The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely. 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
Cancer Sci. 2020 Feb; 111(2): 304–311.
Published online 2019 Dec 27. doi: 10.1111/cas.14266
PMCID: PMC7004529

Cellular senescence and senescence‐associated secretory phenotype via the cGAS‐STING signaling pathway in cancer

Tze Mun Loo

Project for Cellular Senescence, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo Japan

Find articles by Tze Mun Loo

Kenichi Miyata

Project for Cellular Senescence, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo Japan

Find articles by Kenichi Miyata

Yoko Tanaka

Project for Cellular Senescence, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo Japan

Find articles by Yoko Tanaka

Akiko Takahashi

Project for Cellular Senescence, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo Japan PRESTO, Saitama Japan PRIME, AMED, Tokyo Japan Project for Cellular Senescence, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo Japan PRESTO, Saitama Japan PRIME, AMED, Tokyo Japan

Akiko Takahashi, pj.ro.rcfj@ihsahakat.okika .
corresponding author Corresponding author.
* Correspondence
Akiko Takahashi, Project for Cellular Senescence, The Cancer Institute, Japanese Foundation for Cancer Research, 3‐8‐31, Ariake, Koto‐ku, Tokyo 135‐8550, Japan.

1. FEATURES OF CELLULAR SENESCENCE

Cellular senescence is defined as a state of irreversible cell cycle arrest to prevent proliferation of damaged cells and reduce the risk of cancer. Therefore, cellular senescence is believed to be an essential tumor suppression mechanism in vivo. 1 , 2 Hayflick and Moorhead 3 initially mentioned “cellular senescence” after observing that primary human cells have a maximal number of cell proliferations in vitro. This proliferation limit caused by telomere shortening has been termed “replicative senescence.” Similar phenotypes can be induced by activation of certain oncogenes, “oncogene‐induced senescence (OIS),” 4 and a variety of stressors, such as irradiation and oxidative stress. 1 , 5 Furthermore, recent studies have demonstrated that treatment with chemotherapeutic drugs or ionizing radiation provokes “therapy‐induced senescence (TIS)” in tumor cells. 6 , 7 Persistent DNA damage response causes cellular senescence in normal cells, 8 , 9 and induces expression of the cyclin‐dependent kinase (CDK) inhibitors p16 INK4a and p21 WAF1/CIP1 . p16 INK4a inhibits the cyclin D‐CDK4/6 complex, 10 whereas p21 WAF1/CIP1 blocks cyclin E‐CDK2 activity. 11 Both checkpoint proteins block retinoblastoma protein, thereby suppressing expression of E2F target genes and inducing senescent cell cycle arrest (Figure (Figure1 1 ). 12 , 13 , 14 Generally, cellular senescence is a state of essentially irreversible cell cycle arrest; however, several studies have reported that a small population of senescent cells can override the senescent cell cycle arrest and re‐enter the cell cycle under some particular conditions. 15 , 16 Recently, the selective CDK4/6 inhibitors abemaciclib, palbociclib and ribociclib have been shown to induce senescence‐like cell cycle arrest in some cancer cell lines. These effects led to their approval for estrogen receptor‐positive breast cancer treatment. 17 , 18

An external file that holds a picture, illustration, etc. Object name is CAS-111-304-g001.jpg

The characteristics of cellular senescence. Cellular senescence is triggered by various stressors, such as irradiation, reactive oxygen species (ROS), oncogene activation and telomere shortening. The senescent cells show increased expression of p16 INK4a and/or p21 WAF1/CIP1 , senescence‐associated secretory phenotype (SASP) factors, DNA damage foci and senescence‐associated heterochromatin foci (SAHF) formation. In addition, cell proliferation is inhibited and Lamin B1 expression is decreased. Senescent cell cycle arrest is induced by p16 INK4a and/or by p21 WAF1/CIP1

