriptional regulation. In C. elegans, we have previously shown that DAF-16d/f cooperates with DAF-16a to promote longevity. In this study, we generated transgenic strains expressing near-endogenous levels of either daf-16a or daf-16d/f, and examined temporal expression of the isoforms to further define how these isoforms contribute to lifespan regulation.

Results

Here, we show that DAF-16a is sensitive both to changes in gene dosage and to alterations in the level of insulin/IGF-1 signaling. Interestingly, we find that as worms age, the intestinal expression of daf-16d/f but not daf-16a is dramatically upregulated at the level of transcription. Preventing this transcriptional upregulation shortens lifespan, indicating that transcriptional regulation of daf-16d/f promotes longevity. In an RNAi screen of transcriptional regulators, we identify elt-2 (GATA transcription factor) and swsn-1 (core subunit of SWI/SNF complex) as key modulators of daf-16d/f gene expression. ELT-2 and another GATA factor, ELT-4, promote longevity via both DAF-16a and DAF-16d/f while the components of SWI/SNF complex promote longevity specifically via DAF-16d/f.

Conclusions

Our findings indicate that transcriptional control of C. elegans FOXO/daf-16 is an essential regulatory event. Considering the conservation of FOXO across species, our findings identify a new layer of FOXO regulation as a potential determinant of mammalian longevity and age-related diseases such as cancer and diabetes.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​2046-2395-3-5) contains supplementary material, which is available to authorized users.
Ankita Bansal, Eun-Soo Kwon contributed equally to this work.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

AB contributed to the conception and design, data collection and analysis, manuscript writing, critical revision, and final approval of the manuscript. ESK contributed to the concept and design, data collection and analysis, manuscript writing, and final approval of the manuscript. DC contributed to manuscript writing, data analysis, and final approval of the manuscript. HL, MJG, and LTM contributed to data collection and final approval of the manuscript. HAT contributed to the conception and design, financial support, manuscript writing, and final approval of manuscript. All authors read and approved the final manuscript.

Background

The evolutionarily conserved insulin/IGF-1 signaling (IIS) pathway regulates lifespan from worms to mammals and in C. elegans also modulates dauer formation, stress response, and metabolism [14]. The C. elegans IIS pathway consists of an insulin/IGF-1 receptor (DAF-2) [5], a PI 3-kinase (AGE-1/AAP-1) [6, 7] serine/threonine kinases (PDK-1, AKT-1, and AKT-2) [8, 9], and a Forkhead Box O (FOXO) transcription factor (DAF-16) [10, 11]. IIS ultimately results in the AKT-dependent phosphorylation of DAF-16, thereby preventing DAF-16 from entering the nucleus to regulate its target genes [3, 4, 12]. Therefore, the C. elegans FOXO ortholog, DAF-16 is the primary downstream target of the IIS pathway [10, 11].
In contrast to C. elegans, mammals have three closely related FOXO proteins (FOXO1, FOXO3, and FOXO4) that share a high degree of homology, overlapping expression patterns, and common target genes [13, 14]. These FOXOs appear to have discrete functions as each mutant displays a distinct phenotype: Foxo1 null mice die as embryos due to defects in angiogenesis [15], Foxo3 mutants are viable but show age-dependent female sterility [15, 16], and Foxo4 null mutants are viable with no detectable phenotype [15]. Interestingly, conditional somatic deletion of all three Foxo loci results in a cancer-prone phenotype that includes age-progressive thymic cancers and hemangiomas, indicating that Foxo1, Foxo3, and Foxo4 are redundant tumor suppressor genes specifically involved in endothelial growth suppression [17]. Recently, a more distantly related FOXO family member, FOXO6, was shown to regulate gluconeogenesis [18]. Thus, four mammalian FOXO homologs play distinct and overlapping roles in multiple biological processes.
DAF-16, the only C. elegans FOXO ortholog, expresses multiple isoforms (Figure 1). Initial studies on DAF-16 identified three different transcripts, daf-16a1, daf-16a2, and daf-16b. daf-16a1 and daf-16a2 share the same promoter and 11 exons, but alternative splicing at exon 3 results in an insertion of two additional amino acids in the DAF-16a1 protein compared to DAF-16a2 [19, 20]. daf-16b encodes a shorter isoform, expressed from a different promoter and results in a protein that includes the same C-terminal 319 amino acids as the DAF-16a1/a2 isoforms. Recently, we identified a third DAF-16 isoform, daf-16d/f, which is driven by a distinct promoter and initiates approximately 10 kb upstream of daf-16a1/a2. daf-16d/f is encoded by 14 exons, 10 of which are shared with DAF-16a1/a2 and DAF-16b. Studies using isoform-specific RNAi and transgenes have revealed cooperative as well as specific functions for these three DAF-16 isoforms (DAF-16a, DAF-16b, and DAF-16d/f) in the regulation of lifespan, dauer formation, fat storage, and stress resistance [21]. For lifespan regulation, however, we found that only DAF-16a and DAF-16d/f are involved [21]. The relative contribution of each of these two isoforms and regulation of each isoform remains unknown.
To date, much has been learned about the post-translational regulation of FOXO/DAF-16. For example, FOXO/DAF-16 is directly phosphorylated by kinases including AKT-1, AKT-2, SGK-1, Jun-N-terminal kinase (JNK/JNK-1) [2224], Ste20-like protein kinase (MST1/CST-1) [25], and AMP-activated protein kinase (AMPK/AAK-1) [26, 27], and FOXO/DAF-16 interacts with a Serine/threonine-protein phosphatase 4 regulatory subunit, SMK-1 [28]. FOXO/DAF-16 also interacts with 14-3-3 proteins, which modulate the ability of FOXO/DAF-16 to interact with cofactors, including the sirtuin family (SIRT1/SIR-2.1) of NAD-dependent deacetylases [3, 4, 29, 30]. These interactions and direct post-translational modifications function to modulate the activity of FOXO/DAF-16 under different conditions [3, 4].
Despite the growing body of knowledge centered on the post-translational regulation of FOXO/DAF-16, little is known about the transcriptional regulation of daf-16 in C. elegans. To begin to address these issues, we examined the expression of the daf-16a and daf-16d/f isoforms throughout development and adulthood. We found that daf-16d/f expression is dramatically increased at the level of transcription during the young adult stage, and this upregulation of daf-16d/f expression is required for longevity. Furthermore, using an RNAi screen we identified two transcriptional regulators of daf-16d/f expression: elt-2, encoding an essential GATA transcription factor required for intestinal development [31, 32], and swsn-1, encoding a core component of the SWI/SNF chromatin remodeling complex [33, 34]. ELT-2 and another GATA factor, ELT-4, are required for the expression of both daf-16a and daf-16d/f and for the ability of both daf-16a and daf-16d/f to promote longevity. By contrast, SWSN-1 and other SWI/SNF components are required for the upregulation of daf-16d/f expression in the intestine of young adults, but not daf-16a expression. Consistent with this finding, components of the SWI/SNF complex promote longevity via daf-16d/f but not daf-16a. Taken together, our findings reveal that transcriptional regulation of daf-16d/f is an important regulatory event in lifespan determination.

Results and discussion

daf-16has multiple isoforms

daf-16 functions as a central regulator of multiple biological processes including lifespan, development, fat storage, and stress resistance [14]. The C. elegans genome resource, Wormbase (http://​www.​wormbase.​org), predicts eight putative isoforms of daf-16, with each isoform designated by a lowercase letter following the cosmid name for daf-16 (R13H8.1a-h). To verify expression of the daf-16 isoforms, we first analyzed the Gurdon Institute C. elegans expressed sequence tag (EST) database (http://​genomics.​nimr.​mrc.​ac.​uk/​online/​worm-fl-db.​html) [35, 36]. As shown in Additional file 1: Table S1, ESTs were identified that map to daf-16f (R13H8.1f), daf-16d (R13H8.1d), daf-16a (R13H8.1b/c), and daf-16b (R13H8.1a). In addition to ESTs, we examined published capped RNA (Cap-Seq) (Gu et al.[37]), total mRNA, and ribosome-associated mRNA deep sequencing datasets (Stadler et al.[38]). These datasets support the existence of daf-16f, daf-16d, daf-16a1, daf-16a2, and daf-16b. In particular, the Cap-Seq data specifically identified four spliced leader (SL1) sites corresponding to the capped 5’ ends of daf-16f, daf-16d, daf-16a1/a2, and daf-16b, as indicated in green in Figure 1. Therefore, our findings verify the existence of five isoforms in contrast to eight that were predicted initially for DAF-16 - (DAF-f (R13H8.1f), DAF-16d (R13H8.1d), DAF-16a1 (R13H8.1b), DAF-16a2 (R13H8.1c), and DAF-16b (R13H81.a).
Previous RNAi and transgenic studies examined the function of different daf-16 isoforms (daf-16a, b, d/f) [1921], which revealed that daf-16b functions in dauer formation, while daf-16a and daf-16d/f contribute to lifespan regulation. According to our previous studies [21], daf-16a constructs could not rescue the lifespan phenotype of a complete loss of daf-16, and daf-16d transgenes were unstable and did not consistently rescue daf-16 loss-of-function. However, transgenes expressing the daf-16f isoform or daf-16d/f showed similar expression patterns and consistently rescued the complete loss of daf-16[21] (Figure 2). Moreover, based on the CapSeq data, both daf-16f and daf-16d have the same transcription start site. Therefore, we continue to refer to these isoforms as daf-16d/f in this study.

