Movatterモバイル変換


[0]ホーム

URL:


Skip to main content
                                  NCBI home page
Search in PMCSearch
As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsement of, or agreement with, the contents by NLM or the National Institutes of Health.
Learn more:PMC Disclaimer | PMC Copyright Notice
Aging and Disease logo

Longevity-Associated Transcription FactorATF7 Promotes Healthspan by Suppressing Cellular Senescence and Systematic Inflammation

Yaqun Huang1,2,3,#,Ming-Xia Ge1,4,#,Yu-Hong Li1,#,Jing-Lin Li1,2,3,Qin Yu1,Fu-Hui Xiao1,Hong-Shun Ao1,4,Li-Qin Yang1,Ji Li2,3,*,Yonghan He1,*,Qing-Peng Kong1,5,*
1State Key Laboratory of Genetic Resources and Evolution, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
2Department of Dermatology/National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410000, China.
3Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China.
4Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China.
5CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China.
*

Correspondence should be addressed to: Dr. Qing-Peng Kong, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China. E-mail:kongqp@mail.kiz.ac.cn. Dr. Yonghan He, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China. E-mail:heyonghan@mail.kiz.ac.cn. Dr. Ji Li, Xiangya Hospital, Central South University, Changsha, China. Email:liji_xy@csu.edu.cn.

#

These authors contributed equally to this work.

Received 2022 Nov 2; Revision requested 2022 Dec 16; Accepted 2022 Dec 17; Collection date 2023 Aug 1.

Copyright: © 2023 Huang et al.

this is an open access article distributed under the terms of the creative commons attribution license, which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed.

PMCID: PMC10389835  PMID:37163432

Abstract

Aging is characterized by persistent low-grade systematic inflammation, which is largely responsible for the occurrence of various age-associated diseases. We and others have previously reported that long-lived people (such as centenarians) can delay the onset of or even escape certain major age-related diseases. Here, by screening blood transcriptome and inflammatory profiles, we found that long-lived individuals had a relatively lower inflammation level (IL6,TNFα), accompanied by up-regulation of activating transcription factor 7 (ATF7). Interestingly,ATF7 expression was gradually reduced during cellular senescence. Loss ofATF7 induced cellular senescence, while overexpression delayed senescence progress and senescence-associated secretory phenotype (SASP) secretion. We showed that the anti-senescence effects ofATF7 were achieved by inhibiting nuclear factor kappa B (NF-κB) signaling and increasing histone H3K9 dimethylation (H3K9me2). InCaenorhabditis elegans,ATF7 overexpression significantly suppressed aging biomarkers and extended lifespan. Our findings suggest thatATF7 is a longevity-promoting factor that lowers cellular senescence and inflammation in long-lived individuals.

Keywords:ATF7, cellular senescence, H3K9me2, longevity, NF-κB

INTRODUCTION

Aging is a major risk factor for several diseases that limit health span, including neurodegenerative diseases, metabolic syndromes, cancer, and cardiovascular disease [1]. Persistent low-level chronic inflammation associated with aging is largely responsible for these diseases [2,3]. Aging is characterized by various molecular hallmarks, the targeting of which may accelerate or retard normal aging processes. Among them, cellular senescence is closely associated with age-related diseases, as senescent cells (SnCs) secrete a series of senescence-associated secretory phenotype (SASP) factors, resulting in persistent low-grade systemic inflammation [4,5]. Inhibiting cellular senescence and/or age-related inflammation may delay aging and slow the development of age-associated diseases. For example, selective elimination of SnCs (senolytic), or suppression of SASP secretion (senomorphic) via transgenic or pharmaceutical strategies can slow aging and age-related diseases, thereby extending healthy lifespan [6].

Although aging is an irreversible process with progressive loss of physiological integrity, impaired function, and increased vulnerability to death, some long-lived species display a delayed aging phenotype, including long-lived humans [7]. As a paradigm of successful human aging, long-lived individuals (LLIs) can reach an extremely advanced age (>90 years) without developing serious age-related diseases, thus living a healthier and longer lifespan [8]. We previously reported that LLIs exhibit enhanced autophagy-lysosomal activity [9], lower ribosomal biogenesis [10], and superior metabolic profiles [11,12], which may contribute to their healthy aging. Levels of inflammatory signaling increase during aging, which is linked to many age-related diseases [2]. However, centenarian cells show an anti-inflammatory phenotype [13]. Recently, Sayed et al. constructed an inflammatory clock of aging and found that centenarians had a lower inflammatory clock index associated with longevity [5]. In the current study, we found that LLIs displayed lower levels of proinflammatory factors such asIL6 andTNFα. However, how long-lived centenarians gain these survival and healthy aging advantages remains unclear. Based on unbiased transcriptomic and interaction analyses, we identified activating transcription factor 7 (ATF7) as a potential contributor to the survival advantage of LLIs. ATF7 can bind to the cyclic AMP response element (CRE), which belongs to the ATF/CREB superfamily [14-16]. ATF7 activity is growth- and stress-initiated, and its transcriptional function strictly requires phosphorylation by mitogen-activated protein kinases (MAPKs) [17]. In the absence of stress, ATF7 represses the transcription of its target genes, including those encoding factors involved in innate immunity, by recruiting H3K9 di- and tri-methyltransferases G9a, EDET/SET-DB1, and Suv39h1 [18,19]. In the presence of stress, ATF7 is phosphorylated by p38MAPK, leading to the release of ATF7 and methyltransferases from target genes, thereby activating transcription [19]. During aging, the relatively preserved innate immune response overwhelms the more variable adaptive immune response, leading to higher production of proinflammatory mediators [20]. These circulating proinflammatory factors can, in turn, keep the immune system in a state of chronic low-level activation [3].

In this study, we found that relatively low levels of inflammation were accompanied by an up-regulation ofATF7 in LLIs. Intriguingly,ATF7 was not upregulated during the process of cellular aging, but rather was decreased. We demonstrated thatATF7 can suppress cellular senescence and systematic inflammation by modulating nuclear factor kappa B (NF-κB) signaling and dimethylation levels of histone H3K9 (H3K9me2). We also demonstrated thatATF7 extended the lifespan ofCaenorhabditis elegans (C. elegans). These findings suggest thatATF7 may be an important contributor to longevity by suppressing cellular aging and systematic inflammation.

MATERIALS AND METHODS

Study subjects, RNA sequencing (RNA-seq), network construction, and centrality analysis

As described in our prior study [10], we sampled 135 females from longevous families in Hainan Province, southern China, and constructed 76 and 59 sequencing libraries using poly(A) capture and ribosomal RNA depletion methods, respectively. The RNA-seq reads were filtered to obtain clean reads [21], aligned to the human (hg19) andC. elegans (WBcel235) reference genomes [22], and quantified to determine gene abundance [23]. Differentially expressed gene (DEG) analysis was performed using the ‘limma’ package [24], with the effects of library type and cell composition considered (Gene expression ~ 0 + Group + Lymph + Neu + Mid). The ‘removeBatchEffect’ function in ‘limma’ was used to remove the effect of library type on longevous family transcriptome data. The DESeq2 function in the R platform was used to detect DEGs between different experimental groups. A parameter false discovery rate (FDR) cutoff of less than 0.05 was considered statistically significant. Pearson correlation coefficient (r) was used to determine correlations between differently expressed transcription factors (TFs) and immune-related genes, with a Benjamini-Hochberg adjustedP-value cutoff of less than 0.05. The immune-related gene list was downloaded from the ImmPort database (www.immport.org/home) and TFs were obtained from the footprintDB and TRRUST databases (http://floresta.eead.csic.es/footprintdb/index.php;www.grnpedia.org/trrust/). Co-expression network plots were constructed using Cytoscape (v3.9.0) [25] and centrality analysis was conducted using the cytoHubba program in Cytoscape. Functional enrichment analysis was performed using “Metascape” [26] and “DAVID” [27,28]. Raw RNA-seq data of replicative senescent human embryonic lung fibroblasts (WI-38), human neonatal foreskin fibroblasts (HFF and BJ), and oncogene-induced senescent human embryonic lung fibroblasts (IMR-90) were collected from the ‘‘GEO Repository’’ (www.ncbi.nlm.nih.gov/geo/) (accession numbersGSE63577,GSE64553,GSE61130).