Senescent cells show some typical morphological features, such as a flattened shape and vacuolization. 19 As persistent DNA damage signals are critical for induction of cellular senescence, it has been suggested that DNA damage foci could be used to identify senescent cells. Potential targets include accumulation of 53BP1, phosphorylation of histone H2A.X, or other DNA repair markers. 20 In addition, chromatin reorganization occurs in senescent cells, termed senescence‐associated heterochromatin foci (SAHF), which colocalize with trimethylated histone H3 lysine 9 (H3K9me3) and heterochromatin protein 1 and suppress the transcription of proliferation‐related genes. 21 Moreover, downregulation of the nuclear lamina protein Lamin B1 also serves as a marker for senescent cells. 22 Of these, senescence‐associated β‐galactosidase (SA‐β‐gal) staining is the most common and popular marker for identification of senescent cells. 23 SA‐β‐gal activity is based on upregulation of the lysosomal β‐galactosidase gene ( GLB1 ) and is detectable at pH 6. 24 However, SA‐β‐gal activity is not specific for senescent cells. 14 Therefore, a combination of several markers is needed to identify senescent cells.

2. DUAL ROLES OF CELLULAR SENESCENCE IN TUMORIGENESIS

In 2005, several papers reported that cellular senescence occurs in premalignant tumors and benign tumors to prevent cancer development. 25 , 26 , 27 , 28 Coinciding with these reports, it was demonstrated that double knockout of p16 INK4a and p21 WAF1/CIP1 genes increased the rate of cancer development. 29 These data strongly indicated that cellular senescence is an essential tumor suppression mechanism in vivo . Mice have also been generated to visualize the dynamics of p16 INK4a and p21 WAF1/CIP1 expression during the aging process. Real‐time imaging analysis revealed that senescent cells accumulated in benign tumors and throughout the body with age, 30 , 31 suggesting that senescent cells may also have biological roles in aged tissues of the living body.

In the recent decade, many studies have demonstrated that senescent cells secrete a variety of proteins, such as inflammatory cytokines, chemokines, growth factors and MMP. Collectively, this phenotype was termed senescence‐associated secretory phenotype (SASP). 32 , 33 , 34 There is a myriad of physiological activity associated with SASP factors (Figure (Figure2). 2 ). At first, SASP factors secreted from senescent cells were regarded as paracrine and autocrine enhancers of tumor suppressive effects in cellular senescence to prevent the growth of damaged cells. 33 , 34 , 35 In addition to this function, SASP factors can recruit immune cells to clear the senescent cells, which is termed “senescence‐surveillance.” 36 Senescence surveillance results in clearance of the senescent cells and stimulates the local immune reaction to eliminate oncogene‐expressing cells. Thus, SASP factors might contribute to a fail‐safe mechanism of cellular senescence. 15 , 36 Furthermore, it has been reported that some SASP factors can promote tissue repairing under a variety of conditions. Demaria et al 37 demonstrated that platelet‐derived factor‐AA (PDGF‐AA) was secreted from senescent skin fibroblasts in the vicinity of damaged tissues to promote optimal wound healing. In response to liver injury, hepatic stellate cells (HSC) began to proliferate and produce extracellular matrixes for tissue repair before undergoing cellular senescence to prevent fibrosis. 38 , 39 Moreover, these senescent HSC secreted growth factors, thereby promoting cell proliferation for tissue regeneration. In addition to tissue repair, SASP factors are involved in embryonic development of the apical ectodermal ridge and neural roof plate. 40 , 41

An external file that holds a picture, illustration, etc. Object name is CAS-111-304-g002.jpg

The biological function of senescence‐associated secretory phenotype (SASP) factors in physiological and pathological conditions. SASP factors play important roles in common physiological conditions, as shown by the red arrows, such as wound healing (PDGF‐AA, platelet‐derived growth factor‐AA), embryonic development (TGF‐β, transforming growth factor‐β; Wnt) and immune recruitment (CXCL1, CCL2 and CCL5). However, SASP factors also induce tumor progression and migration (MMP; IL‐1β, interleukin‐1β; VEGF, vascular endothelial growth factor) and suppression of anti–tumor immunity (PGE 2 , prostaglandin E2) under pathological conditions, as shown by the blue arrows. Moreover, SASP factors induce and maintain senescence cell cycle arrest through paracrine and autocrine factors