Effects of altering DAF-16a dosage on lifespan

Dissecting the relative contributions of DAF-16d/f and DAF-16a in the regulation of longevity has been challenging due to incomplete knockdown by isoform-specific RNAi [21] and the lack of isoform-specific mutants. This is further complicated by the existence of a dose-dependent effect of daf-16a and daf-16d/f transgenes [21]. To expand our analysis of the daf-16 dosage effect on lifespan, we first generated a new low-copy daf-16a transgene by microparticle bombardment, which we refer to as daf-16a:gfp HT . In addition, we chose three commonly used high-copy daf-16a transgenes, referred to as daf-16a::gfp CF derived from strain CF1407 [20], daf-16a::gfp TJ from TJ356 [39], and daf-16a::gfp GR from RX86 [40]. For all the analyses that follow, we first placed each daf-16 transgene into the same genetic background which is a null allele of the endogenous daf-16 locus [daf-16(mgDf50)].
To compare the levels of daf-16a in the different transgenic strains, we measured the level of daf-16a expression from each transgene in the daf-16(mgDf50); daf-2(e1370) background and compared it to the endogenous level of daf-16a in daf-2(e1370) mutants (Figure 2A, Additional file 1: Table S2). The level of daf-16a expressed from the newly generated daf-16a HT transgenic strain was approximately two-fold higher than the endogenous daf-16a in the daf-2(e1370) mutant, while the commonly used daf-16a transgenes were expressed at 10- to 400-fold higher levels than endogenous daf-16a. These results show the following order for levels of the daf-16a transcript: endogenous daf-16a < HT < CF < TJ < < GR. Consistent with the mRNA expression data, the level of DAF-16a protein from the transgenic strains also increases (Additional file 1: Figure S1). The GFP intensity in the four transgenic strains correlated with the level of expression of DAF-16a (data not shown).
Next, we determined the nuclear:cytosolic ratio of DAF-16a in the transgenic strains expressing different levels of DAF-16a (Additional file 1: Figure S2) in daf-2(e1370) background. DAF-16a was almost entirely in the nucleus when expressed at low levels from the daf-16a::gfp HT transgene. By contrast, DAF-16a was mainly cytosolic in the daf-2(e1370); daf-16a GR strain, which shows the highest level of DAF-16a expression, while the daf-2(e1370); daf-16a::gfp CF and daf-2(e1370); daf-16a::gfp TJ strains showed an intermediate nuclear localization of DAF-16a. Importantly, since these different transgenic strains were generated with different methods and may be integrated at different locations, this indicates that it is the level of mRNA and protein of DAF-16a that determines its regulatory ability.
We reasoned that if a daf-16a transgene is expressed at close to endogenous levels, the daf-16a transgenic animals should not live longer than wild-type animals expressing endogenous levels of all three DAF-16 isoforms. Interestingly, our low-copy daf-16a::gfp HT transgenic worms lived shorter than wild-type (Figure 2B, Additional file 1: Table S3), suggesting that low level expression of daf-16a alone cannot replace loss of the endogenous daf-16 for lifespan regulation. However, the three high-copy daf-16a transgenic strains lived longer than wild type (Figure 2B, Additional file 1: Table S3). Therefore, these data suggest that the level of DAF-16a expressed from the daf-16a::gfp CF , daf-16a::gfp TJ , and daf-16a::gfp GR transgenes exceeds the inhibitory capacity of the IIS pathway. This gene dosage effect was even more pronounced when each daf-16a transgene was analyzed in the daf-16(mgDf50); daf-2(e1370) double mutant background (Figure 2C, Additional file 1: Table S3). As shown in Figure 2B and consistent with previous findings [1921], when expressed at low levels from a daf-16a::gfp HT transgene, DAF-16a cannot fully compensate for a complete loss of DAF-16 function (Figure 2B) [21]. An increase in lifespan was observed with increased expression of DAF-16a (daf-16a::gfp CF , daf-16a::gfp TJ ). However, as the dosage of DAF-16a continued to increase (for example, daf-16a::gfp GR ), a reduction in lifespan was seen (Figure 2C, Additional file 1: Table S3). Consistent with this finding, unlike the other daf-16a transgenic strains, the daf-16a::gfp GR showed severely delayed development and reduced fecundity, suggesting that extremely high levels of DAF-16a are toxic (Additional file 1: Figure S3). Together, these findings suggest that the three commonly used strains for analyses of DAF-16:GFP do not accurately reflect the endogenous function of DAF-16a in lifespan regulation. Furthermore, these results indicate that low-copy daf-16a strains are more relevant for functional analyses as well as for DAF-16 localization assays (Additional file 1: Figures S2).
Next, we examined the low-copy daf-16d/f::gfp HT transgene [21], which had approximately four-fold higher transcript level than endogenous daf-16d/f levels in the daf-2(e1370) mutant background (Additional file 1: Table S2). Unlike high-copy daf-16a and daf-16d/f transgenic worms [21], these daf-16(mgDf50); daf-16d/f::gfp HT transgenic worms did not live longer than wild-type (Figure 2B, Additional file 1: Table S3), suggesting that DAF-16d/f activity in the low-copy daf-16d/f transgenic strain is regulated within the inhibitory capacity of the IIS pathway. Even though the strains were generated by different methods, we suggest that it is the level of the DAF-16 isoform protein that leads to an environment that does not accurately reflect endogenous levels of DAF-16.

DAF-16d/f and DAF-16a respond differently to changes in IIS

The low-copy daf-16 isoform transgenes were crossed into the background of another widely used daf-2 allele, daf-2(e1368), which exhibits weaker longevity and dauer arrest phenotypes than daf-2(e1370)[5, 41]. Based on our lifespan data in the daf-16(mgDf50); daf-2(e1370) background, we expected that the daf-16d/f::gfp HT transgene would be sufficient to fully complement the lifespan of a daf-16(mgDf50); daf-2(e1368) double mutant. Yet, to our surprise, daf-16(mgDf50); daf-2(e1368); daf-16d/f::gfp HT transgenic worms lived significantly shorter than the daf-2(e1368) single mutant (Figure 3A, Additional file 1: Figure S4, Additional file 1: Table S3, Additional file 1: Table S4). In contrast, the daf-16a::gfp HT transgene rescued lifespan to similar levels in both daf-2(e1368) and daf-2(e1370) mutant backgrounds (Additional file 1: Figure S4, Additional file 1: Table S3, Additional file 1: Table S4).
To further test the effect of DAF-16d/f in the daf-2(e1370) and daf-2(e1368) mutant backgrounds, we examined dauer formation as an additional output of DAF-16 activity. At the restrictive temperature of 25°C, both daf-2(e1370) and daf-2(1368) form 100% dauers whereas daf-16(mgDf50); daf-2(e1370) and daf-16(mgDf50); daf-2(e1368) double mutants do not form any dauers. However, once again, the DAF-16d/f transgene had different effects in the two daf-2 alleles: daf-16(mgDf50); daf-2(e1368); daf-16d/f::gfp HT transgenic worms formed approximately 39% dauers, whereas daf-16(mgDf50); daf-2(e1370); daf-16d/f::gfp HT worms formed approximately 98% dauers at 25°C (Additional file 1: Figure S5A). At the semi-permissive temperature of 20°C, a similar trend was observed. This is in contrast to daf-16a transgenic worms where dauer formation was comparable in both daf-2 mutant backgrounds (Figure 3B, Additional file 1: Figure S5).
Examining the DAF-16 nuclear/cytosolic localization, DAF-16d/f was enriched in the nucleus in the daf-2(e1370) background, but more cytosolic in the daf-2(e1368) background (Figure 3C,D). In contrast, DAF-16a was primarily enriched in the nucleus in both daf-2(e1368) and daf-2(e1370) backgrounds (Figure 3C,D). Thus, the DAF-16 localization data correlated with both functional outputs - lifespan and dauer formation. Whereas DAF-16a is activated to a similar level in both alleles, DAF-16d/f is less active in the daf-2(e1368) mutant than in the daf-2(e1370) mutant. Therefore, these data show that DAF-16a and DAF-16d/f exhibit different thresholds for inhibition by the upstream IIS pathway.

Transcription of daf-16d/fis upregulated during aging

Although the spatial expression patterns of daf-16 have been extensively investigated [19, 20, 39], the temporal expression of daf-16 has not been studied. We monitored the expression of the daf-16 isoform::gfp strains throughout development and for the first few days of adulthood. Interestingly, as shown in Additional file 1: Figure S6, the GFP intensity of the daf-16d/f::gfp transgenic worms dramatically increased in the adult stage compared to larval stages.
Next, we asked if the increase in GFP intensity in adult daf-16d/f::gfp transgenic animals correlates with the level of the endogenous transcript. Using qRT-PCR, we found that each of the daf-16 isoforms was expressed at a constant level throughout larval development. However, as worms aged, the daf-16d/f transcript dramatically increased in both wild-type and long-lived daf-2(e1370) mutants, while the daf-16a transcript increased only slightly (Figure 4A,B, Additional file 1: Figure S7-S9). To further confirm this result, we tested the upregulation of DAF-16a vs DAF-16d/f at the protein level. Since it is not possible to distinguish differences in endogenous levels of DAF-16a and DAF-16d/f protein, we were limited to examining changes in DAF-16 protein levels in strains that only bear one DAF-16 isoform (daf-2;daf-16;daf-16a::gfp or daf-2;daf-16;daf-16d/f::gfp). Using these transgenic strains, consistent with our mRNA data, the age-dependent increase in the levels of DAF-16d/f was also observed at the protein level (Figure 4C,D).
The qRT-PCR assay is a read-out of the steady-state level of mRNA, but the DAF-16::GFP fusion fluorescence is the product of both transcriptional and post-transcriptional regulation. Therefore, to measure the transcriptional promoter activity of each daf-16 isoform, we generated daf-16 promoter::GFP fusions (Pdaf-16::gfp). The promoter of each daf-16 isoform was placed upstream of the GFP open reading frame fused to the unc-54 3′UTR, which is known to be absent of temporal regulatory elements [42]. We reasoned that if increased transcription is responsible for the elevated mRNA level in young adults, then the GFP intensity should become brighter in the promoter::gfp transgenic worms as the worms age. In the Pdaf-16a::gfp worms, the GFP signal throughout the body increased slightly with age. In contrast, in Pdaf-16d/f::gfp worms, GFP expression increased dramatically throughout the intestine (data not shown Figure 4E). Therefore, across multiple independent experiments we found that transcription of daf-16, particularly daf-16d/f, was regulated in an age-dependent manner.

daf-16d/ftranscript level in the early adult determines lifespan

daf-16 expression in the intestine is critical for lifespan regulation in C. elegans[21, 43], and our temporal and spatial expression data indicate that daf-16d/f mRNA is dramatically upregulated in the intestine as worms age. To determine the importance of DAF-16 upregulation in aging worms, we used RNAi to maintain daf-16d/f expression in the adult stage at a level comparable to that in the L4 stage, thereby preventing the upregulation of DAF-16d/f during aging. To achieve the correct knockdown level, we prepared serial-dilutions of daf-16 RNAi bacteria (from no dilution to 64-fold dilution) and fed daf-16(mgDf50); daf-2(e1370); daf-16d/f::gfp HT worms from the L4 stage to day 2 adults. After 2 days of growth on the daf-16 RNAi plates, worms were transferred to control RNAi plates. We found that worms grown on four-fold to 16-fold diluted daf-16 RNAi bacteria maintained daf-16d/f expression at levels similar to that of the L4 worms (Additional file 1: Figure S10). Importantly, the lifespan of daf-16(mgDf50); daf-2(e1370); daf-16d/f::gfp HT worms fed on the four-fold to 16-fold diluted daf-16 RNAi bacteria was reduced as compared to animals grown on control bacteria (Figure 4F, Additional file 1: Table S5). Therefore, the upregulation of daf-16d/f gene expression at the young adult stage is an important determinant of C. elegans lifespan.