Cell culture

Human dermal fibroblasts (HDFs) were obtained from circumcised foreskins of healthy human donors aged 5-20 years in Xiangya Hospital, Central South University, as described previously [29,30]. Human embryonic lung fibroblasts IMR-90 and HEL were purchased from the American Type Culture Collection (ATCC) and Kunming Cell Bank (Chinese Academy of Sciences), respectively. The HDF, HEL, and IMR-90 cells were cultured in Dulbecco’s Modified Eagle Medium (DMEM, C11995500BT, Gibco, USA) or Minimum Essential Medium (MEM, C11095500BT, Gibco, USA) supplemented with 10% fetal bovine serum (35-076-CV, Gibco, USA) and 1% penicillin/streptomycin (15140-122, Gibco, USA) in a 37 °C/5% CO2 humidified incubator. Human embryonic kidney HEK-293T cells were purchased from ATCC and cultured in DMEM (C11995500BT, Gibco, USA) supplemented with 10% fetal bovine serum (04-001-1ACS, BI, Israel) and 1% penicillin/streptomycin (15140-122, Gibco, USA) in a 37 °C/5% CO2 humidified incubator.

Plasmids and lentiviral infection

ATF7 overexpression plasmid was constructed using the pCDH-MSCV-E2F-eGFP lentiviral vector.ATF7 short hairpin RNA (shRNA) (target sequence: CGAAGAA CTCACTTCTCAGAA; CCGAACTGACTCAGTCATC AT) plasmids were constructed using the pLKO.1 lentiviral vector. Plasmids were transfected into HEK-293T cells, with the collected lentiviruses then transduced into fibroblasts (HDF, IMR-90). Cells were subsequently cultured in selected media supplemented with 2 μg/mL puromycin (ant-pr-1, InvivoGen, USA).

Senescence-associated beta-galactosidase (SA-β-gal) staining

SA-β-gal staining was performed using a Senescence Cells Histochemical Staining Kit (CS0030-1kt, Sigma-Aldrich, USA) according to the manufacturer’s instructions. Briefly, cells were washed with phosphate-buffered saline (PBS) and fixed at room temperature for 7 min, then incubated in staining working solution overnight at 37 °C. Cells were visualized using a Nikon eclipse Ti inverted microscope and a minimum of 200 cells were counted.

Quantitative real-time polymerase chain reaction (qRT-PCR)

Total RNA was extracted using TRIzol reagent (15596018, Invitrogen, USA) and reverse transcribed into cDNA using a RevertAid First Strand cDNA Synthesis Kit (K1622, Thermo Fisher Scientific, USA) according to the manufacturer’s protocols. qRT-PCR was performed using 2 × Tsingke® Master qPCR Mix (TSE201, Tsingke, China), with actin as the endogenous normalization control. Gene expression levels were calculated using the 2-ΔΔCt method [31]. All primers used are presented inSupplementary Table 1.

Western blotting

Cells were lysed using RIPA lysis buffer (P0013B, Beyotime, China) supplemented with 1% phenylmethylsulfonyl fluoride (ST506, Beyotime, China) on ice. Protein extract (30 μg) was separated by sodium dodecyl-sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and subsequently transferred to polyvinylidene fluoride (PVDF) membranes (162-0177, Bio-Rad, USA). The membranes were incubated with appropriate primary antibodies at 4 °C overnight, then with horseradish peroxidase (HRP)-labeled goat anti-rabbit or anti-mouse immunoglobulin G (IgG) (H + L) secondary antibodies at room temperature for 1 h. Protein bands were visualized using an ECL Detection Kit (UEL-S6009L, US Everbright, China). Quantification of blots was performed using ImageJ software. Detailed information on primary antibodies used in western blotting is provided inSupplementary Table 2.

EdU incorporation assay

Cell proliferation was analyzed using a Cell-Light EdU Apollo567In Vitro Kit (C10310-1, Ribobio, China). Briefly, cells (8 × 103) were seeded into each well of a 96-well plate and incubated for 24 h with 5 μM EdU at 37 °C. The cells were then added with 1 × reaction buffer for 30 min. Images were captured using a Cytation 5 Cell Imaging Multi-Mode Reader (BioTek).

Cellular reactive oxygen species (ROS) detection

Cellular ROS detection was performed using CellROX® Oxidative Stress Reagent (C10422, Thermo Fisher Scientific, USA) according to the manufacturer’s instructions. Briefly, cells (8 × 103) were seeded into each well of a 96-well plate, followed by the addition of CellROX® Reagent at a final concentration of 10 μM and incubation for 30 min at 37 °C. Images were captured using a Cytation 5 Cell Imaging Multi-Mode Reader (BioTek). Quantification was performed using ImageJ software.

Cytokine array

Cells were seeded into 6-cm dishes and washed and incubated in serum-free DMEM in a 37 °C/5% CO2 humidified incubator. After 24 h, the conditioned medium (CM) was collected, filtered (0.2 μm), and frozen at -80 °C. The CM was analyzed using an antibody array (QAH-CYT-1, RayBiotech, USA) according to the manufacturer’s instructions. The array was used to detect 20 cytokines and chemokines, including IL1A, IL1B, IL2, IL4, IL5, IL6, IL8 (CXCL8), IL10, IL12, IL13, IFNγ, TNFα, GM-CSF, GROα/β/γ, MCP1 (CCL2), MIP1a (CCL3), MIP1b (CCL4), MMP9, VEGFA, and RANTES (CCL5). The quantity of secreted cytokines was measured by adding 90 μL of concentrated conditioned medium to the array, followed by overnight incubation at 4 °C. After washing with wash buffer, biotinylated primary antibodies were added and incubated at room temperature for 2 h. After washing again with wash buffer, Cy3 equivalent dye-conjugated streptavidin was added for 1 h. The chip was scanned using an InnoScan 300 Microarray Scanner (Innopsys, France). Cytokines were quantified according to the standard curve calibrated from the same array.

Chromatin immunoprecipitation (ChIP) analysis

ChIP analysis was performed using a SimpleChIP® Enzymatic Chromatin IP Kit (9003, Cell Signaling Technology, USA) according to the manufacturer’s instructions. Chromatin was crosslinked with 1% formaldehyde in cell medium at room temperature for 10 min. Glycine was then added to the medium to terminate the reaction. After washing in ice-cold PBS, cells were lysed and sonicated into 150-900-bp fragments. Immunoprecipitation was performed overnight at 4 °C with anti-histone H3K9me2 and anti-histone H3. Normal rabbit IgG was used as a negative control. The immunocomplexes were washed using ChIP-Grade Protein G Magnetic Beads (9006, Cell Signaling Technology, USA) and incubated at 65 °C overnight following the addition of 5 M NaCl and Proteinase K to reverse the crosslinks. Free precipitated DNA was further purified. The DNA samples were used for qRT-PCR. All primers are presented inSupplementary Table 3.

Co-immunoprecipitation

The HDF cells were lysed in IP-lysis buffer (50 mM Tris-HCl, 150 nM NaCl, 1 mM ethylenediaminetetraacetic acid, 0.2% NP40, and 10% glycerol) for 30 min on ice. After centrifugation at 12 500 rpm and 4 °C for 15 min, the supernatants were incubated in anti-ATF7 or control IgG. The immunocomplexes were then collected using Protein-A-agarose beads (11134515001, Roche, Switzerland) and subjected to western blotting with anti-G9a antibodies.