In contrast to the beneficial effects of SASP in the living body, long‐term survival of senescent cells also drives age‐related diseases, such as atherosclerosis, neuropsychiatric disorders, chronic nephritis and cancer. 42 During the aging process, declining immune function leads to failures in senescence surveillance, resulting in accumulation of senescent cells. 43 Therefore, prolonged secretion of SASP factors, such as interleukin (IL)‐1β and MMP, could drive cancer development. 44 , 45 , 46 Intriguingly, it has been reported that upregulation of IL‐8 in senescent carcinoma‐associated fibroblasts promotes invasion and metastasis of pancreatic cancer cells. 47 Our previous studies have established that IL‐1β secreted from senescent HSC in the liver plays an essential role in promoting obesity‐associated hepatocellular carcinoma (HCC) in the obese mice model. Increased levels of deoxycholic acid (DCA), a gut bacterial metabolite derived from altered gut microbiota in obesity, induced cellular senescence and production of SASP factors in HSC, thereby promoting development of obesity‐associated HCC. 45 Moreover, it was found that expression of COX‐2, a rate‐limiting enzyme involved in prostaglandin biosynthesis, significantly increased in senescent HSC. COX‐2 induces overproduction of prostaglandin E 2 (PGE 2 ), resulting in suppression of anti–tumor immunity and progression of obesity‐associated HCC. 48 Remarkably, cellular senescence has beneficial and harmful effects on cancer development via SASP.

3. MOLECULAR MECHANISM OF SENESCENCE‐ASSOCIATED SECRETORY PHENOTYPE INDUCTION

According to previous reports, numerous mechanisms have been implicated in the regulation of SASP factors. First, the transcription factors nuclear factor κB (NF‐κB) and CCAAT/enhancer‐binding protein (C/EBP‐β) were identified as positive regulators of SASP factor expression. 33 , 34 , 49 Second, elevated IL‐1α in the early stage of cellular senescence initiates the inflammatory signaling cascade and forms a positive feedback system to amplify SASP signaling. 50

Notably, gene expression of SASP factors is regulated by epigenetic mechanisms. It has been previously reported that persistent DNA damage leads to proteasomal degradation of the major histone H3K9 dimethyltransferases, G9a and GLP, causing increased SASP factor genes in senescent cells. 51 Other epigenetic regulators, such as macroH2A1, high mobility group box 2 (HMGB2), mixed‐lineage leukemia 1 (MLL1) and bromodomain‐containing protein 4 (BRD4), have been demonstrated as regulators of SASP factor gene expression in response to DNA damage. 52 , 53 , 54 , 55 In addition, the nutrient sensing pathways, mTOR and NAD, are involved in SASP regulation. 56 , 57 , 58

Recently, it has become apparent that toll‐like receptors (TLR), an innate immune receptor, can trigger SASP induction. 48 , 59 , 60 TLR are pattern‐recognition receptors that can recognize microbe‐specific molecular signatures, known as pathogen‐associated molecular patterns (PAMP) and self‐derived molecules from damaged cells, known as damage‐associated molecules patterns (DAMP). 61 A recent study by Hari et al indicated that the acute‐phase serum amyloids A1 and A2 function as DAMP and are recognized by TLR2. Thereafter, TLR2 initiates the inflammatory signaling cascade and induces SASP. 60 It has also been reported that TLR2 is stimulated by lipoteichoic acid (LTA), a component of the cell wall of gram‐positive bacteria. TLR2 stimulation induces expression of SASP factors through the NF‐κB signaling pathway to promote development of obesity‐associated HCC. 48 In concordance with these findings, HMGB1 was also recognized by TLR4, resulting in increased SASP factor secretion. 59 Taken together, these results indicate that senescent cells secrete pro–inflammatory factors in response to genotoxic stress and this may be associated with pathobiological effects in the cancer microenvironment.