The ELT-2 transcription factor promote longevity by regulating both daf-16a and daf-16d/fexpression

Our findings suggest that transcriptional regulators expressed in the intestine activate daf-16d/f transcription in an age-dependent manner. To identify regulator(s) of daf-16d/f gene expression, we performed an RNAi screen of 892 transcriptional regulators using Pdaf-16d/f::gfp transgenic worms. RNAi of two transcriptional regulators reduced Pdaf-16d/f::gfp expression: elt-2, a GATA transcription factor; and swsn-1, a core component of the SWI/SNF chromatin remodeling complex (Figure 5A, B,C). elt-2 is essential for intestinal development [31, 32, 44, 45] as well as innate immune responses to bacterial and fungal infection [46] and directly regulates the expression of >80% of intestinal genes, including genes that are downstream of daf-16 (dod gene) [32].
Because worms fed with elt-2 RNAi from hatching develop an abnormal intestinal structure and die within a few days, we used post-developmental RNAi to silence elt-2 beginning at the L4 stage when intestinal development is complete. Indeed, post-developmental elt-2 RNAi treatment also reduced Pdaf-16d/f::gfp expression (Figure 5B). When we examined the endogenous daf-16 transcripts, we found that post-developmental elt-2 RNAi treatment reduced the expression of endogenous daf-16d/f compared to control RNAi (Figure 5C). In contrast, the Pdaf-16a::gfp was only marginally downregulated on elt-2 RNAi at day 2 and endogenous daf-16a expression was not changed, likely due to the fact that its intestinal expression is only a minor portion of total daf-16a (Additional file 1: Figure S11) [21]. Accordingly and consistent with a recent report [47], elt-2 RNAi reduced the lifespan of the daf-2(e1370) single mutant (expressing endogenous daf-16), as well as daf-16(mgDf50); daf-2(e1370); daf-16a::gfp HT and daf-16(mgDf50); daf-2(e1370); daf-16d/f::gfp HT transgenic worms (Figure 6A, C, D, Additional file 1: Table S6). Relative to the control RNAi, elt-2 RNAi did not shorten the lifespan of a daf-16(mgDf50); daf-2(e1370) mutant (Figure 6B), suggesting that elt-2 RNAi shortens lifespan by reducing intestinal daf-16 gene expression and not by a daf-16-independent pathway.
We analyzed several additional GATA transcription factors for a role in daf-16 transcriptional regulation and longevity, including elt-4 and elt-7, which are expressed in the intestine [44], and elt-3, elt-5, and elt-6, which were previously implicated in lifespan regulation [48]. Only elt-4 RNAi reduced the expression of the Pdaf-16d/f::gfp transcriptional reporter (Additional file 1: Figure S13), though less dramatically than elt-2 RNAi. Furthermore, only elt-4 RNAi reduced the lifespan of both daf-16(mgDf50); daf-2(e1370); daf-16a::gfp HT and daf-16(mgDf50); daf-2(e1370); daf-16d/f::gfp HT transgenic worms (Figure 6E–H, Additional file 1: Table S6). Neither Pdaf-16d/f::gfp expression nor lifespan was affected by elt-7, elt-3, elt-5, or elt-6 RNAi (Additional file 1: Figure S13 and Additional file 1: Table S6). These data are consistent with a recent study [49] suggesting that the previously reported role of ELT-3 in lifespan regulation [48] should be re-evaluated. Our findings indicate that the ELT-2 and ELT-4 GATA factors promote longevity by regulating intestinal daf-16 expression.

SWI/SNF determines lifespan by regulating daf-16d/fexpression

The second positive clone we obtained from our screen was swsn-1, which encodes a core component of the highly conserved SWI/SNF nucleosome-remodeling complex [50]. In C. elegans, SWI/SNF is required for asymmetric cell division [51] and differentiation [52]. Prior to the L4 stage, Pdaf-16d/f::gfp expression was unaffected by swsn-1 RNAi (Figure 5A,B). In adult worms, however, the upregulation of GFP in the mid-intestine is severely reduced by swsn-1 RNAi (Figure 5B). Expression of endogenous daf-16d/f was also reduced in animals exposed to swsn-1 RNAi (Figure 5C, Additional file 1: Figure S12B). Interestingly, swsn-1 RNAi did not significantly change the expression of Pdaf-16a::gfp (Additional file 1: Figure S11C), suggesting that SWSN-1 regulates the temporal and spatial expression of daf-16d/f, specifically. RNAi targeting snfc-5 and swsn-2.1, core and accessory subunits of SWI/SNF, respectively, also reduced the temporal upregulation of daf-16d/f expression with age (Additional file 1: Figure S12) but did not change daf-16a expression, indicating that the SWI/SNF complex specifically controls daf-16d/f expression.
We next asked if SWI/SNF plays a role in lifespan regulation. Both swsn-1 and snfc-5 are essential genes [51] and hatched larvae grown on swsn-1 or snfc-5 RNAi develop into sick adults with pleiotropic defects [51, 53]. To avoid these complications in lifespan analysis, we used post-developmental (L4) RNAi to silence swsn-1. However, Pdaf-16d/f::gfp expression was largely unaffected by post-developmental swsn-1 RNAi (data not shown) and we observed only a marginal reduction of lifespan in daf-16(mgDf50); daf-2(e1370); daf-16d/f::gfp HT transgenic worms (Figure 6D, Additional file 1: Table S6), suggesting that post-developmental (L4) RNAi does not efficiently silence swsn-1. Worms exposed to swsn-2.1 RNAi from hatching, however, were much healthier than animals grown on swsn-1 or snfc-5 RNAi, which allowed us to assess the effect of SWI/SNF on lifespan. As shown in Figure 6H and Additional file 1: Table S6, swsn-2.1 RNAi shortened the lifespan of daf-16(mgDf50); daf-2(e1370); daf-16d/f::gfp HT worms, but only modestly reduced the lifespan of daf-2(e1370) or daf-16(mgDf50); daf-2(e1370); daf-16a::gfp HT animals (Figure 6F,G, Additional file 1: Table S6). Interestingly, a very recent paper found that SWI/SNF interacts with DAF-16 [54]. These studies revealed that a SWI/SNF-DAF-16 complex co-localizes at the promoters of DAF-16 direct targets and activates genes important for lifespan, dauer formation, and stress resistance [54]. Therefore, these results support a biochemical interaction of DAF-16 and SWI/SNF. Taken together, our data are consistent with a model in which the SWI/SNF complex directly interacts with DAF-16 to specifically promote longevity by regulating daf-16d/f expression.

Conclusion

In C. elegans, the single FOXO family member, DAF-16 regulates lifespan, metabolism, development, and stress resistance by generating multiple isoforms including DAF-16d/f, DAF-16a, and DAF-16b [21]. Comparing the relative abundances of the different isoforms revealed that daf-16d/f is the most abundant isoform in early adulthood and that transcription of daf-16d/f mRNA is dramatically increased in the intestine as animals age. These findings are in agreement with previous studies showing that DAF-16 expression in the intestine is important for lifespan regulation and that lifespan is determined in adulthood [43, 55]. Therefore, the temporal control of daf-16d/f transcription in the intestine is a critical determinant of longevity.
Importantly, temporal regulation of FOXO expression is conserved in mammals. In rats, FOXO3 and FOXO4 transcript are undetectable in very young animals but increase as animals age in the duodenum [56], and human FOXO1 mRNA is significantly enriched in muscle samples from old individuals [57]. It will be particularly interesting to determine whether the age-dependent increase in mammalian FOXO expression are regulated at the level of transcription, as we have shown for daf-16d/f in C. elegans.
In worms, flies, and mammals, FOXO is the primary target of the IIS pathway [2, 58]. In C. elegans, the DAF-2 IIS receptor has been extensively studied by genetic analysis. The daf-2(e1370) allele harbors a mutation in the tyrosine kinase domain, whereas daf-2(e1368) has a mutation in the ligand-binding domain [5]. These daf-2 alleles show phenotypic differences, with daf-2(e1370) displaying longer lifespan and stronger dauer arrest compared to daf-2(1368) [41, 58], indicating that the IIS pathway is more active in daf-2(e1368) than in daf-2(e1370). Our lifespan, dauer, and DAF-16 nuclear localization data in these daf-2 mutants revealed that daf-16d/f and daf-16a respond differently to changes in the levels of IIS. In both mutant backgrounds, DAF-16a is predominantly nuclear indicating that DAF-16a is activated to a similar level in both daf-2 mutants. However, DAF-16d/f is nuclear only in the daf-2(e1370) background and therefore appears to be more active in this background. This suggests that a small decrease in IIS activates DAF-16a, whereas large decreases in IIS will activate DAF-16d/f and could add another layer of regulation by the different DAF-16 isoforms.
Using an RNAi screen, we identified the GATA factor ELT-2 and the SWI/SNF subunit SWSN-1 as factors required for daf-16d/f expression. Our functional analysis reveals that the intestinal GATA transcription factors, ELT-2 and ELT-4, regulate daf-16 expression and, in turn, longevity. Our findings are consistent with the work of McGhee et al. [32], who identified multiple GATA binding sites upstream of both daf-16a and daf-16d/f and proposed that intestinal GATA factors, particularly ELT-2, might regulate the expression of daf-16. Moreover, the promoters of many DAF-16 targets also have GATA binding sites [32, 48, 59], suggesting that ELT-2/ELT-4 may cooperate with DAF-16 to regulate longevity genes in the intestine.
Recent studies suggest that epigenetic control of gene expression is important for the aging process [60]. In C. elegans, IIS-dependent and IIS-independent epigenetic modifications have been linked to longevity [6164]. Our study shows that SWI/SNF regulates lifespan by promoting the age-dependent activity of a FOXO gene. Importantly, components of the SWI/SNF complex are required for the age-dependent upregulation of daf-16d/f but not daf-16a. SWSN-2.1 is homologous to mammalian BAF60, a subunit known to interact with a number of transcriptional activators [65, 66]. BAF60, has three variants (a, b, c) [66] and BAF60c has been shown to interact with the GATA4 transcription factor to promote differentiation in early mouse heart development [67]. We envision a similar role in stimulation of daf-16d/f transcription in the adult intestine. For example, ELT-2/ELT-4 GATA or other transcription factor(s) together with SWSN-2.1/BAF60 may interact with the daf-16d/f promoter during development. At the young adult stage, an intestine-specific developmental cue may stimulate the SWSN-2.1-dependent recruitment of the core SWI/SNF machinery to remodel the daf-16d/f promoter and activate daf-16d/f transcription.
We have shown here and elsewhere [21] that daf-16a and daf-16d/f are sensitive to changes in gene dosage. For daf-16d/f, mRNA and protein are dramatically upregulated with age, whereas daf-16a mRNA only minimally changes with age, and preventing the upregulation of daf-16d/f in adults shortens lifespan. ELT-2 and ELT-4 are likely to be direct regulators of DAF-16. Indeed, multiple GATA binding sites exist upstream of both daf-16a and daf-16d/f contain (data not shown). Based on our finding that SWI/SNF specifically regulates the expression of daf-16d/f but not daf-16a, we propose that SWI/SNF modifies the activity of the daf-16d/f promoter but not the daf-16a promoter. Taken together, and consistent with our findings, we suggest a model where daf-16d/f plays a more prominent role in lifespan regulation than daf-16a (Figure 7).
C. elegans has proved to be an invaluable model system for understanding the regulation and biological functions of FOXO transcription factors. The mechanisms that control FOXO proteins at a post-translational level are remarkably conserved from worms to mammals. Our findings indicate that transcriptional control of daf-16/FOXO is also an essential regulatory event. As there is increasing evidence that FOXOs are required for human longevity [6873], our findings will advance our understanding of the regulation and function of DAF-16/mammalian FOXO in aging and age-dependent diseases including cancer and diabetes.

Methods

Strain maintenance

All strains were maintained and handled as described [74]. Animals were grown on standard NGM plates at 15°C using standard C. elegans techniques, unless otherwise indicated. Mutants used in this study included, LGI: daf-16(mgDf50); LGIII: daf-2(e1368, e1370); unc-119(ed3). All transgenic worms have an unc-119(ed3) mutation rescued by an unc-119 + co-transformation marker. Transgenic strains generated and/or used in this study are listed in Additional file 1: Table S8.

Strain construction

i)
daf-2(e1368) unc-119(ed3) double mutants
 
Both genes map to Chromosome II but are 13 map units apart. daf-2(e1368) males were mated to unc-119(ed3) hermaphrodites. Approximately 20 F1 progeny were picked onto individual plates and allowed to produce progeny at 25°C. F2 daf-2(e1368) dauers were selected from each plate and allowed to recover at 15°C. From the F3 progeny, Unc worms were selected, generating daf-2(e1368); unc-119(ed3).
ii) daf-16(mgDf50) ; daf-2(e1368) unc-119(ed3) mutants
daf-16(mgDf50); daf-2(e1368) males were mated to daf-2(e1368); unc-119(ed3) hermaphrodites. Approximately 20 F1 progeny were picked onto individual plates and allowed to have progeny at 25°C. From the F2 progeny on each plate, Unc Non-dauer worms were selected and tested for the daf-16(mgDf50) mutation by PCR. Primer sequences are listed in Additional file 1: Table S7. daf-16(mgDf50); daf-2(e1370); unc-119(ed3) mutants were used for microparticle bombardment [75] to generate daf-16 isoform transgenic worms.