Cleavage Under Targets and Tagmentation (CUT&Tag) assay

CUT&Tag assay was performed using a Hyperactive® Universal CUT&Tag Assay Kit for Illumina (TD903, Vazyme, China) according to the manufacturer’s instructions. Briefly, 1 × 105 cells were collected, washed in wash buffer, and added to 10 μL of activated ConA Beads. Bead-bound cells were incubated overnight at 4 °C with anti-histone H3K9me2. Normal rabbit IgG was used as a negative control. The cells were washed and incubated with rabbit secondary antibodies at room temperature for 1 h. After washing, the cells were incubated with pA/G-Tnp at room temperature for 1 h, then with TruePrep Tagment Buffer L (TTBL) and Dig-300 buffer for fragmentation at 37 °C for 1 h. The released chromatin fragments were extracted, and libraries were generated using TruePrep® Index Kit V2 for Illumina (TD202, Vazyme, China). Reads were aligned to the human reference genome hg19 using Bowtie2 (v.2.4.1) [32]. The ‘picard.jar’ program in GATK (v4.1.8.0) was used to remove duplicated reads [33]. For each replicate, peaks were first called using MACS2 (v.2.1.2) [34] with the parameters: -q 0.05 -B. Differential binding analysis of peaks was performed using the Bioconductor package ‘DiffBind’. The ‘annotatedPeak’ function in the R package ‘CHIPseeker’ [35] was used to annotate peaks. Peaks were visualized using Integrative Genomics Viewer (IGV) [36].

Lifespan assay

AllC. elegans lifespan experiments were conducted at 20 °C in accordance with standard protocols using HT115/RNA interference (RNAi) (with addition of Amp). Briefly, worms were branched to obtain synchronized eggs, which were cultured to hatch and used for lifespan assays. At L4 molting, animals were transferred to plates containing 20 M 5-fluoro-2’-deoxyuridine (FUDR), which kills their progeny at the embryo stage but does not significantly affect lifespan. The plates were refreshed daily. Approximately 50 worms were used for each treatment. The first day of adulthood was recorded as day 1 and dead worms were counted from day 5. Worms that did not respond to nose touch were picked out of the bacterial lawn, and worms that did not move were scored as dead.

Body bending assay

Worms were pretreated as in the lifespan assays. On the day of examination, 100 μL of M9 buffer was pipetted onto the surface of an empty 35-mm NGM plate. A single worm was added to the buffer and allowed 30 s to recover from the transfer. The number of thrashing movements over a 30-s period was counted (four times per worm, 15-20 worms per treatment). One worm from one treatment was counted, then one worm from another treatment was counted, and so on, with the cycle repeated to avoid environmental or operational interference.

Lipofuscin accumulation

Autofluorescence of intestinal lipofuscin was measured as an index of senescence in day 10 adults. Worms were randomly selected from each bacterial lawn and washed three times with M9 buffer. The worms were then placed on 5% agar pads coated with 10 mM sodium azide in M9 buffer to initiate paralysis. Lipofuscin autofluorescence images were taken using blue excitation light (405-488 nm) with 4’,6-diamidino-2-phenylindole (DAPI) and laser confocal scanning microscopy. Fluorescence was quantified using ImageJ software to determine lipofuscin levels. Three independent experiments were performed with more than 30 worms for each bacterial species each day.

Pharyngeal pumping

Wild-type andATF7-overexpressing worms were synchronized by timed oviposition onto an empty vector (control) andATF7 double-stranded RNA (dsRNA)-expressing HT115. Day 1 adult worms were transferred to control and RNAi plates with FUDR. Pumping rates were determined on day 10 of adulthood. Terminal pharyngeal bulb pumps were counted at 30-s intervals for each worm on the bacterial lawn. The pumping rate per minute was averaged for 10 worms per treatment. Pumping rates were assessed in three independent replicate experiments.

RNAi in C. elegans

RNAiATF7 was generated using primers GAATTCC TGCAGCCCCGCGTCCAATTGATGGGC and ACGC GTGGATCCCCCCACAGATAACAATTTGGAAATGTCC and linked to L4440 by infusion (SmaI). TheE. coli strain HT115 (DE3) expressingATF7 dsRNA was grown overnight at 37 °C in Luria-Bertani (LB) broth containing 100 μg/mL ampicillin, then spread on nematode growth medium (NGM) plates containing 100 μg/mL ampicillin and 1 mM isopropyl 1-thio-β-D-galactopyranoside (IPTG). The RNAi-expressing bacteria were then grown at 25 °C overnight. Synchronized L4 larvae were placed on the plates at 20 °C. RNAi was induced from L4 unless otherwise noted. L4 adult animals were used for further experiments.

ATF7 overexpression in C. elegans

ATF7 cDNA expression was driven by anATF7 promoter andATF7 cDNA insertion was verified by sequencing. The primers used for cloningATF7 were:ATF7 promoter-F: GTCGACTCTAGAGGATCCCCTT TTGGAGCCG CCAAAGATCG;ATF7 promoter-R: CGACCGACATA ATTGTGTTGGACGATTGCTGA;ATF7 cDNA -F: CAACACAATTATGTCGGTCGTAACAACGA;ATF7 cDNA-R: TGGGTCCTTTGGCCAATCCCTTGGAGT TT GGGTAATTTCAGTTGC. The cDNA and promoter ofATF7 were then linked to pPD95_75 by infusion (SmaI). The plasmid was injected at a concentration of 50 ng/μL into N2 worms using a Pmyo-3::mCherry::unc-54 (pCFJ104) expressing plasmid as a co-injection marker.

Statistical analysis

Statistical analyses were conducted using GraphPad Prism v9 (GraphPad Software). Data are presented as means ± standard error of the mean (SEM). Normal distribution of data was determined using the Shapiro-Wilk test. Comparisons were performed using two-tailed Student’st-test or Mann-Whitney U test. Survival curves were compared, andP-values were calculated using log-rank (Mantel-Cox) analysis.P < 0.05 was defined as statistically significant.

RESULTS

Centenarians display lower inflammation with possible ATF7 involvement

Peripheral blood mononuclear cells (PBMCs) from centenarians show reducedIL6 mRNA levels [13]. To determine overall gene expression changes in proinflammatory factors in LLIs, we analyzed RNA-seq data in a cohort of longevous families [10]. In total, 2 870 significant DEGs were identified between LLIs and younger controls (YCs) (P < 0.05) (Fig. 1A). Among the DEGs, a portion of major proinflammatory factors, includingIL6,CXCL8 (IL8),TNF,IGFBP4, andIGFBP6 [37-39], were down-regulated in LLIs compared to YCs (Fig. 1B). Consistent with the RNA-seq results, cytokine array showed that LLIs had significantly lower levels of MMP9 and CCL4 (MIP1b) and lower levels of proinflammatory factors, including IL6, IL1A, and TNFα, compared to the sex-matched YCs (Fig. 1C). As proinflammatory factors are primarily regulated at the transcriptional level, we focused on differentially expressed TF genes that may have both anti-inflammatory and potentially anti-aging functions. We performed network centrality analysis to prioritize candidate TF genes [40,41], and identified five TF genes related to inflammatory regulation (Fig. 1D). Among them,ELF1,IRF3,ZNF410, andARNT may promote the development of inflammation or senescence [42-45].ATF7 not only plays a repressive role in innate immunity [18], but also responds to intrinsic and extrinsic stresses [17], and thus was chosen as the candidate gene (Fig. 1B, D). We confirmed the expression level ofATF7 in LLIs and YCs using qRT-PCR (Supplementary Fig. 1A) and explored its potential roles and mechanisms in the following experiments.

Figure 1.

Figure 1.