4. A NOVEL PATHWAY FOR SENESCENCE‐ASSOCIATED SECRETORY PHENOTYPE INDUCTION: cGAS‐STING SIGNALING

The antiviral cyclic GMP‐AMP synthase (cGAS)‐stimulator of interferon genes (STING) signaling pathway has recently been identified as a SASP regulator. 62 , 63 , 64 cGAS was originally observed as a cytosolic DNA sensor that recognizes pathogenic DNA and induces innate immune responses. Once cGAS detects pathogenic DNA, it generates cyclic GMP‐AMP (cGAMP). In turn, cGAMP acts as the second messenger that stimulates STING, leading to recruitment and autophosphorylation of TANK‐binding kinase 1 (TBK1). Thereafter, TBK1 phosphorylates interferon‐regulatory factor 3 (IRF3) transcription factor, resulting in translocation of IRF3 from the cytosol to the nucleus and stimulating transcription of type‐I interferon (IFN). TBK1 also activates NF‐κB to induce expression of IFN and inflammatory cytokines. 65 , 66 , 67

The emerging evidence indicates that the cGAS‐STING pathway detects self‐derived DNA fragments and activates SASP in senescent cells (Figure (Figure3 3 ). 62 , 64 There are several possibilities for the candidate ligands that activate the DNA‐sensing machinery underlying cellular senescence. Downregulation of Lamin B1 leads to collapse of the nuclear envelope, which can trigger release of chromatin fragments from the nucleus to the cytosol, termed cytoplasmic chromatin fragments (CCF). 68 DNA damage promotes the production of cytosolic DNA, such as CCF, mtDNA, cDNA, micronuclei and nuclear buds. 69 , 70 However, the mechanism of cytosolic DNA accumulation in the cytoplasm of senescent cells has remained unclear. Therefore, we focused on this molecular mechanism. In normal cells, the cytoplasmic DNases, DNase2 and TREX1, degrade the double‐stranded and single‐stranded DNA in the cytoplasm to prevent activation of the innate immune response. 71 , 72 However, downregulation of DNase2 and TREX1, which are transcriptional targets of the E2F complex, results in accumulation of cytosolic DNA, promoting SASP in senescent cells. 73

An external file that holds a picture, illustration, etc. Object name is CAS-111-304-g003.jpg

The cGAS‐STING pathway in senescence‐associated secretory phenotype (SASP) regulation. Various stressors such as reactive oxygen species (ROS) or UV irradiation cause accumulation of DNA fragments from nucleic double‐strand breaks (DSB), termed cytoplasmic chromatin fragments (CCF), in senescent cells. Damaged mitochondria release mtDNA into the cytoplasm. Long‐interspersed element‐1 (LINE‐1) transcription is upregulated, thereby promoting cDNA production in senescent cells. These DNA fragments are recognized by cGAS to generate 2′3′‐cyclic GMP‐AMP (2′3′‐cGAMP). 2′3′‐cGAMP activates both STING and TANK‐binding kinase 1 (TBK1), resulting in phosphorylation of IRF3. 2′3′‐cGAMP also activates IκBa. These transcription factors enter the nucleus and induce expression of type‐I interferon (IFN) and inflammatory cytokines. Both IRF3 and NF‐κB induce senescence‐associated secretory phenotype (SASP) factors such as IFN‐β, IL‐6 and IL‐8, which are known to induce ROS and maintain cellular senescence

De Cecco et al 74 reported high transcription levels of the long‐interspersed element‐1 (LINE‐1, also known as L1), a retrotransposable element, in senescent cells. LINE‐1 possesses high reverse transcriptase activity, which can transcribe mRNA to cDNA in the cytosol. 75 Therefore, increases in LINE‐1 transcription during senescence facilitates accumulation of cDNA in the cytoplasm and triggers cGAS‐STING signaling to produce SASP factors.

We propose that senescent cells secrete not only inflammatory proteins but also small extracellular vesicles, such as exosomes. 76 , 77 Interestingly, cGAMP is also secreted from cancer cells via SLC19A1, which induces activation of the STING pathway in recipient cells. 78 , 79 There is a possibility that senescent cells secrete cGAMP to promote the paracrine innate immune response via SLC19A1.

5. THE cGAS‐STING PATHWAY AND CANCER

Chronic inflammation caused by SASP via the cGAS‐STING pathway is crucial for the development of HCC in the obesity‐induced liver cancer model. 73 Knocking out the STING gene blunted SASP factor production in HSC and attenuated the development of obesity‐associated HCC in the mouse model. 73 However, Dou et al 68 demonstrated that inhibiting the cGAS‐STING signaling pathway impaired the immuno‐surveillance of senescent hepatocytes and premalignant hepatocytes, resulting in tumorigenesis in the liver. Accordingly, function of the cGAS‐STING pathway appears to depend on the biological context (Figure (Figure4). 4 ). Although short‐term exposure to SASP factors may encourage immuno‐surveillance and prevent tumorigenesis, persistent exposure to SASP factors may cause tissue damage and chronic inflammation linked to tumor growth.