DNA construction for daf-16isoform specific GFP and RNAi

To generate daf-16 isoform::gfp constructs, each cDNA encoding daf-16a, daf-16d/f was cloned into the pGEM-T vector. The clones were then verified by sequencing. For the daf-16a isoform promoter construct, a SalI/BamHI fragment containing 6.0 kb upstream of daf-16a was cloned into pPD95.75 (GFP containing plasmid). For the daf-16d/f isoform promoter construct, a SphI/BamHI fragment containing 4.1 kb upstream of daf-16d/f was cloned into pPD95.75 (GFP containing plasmid). We then generated full-length daf-16a1 cDNAs using mutagenic primers with BamHI/SmaI restriction enzyme sites, which were subcloned into pPD95.75 with the upstream promoter fragment. For daf-16d/f, since we were unable to amplify the daf-16d/f specific cDNA, we generated a construct which could generate both DAF-16d1 and DAF-16d/f by subcloning the 4.1 kb upstream genomic DNA next to the 5’ putative start codon of daf-16d1 cDNA in frame [21].
To allow the constructs to be compatible with ballistic transformation, the unc-119 + gene was introduced into each vector using PCR redirected DNA recombination method [76]. To generate daf-16 isoform specific RNAi vector, NheI/HindIII fragments covering isoform specific/overlapping cDNA were cloned into L4440 vector [21]. The primers used for the PCR analysis are listed in Additional file 1: Table S7.

RNA isolation and real-time PCR

Growing samples

To compare the expression of daf-16 isoforms in developmental stages, worms were synchronized by bleaching, followed by tranferring eggs on plates seeded with OP50 bacteria. Worms were grown until the L1, L2, L3, and L4 stage and then harvested. To obtain aged worms, L4 stage worms were transferred to FuDR plates with final concentration of 0.1 mg/mL [77] seeded with OP50 bacteria. On days 1, 2, 5, and 10, worms were harvested and frozen at -80°C. To compare the expression of daf-16a in L4 stage, in Figure 2, worms were synchronized by bleaching, followed by L1 arrest in M9 buffer. The following day, the L1 worms were placed on plates seeded with OP50 bacteria and further incubated at 15°C until worms reached the L4 stage.

RNA preparation

Total RNA was isolated using acidic phenol (Sigma). Briefly, worms were washed off the plates using ice cold M9 buffer, followed by three additional washes. Next, 0.5 mL of AE buffer (acetic acid, EDTA), 0.1 mL of 10% SDS, and 0.5 mL of phenol were added and the mixture and vortexed vigorously for 1 min followed by incubation at 65°C for 4 min. The RNA was then purified by phenol:chloroform extraction followed by ethanol precipitation. The concentration and the purity of the RNA were determined by measuring the absorbance at 260/280 nm. To further determine the quality of the RNA, both the ribosomal 28 S and 18 S were visually inspected on an agarose gel. For Figure 4A and B, total RNA was purified from approximately 100 worms using Direct-zolTM RNA MiniPrep kit (Zymo Research), as described by the manufacturer. cDNA was synthesized using total RNA and the SuperScript cDNA synthesis kit (Invitrogen, USA). Gene expression levels were then determined by real time PCR using the PowerSYBR® Green PCR Master Mix and StepOnePlus Real-Time PCR System (Applied Biosystems, USA). Relative gene expression was compared to actin as an internal control. Primers used are listed in Additional file 1: Table S7.

Western blots

All strains were allowed to grow at 15°C. For Additional file 1: Figure S1, approximately 100 L4 stage worms were collected for each of the transgenic strain in 10 to 15 mL of M9 buffer. Worms were then washed once with M9. An equal amount of SDS containing loading buffer was added to the worm pellets and samples were immediately boiled to lyse the worms. Samples were cooled, centrifuged briefly, and the supernatant was loaded onto the gel. For the western blot analyses in Figure 4C and D, equal amounts (10 mL) of worms were used. For each experiment, the lysates were resolved on a 10% polyacrylamide gel by SDS-PAGE. Proteins were transferred to a nitrocellulose membrane and probed with antibody against DAF-16 [78] (1:2,500 dilution). The membrane was then reprobed with anti -α-tubulin (1:8,000 dilution).

Growth assays

All the strains were grown at 15°C and five L4s were picked onto plates at 15°C. The plates were left undisturbed for 7 days and photos of each of the plates were taken using Nikon Coolpix995 camera to compare the growth of the strains.

Lifespan assays

All lifespan analyses were performed at 20°C. Strains were semi-synchronized by allowing gravid adults to lay eggs overnight and then removing the adult worms. Worms were grown for several days until they reached the young adult stage at 15°C. Approximately 150 young adult worms were transferred to five freshly seeded plates containing FuDR to a final concentration of 0.1 mg/mL [77]. Worms were then scored as dead or alive by tapping them with a platinum wire every 2 to 3 days. Worms that died from vulval bursting were censored. Day 1 of the lifespan was when worms were transferred to the FuDR plate. Statistical analyses were done using the standard chi-squared-based log rank test. Lifespans were also verified on non-FuDR NGM plates (Additional file 1: Figure S15).

DAF-16::GFP analysis

For measuring the nuclear:cytosolic ratio (Additional file 1: Figure S2 and Figure 3), worms were grown at 15°C until they reached young adult stage. Then, worms were mounted on glass slides in 50 mM sodium azide and visualized using Zeiss Axioscope 2+ microscope. Fluorescent images were taken using OpenLab.3.1.7 software with a Hamamatsu camera. For the quantification, only the region above intestinal cells in the pharynx was considered. Using ImageJ software, the fluorescence was quantified by measuring the pixel intensity in the nuclear versus cytosolic region. The ratio between the respective intensities was then calculated and plotted. This was repeated twice with approximately 14 to 15 worms in each assay.

RNAi screen

RNAi knockdown by feeding was performed essentially as described in Timmons et al. [79] using a library of clones representing 892 of the predicted 937 C. elegans transcription factors (MacNeil and Walhout, unpublished). Briefly, 50 μL of an overnight culture of HT115 bacteria carrying RNAi clones was used to inoculate 1 mL LB supplemented with 50 μg/mL ampicillin. Cultures were grown, with shaking, for 6 h at 37°C. Bacteria were pelleted and resuspended in 10% of the original volume. NGM plates containing 5 mM IPTG were seeded with concentrated bacteria and allowed to dry. Synchronized L1 larvae were used to seed prepared RNAi plates. Animals were visually examined for changes in GFP expression during the adult stage.

Availability of supporting data

The datasets- supporting figures for the results of this article are included as additional files.

Acknowledgements

We are grateful to members of the Tissenbaum laboratory for advice and critical comments on the manuscript and Nina Bhabhalia for technical support. We thank Marian Walhout for sharing the transcription regulator library prior to publication. Some of the strains were kindly provided by the Caenorhabditis Genetics Center which is funded by NIH Office of Research Infrastructure Programs (P40 OD010440). HAT is a William Randolph Hearst Investigator. This project was funded in part by grants from the National Institute of Aging (AG025891 and AG031237) and an endowment from the William Randolph Hearst Foundation.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.
The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.
The Creative Commons Public Domain Dedication waiver (https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