Identification ofATF7 in DEGs between LLIs and YCs. (A) Volcano plot of DEGs identified based onP-value and fold-change of genes with expression differences between all LLI and YC transcripts (n = 80, 55). (B) Expression of representative pro-inflammatory factor andATF7 expression levels in LLI and YC transcripts (n = 80 and 55 for LLI and YC, respectively). (C) Proteins in plasma collected from LLIs and YCs, measured by cytokine array (n = 24 and 25 for LLI and YC, respectively). (D) Rank of local- and global-based method values of candidate TF genes. LLIs: long-lived individuals; YCs: younger controls; MNC, Maximum neighborhood component; FC, fold-change.

Manipulation of ATF7 alters cellular senescence progression

AsATF7 was upregulated in LLIs compared to YCs, we investigated whether it affects cellular senescence, one of the major hallmarks of aging. Unlike the case in LLIs, the protein level of ATF7 was progressively down-regulated during serial passage in HDF cells, accompanied by the up-regulation of p16 (CDKN2A), a marker of cellular senescence (Fig. 2A). Similarly,ATF7 was down-regulated in senescent WI-38 fibroblasts, HFF and BJ fibroblasts, and IMR-90 fibroblasts compared to non-senescent cells (Supplementary Fig. 1B) (online published data) [46-48]. The downregulation of ATF7 was partly due to increased phosphorylation by p38MAPK (Supplementary Fig. 2A), which can cause instability and degradation [49,50]. Notably, we found that application of SB203580, an inhibitor of p38MAPK, decreased the degradation of the ATF7 protein (Supplementary Fig. 2B).

Figure 2.

Figure 2.

Manipulation ofATF7 expression changes cellular senescence progression in HDFs. (A) ATF7 protein levels were assessed during serial passage by western blotting (n = 2 biological replicates). (B) Western blot analysis of protein levels of ATF7, p16, H3K9me2 in control andATF7-overexpressing HDFs (n = 3 biological replicates). (C) SA-β-Gal-staining cells in control andATF7-overexpressing HDFs. Typical images are presented on top and quantitative result is presented at bottom (n = 3 biological replicates, scale bar, 200 μm). (D) qRT-PCR analysis of mRNA levels ofATF7,p21, andp16 in control andATF7-knockdown HDFs (n = 4 biological replicates). (E) Western blot analysis of protein levels of ATF7, p16, and H3K9me2 in control andATF7-knockdown HDFs (n = 3 biological replicates). (F) SA-β-Gal-staining cells in control andATF7-knockdown HDFs. Typical images are presented on the left and quantitative result is presented on the right (n = 3 biological replicates, scale bar, 200 μm). (G) EdU-staining in control andATF7-knockdown HDFs. Typical images are presented on the left and quantitative result is presented on the right (n = 3 biological replicates, scale bar, 200 μm). (H) Cellular ROS detected in control andATF7-overexpressing (left, n = 4 biological replicates) or knockdown (right, n = 3 biological replicates) HDFs. P, passage; OE, overexpression; KD, knockdown; MSI, Mean signal intensity. Statistical analyses were performed using two-sided Student’st-test.

Next, we examined the effect ofATF7 on HDF senescence. Overexpression ofATF7 significantly decreased senescence markers, including p16 (CDKN2A) protein levels and SA-β-gal-staining positive cells, and increased H3K9me2 levels compared to control HDFs (Fig. 2B-C). In contrast, knockdown ofATF7 with shRNA reversed the above senescence markers (Fig. 2D-F) and inhibited DNA synthesis indicated by EdU staining (Fig. 2G). These results were validated in the IMR-90 fibroblasts (Supplementary Fig. 3A-B), suggesting a suppressive role ofATF7 in cellular senescence. As DNA damage and ROS are primarily responsible for cellular senescence, we tested ROS levels inATF7-overexpressed or down-regulated cells. As shown inFig. 2H, compared to untreated control HDFs, overexpression and down-regulation ofATF7 significantly decreased and increased ROS levels in HDFs, respectively.

ATF7 represses secretion of SASP

SnCs affect surrounding cells/tissues by secreting SASP, which consists of inflammatory factors, extracellular modifiers, and growth factors [51]. To test whetherATF7 regulates SASP and to identify the pathways involved in senescence followingATF7 manipulation, we performed transcriptome analysis ofATF7-overexpressing and control HDFs. In total, 827 DEGs were identified, including 534 down-regulated and 293 up-regulated DEGs in theATF7-overexpressing HDFs relative to the control cells. KEGG pathway enrichment analysis indicated that the down-regulated DEGs were enriched in inflammatory pathways, including cytokine-cytokine receptor interaction, PI3K-Akt signaling pathway, MAPK signaling pathway, JAK-STAT signaling pathway, and NF-κB signaling pathway (Fig. 3A). Up-regulated DEGs were enriched in the TGF-β signaling pathway (Fig. 3B), among whichID1,ID2,ID3,ID4,SMAD6,SMAD7, andSMAD9 are negative regulators of inflammation [52-55]. These findings suggest thatATF7 may play an inhibitory role in modulating HDF inflammation. SnC-secreted SASP is responsible for multiple age-related pathologies [56]. Based on transcriptome data, 13 down-regulated SASP factors, together with upstream inflammation-regulatory genes (e.g.,STAT3,NFKB2, andRELB), were confirmed using qRT-PCR (Fig. 3C, D). In addition,ATF7 overexpression significantly reduced the mRNA expression levels ofIL8 andGM-CSF (Fig. 3D). In contrast, compared to the control cells, knockdown ofATF7 up-regulated several SASP factors and down-regulated genes in the TGF-β signaling pathway, i.e.,ID1,ID2,SMAD6, andSMAD7 (Supplementary Fig. S4A-C). To verify the inhibitory role ofATF7 on SASP, we analyzed SASP release in culture medium using antibody arrays. As shown inFig. 3E, a panel of inflammatory factors, including IL6, IL8, TNFα, GM-CSF, IL2, IL5, and IFNγ, were significantly decreased inATF7-overexpressing HDFs compared to the controls.

Figure 3.

Figure 3.

ATF7 represses SASP secretion in HDFs. (A) KEGG pathway enrichment analysis of down-regulated DEGs between control andATF7-overexpressing HDFs. (B) KEGG pathway enrichment analysis of up-regulated DEGs between control andATF7-overexpressing HDFs. (C) Heatmap of SASP genes forATF7-overexpressing HDFs relative to the control group from RNA-seq datasets (n = 3 biological replicates). (D) qRT-PCR analysis of mRNA levels of SASP genes and inflammatory pathway genes inATF7-overexpressing HDFs relative to the control (n = 3 biological replicates). (E) SASP and inflammatory factors in serum-free CM collected from control andATF7-overexpressing HDFs, measured by cytokine array (n = 4 biological replicates). OE, overexpression. Statistical analyses were performed using two-sided Student’st-test.

Notably, the release of TNFα, GM-CSF, and IL2 was markedly inhibited (>50% reduction) and IL5 was completely inhibited in the HDFs withATF7 overexpression relative to the control cells (Fig. 3E). The effect ofATF7 on suppressing SASP in SnCs mimicked that in LLIs, in whichATF7 up-regulation was accompanied by lower levels of SASP factors compared to YCs (Fig. 1B-C). These findings suggest thatATF7 can repress cellular senescence and SASP secretion in SnCs and may contribute, at least partially, to lower inflammation in LLIs.

ATF7 restrains SASP by inhibiting NF-κB signaling and modulating H3K9me2

To determine howATF7 inhibits SASP, we focused on the inflammatory pathways enriched in KEGG analysis. Based on immunoblotting, we found that the phosphorylation level of P65, a canonical indicator of NF-κB signaling activity, was significantly decreased inATF7-overexpressing HDFs compared to control HDFs (Supplementary Fig. S5A). NF-κB is a recognized regulator of SASP production [6,57]. Therefore, we investigated upstream signals in this pathway and found that the phosphorylation levels of P65 and IKKα/β were diminished inATF7-overexpressed cells compared to the control cells, whereas the total protein levels of P65 and IKKα/β remained unchanged (Fig. 4A, B).