An external file that holds a picture, illustration, etc. Object name is CAS-111-304-g004.jpg

The cGAS‐STING pathway plays two different roles in liver tumorigenesis. DNA damage induces cellular senescence in hepatic stellate cells (HSC) and hepatocytes in the liver. Accumulation of cytosolic DNA leads to activation of the cGAS‐STING pathway, resulting in production of senescence‐associated secretory phenotype (SASP) factors. Short‐term exposure to SASP factors drives recruitment of immune cells to clear pre–malignant cells and senescent cells, thereby preventing tumorigenesis. However, long‐term exposure to SASP factors generates chronic inflammation and promotes tumorigenesis in obese mice

The importance of STING‐associated inflammation has also been reported in lung cancer. Kitajima et al provided evidence that STING expression was silenced in KRAS‐LKB1 mutant lung cancer. LKB1 is the upstream activator of AMPK. Double mutation of KRAS and LKB1 genes in lung cancer resulted in failure to respond to immune checkpoint blockade. 80 Reinduction of LKB1 and STING restored the immune checkpoint blockade response and promoted T‐cell chemotaxis. Collectively, production of SASP factors via the cGAS‐STING signaling pathway plays a prominent role in promoting or inhibiting tumorigenesis depending on the biological context in the cancer microenvironment.

6. CONCLUSION

In this review, we illustrated that SASP factors can induce immuno‐surveillance and act as a tumor suppression mechanism. Conversely, SASP factors may provoke both tumorigenesis and age‐related diseases due to exposure to chronic inflammation. Many researchers have attempted to establish senolytic drugs to eliminate harmful senescent cells from aged tissues, thereby preventing cancer and age‐related pathologies. Several senolytic drugs, such as ABT‐737, BET inhibitors and dasatinib plus quercetin, can selectively induce cell death in senescent cells and effectively improve some symptoms of age‐related disease, including cancer, by attenuating SASP factor production. 81 However, the off‐target effects of these senolytic molecules are still unknown. In addition, evaluating the efficiency of senolytic therapies in the living body is an important factor to optimize treatment. Thus, we need to carefully consider the clinical application of senolytic drugs for cancer treatment and prevention of age‐related disorders. The cGAS‐STING pathway may be an alternative potential therapeutic target to control cancer. 82 Based on the role of the cGAS‐STING pathway in facilitating anti–tumor immunity and SASP induction, modulation of its activity would be beneficial for cancer immune therapy and to inhibit cancer development. Finally, to extend healthy lifespans via effective therapeutic strategies, significantly more research is necessary to investigate the molecular mechanisms of SASP regulation.

DISCLOSURE

The authors have no conflict of interest.

ACKNOWLEDGMENTS

We are grateful to all members of the Takahashi laboratorty for their helpful comments on this manuscript. This work was supported in part by grants from the Japan Science and Technology Agency (JST)‐PRESTO under grant number JPMJPR17H7, from the Japan Agency of Medical Research and Development (AMED) under grant number 19gm6110023h0001 and from the Japan Society for the Promotion of Science (JSPS) under grant numbers 18K15073, 18K15254 and 19H03507, the Princess Takamatsu Cancer Research Fund, the Vehicle Racing Commemorative Foundation and the Takeda Science Foundation. This research was also supported by the Research Fellowships for Young Scientists from JSPS under grant number 19J00796.