AB contributed to the conception and design, data collection and analysis, manuscript writing, critical revision, and final approval of the manuscript. ESK contributed to the concept and design, data collection and analysis, manuscript writing, and final approval of the manuscript. DC contributed to manuscript writing, data analysis, and final approval of the manuscript. HL, MJG, and LTM contributed to data collection and final approval of the manuscript. HAT contributed to the conception and design, financial support, manuscript writing, and final approval of manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Bluher M, Kahn BB, Kahn CR: Extended longevity in mice lacking the insulin receptor in adipose tissue. Science. 2003, 299: 572-574. 10.1126/science.1078223.CrossRefPubMed Bluher M, Kahn BB, Kahn CR: Extended longevity in mice lacking the insulin receptor in adipose tissue. Science. 2003, 299: 572-574. 10.1126/science.1078223.CrossRefPubMed
3.
Zurück zum Zitat Landis JN, Murphy CT: Integration of diverse inputs in the regulation of Caenorhabditis elegans DAF-16/FOXO. Dev Dyn. 2010, 239: 1405-1412.PubMed Landis JN, Murphy CT: Integration of diverse inputs in the regulation of Caenorhabditis elegans DAF-16/FOXO. Dev Dyn. 2010, 239: 1405-1412.PubMed
4.
Zurück zum Zitat Yen K, Narasimhan SD, Tissenbaum HA: DAF-16/Forkhead box O transcription factor: many paths to a single Fork(head) in the road. Antioxid Redox Signal. 2011, 14: 623-634. 10.1089/ars.2010.3490.PubMedCentralCrossRefPubMed Yen K, Narasimhan SD, Tissenbaum HA: DAF-16/Forkhead box O transcription factor: many paths to a single Fork(head) in the road. Antioxid Redox Signal. 2011, 14: 623-634. 10.1089/ars.2010.3490.PubMedCentralCrossRefPubMed
5.
Zurück zum Zitat Kimura KD, Tissenbaum HA, Liu Y, Ruvkun G: daf-2, an insulin receptor-like gene that regulates longevity and diapause in Caenorhabditis elegans. Science. 1997, 277: 942-946. 10.1126/science.277.5328.942.CrossRefPubMed Kimura KD, Tissenbaum HA, Liu Y, Ruvkun G: daf-2, an insulin receptor-like gene that regulates longevity and diapause in Caenorhabditis elegans. Science. 1997, 277: 942-946. 10.1126/science.277.5328.942.CrossRefPubMed
6.
Zurück zum Zitat Morris JZ, Tissenbaum HA, Ruvkun G: A phosphatidylinositol-3-OH kinase family member regulating longevity and diapause in Caenorhabditis elegans. Nature. 1996, 382: 536-539. 10.1038/382536a0.CrossRefPubMed Morris JZ, Tissenbaum HA, Ruvkun G: A phosphatidylinositol-3-OH kinase family member regulating longevity and diapause in Caenorhabditis elegans. Nature. 1996, 382: 536-539. 10.1038/382536a0.CrossRefPubMed
7.
Zurück zum Zitat Wolkow CA, Munoz MJ, Riddle DL, Ruvkun G: Insulin receptor substrate and p55 orthologous adaptor proteins function in the Caenorhabditis elegans daf-2/insulin-like signaling pathway. J Biol Chem. 2002, 277: 49591-49597. 10.1074/jbc.M207866200.CrossRefPubMed Wolkow CA, Munoz MJ, Riddle DL, Ruvkun G: Insulin receptor substrate and p55 orthologous adaptor proteins function in the Caenorhabditis elegans daf-2/insulin-like signaling pathway. J Biol Chem. 2002, 277: 49591-49597. 10.1074/jbc.M207866200.CrossRefPubMed
8.
Zurück zum Zitat Paradis S, Ailion M, Toker A, Thomas JH, Ruvkun G: A PDK1 homolog is necessary and sufficient to transduce AGE-1 PI3 kinase signals that regulate diapause in Caenorhabditis elegans. Genes Dev. 1999, 13: 1438-1452. 10.1101/gad.13.11.1438.PubMedCentralCrossRefPubMed Paradis S, Ailion M, Toker A, Thomas JH, Ruvkun G: A PDK1 homolog is necessary and sufficient to transduce AGE-1 PI3 kinase signals that regulate diapause in Caenorhabditis elegans. Genes Dev. 1999, 13: 1438-1452. 10.1101/gad.13.11.1438.PubMedCentralCrossRefPubMed
9.
Zurück zum Zitat Paradis S, Ruvkun G: Caenorhabditis elegans Akt/PKB transduces insulin receptor-like signals from AGE-1 PI3 kinase to the DAF-16 transcription factor. Genes Dev. 1998, 12: 2488-2498. 10.1101/gad.12.16.2488.PubMedCentralCrossRefPubMed Paradis S, Ruvkun G: Caenorhabditis elegans Akt/PKB transduces insulin receptor-like signals from AGE-1 PI3 kinase to the DAF-16 transcription factor. Genes Dev. 1998, 12: 2488-2498. 10.1101/gad.12.16.2488.PubMedCentralCrossRefPubMed
10.
Zurück zum Zitat Lin K, Dorman JB, Rodan A, Kenyon C: daf-16: An HNF-3/forkhead family member that can function to double the life-span of Caenorhabditis elegans. Science. 1997, 278: 1319-1322. 10.1126/science.278.5341.1319.CrossRefPubMed Lin K, Dorman JB, Rodan A, Kenyon C: daf-16: An HNF-3/forkhead family member that can function to double the life-span of Caenorhabditis elegans. Science. 1997, 278: 1319-1322. 10.1126/science.278.5341.1319.CrossRefPubMed
11.
Zurück zum Zitat Ogg S, Paradis S, Gottlieb S, Patterson GI, Lee L, Tissenbaum HA, Ruvkun G: The Fork head transcription factor DAF-16 transduces insulin-like metabolic and longevity signals in C. elegans. Nature. 1997, 389: 994-999. 10.1038/40194.CrossRefPubMed Ogg S, Paradis S, Gottlieb S, Patterson GI, Lee L, Tissenbaum HA, Ruvkun G: The Fork head transcription factor DAF-16 transduces insulin-like metabolic and longevity signals in C. elegans. Nature. 1997, 389: 994-999. 10.1038/40194.CrossRefPubMed
12.
Zurück zum Zitat Hertweck M, Gobel C, Baumeister R: C. elegans SGK-1 is the critical component in the Akt/PKB kinase complex to control stress response and life span. Dev Cell. 2004, 6: 577-588. 10.1016/S1534-5807(04)00095-4.CrossRefPubMed Hertweck M, Gobel C, Baumeister R: C. elegans SGK-1 is the critical component in the Akt/PKB kinase complex to control stress response and life span. Dev Cell. 2004, 6: 577-588. 10.1016/S1534-5807(04)00095-4.CrossRefPubMed
14.
Zurück zum Zitat Furuyama T, Nakazawa T, Nakano I, Mori N: Identification of the differential distribution patterns of mRNAs and consensus binding sequences for mouse DAF-16 homologues. Biochem J. 2000, 349: 629-634. 10.1042/0264-6021:3490629.PubMedCentralCrossRefPubMed Furuyama T, Nakazawa T, Nakano I, Mori N: Identification of the differential distribution patterns of mRNAs and consensus binding sequences for mouse DAF-16 homologues. Biochem J. 2000, 349: 629-634. 10.1042/0264-6021:3490629.PubMedCentralCrossRefPubMed
15.
Zurück zum Zitat Hosaka T, Biggs WH, Tieu D, Boyer AD, Varki NM, Cavenee WK, Arden KC: Disruption of forkhead transcription factor (FOXO) family members in mice reveals their functional diversification. Proc Natl Acad Sci U S A. 2004, 101: 2975-2980. 10.1073/pnas.0400093101.PubMedCentralCrossRefPubMed Hosaka T, Biggs WH, Tieu D, Boyer AD, Varki NM, Cavenee WK, Arden KC: Disruption of forkhead transcription factor (FOXO) family members in mice reveals their functional diversification. Proc Natl Acad Sci U S A. 2004, 101: 2975-2980. 10.1073/pnas.0400093101.PubMedCentralCrossRefPubMed
16.
Zurück zum Zitat Castrillon DH, Miao L, Kollipara R, Horner JW, DePinho RA: Suppression of ovarian follicle activation in mice by the transcription factor Foxo3a. Science. 2003, 301: 215-218. 10.1126/science.1086336.CrossRefPubMed Castrillon DH, Miao L, Kollipara R, Horner JW, DePinho RA: Suppression of ovarian follicle activation in mice by the transcription factor Foxo3a. Science. 2003, 301: 215-218. 10.1126/science.1086336.CrossRefPubMed
17.
Zurück zum Zitat Paik JH, Kollipara R, Chu G, Ji H, Xiao Y, Ding Z, Miao L, Tothova Z, Horner JW, Carrasco DR, Jiang S, Gilliland DG, Chin L, Wong WH, Castrillon DH: FoxOs are lineage-restricted redundant tumor suppressors and regulate endothelial cell homeostasis. Cell. 2007, 128: 309-323. 10.1016/j.cell.2006.12.029.PubMedCentralCrossRefPubMed Paik JH, Kollipara R, Chu G, Ji H, Xiao Y, Ding Z, Miao L, Tothova Z, Horner JW, Carrasco DR, Jiang S, Gilliland DG, Chin L, Wong WH, Castrillon DH: FoxOs are lineage-restricted redundant tumor suppressors and regulate endothelial cell homeostasis. Cell. 2007, 128: 309-323. 10.1016/j.cell.2006.12.029.PubMedCentralCrossRefPubMed
18.
Zurück zum Zitat Kim DH, Perdomo G, Zhang T, Slusher S, Lee S, Phillips BE, Fan Y, Giannoukakis N, Gramignoli R, Strom S, Ringquist S, Dong HH: FoxO6 integrates insulin signaling with gluconeogenesis in the liver. Diabetes. 2011, 60: 2763-2774. 10.2337/db11-0548.PubMedCentralCrossRefPubMed Kim DH, Perdomo G, Zhang T, Slusher S, Lee S, Phillips BE, Fan Y, Giannoukakis N, Gramignoli R, Strom S, Ringquist S, Dong HH: FoxO6 integrates insulin signaling with gluconeogenesis in the liver. Diabetes. 2011, 60: 2763-2774. 10.2337/db11-0548.PubMedCentralCrossRefPubMed
19.
Zurück zum Zitat Lee RY, Hench J, Ruvkun G: Regulation of C. elegans DAF-16 and its human ortholog FKHRL1 by the daf-2 insulin-like signaling pathway. Curr Biol. 2001, 11: 1950-1957. 10.1016/S0960-9822(01)00595-4.CrossRefPubMed Lee RY, Hench J, Ruvkun G: Regulation of C. elegans DAF-16 and its human ortholog FKHRL1 by the daf-2 insulin-like signaling pathway. Curr Biol. 2001, 11: 1950-1957. 10.1016/S0960-9822(01)00595-4.CrossRefPubMed
20.
Zurück zum Zitat Lin K, Hsin H, Libina N, Kenyon C: Regulation of the Caenorhabditis elegans longevity protein DAF-16 by insulin/IGF-1 and germline signaling. Nat Genet. 2001, 28: 139-145. 10.1038/88850.CrossRefPubMed Lin K, Hsin H, Libina N, Kenyon C: Regulation of the Caenorhabditis elegans longevity protein DAF-16 by insulin/IGF-1 and germline signaling. Nat Genet. 2001, 28: 139-145. 10.1038/88850.CrossRefPubMed
21.
22.
Zurück zum Zitat Essers MA, Weijzen S, de Vries-Smits AM, Saarloos I, de Ruiter ND, Bos JL, Burgering BM: FOXO transcription factor activation by oxidative stress mediated by the small GTPase Ral and JNK. EMBO J. 2004, 23: 4802-4812. 10.1038/sj.emboj.7600476.PubMedCentralCrossRefPubMed Essers MA, Weijzen S, de Vries-Smits AM, Saarloos I, de Ruiter ND, Bos JL, Burgering BM: FOXO transcription factor activation by oxidative stress mediated by the small GTPase Ral and JNK. EMBO J. 2004, 23: 4802-4812. 10.1038/sj.emboj.7600476.PubMedCentralCrossRefPubMed