Figure 4.

Figure 4.

ATF7 inhibits SASP secretion by inhibiting NF-κB signaling and modulating H3K9me2 in HDFs. (A) Western blot analysis of NF-κB signaling pathway activation in control andATF7-overexpressing HDFs (n = 4 biological replicates). (B) Qualification results of (A). (C) Co-immunoprecipitation of ATF7 and G9a. Cell lysates of HDFs were immunoprecipitated with anti-ATF7 antibody or control IgG, with immunocomplexes then subjected to western blotting using G9a (n = 2 biological replicates). (D) ChIP assay with qRT-PCR detection of H3K9me2 abundance in SASP genes at ATF7-binding sites of promoter regions in control andATF7-overexpressing HDFs (n = 4 biological replicates forIL6 andp16, n = 5 biological replicates for other genes). OE, overexpression. Statistical analyses were performed using two-sided Student’st-test.

In addition to its role in regulating NF-κB signaling,ATF7 is also reported to regulate inflammatory gene expression epigenetically [18,50]. As shown inFig. 2B,ATF7 overexpression up-regulated H3K9me2, which negatively regulates SASP factorsIL6 andIL8 [58]. To investigate whetherATF7 regulates SASP through H3K9me2, we performed chromatin immunoprecipitation plus qRT-PCR analysis using H3K9me2 antibodies. As shown inFig. 4C andSupplementary Fig. 5B, co-immunoprecipitation revealed that ATF7 could directly bind G9a, which regulates H3K9 dimethylation. These results suggest that ATF7 may recruit G9a to regulate H3K9me2 and silence its target genes, as reported previously [18,50]. The ChIP-qRT-PCR analysis also showed thatATF7 overexpression up-regulated H3K9me2 levels around the promotor regions of several SASP factors, includingIL8,IL6,CXCL1,CXCL2,CXCL3,CCL8, andTNF, as well as the upstream regulatorSTAT1, but not p16 (Fig. 4D). These data were validated using the CUT&Tag assay, which showed a high signal-to-noise ratio (Supplementary Fig. 6) [59]. Thus, these results suggest thatATF7 suppresses SASP via both NF-κB and H3K9me2.

Manipulation of ATF7 affects lifespan and survival in C. elegans

As overexpression ofATF7 delayed cellular senescence, suppressed SASP productionin vitro, and was associated with longevity in LLIs, we wondered whether it may function to promote lifespan. We tested this usingC. elegans, a model species widely used to determine molecular mechanisms of lifespan [60]. As shown inFig. 5, transgenicATF7 overexpression extended theC. elegans lifespan by 10.3% compared to the control, whereas RNAi knockdown ofATF7 decreased lifespan by 17.2% (Fig. 5B). Consistently, compared to the control, overexpression ofATF7 reduced intestinal lipofuscin and promoted body locomotion and pharyngeal pumping (Fig. 5C, D), applied as aging markers inC. elegans [61]. In contrast, knockdown ofATF7 accelerated aging inC. elegans compared to the control group (Fig. 5C, D).

Figure 5.

Figure 5.

Manipulation ofATF7 affects lifespan and survival inC. elegans. (A) qRT-PCR analysis ofATF7 mRNA levels in N2 andATF7-overexpressing or RNAi worms (n = 3 biological replicates). (B) Lifespan analysis of N2 andATF7-overexpressing or RNAi worms from (A) (n = 3 biological replicates). (C, D) Lipofuscin accumulation assay (C, n = 3 biological replicates, scale bar, 100 μm), body bending assay (D, left, n = 53/group for 3 biological replicates), and pharyngeal pumping assay (D, right, n = 30/group for 3 biological replicates) in N2 andATF7-overexpressing or RNAi worms. (E) Gene Ontology (GO) enrichment analysis of down-regulated DEGs between N2 andATF7 overexpressing worms. (F) GO enrichment analysis of up-regulated DEGs between N2 andATF7-RNAi worms. (G) (H) Survival analysis of N2 andATF7-overexpressing or RNAi worms exposed toP. aeruginosa PA14. Worms were fed with PA14 for 2 days (g) or 10 days (h) after reaching adulthood (n = 3 biological replicates/group). OE, overexpression. Statistical analyses were performed using two-sided Student’st-test.

To explore howATF7 regulates lifespan and aging inC. elegans, we performed RNA-seq analysis ofATF7-overexpressed or silencedC. elegans and controlC. elegans. Gene Ontology (GO) enrichment analysis showed that the up-regulated genes inATF7-overexpressedC. elegans relative to controlC. elegans were mainly enriched in innate immune response, whereas the down-regulated genes inATF7-silencedC. elegans were mainly enriched in innate immune response and defense responses against gram-positive bacteria (Fig. 5E, F). The downregulation and up-regulation of innate immune-related genes inATF7-silenced and overexpressing worms support the regulatory role ofATF7 in innate immunity [18,62,63]. Moreover, most up-regulated DEG orthologs in humans play roles in modulating immune function, depending on different physiological/pathophysiological contexts (Supplementary Table 4). Importantly, asATF7 is closely associated with innate immunity, we suspect it may influence the survival of pathogen-challengedC. elegans. Indeed, in the presence ofPseudomonas aeruginosa (PA14), transgenicATF7 overexpression enhancedC. elegans survival, whereas RNAi knockdown ofATF7 reduced survival (Fig. 5G, H). Overall, these results suggest thatATF7 can extend the lifespan and promote healthy aging inC. elegans, in part by regulating innate immunity genes.

DISCUSSION

Persistent systemic chronic inflammation can lead to a series of age-related diseases [2,3]. Inflammaging is a well-recognized source of age-related inflammation [3]. As an important contributor to inflammagingin vitro andin vivo, SnC-secreted SASP can cause several age-related diseases by maintaining a proinflammatory internal microenvironment [64]. Accumulating evidence suggests that attenuating SASP production and age-related inflammation can alleviate age-related diseases and extend healthspan [2,65-67]. Our previous study and others have shown that LLIs can delay (or avoid) the onset of major age-related diseases [68-71]. We therefore wondered whether this survival advantage is achieved by maintaining better levels of inflammation. Here, by screening a panel of inflammatory factors, we found relatively low levels of inflammation in our longevity cohort. We also identified theATF7 transcriptional factor as a key upstream regulator in maintaining lower inflammation in LLIs. Importantly, manipulation ofATF7 modulated cellular senescence andC. elegans lifespan, suggesting it may be a novel and conserved contributor for longevity across various species, including humans.

In the current study,ATF7 was highly expressed in LLIs, but its expression was gradually lost during cellular senescence, suggesting it may be a longevity-associated factor rather than an aging-related factor. As a transcription repressor, ATF7 is involved in the regulation of cell cycle progression [72,73], heterochromatin formation [16,18,74], telomere shortening [49,75], and innate immunity [18,62,63,76]. Our results showed that overexpression ofATF7 suppressed cellular senescence and various inflammatory factors, indicating a possible role in delaying aging in individuals. Indeed,ATF7 has been reported to affect aging in mice, with its depletion found to promote senescence of embryonic fibroblasts and shorten lifespan [77]. Here, we provide evidence thatATF7 acts as a longevity-promoting factor in humans by inhibiting cellular senescence and SASP secretion.

Cellular senescence can be induced by various stimuli, including DNA damage, oncogene activation, oxidative stress, chemotherapy, mitochondrial dysfunction, and epigenetic changes [78]. Our data showed thatATF7-dependent senescence may be mediated through its regulation of oxidative stress and epigenetic changes (Fig. 6). SASP factors are primarily regulated by transcription [79]. Upstream regulators of SASP, such as p38MAPK, JAK2/3, TGF-β, and PI3K-Akt, converge into two transcriptional mechanisms, i.e., the NF-κB pathway and C/EBP family [6]. While our results showed thatATF7 significantly represses NF-κB activity, whether C/EBP is involved remains to be investigated.