Notes

Loo TM, Miyata K, Tanaka Y, Takahashi A. Cellular senescence and senescence‐associated secretory phenotype via the cGAS‐STING signaling pathway in cancer . Cancer Sci . 2020; 111 :304–311. 10.1111/cas.14266 [ PMC free article ] [ PubMed ] [ CrossRef ] [ Google Scholar ]

REFERENCES

1. Campisi J. Aging, cellular senescence, and cancer . Annu Rev Physiol . 2013; 75 :685‐705. [ PMC free article ] [ PubMed ] [ Google Scholar ]
2. Munoz‐Espin D, Serrano M. Cellular senescence: from physiology to pathology . Nat Rev Mol Cell Biol . 2014; 15 :482‐496. [ PubMed ] [ Google Scholar ]
3. Hayflick L, Moorhead PS. The serial cultivation of human diploid cell strains . Exp Cell Res . 1961; 25 :585‐621. [ PubMed ] [ Google Scholar ]
4. Serrano M, Lee H, Chin L, Cordon‐Cardo C, Beach D, DePinho RA. Role of the INK4a locus in tumor suppression and cell mortality . Cell . 1996; 85 :27‐37. [ PubMed ] [ Google Scholar ]
5. McHugh D, Gil J. Senescence and aging: causes, consequences, and therapeutic avenues . J Cell Biol . 2018; 217 :65‐77. [ PMC free article ] [ PubMed ] [ Google Scholar ]
6. Ewald JA, Desotelle JA, Wilding G, Jarrard DF. Therapy‐induced senescence in cancer . J Natl Cancer Inst . 2010; 102 :1536‐1546. [ PMC free article ] [ PubMed ] [ Google Scholar ]
7. Fan DNY, Schmitt CA. Genotoxic stress‐induced senescence . Methods Mol Biol . 1896; 2019 :93‐105. [ PubMed ] [ Google Scholar ]
8. Bartkova J, Rezaei N, Liontos M, et al. Oncogene‐induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints . Nature . 2006; 444 :633‐637. [ PubMed ] [ Google Scholar ]
9. Di Micco R, Fumagalli M, Cicalese A, et al. Oncogene‐induced senescence is a DNA damage response triggered by DNA hyper‐replication . Nature . 2006; 444 :638‐642. [ PubMed ] [ Google Scholar ]
10. Ohtani N, Zebedee Z, Huot TJ, et al. Opposing effects of Ets and Id proteins on p16INK4a expression during cellular senescence . Nature . 2001; 409 :1067‐1070. [ PubMed ] [ Google Scholar ]
11. Brugarolas J, Chandrasekaran C, Gordon JI, Beach D, Jacks T, Hannon GJ. Radiation‐induced cell cycle arrest compromised by p21 deficiency . Nature . 1995; 377 :552‐557. [ PubMed ] [ Google Scholar ]
12. Maehara K, Yamakoshi K, Ohtani N, et al. Reduction of total E2F/DP activity induces senescence‐like cell cycle arrest in cancer cells lacking functional pRB and p53 . J Cell Biol . 2005; 168 :553‐560. [ PMC free article ] [ PubMed ] [ Google Scholar ]
13. Takahashi A, Ohtani N, Yamakoshi K, et al. Mitogenic signalling and the p16INK4a‐Rb pathway cooperate to enforce irreversible cellular senescence . Nat Cell Biol . 2006; 8 :1291‐1297. [ PubMed ] [ Google Scholar ]
14. Imai Y, Takahashi A, Hanyu A, et al. Crosstalk between the Rb pathway and AKT signaling forms a quiescence‐senescence switch . Cell Rep . 2014; 7 :194‐207. [ PubMed ] [ Google Scholar ]
15. Lee S, Schmitt CA. The dynamic nature of senescence in cancer . Nat Cell Biol . 2019; 21 :94‐101. [ PubMed ] [ Google Scholar ]
16. Gorgoulis V, Adams PD, Alimonti A, et al. Cellular senescence: defining a path forward . Cell . 2019; 179 :813‐827. [ PubMed ] [ Google Scholar ]
17. Pernas S, Tolaney SM, Winer EP, Goel S. CDK4/6 inhibition in breast cancer: current practice and future directions . Ther Adv Med Oncol . 2018; 10 :1758835918786451. [ PMC free article ] [ PubMed ] [ Google Scholar ]