23.
Zurück zum Zitat Oh SW, Mukhopadhyay A, Svrzikapa N, Jiang F, Davis RJ, Tissenbaum HA: JNK regulates lifespan in Caenorhabditis elegans by modulating nuclear translocation of forkhead transcription factor/DAF-16. Proc Natl Acad Sci USA. 2005, 102: 4494-4499. 10.1073/pnas.0500749102.PubMedCentralCrossRefPubMed Oh SW, Mukhopadhyay A, Svrzikapa N, Jiang F, Davis RJ, Tissenbaum HA: JNK regulates lifespan in Caenorhabditis elegans by modulating nuclear translocation of forkhead transcription factor/DAF-16. Proc Natl Acad Sci USA. 2005, 102: 4494-4499. 10.1073/pnas.0500749102.PubMedCentralCrossRefPubMed
24.
Zurück zum Zitat Wang MC, Bohmann D, Jasper H: JNK extends life span and limits growth by antagonizing cellular and organism-wide responses to insulin signaling. Cell. 2005, 121: 115-125. 10.1016/j.cell.2005.02.030.CrossRefPubMed Wang MC, Bohmann D, Jasper H: JNK extends life span and limits growth by antagonizing cellular and organism-wide responses to insulin signaling. Cell. 2005, 121: 115-125. 10.1016/j.cell.2005.02.030.CrossRefPubMed
25.
Zurück zum Zitat Lehtinen MK, Yuan Z, Boag PR, Yang Y, Villen J, Becker EB, DiBacco S, de la Iglesia N, Gygi S, Blackwell TK, Bonni A: A conserved MST-FOXO signaling pathway mediates oxidative-stress responses and extends life span. Cell. 2006, 125: 987-1001. 10.1016/j.cell.2006.03.046.CrossRefPubMed Lehtinen MK, Yuan Z, Boag PR, Yang Y, Villen J, Becker EB, DiBacco S, de la Iglesia N, Gygi S, Blackwell TK, Bonni A: A conserved MST-FOXO signaling pathway mediates oxidative-stress responses and extends life span. Cell. 2006, 125: 987-1001. 10.1016/j.cell.2006.03.046.CrossRefPubMed
26.
Zurück zum Zitat Curtis R, O’Connor G, DiStefano PS: Aging networks in Caenorhabditis elegans: AMP-activated protein kinase (aak-2) links multiple aging and metabolism pathways. Aging Cell. 2006, 5: 119-126. 10.1111/j.1474-9726.2006.00205.x.CrossRefPubMed Curtis R, O’Connor G, DiStefano PS: Aging networks in Caenorhabditis elegans: AMP-activated protein kinase (aak-2) links multiple aging and metabolism pathways. Aging Cell. 2006, 5: 119-126. 10.1111/j.1474-9726.2006.00205.x.CrossRefPubMed
27.
Zurück zum Zitat Greer EL, Dowlatshahi D, Banko MR, Villen J, Hoang K, Blanchard D, Gygi SP, Brunet A: An AMPK-FOXO pathway mediates longevity induced by a novel method of dietary restriction in C. elegans. Curr Biol. 2007, 17: 1646-1656. 10.1016/j.cub.2007.08.047.PubMedCentralCrossRefPubMed Greer EL, Dowlatshahi D, Banko MR, Villen J, Hoang K, Blanchard D, Gygi SP, Brunet A: An AMPK-FOXO pathway mediates longevity induced by a novel method of dietary restriction in C. elegans. Curr Biol. 2007, 17: 1646-1656. 10.1016/j.cub.2007.08.047.PubMedCentralCrossRefPubMed
28.
Zurück zum Zitat Wolff S, Ma H, Burch D, Maciel GA, Hunter T, Dillin A: SMK-1, an essential regulator of DAF-16-mediated longevity. Cell. 2006, 124: 1039-1053. 10.1016/j.cell.2005.12.042.CrossRefPubMed Wolff S, Ma H, Burch D, Maciel GA, Hunter T, Dillin A: SMK-1, an essential regulator of DAF-16-mediated longevity. Cell. 2006, 124: 1039-1053. 10.1016/j.cell.2005.12.042.CrossRefPubMed
29.
Zurück zum Zitat Berdichevsky A, Viswanathan M, Horvitz HR, Guarente L: C. elegans SIR-2.1 interacts with 14-3-3 proteins to activate DAF-16 and extend life span. Cell. 2006, 125: 1165-1177. 10.1016/j.cell.2006.04.036.CrossRefPubMed Berdichevsky A, Viswanathan M, Horvitz HR, Guarente L: C. elegans SIR-2.1 interacts with 14-3-3 proteins to activate DAF-16 and extend life span. Cell. 2006, 125: 1165-1177. 10.1016/j.cell.2006.04.036.CrossRefPubMed
30.
Zurück zum Zitat Wang Y, Oh SW, Deplancke B, Luo J, Walhout AJ, Tissenbaum HA: C. elegans 14-3-3 proteins regulate life span and interact with SIR-2.1 and DAF-16/FOXO. Mech Ageing Dev. 2006, 127: 741-747. 10.1016/j.mad.2006.05.005.CrossRefPubMed Wang Y, Oh SW, Deplancke B, Luo J, Walhout AJ, Tissenbaum HA: C. elegans 14-3-3 proteins regulate life span and interact with SIR-2.1 and DAF-16/FOXO. Mech Ageing Dev. 2006, 127: 741-747. 10.1016/j.mad.2006.05.005.CrossRefPubMed
31.
Zurück zum Zitat Fukushige T, Hawkins MG, McGhee JD: The GATA-factor elt-2 is essential for formation of the Caenorhabditis elegans intestine. Dev Biol. 1998, 198: 286-302.PubMed Fukushige T, Hawkins MG, McGhee JD: The GATA-factor elt-2 is essential for formation of the Caenorhabditis elegans intestine. Dev Biol. 1998, 198: 286-302.PubMed
32.
Zurück zum Zitat McGhee JD, Fukushige T, Krause MW, Minnema SE, Goszczynski B, Gaudet J, Kohara Y, Bossinger O, Zhao Y, Khattra J, Hirst M, Jones SJ, Marra MA, Ruzanov P, Warner A, Zapf R, Moerman DG, Kalb JM: ELT-2 is the predominant transcription factor controlling differentiation and function of the C. elegans intestine, from embryo to adult. Dev Biol. 2009, 327: 551-565. 10.1016/j.ydbio.2008.11.034.PubMedCentralCrossRefPubMed McGhee JD, Fukushige T, Krause MW, Minnema SE, Goszczynski B, Gaudet J, Kohara Y, Bossinger O, Zhao Y, Khattra J, Hirst M, Jones SJ, Marra MA, Ruzanov P, Warner A, Zapf R, Moerman DG, Kalb JM: ELT-2 is the predominant transcription factor controlling differentiation and function of the C. elegans intestine, from embryo to adult. Dev Biol. 2009, 327: 551-565. 10.1016/j.ydbio.2008.11.034.PubMedCentralCrossRefPubMed
33.
Zurück zum Zitat Martens JA, Winston F: Recent advances in understanding chromatin remodeling by Swi/Snf complexes. Curr Opin Genet Dev. 2003, 13: 136-142. 10.1016/S0959-437X(03)00022-4.CrossRefPubMed Martens JA, Winston F: Recent advances in understanding chromatin remodeling by Swi/Snf complexes. Curr Opin Genet Dev. 2003, 13: 136-142. 10.1016/S0959-437X(03)00022-4.CrossRefPubMed
34.
Zurück zum Zitat Phelan ML, Sif S, Narlikar GJ, Kingston RE: Reconstitution of a core chromatin remodeling complex from SWI/SNF subunits. Mol Cell. 1999, 3: 247-253. 10.1016/S1097-2765(00)80315-9.CrossRefPubMed Phelan ML, Sif S, Narlikar GJ, Kingston RE: Reconstitution of a core chromatin remodeling complex from SWI/SNF subunits. Mol Cell. 1999, 3: 247-253. 10.1016/S1097-2765(00)80315-9.CrossRefPubMed
35.
Zurück zum Zitat Gilchrist MJ, Zorn AM, Voigt J, Smith JC, Papalopulu N, Amaya E: Defining a large set of full-length clones from a Xenopus tropicalis EST project. Dev Biol. 2004, 271: 498-516. 10.1016/j.ydbio.2004.04.023.CrossRefPubMed Gilchrist MJ, Zorn AM, Voigt J, Smith JC, Papalopulu N, Amaya E: Defining a large set of full-length clones from a Xenopus tropicalis EST project. Dev Biol. 2004, 271: 498-516. 10.1016/j.ydbio.2004.04.023.CrossRefPubMed
36.
Zurück zum Zitat Gorodkin J, Cirera S, Hedegaard J, Gilchrist MJ, Panitz F, Jorgensen C, Scheibye-Knudsen K, Arvin T, Lumholdt S, Sawera M, Green T, Nielsen BJ, Havgaard JH, Rosenkilde C, Wang J, Li H, Li R, Liu B, Hu S, Dong W, Li W, Yu J, Wang J, Staefeldt HH, Wernersson R, Madsen LB, Thomsen B, Hornshoj H, Bujie Z, Wang X, et al: Porcine transcriptome analysis based on 97 non-normalized cDNA libraries and assembly of 1,021,891 expressed sequence tags. Genome Biol. 2007, 8: R45-10.1186/gb-2007-8-4-r45.PubMedCentralCrossRefPubMed Gorodkin J, Cirera S, Hedegaard J, Gilchrist MJ, Panitz F, Jorgensen C, Scheibye-Knudsen K, Arvin T, Lumholdt S, Sawera M, Green T, Nielsen BJ, Havgaard JH, Rosenkilde C, Wang J, Li H, Li R, Liu B, Hu S, Dong W, Li W, Yu J, Wang J, Staefeldt HH, Wernersson R, Madsen LB, Thomsen B, Hornshoj H, Bujie Z, Wang X, et al: Porcine transcriptome analysis based on 97 non-normalized cDNA libraries and assembly of 1,021,891 expressed sequence tags. Genome Biol. 2007, 8: R45-10.1186/gb-2007-8-4-r45.PubMedCentralCrossRefPubMed
37.
Zurück zum Zitat Gu W, Lee HC, Chaves D, Youngman EM, Pazour GJ, Conte D Jr, Mello CC: CapSeq and CIP-TAP identify Pol II start sites and reveal capped small RNAs as C. elegans piRNA precursors. Cell. 2012, 151: 1488-1500. 10.1016/j.cell.2012.11.023.PubMedCentralCrossRefPubMed Gu W, Lee HC, Chaves D, Youngman EM, Pazour GJ, Conte D Jr, Mello CC: CapSeq and CIP-TAP identify Pol II start sites and reveal capped small RNAs as C. elegans piRNA precursors. Cell. 2012, 151: 1488-1500. 10.1016/j.cell.2012.11.023.PubMedCentralCrossRefPubMed
38.
Zurück zum Zitat Stadler M, Artiles K, Pak J, Fire A: Contributions of mRNA abundance, ribosome loading, and post- or peri-translational effects to temporal repression of C. elegans heterochronic miRNA targets. Genome Res. 2012, 22: 2418-2426. 10.1101/gr.136515.111.PubMedCentralCrossRefPubMed Stadler M, Artiles K, Pak J, Fire A: Contributions of mRNA abundance, ribosome loading, and post- or peri-translational effects to temporal repression of C. elegans heterochronic miRNA targets. Genome Res. 2012, 22: 2418-2426. 10.1101/gr.136515.111.PubMedCentralCrossRefPubMed
39.
Zurück zum Zitat Henderson ST, Johnson TE: daf-16 integrates developmental and environmental inputs to mediate aging in the nematode Caenorhabditis elegans. Curr Biol. 2001, 11: 1975-1980. 10.1016/S0960-9822(01)00594-2.CrossRefPubMed Henderson ST, Johnson TE: daf-16 integrates developmental and environmental inputs to mediate aging in the nematode Caenorhabditis elegans. Curr Biol. 2001, 11: 1975-1980. 10.1016/S0960-9822(01)00594-2.CrossRefPubMed
40.
Zurück zum Zitat Padmanabhan S, Mukhopadhyay A, Narasimhan S, Tesz G, Czech MP, Tissenbaum HA: A PP2A Regulatory subunit regulates C. elegans Insulin/IGF-1 signaling by modulating AKT-1 phosphorylation. Cell. 2009, 136: 939-951. 10.1016/j.cell.2009.01.025.PubMedCentralCrossRefPubMed Padmanabhan S, Mukhopadhyay A, Narasimhan S, Tesz G, Czech MP, Tissenbaum HA: A PP2A Regulatory subunit regulates C. elegans Insulin/IGF-1 signaling by modulating AKT-1 phosphorylation. Cell. 2009, 136: 939-951. 10.1016/j.cell.2009.01.025.PubMedCentralCrossRefPubMed