Figure 6.

Figure 6.

Schematic role ofATF7 in senescence process.ATF7 influences senescence via three pathways. First,ATF7 represses activation of NF-κB signaling. Second,ATF7 silences target genes, including SASP-related genes, by recruiting histone H3K9 dimethyltransferase G9a. These two pathways synergistically decrease SASP factor production. Third,ATF7 directly inhibits ROS production, thus repressing senescence. These pathways may synergistically lead to lifespan extension inC. elegans and humans.

ATF7 is also reported to suppress a subset of genes associated with innate immunity by recruiting the H3K9 dimethyltransferase G9a [18]. For example, high H3K9me2 levels around the promoters of SASP factorsIL6 andIL8 have been shown to decrease their induction [58]. Our data indicated that overexpression ofATF7 markedly increased H3K9me2 levels, and ATF7 directly bound to and recruited G9a to dimethylate H3K9, further suppressing genes encoding SASP factors (Fig. 6). These findings suggest thatATF7 plays a role in delaying cellular senescence and inhibiting SASP production, which may function to regulate individual aging. Furthermore, our results showed that overexpression and depletion ofATF7 delayed and accelerated aging, and extended and shortenedC. elegans lifespan, respectively. However, the exact mechanism by whichATF7 protectsC. elegans from aging has not yet been determined as the immune system ofC. elegans differs from that of humans, although preliminary RNA-seq data suggest that innate immune functions may be involved. Taken together, our results suggest thatATF7 plays an important role in promoting healthy aging and longevity by regulating cellular senescence and inflammatory genes (Fig. 6).

In conclusion, we identified an up-regulation of transcription factorATF7 in LLIs and demonstrated its role in promoting longevity in a model organism.ATF7 inhibited senescence and SASP factors by suppressing NF-κB pathway activity and increasing H3K9 dimethylation, suggesting that it may inhibit persistent systemic chronic inflammation and promote lifespan in LLIs. However, further research is necessary to determine whetherATF7 may be a target for reducing age-related pathology associated with chronic inflammation.

Supplementary Materials

The Supplementary data can be found online at:www.aginganddisease.org/EN/10.14336/AD.2022.1217.

AD-14-4-1374-s.pdf (630KB, pdf)

Acknowledgments

This work was supported by grants from the National Key R&D Program of China (2018YFC2000400, 2018 YFE0203700, 2021YFF1201200). Additional support was received from the CAS Project for Young Scientists in Basic Research (YSBR-076), Key Research Program (KFZD-SW-221), West Light Foundation (to F.-H.X), and Pioneer Hundred Talents Program (to Y.H) of the Chinese Academy of Sciences, National Natural Science Foundation of China (82071595, 82171558), Basic Research Projects of Yunnan Province (202101 AT070299, 202101AS070058, 202201AS0700 80, 202101AS070137), High-level Talent Promotion and Training Project of Kunming (Spring City Plan; 2020SCP001), Yunling Scholar of Yunnan Province, and Project Program of National Clinical Research Center for Geriatric Disorders (Xiangya Hospital, 2021LNJJ03). We thank Z. Wang for assistance with the experiment.

Footnotes

Conflict of interest statement

The authors declare no conflicts of interest.