41.
Zurück zum Zitat Gems D, Sutton AJ, Sundermeyer ML, Albert PS, King KV, Edgley ML, Larsen PL, Riddle DL: Two pleiotropic classes of daf-2 mutation affect larval arrest, adult behavior, reproduction and longevity in Caenorhabditis elegans. Genetics. 1998, 150: 129-155.PubMedCentralPubMed Gems D, Sutton AJ, Sundermeyer ML, Albert PS, King KV, Edgley ML, Larsen PL, Riddle DL: Two pleiotropic classes of daf-2 mutation affect larval arrest, adult behavior, reproduction and longevity in Caenorhabditis elegans. Genetics. 1998, 150: 129-155.PubMedCentralPubMed
42.
Zurück zum Zitat Wightman B, Burglin TR, Gatto J, Arasu P, Ruvkun G: Negative regulatory sequences in the lin-14 3'-untranslated region are necessary to generate a temporal switch during Caenorhabditis elegans development. Genes Dev. 1991, 5: 1813-1824. 10.1101/gad.5.10.1813.CrossRefPubMed Wightman B, Burglin TR, Gatto J, Arasu P, Ruvkun G: Negative regulatory sequences in the lin-14 3'-untranslated region are necessary to generate a temporal switch during Caenorhabditis elegans development. Genes Dev. 1991, 5: 1813-1824. 10.1101/gad.5.10.1813.CrossRefPubMed
43.
Zurück zum Zitat Libina N, Berman JR, Kenyon C: Tissue-specific activities of C. elegans DAF-16 in the regulation of lifespan. Cell. 2003, 115: 489-502. 10.1016/S0092-8674(03)00889-4.CrossRefPubMed Libina N, Berman JR, Kenyon C: Tissue-specific activities of C. elegans DAF-16 in the regulation of lifespan. Cell. 2003, 115: 489-502. 10.1016/S0092-8674(03)00889-4.CrossRefPubMed
45.
Zurück zum Zitat Racher H, Hansen D: Translational control in the C. elegans hermaphrodite germ line. Genome. 2010, 53: 83-102. 10.1139/G09-090.CrossRefPubMed Racher H, Hansen D: Translational control in the C. elegans hermaphrodite germ line. Genome. 2010, 53: 83-102. 10.1139/G09-090.CrossRefPubMed
46.
Zurück zum Zitat Kerry S, Tekippe M, Gaddis NC, Aballay A: GATA transcription factor required for immunity to bacterial and fungal pathogens. PLoS One. 2006, 1: e77-10.1371/journal.pone.0000077.PubMedCentralCrossRefPubMed Kerry S, Tekippe M, Gaddis NC, Aballay A: GATA transcription factor required for immunity to bacterial and fungal pathogens. PLoS One. 2006, 1: e77-10.1371/journal.pone.0000077.PubMedCentralCrossRefPubMed
47.
Zurück zum Zitat Zhang P, Judy M, Lee SJ, Kenyon C: Direct and indirect gene regulation by a life-extending FOXO protein in C. elegans: roles for GATA factors and lipid gene regulators. Cell Metab. 2013, 17: 85-100. 10.1016/j.cmet.2012.12.013.PubMedCentralCrossRefPubMed Zhang P, Judy M, Lee SJ, Kenyon C: Direct and indirect gene regulation by a life-extending FOXO protein in C. elegans: roles for GATA factors and lipid gene regulators. Cell Metab. 2013, 17: 85-100. 10.1016/j.cmet.2012.12.013.PubMedCentralCrossRefPubMed
48.
Zurück zum Zitat Budovskaya YV, Wu K, Southworth LK, Jiang M, Tedesco P, Johnson TE, Kim SK: An elt-3/elt-5/elt-6 GATA transcription circuit guides aging in C. elegans. Cell. 2008, 134: 291-303. 10.1016/j.cell.2008.05.044.CrossRefPubMed Budovskaya YV, Wu K, Southworth LK, Jiang M, Tedesco P, Johnson TE, Kim SK: An elt-3/elt-5/elt-6 GATA transcription circuit guides aging in C. elegans. Cell. 2008, 134: 291-303. 10.1016/j.cell.2008.05.044.CrossRefPubMed
49.
Zurück zum Zitat Tonsaker T, Pratt RM, McGhee JD: Re-evaluating the role of ELT-3 in a GATA transcription factor circuit proposed to guide aging in C. elegans. Mech Ageing Dev. 2012, 133: 50-53. 10.1016/j.mad.2011.09.006.CrossRefPubMed Tonsaker T, Pratt RM, McGhee JD: Re-evaluating the role of ELT-3 in a GATA transcription factor circuit proposed to guide aging in C. elegans. Mech Ageing Dev. 2012, 133: 50-53. 10.1016/j.mad.2011.09.006.CrossRefPubMed
50.
Zurück zum Zitat Euskirchen G, Auerbach RK, Snyder M: SWI/SNF chromatin-remodeling factors: multiscale analyses and diverse functions. J Biol Chem. 2012, 287: 30897-30905. 10.1074/jbc.R111.309302.PubMedCentralCrossRefPubMed Euskirchen G, Auerbach RK, Snyder M: SWI/SNF chromatin-remodeling factors: multiscale analyses and diverse functions. J Biol Chem. 2012, 287: 30897-30905. 10.1074/jbc.R111.309302.PubMedCentralCrossRefPubMed
51.
Zurück zum Zitat Sawa H, Kouike H, Okano H: Components of the SWI/SNF complex are required for asymmetric cell division in C. elegans. Mol Cell. 2000, 6: 617-624. 10.1016/S1097-2765(00)00060-5.CrossRefPubMed Sawa H, Kouike H, Okano H: Components of the SWI/SNF complex are required for asymmetric cell division in C. elegans. Mol Cell. 2000, 6: 617-624. 10.1016/S1097-2765(00)00060-5.CrossRefPubMed
52.
Zurück zum Zitat Hayes GD, Riedel CG, Ruvkun G: The Caenorhabditis elegans SOMI-1 zinc finger protein and SWI/SNF promote regulation of development by the mir-84 microRNA. Genes Dev. 2011, 25: 2079-2092. 10.1101/gad.17153811.PubMedCentralCrossRefPubMed Hayes GD, Riedel CG, Ruvkun G: The Caenorhabditis elegans SOMI-1 zinc finger protein and SWI/SNF promote regulation of development by the mir-84 microRNA. Genes Dev. 2011, 25: 2079-2092. 10.1101/gad.17153811.PubMedCentralCrossRefPubMed
53.
Zurück zum Zitat Cui M, Fay DS, Han M: lin-35/Rb cooperates with the SWI/SNF complex to control Caenorhabditis elegans larval development. Genetics. 2004, 167: 1177-1185. 10.1534/genetics.103.024554.PubMedCentralCrossRefPubMed Cui M, Fay DS, Han M: lin-35/Rb cooperates with the SWI/SNF complex to control Caenorhabditis elegans larval development. Genetics. 2004, 167: 1177-1185. 10.1534/genetics.103.024554.PubMedCentralCrossRefPubMed
54.
Zurück zum Zitat Riedel CG, Dowen RH, Lourenco GF, Kirienko NV, Heimbucher T, West JA, Bowman SK, Kingston RE, Dillin A, Asara JM, Ruvkun G: DAF-16 employs the chromatin remodeller SWI/SNF to promote stress resistance and longevity. Nat Cell Biol. 2013, 15: 491-501. 10.1038/ncb2720.PubMedCentralCrossRefPubMed Riedel CG, Dowen RH, Lourenco GF, Kirienko NV, Heimbucher T, West JA, Bowman SK, Kingston RE, Dillin A, Asara JM, Ruvkun G: DAF-16 employs the chromatin remodeller SWI/SNF to promote stress resistance and longevity. Nat Cell Biol. 2013, 15: 491-501. 10.1038/ncb2720.PubMedCentralCrossRefPubMed
55.
Zurück zum Zitat Dillin A, Crawford DK, Kenyon C: Timing requirements for insulin/IGF-1 signaling in C. elegans. Science. 2002, 298: 830-834. 10.1126/science.1074240.CrossRefPubMed Dillin A, Crawford DK, Kenyon C: Timing requirements for insulin/IGF-1 signaling in C. elegans. Science. 2002, 298: 830-834. 10.1126/science.1074240.CrossRefPubMed
56.
Zurück zum Zitat Huang P, Zhou ZQ, Huang RH, Zhou B, Wei QW, Shi FX: Age-dependent expression of forkhead box O proteins in the duodenum of rats. J Zhejiang Univ Sci B. 2011, 12: 730-735.PubMedCentralCrossRefPubMed Huang P, Zhou ZQ, Huang RH, Zhou B, Wei QW, Shi FX: Age-dependent expression of forkhead box O proteins in the duodenum of rats. J Zhejiang Univ Sci B. 2011, 12: 730-735.PubMedCentralCrossRefPubMed
57.
Zurück zum Zitat Buford TW, Cooke MB, Shelmadine BD, Hudson GM, Redd LL, Willoughby DS: Differential gene expression of FoxO1, ID1, and ID3 between young and older men and associations with muscle mass and function. Aging Clin Exp Res. 2011, 23: 170-174. 10.1007/BF03324957.PubMedCentralCrossRefPubMed Buford TW, Cooke MB, Shelmadine BD, Hudson GM, Redd LL, Willoughby DS: Differential gene expression of FoxO1, ID1, and ID3 between young and older men and associations with muscle mass and function. Aging Clin Exp Res. 2011, 23: 170-174. 10.1007/BF03324957.PubMedCentralCrossRefPubMed
58.
Zurück zum Zitat Barbieri M, Bonafe M, Franceschi C, Paolisso G: Insulin/IGF-I-signaling pathway: an evolutionarily conserved mechanism of longevity from yeast to humans. Am J Physiol Endocrinol Metab. 2003, 285: E1064-E1071.CrossRefPubMed Barbieri M, Bonafe M, Franceschi C, Paolisso G: Insulin/IGF-I-signaling pathway: an evolutionarily conserved mechanism of longevity from yeast to humans. Am J Physiol Endocrinol Metab. 2003, 285: E1064-E1071.CrossRefPubMed
59.
Zurück zum Zitat Murphy CT, McCarroll SA, Bargmann CI, Fraser A, Kamath RS, Ahringer J, Li H, Kenyon C: Genes that act downstream of DAF-16 to influence the lifespan of Caenorhabditis elegans. Nature. 2003, 424: 277-283. 10.1038/nature01789.CrossRefPubMed Murphy CT, McCarroll SA, Bargmann CI, Fraser A, Kamath RS, Ahringer J, Li H, Kenyon C: Genes that act downstream of DAF-16 to influence the lifespan of Caenorhabditis elegans. Nature. 2003, 424: 277-283. 10.1038/nature01789.CrossRefPubMed
61.
Zurück zum Zitat Greer EL, Maures TJ, Hauswirth AG, Green EM, Leeman DS, Maro GS, Han S, Banko MR, Gozani O, Brunet A: Members of the H3K4 trimethylation complex regulate lifespan in a germline-dependent manner in C. elegans. Nature. 2010, 466: 383-387. 10.1038/nature09195.PubMedCentralCrossRefPubMed Greer EL, Maures TJ, Hauswirth AG, Green EM, Leeman DS, Maro GS, Han S, Banko MR, Gozani O, Brunet A: Members of the H3K4 trimethylation complex regulate lifespan in a germline-dependent manner in C. elegans. Nature. 2010, 466: 383-387. 10.1038/nature09195.PubMedCentralCrossRefPubMed
62.
Zurück zum Zitat Greer EL, Maures TJ, Ucar D, Hauswirth AG, Mancini E, Lim JP, Benayoun BA, Shi Y, Brunet A: Transgenerational epigenetic inheritance of longevity in Caenorhabditis elegans. Nature. 2011, 479: 365-371. 10.1038/nature10572.PubMedCentralCrossRefPubMed Greer EL, Maures TJ, Ucar D, Hauswirth AG, Mancini E, Lim JP, Benayoun BA, Shi Y, Brunet A: Transgenerational epigenetic inheritance of longevity in Caenorhabditis elegans. Nature. 2011, 479: 365-371. 10.1038/nature10572.PubMedCentralCrossRefPubMed