References

  • [1].Kennedy BK, Berger SL, Brunet A, Campisi J, Cuervo AM, Epel ES, et al. (2014). Geroscience: linking aging to chronic disease. Cell, 159:709-713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Goldberg EL, Dixit VD (2015). Drivers of age-related inflammation and strategies for healthspan extension. Immunol Rev, 265:63-74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Freund A, Orjalo AV, Desprez PY, Campisi J (2010). Inflammatory networks during cellular senescence: causes and consequences. Trends Mol Med, 16:238-246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G (2013). The hallmarks of aging. Cell, 153:1194-1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Sayed N, Huang Y, Nguyen K, Krejciova-Rajaniemi Z, Grawe AP, Gao T, et al. (2021). An inflammatory aging clock (iAge) based on deep learning tracks multimorbidity, immunosenescence, frailty and cardiovascular aging. Nat Aging, 1:598-615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Sun Y, Coppé JP, Lam EW (2018). Cellular Senescence: The Sought or the Unwanted?Trends Mol Med, 24:871-885. [DOI] [PubMed] [Google Scholar]
  • [7].Tian X, Seluanov A, Gorbunova V (2017). Molecular Mechanisms Determining Lifespan in Short- and Long-Lived Species. Trends Endocrinol Metab, 28:722-734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Engberg H, Oksuzyan A, Jeune B, Vaupel JW, Christensen K (2009). Centenarians--a useful model for healthy aging? A 29-year follow-up of hospitalizations among 40,000 Danes born in 1905. Aging Cell, 8:270-276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Xiao FH, Chen XQ, Yu Q, Ye Y, Liu YW, Yan D, et al. (2018). Transcriptome evidence reveals enhanced autophagy-lysosomal function in centenarians. Genome Res, 28:1601-1610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Xiao FH, Yu Q, Deng ZL, Yang K, Ye Y, Ge MX, et al. (2022). ETS1 acts as a regulator of human healthy aging via decreasing ribosomal activity. Sci Adv, 8:eabf2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].He YH, Pu SY, Xiao FH, Chen XQ, Yan DJ, Liu YW, et al. (2016). Improved lipids, diastolic pressure and kidney function are potential contributors to familial longevity: a study on 60 Chinese centenarian families. Sci Rep, 6:21962. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Li GH, Han F, Xiao FH, Gu KS, Shen Q, Xu W, et al. (2022). System-level metabolic modeling facilitates unveiling metabolic signature in exceptional longevity. Aging Cell, 21:e13595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Storci G, De Carolis S, Papi A, Bacalini MG, Gensous N, Marasco E, et al. (2019). Genomic stability, anti-inflammatory phenotype, and up-regulation of the RNAseH2 in cells from centenarians. Cell Death Differ, 26:1845-1858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Hai TW, Liu F, Coukos WJ, Green MR (1989). Transcription factor ATF cDNA clones: an extensive family of leucine zipper proteins able to selectively form DNA-binding heterodimers. Genes Dev, 3:2083-2090. [DOI] [PubMed] [Google Scholar]
  • [15].Maekawa T, Sakura H, Kanei-Ishii C, Sudo T, Yoshimura T, Fujisawa J, et al. (1989). Leucine zipper structure of the protein CRE-BP1 binding to the cyclic AMP response element in brain. Embo j, 8:2023-2028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Gaire M, Chatton B, Kedinger C (1990). Isolation and characterization of two novel, closely related ATF cDNA clones from HeLa cells. Nucleic Acids Res, 18:3467-3473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Walczynski J, Lyons S, Jones N, Breitwieser W (2014). Sensitisation of c-MYC-induced B-lymphoma cells to apoptosis by ATF2. Oncogene, 33:1027-1036. [DOI] [PubMed] [Google Scholar]
  • [18].Yoshida K, Maekawa T, Zhu Y, Renard-Guillet C, Chatton B, Inoue K, et al. (2015). The transcription factor ATF7 mediates lipopolysaccharide-induced epigenetic changes in macrophages involved in innate immunological memory. Nat Immunol, 16:1034-1043. [DOI] [PubMed] [Google Scholar]
  • [19].Maekawa T, Kim S, Nakai D, Makino C, Takagi T, Ogura H, et al. (2010). Social isolation stress induces ATF-7 phosphorylation and impairs silencing of the 5-HT 5B receptor gene. Embo j, 29:196-208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Fulop T, Larbi A, Dupuis G, Le Page A, Frost EH, Cohen AA, et al. (2017). Immunosenescence and Inflamm-Aging As Two Sides of the Same Coin: Friends or Foes?Front Immunol, 8:1960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Chen S, Zhou Y, Chen Y, Gu J (2018). fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics, 34:i884-i890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Kim D, Paggi JM, Park C, Bennett C, Salzberg SL (2019). Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat Biotechnol, 37:907-915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Liao Y, Smyth GK, Shi W (2014). featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics, 30:923-930. [DOI] [PubMed] [Google Scholar]
  • [24].Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. (2015). limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res, 43:e47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. (2003). Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res, 13:2498-2504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, et al. (2019). Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun, 10:1523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Huang da W, Sherman BT, Lempicki RA (2009). Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res, 37:1-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Huang da W, Sherman BT, Lempicki RA (2009). Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc, 4:44-57. [DOI] [PubMed] [Google Scholar]
  • [29].Li J, Tang H, Hu X, Chen M, Xie H (2010). Aquaporin-3 gene and protein expression in sun-protected human skin decreases with skin ageing. Australas J Dermatol, 51:106-112. [DOI] [PubMed] [Google Scholar]
  • [30].Lei D, Huang Y, Xie H, Yi Y, Long J, Lin S, et al. (2018). Fluorofenidone inhibits UV-A induced senescence in human dermal fibroblasts via the mammalian target of rapamycin-dependent SIRT1 pathway. J Dermatol, 45:791-798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Livak KJ, Schmittgen TD (2001). Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods, 25:402-408. [DOI] [PubMed] [Google Scholar]
  • [32].Langmead B, Salzberg SL (2012). Fast gapped-read alignment with Bowtie 2. Nat Methods, 9:357-359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].DePristo MA, Banks E, Poplin R, Garimella KV, Maguire JR, Hartl C, et al. (2011). A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet, 43:491-498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Feng J, Liu T, Qin B, Zhang Y, Liu XS (2012). Identifying ChIP-seq enrichment using MACS. Nat Protoc, 7:1728-1740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Yu G, Wang LG, He QY (2015). ChIPseeker: an R/Bioconductor package for ChIP peak annotation, comparison and visualization. Bioinformatics, 31:2382-2383. [DOI] [PubMed] [Google Scholar]
  • [36].Robinson JT, Thorvaldsdóttir H, Winckler W, Guttman M, Lander ES, Getz G, et al. (2011). Integrative genomics viewer. Nat Biotechnol, 29:24-26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Santoro A, Bientinesi E, Monti D (2021). Immunosenescence and inflammaging in the aging process: age-related diseases or longevity?Ageing Res Rev, 71:101422. [DOI] [PubMed] [Google Scholar]
  • [38].Chen C, Shi L, Li Y, Wang X, Yang S (2016). Disease-specific dynamic biomarkers selected by integrating inflammatory mediators with clinical informatics in ARDS patients with severe pneumonia. Cell Biol Toxicol, 32:169-184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [39].Liso A, Capitanio N, Gerli R, Conese M (2018). From fever to immunity: A new role for IGFBP-6?J Cell Mol Med, 22:4588-4596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Zhou S, Lu H, Xiong M (2021). Identifying Immune Cell Infiltration and Effective Diagnostic Biomarkers in Rheumatoid Arthritis by Bioinformatics Analysis. Front Immunol, 12:726747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Lee G, Kim YY, Jang H, Han JS, Nahmgoong H, Park YJ, et al. (2022). SREBP1c-PARP1 axis tunes anti-senescence activity of adipocytes and ameliorates metabolic imbalance in obesity. Cell Metab, 34:702-718.e705. [DOI] [PubMed] [Google Scholar]
  • [42].Di Micco R, Krizhanovsky V, Baker D, d'Adda di Fagagna F (2021). Cellular senescence in ageing: from mechanisms to therapeutic opportunities. Nat Rev Mol Cell Biol, 22:75-95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Benanti JA, Williams DK, Robinson KL, Ozer HL, Galloway DA (2002). Induction of extracellular matrix-remodeling genes by the senescence-associated protein APA-1. Mol Cell Biol, 22:7385-7397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Yao JJ, Liu Y, Lacorazza HD, Soslow RA, Scandura JM, Nimer SD, et al. (2007). Tumor promoting properties of the ETS protein MEF in ovarian cancer. Oncogene, 26:4032-4037. [DOI] [PubMed] [Google Scholar]
  • [45].Bourner LA, Muro I, Cooper AM, Choudhury BK, Bailey AO, Russell WK, et al. (2022). AhR promotes phosphorylation of ARNT isoform 1 in human T cell malignancies as a switch for optimal AhR activity. Proc Natl Acad Sci U S A, 119:e2114336119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Marthandan S, Baumgart M, Priebe S, Groth M, Schaer J, Kaether C, et al. (2016). Conserved Senescence Associated Genes and Pathways in Primary Human Fibroblasts Detected by RNA-Seq. PLoS One, 11:e0154531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Marthandan S, Priebe S, Groth M, Guthke R, Platzer M, Hemmerich P, et al. (2015). Hormetic effect of rotenone in primary human fibroblasts. Immun Ageing, 12:11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Herranz N, Gallage S, Mellone M, Wuestefeld T, Klotz S, Hanley CJ, et al. (2015). mTOR regulates MAPKAPK2 translation to control the senescence-associated secretory phenotype. Nat Cell Biol, 17:1205-1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Liu B, Maekawa T, Yoshida K, Ly NH, Inoue K, Hasegawa A, et al. (2019). Telomere shortening by transgenerational transmission of TNF-α-induced TERRA via ATF7. Nucleic Acids Res, 47:283-298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Yoshida K, Maekawa T, Ly NH, Fujita SI, Muratani M, Ando M, et al. (2020). ATF7-Dependent Epigenetic Changes Are Required for the Intergenerational Effect of a Paternal Low-Protein Diet. Mol Cell, 78:445-458.e446. [DOI] [PubMed] [Google Scholar]