63.
Zurück zum Zitat Jin C, Li J, Green CD, Yu X, Tang X, Han D, Xian B, Wang D, Huang X, Cao X, Yan Z, Hou L, Shukeir N, Khaitovich P, Chen CD, Zhang H, Jenuwein T: Histone demethylase UTX-1 regulates C. elegans life span by targeting the insulin/IGF-1 signaling pathway. Cell Metab. 2011, 14: 161-172. 10.1016/j.cmet.2011.07.001.CrossRefPubMed Jin C, Li J, Green CD, Yu X, Tang X, Han D, Xian B, Wang D, Huang X, Cao X, Yan Z, Hou L, Shukeir N, Khaitovich P, Chen CD, Zhang H, Jenuwein T: Histone demethylase UTX-1 regulates C. elegans life span by targeting the insulin/IGF-1 signaling pathway. Cell Metab. 2011, 14: 161-172. 10.1016/j.cmet.2011.07.001.CrossRefPubMed
64.
Zurück zum Zitat Maures TJ, Greer EL, Hauswirth AG, Brunet A: The H3K27 demethylase UTX-1 regulates C. elegans lifespan in a germline-independent, insulin-dependent manner. Aging Cell. 2011, 10: 980-990. 10.1111/j.1474-9726.2011.00738.x.PubMedCentralCrossRefPubMed Maures TJ, Greer EL, Hauswirth AG, Brunet A: The H3K27 demethylase UTX-1 regulates C. elegans lifespan in a germline-independent, insulin-dependent manner. Aging Cell. 2011, 10: 980-990. 10.1111/j.1474-9726.2011.00738.x.PubMedCentralCrossRefPubMed
65.
Zurück zum Zitat Debril MB, Gelman L, Fayard E, Annicotte JS, Rocchi S, Auwerx J: Transcription factors and nuclear receptors interact with the SWI/SNF complex through the BAF60c subunit. J Biol Chem. 2004, 279: 16677-16686. 10.1074/jbc.M312288200.CrossRefPubMed Debril MB, Gelman L, Fayard E, Annicotte JS, Rocchi S, Auwerx J: Transcription factors and nuclear receptors interact with the SWI/SNF complex through the BAF60c subunit. J Biol Chem. 2004, 279: 16677-16686. 10.1074/jbc.M312288200.CrossRefPubMed
67.
Zurück zum Zitat Lickert H, Takeuchi JK, Von Both I, Walls JR, McAuliffe F, Adamson SL, Henkelman RM, Wrana JL, Rossant J, Bruneau BG: Baf60c is essential for function of BAF chromatin remodelling complexes in heart development. Nature. 2004, 432: 107-112. 10.1038/nature03071.CrossRefPubMed Lickert H, Takeuchi JK, Von Both I, Walls JR, McAuliffe F, Adamson SL, Henkelman RM, Wrana JL, Rossant J, Bruneau BG: Baf60c is essential for function of BAF chromatin remodelling complexes in heart development. Nature. 2004, 432: 107-112. 10.1038/nature03071.CrossRefPubMed
68.
Zurück zum Zitat Anselmi CV, Malovini A, Roncarati R, Novelli V, Villa F, Condorelli G, Bellazzi R, Puca AA: Association of the FOXO3A locus with extreme longevity in a southern Italian centenarian study. Rejuvenation Res. 2009, 12: 95-104. 10.1089/rej.2008.0827.CrossRefPubMed Anselmi CV, Malovini A, Roncarati R, Novelli V, Villa F, Condorelli G, Bellazzi R, Puca AA: Association of the FOXO3A locus with extreme longevity in a southern Italian centenarian study. Rejuvenation Res. 2009, 12: 95-104. 10.1089/rej.2008.0827.CrossRefPubMed
69.
Zurück zum Zitat Flachsbart F, Caliebe A, Kleindorp R, Blanche H, von Eller-Eberstein H, Nikolaus S, Schreiber S, Nebel A: Association of FOXO3A variation with human longevity confirmed in German centenarians. Proc Natl Acad Sci USA. 2009, 106: 2700-2705. 10.1073/pnas.0809594106.PubMedCentralCrossRefPubMed Flachsbart F, Caliebe A, Kleindorp R, Blanche H, von Eller-Eberstein H, Nikolaus S, Schreiber S, Nebel A: Association of FOXO3A variation with human longevity confirmed in German centenarians. Proc Natl Acad Sci USA. 2009, 106: 2700-2705. 10.1073/pnas.0809594106.PubMedCentralCrossRefPubMed
70.
Zurück zum Zitat Li Y, Wang WJ, Cao H, Lu J, Wu C, Hu FY, Guo J, Zhao L, Yang F, Zhang YX, Li W, Zheng GY, Cui H, Chen X, Zhu Z, He H, Dong B, Mo X, Zeng Y, Tian XL: Genetic association of FOXO1A and FOXO3A with longevity trait in Han Chinese populations. Hum Mol Genet. 2009, 18: 4897-4904. 10.1093/hmg/ddp459.PubMedCentralCrossRefPubMed Li Y, Wang WJ, Cao H, Lu J, Wu C, Hu FY, Guo J, Zhao L, Yang F, Zhang YX, Li W, Zheng GY, Cui H, Chen X, Zhu Z, He H, Dong B, Mo X, Zeng Y, Tian XL: Genetic association of FOXO1A and FOXO3A with longevity trait in Han Chinese populations. Hum Mol Genet. 2009, 18: 4897-4904. 10.1093/hmg/ddp459.PubMedCentralCrossRefPubMed
71.
Zurück zum Zitat Malovini A, Illario M, Iaccarino G, Villa F, Ferrario A, Roncarati R, Anselmi CV, Novelli V, Cipolletta E, Leggiero E, Orro A, Rusciano MR, Milanesi L, Maione AS, Condorelli G, Bellazzi R, Puca AA: Association study on long-living individuals from Southern Italy identifies rs10491334 in the CAMKIV gene that regulates survival proteins. Rejuvenation Res. 2011, 14: 283-291. 10.1089/rej.2010.1114.CrossRefPubMed Malovini A, Illario M, Iaccarino G, Villa F, Ferrario A, Roncarati R, Anselmi CV, Novelli V, Cipolletta E, Leggiero E, Orro A, Rusciano MR, Milanesi L, Maione AS, Condorelli G, Bellazzi R, Puca AA: Association study on long-living individuals from Southern Italy identifies rs10491334 in the CAMKIV gene that regulates survival proteins. Rejuvenation Res. 2011, 14: 283-291. 10.1089/rej.2010.1114.CrossRefPubMed
72.
Zurück zum Zitat Soerensen M, Dato S, Christensen K, McGue M, Stevnsner T, Bohr VA, Christiansen L: Replication of an association of variation in the FOXO3A gene with human longevity using both case–control and longitudinal data. Aging Cell. 2010, 9: 1010-1017. 10.1111/j.1474-9726.2010.00627.x.PubMedCentralCrossRefPubMed Soerensen M, Dato S, Christensen K, McGue M, Stevnsner T, Bohr VA, Christiansen L: Replication of an association of variation in the FOXO3A gene with human longevity using both case–control and longitudinal data. Aging Cell. 2010, 9: 1010-1017. 10.1111/j.1474-9726.2010.00627.x.PubMedCentralCrossRefPubMed
73.
Zurück zum Zitat Willcox BJ, Donlon TA, He Q, Chen R, Grove JS, Yano K, Masaki KH, Willcox DC, Rodriguez B, Curb JD: FOXO3A genotype is strongly associated with human longevity. Proc Natl Acad Sci USA. 2008, 105: 13987-13992. 10.1073/pnas.0801030105.PubMedCentralCrossRefPubMed Willcox BJ, Donlon TA, He Q, Chen R, Grove JS, Yano K, Masaki KH, Willcox DC, Rodriguez B, Curb JD: FOXO3A genotype is strongly associated with human longevity. Proc Natl Acad Sci USA. 2008, 105: 13987-13992. 10.1073/pnas.0801030105.PubMedCentralCrossRefPubMed
75.
Zurück zum Zitat Praitis V: Creation of transgenic lines using microparticle bombardment methods. Methods Mol Biol. 2006, 351: 93-107.PubMed Praitis V: Creation of transgenic lines using microparticle bombardment methods. Methods Mol Biol. 2006, 351: 93-107.PubMed
76.
Zurück zum Zitat Datsenko KA, Wanner BL: One-step inactivation of chromosomal genes in Escherichia coli K-12 using PCR products. Proc Natl Acad Sci USA. 2000, 97: 6640-6645. 10.1073/pnas.120163297.PubMedCentralCrossRefPubMed Datsenko KA, Wanner BL: One-step inactivation of chromosomal genes in Escherichia coli K-12 using PCR products. Proc Natl Acad Sci USA. 2000, 97: 6640-6645. 10.1073/pnas.120163297.PubMedCentralCrossRefPubMed
77.
Zurück zum Zitat Hosono R, Mitsui Y, Sato Y, Aizawa S, Miwa J: Life span of the wild and mutant nematode Caenorhabditis elegans. Effects of sex, sterilization, and temperature. Exp Gerontol. 1982, 17: 163-172. 10.1016/0531-5565(82)90052-3.CrossRefPubMed Hosono R, Mitsui Y, Sato Y, Aizawa S, Miwa J: Life span of the wild and mutant nematode Caenorhabditis elegans. Effects of sex, sterilization, and temperature. Exp Gerontol. 1982, 17: 163-172. 10.1016/0531-5565(82)90052-3.CrossRefPubMed
78.
Zurück zum Zitat Oh SW, Mukhopadhyay A, Dixit BL, Raha T, Green MR, Tissenbaum HA: Identification of direct DAF-16 targets controlling longevity, metabolism and diapause by chromatin immunoprecipitation. Nat Genet. 2006, 38: 251-257. 10.1038/ng1723.CrossRefPubMed Oh SW, Mukhopadhyay A, Dixit BL, Raha T, Green MR, Tissenbaum HA: Identification of direct DAF-16 targets controlling longevity, metabolism and diapause by chromatin immunoprecipitation. Nat Genet. 2006, 38: 251-257. 10.1038/ng1723.CrossRefPubMed
79.
Zurück zum Zitat Timmons L, Court DL, Fire A: Ingestion of bacterially expressed dsRNAs can produce specific and potent genetic interference in Caenorhabditis elegans. Gene. 2001, 263: 103-112. 10.1016/S0378-1119(00)00579-5.CrossRefPubMed Timmons L, Court DL, Fire A: Ingestion of bacterially expressed dsRNAs can produce specific and potent genetic interference in Caenorhabditis elegans. Gene. 2001, 263: 103-112. 10.1016/S0378-1119(00)00579-5.CrossRefPubMed
Metadaten
Titel
Transcriptional regulation of Caenorhabditis elegansFOXO/DAF-16 modulates lifespan
verfasst von
Ankita Bansal
Eun-Soo Kwon
Darryl Conte Jr
Haibo Liu
Michael J Gilchrist
Lesley T MacNeil
Heidi A Tissenbaum
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
Longevity & Healthspan / Ausgabe 1/2014
Elektronische ISSN: 2046-2395
DOI
https://doi.org/10.1186/2046-2395-3-5

Weitere Artikel der Ausgabe 1/2014

Longevity & Healthspan 1/2014 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Wo hapert es noch bei der Umsetzung der POMGAT-Leitlinie?

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Das Risiko für Vorhofflimmern in der Bevölkerung steigt

02.05.2024 Vorhofflimmern Nachrichten

Das Risiko, im Lauf des Lebens an Vorhofflimmern zu erkranken, ist in den vergangenen 20 Jahren gestiegen: Laut dänischen Zahlen wird es drei von zehn Personen treffen. Das hat Folgen weit über die Schlaganfallgefährdung hinaus.

VHF-Ablation nützt wohl nur bei reduzierter Auswurfleistung

02.05.2024 Ablationstherapie Nachrichten

Ob die Katheterablation von Vorhofflimmern bei Patienten mit Herzinsuffizienz die Komplikationsraten senkt, scheint davon abzuhängen, ob die Auswurfleistung erhalten ist oder nicht. Das legen die Ergebnisse einer Metaanalyse nahe.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.