  • [51].Faget DV, Ren Q, Stewart SA (2019). Unmasking senescence: context-dependent effects of SASP in cancer. Nat Rev Cancer, 19:439-453. [DOI] [PubMed] [Google Scholar]
  • [52].Tominaga K, Suzuki HI (2019). TGF-β Signaling in Cellular Senescence and Aging-Related Pathology. Int J Mol Sci, 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Gregson CL, Bergen DJM, Leo P, Sessions RB, Wheeler L, Hartley A, et al. (2020). A Rare Mutation in SMAD9 Associated With High Bone Mass Identifies the SMAD-Dependent BMP Signaling Pathway as a Potential Anabolic Target for Osteoporosis. J Bone Miner Res, 35:92-105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Lin L, Zhou Z, Zheng L, Alber S, Watkins S, Ray P, et al. (2008). Cross talk between Id1 and its interactive protein Dril1 mediate fibroblast responses to transforming growth factor-beta in pulmonary fibrosis. Am J Pathol, 173:337-346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Cao Y, Liu X, Zhang W, Deng X, Zhang H, Liu Y, et al. (2009). TGF-beta repression of Id2 induces apoptosis in gut epithelial cells. Oncogene, 28:1089-1098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Lecot P, Alimirah F, Desprez PY, Campisi J, Wiley C (2016). Context-dependent effects of cellular senescence in cancer development. Br J Cancer, 114:1180-1184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Freund A, Patil CK, Campisi J (2011). p38MAPK is a novel DNA damage response-independent regulator of the senescence-associated secretory phenotype. Embo j, 30:1536-1548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Takahashi A, Imai Y, Yamakoshi K, Kuninaka S, Ohtani N, Yoshimoto S, et al. (2012). DNA damage signaling triggers degradation of histone methyltransferases through APC/C(Cdh1) in senescent cells. Mol Cell, 45:123-131. [DOI] [PubMed] [Google Scholar]
  • [59].Kaya-Okur HS, Wu SJ, Codomo CA, Pledger ES, Bryson TD, Henikoff JG, et al. (2019). CUT&Tag for efficient epigenomic profiling of small samples and single cells. Nat Commun, 10:1930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Antebi A (2007). Genetics of aging in Caenorhabditis elegans. PLoS Genet, 3:1565-1571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Tao M, Li R, Zhang Z, Wu T, Xu T, Zogona D, et al. (2022). Vitexin and Isovitexin Act Through Inhibition of Insulin Receptor to Promote Longevity and Fitness in Caenorhabditis elegans. Mol Nutr Food Res:e2100845. [DOI] [PubMed] [Google Scholar]
  • [62].Shivers RP, Pagano DJ, Kooistra T, Richardson CE, Reddy KC, Whitney JK, et al. (2010). Phosphorylation of the conserved transcription factor ATF-7 by PMK-1 p38 MAPK regulates innate immunity in Caenorhabditis elegans. PLoS Genet, 6:e1000892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Fletcher M, Tillman EJ, Butty VL, Levine SS, Kim DH (2019). Global transcriptional regulation of innate immunity by ATF-7 in C. elegans. PLoS Genet, 15:e1007830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Nardini C, Moreau JF, Gensous N, Ravaioli F, Garagnani P, Bacalini MG (2018). The epigenetics of inflammaging: The contribution of age-related heterochromatin loss and locus-specific remodelling and the modulation by environmental stimuli. Semin Immunol, 40:49-60. [DOI] [PubMed] [Google Scholar]
  • [65].Youm YH, Grant RW, McCabe LR, Albarado DC, Nguyen KY, Ravussin A, et al. (2013). Canonical Nlrp3 inflammasome links systemic low-grade inflammation to functional decline in aging. Cell Metab, 18:519-532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Lim H, Park H, Kim HP (2015). Effects of flavonoids on senescence-associated secretory phenotype formation from bleomycin-induced senescence in BJ fibroblasts. Biochem Pharmacol, 96:337-348. [DOI] [PubMed] [Google Scholar]
  • [67].Kim HP (2022). The Long Search for Pharmacologically Useful Anti-Inflammatory Flavonoids and Their Action Mechanisms: Past, Present, and Future. Biomol Ther (Seoul), 30:117-125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].He YH, Zhang YX, Yang LQ, Liao XP, Zhang QY, Cai WW, et al. (2014). Assessment of the health status of centenarians in the south of China: a cross-sectional study. J Am Geriatr Soc, 62:1402-1404. [DOI] [PubMed] [Google Scholar]
  • [69].Hitt R, Young-Xu Y, Silver M, Perls T (1999). Centenarians: the older you get, the healthier you have been. Lancet, 354:652. [DOI] [PubMed] [Google Scholar]
  • [70].Gessert CE, Elliott BA, Haller IV (2002). Dying of old age: an examination of death certificates of Minnesota centenarians. J Am Geriatr Soc, 50:1561-1565. [DOI] [PubMed] [Google Scholar]
  • [71].Selim AJ, Fincke G, Berlowitz DR, Miller DR, Qian SX, Lee A, et al. (2005). Comprehensive health status assessment of centenarians: results from the 1999 large health survey of veteran enrollees. J Gerontol A Biol Sci Med Sci, 60:515-519. [DOI] [PubMed] [Google Scholar]
  • [72].Hasegawa H, Ishibashi K, Kubota S, Yamaguchi C, Yuki R, Nakajo H, et al. (2014). Cdk1-mediated phosphorylation of human ATF7 at Thr-51 and Thr-53 promotes cell-cycle progression into M phase. PLoS One, 9:e116048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Schaeffer E, Vigneron M, Sibler AP, Oulad-Abdelghani M, Chatton B, Donzeau M (2015). ATF7 is stabilized during mitosis in a CDK1-dependent manner and contributes to cyclin D1 expression. Cell Cycle, 14:2655-2666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Liu B, Maekawa T, Chatton B, Ishii S (2016). In utero TNF-α treatment induces telomere shortening in young adult mice in an ATF7-dependent manner. FEBS Open Bio, 6:56-63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Maekawa T, Liu B, Nakai D, Yoshida K, Nakamura KI, Yasukawa M, et al. (2018). ATF7 mediates TNF-α-induced telomere shortening. Nucleic Acids Res, 46:4487-4504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Wu Z, Isik M, Moroz N, Steinbaugh MJ, Zhang P, Blackwell TK (2019). Dietary Restriction Extends Lifespan through Metabolic Regulation of Innate Immunity. Cell Metab, 29:1192-1205.e1198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Maekawa T, Liu B, Liu Y, Yoshida K, Muratani M, Chatton B, et al. (2019). Stress-induced and ATF7-dependent epigenetic change influences cellular senescence. Genes Cells, 24:627-635. [DOI] [PubMed] [Google Scholar]
  • [78].Hernandez-Segura A, Nehme J, Demaria M (2018). Hallmarks of Cellular Senescence. Trends Cell Biol, 28:436-453. [DOI] [PubMed] [Google Scholar]
  • [79].Coppé JP, Patil CK, Rodier F, Sun Y, Muñoz DP, Goldstein J, et al. (2008). Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol, 6:2853-2868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Rittchen S, Heinemann A (2019). Therapeutic Potential of Hematopoietic Prostaglandin D(2) Synthase in Allergic Inflammation. Cells, 8:619. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Méndez-García LA, Escobedo G, Minguer-Uribe AG, Viurcos-Sanabria R, Aguayo-Guerrero JA, Carrillo-Ruiz JD, et al. (2022). Role of the renin-angiotensin system in the development of COVID-19-associated neurological manifestations. Front Cell Neurosci, 16:977039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Brisson L, Carrier A (2015). A novel actor in antitumoral immunity: The thymus-specific serine protease TSSP/PRSS16 involved in CD4(+) T-cell maturation. Oncoimmunology, 4:e1026536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Xie Z, Meng J, Kong W, Wu Z, Lan F, Narengaowa, et al. (2022). Microglial cathepsin E plays a role in neuroinflammation and amyloid β production in Alzheimer's disease. Aging Cell, 21:e13565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Weng HJ, Tsai TF (2021). ABCB1 in dermatology: roles in skin diseases and their treatment. J Mol Med (Berl), 99:1527-1538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Fleming TJ, Schrankel CS, Vyas H, Rosenblatt HD, Hamdoun A (2021). CRISPR/Cas9 mutagenesis reveals a role for ABCB1 in gut immune responses to Vibrio diazotrophicus in sea urchin larvae. J Exp Biol, 224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Mao H, Jia J, Sheng J, Zhang S, Huang K, Li H, et al. (2021). Protective and anti-inflammatory role of REG1A in inflammatory bowel disease induced by JAK/STAT3 signaling axis. Int Immunopharmacol, 92:107304. [DOI] [PubMed] [Google Scholar]
  • [87].Houzelstein D, Reyes-Gomez E, Maurer M, Netter P, Higuet D (2013). Expression patterns suggest that despite considerable functional redundancy, galectin-4 and -6 play distinct roles in normal and damaged mouse digestive tract. J Histochem Cytochem, 61:348-361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Li N, Wang N, He W, Feng Y, Qiu Q, Qiu H, et al. (2022). The suppressive functions of Rora in B lineage cell proliferation and BCR/ABL1-induced B-ALL pathogenesis. Int J Biol Sci, 18:2277-2291. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

The Supplementary data can be found online at:www.aginganddisease.org/EN/10.14336/AD.2022.1217.

AD-14-4-1374-s.pdf (630KB, pdf)

Articles from Aging and Disease are provided here courtesy ofJKL International LLC

ACTIONS

RESOURCES


[8]ページ先頭

©2009-2025 Movatter.jp