18. Klein ME, Kovatcheva M, Davis LE, Tap WD, Koff A. CDK4 /6 inhibitors: the mechanism of action may not be as simple as once thought . Cancer Cell . 2018; 34 :9–20. [ PMC free article ] [ PubMed ] [ Google Scholar ]
19. Kuilman T, Michaloglou C, Mooi WJ, Peeper DS. The essence of senescence . Genes Dev . 2010; 24 :2463‐2479. [ PMC free article ] [ PubMed ] [ Google Scholar ]
20. Hernandez‐Segura A, Nehme J, Demaria M. Hallmarks of cellular senescence . Trends Cell Biol . 2018; 28 :436‐453. [ PubMed ] [ Google Scholar ]
21. Narita M, Nunez S, Heard E, et al. Rb‐mediated heterochromatin formation and silencing of E2F target genes during cellular senescence . Cell . 2003; 113 :703‐716. [ PubMed ] [ Google Scholar ]
22. Freund A, Laberge RM, Demaria M, Campisi J. Lamin B1 loss is a senescence‐associated biomarker . Mol Biol Cell . 2012; 23 :2066‐2075. [ PMC free article ] [ PubMed ] [ Google Scholar ]
23. Dimri GP, Lee X, Basile G, et al. A biomarker that identifies senescent human cells in culture and in aging skin in vivo . Proc Natl Acad Sci USA . 1995; 92 :9363‐9367. [ PMC free article ] [ PubMed ] [ Google Scholar ]
24. Lee BY, Han JA, Im JS, et al. Senescence‐associated beta‐galactosidase is lysosomal beta‐galactosidase . Aging Cell . 2006; 5 :187‐195. [ PubMed ] [ Google Scholar ]
25. Michaloglou C, Vredeveld LC, Soengas MS, et al. BRAFE600‐associated senescence‐like cell cycle arrest of human naevi . Nature . 2005; 436 :720‐724. [ PubMed ] [ Google Scholar ]
26. Braig M, Lee S, Loddenkemper C, et al. Oncogene‐induced senescence as an initial barrier in lymphoma development . Nature . 2005; 436 :660‐665. [ PubMed ] [ Google Scholar ]
27. Chen Z, Trotman LC, Shaffer D, et al. Crucial role of p53‐dependent cellular senescence in suppression of Pten‐deficient tumorigenesis . Nature . 2005; 436 :725‐730. [ PMC free article ] [ PubMed ] [ Google Scholar ]
28. Collado M, Gil J, Efeyan A, et al. Tumour biology: senescence in premalignant tumours . Nature . 2005; 436 :642. [ PubMed ] [ Google Scholar ]
29. Takeuchi S, Takahashi A, Motoi N, et al. Intrinsic cooperation between p16INK4a and p21Waf1/Cip1 in the onset of cellular senescence and tumor suppression in vivo . Cancer Res . 2010; 70 :9381‐9390. [ PubMed ] [ Google Scholar ]
30. Yamakoshi K, Takahashi A, Hirota F, et al. Real‐time in vivo imaging of p16Ink4a reveals cross talk with p53 . J Cell Biol . 2009; 186 :393‐407. [ PMC free article ] [ PubMed ] [ Google Scholar ]
31. Ohtani N, Imamura Y, Yamakoshi K, et al. Visualizing the dynamics of p21(Waf1/Cip1) cyclin‐dependent kinase inhibitor expression in living animals . Proc Natl Acad Sci USA . 2007; 104 :15034‐15039. [ PMC free article ] [ PubMed ] [ Google Scholar ]
32. Wajapeyee N, Serra RW, Zhu X, Mahalingam M, Green MR. Oncogenic BRAF induces senescence and apoptosis through pathways mediated by the secreted protein IGFBP7 . Cell . 2008; 132 :363‐374. [ PMC free article ] [ PubMed ] [ Google Scholar ]
33. Kuilman T, Michaloglou C, Vredeveld LC, et al. Oncogene‐induced senescence relayed by an interleukin‐dependent inflammatory network . Cell . 2008; 133 :1019‐1031. [ PubMed ] [ Google Scholar ]
34. Acosta JC, O'Loghlen A, Banito A, et al. Chemokine signaling via the CXCR2 receptor reinforces senescence . Cell . 2008; 133 :1006‐1018. [ PubMed ] [ Google Scholar ]
35. Acosta JC, Banito A, Wuestefeld T, et al. A complex secretory program orchestrated by the inflammasome controls paracrine senescence . Nat Cell Biol . 2013; 15 :978‐990. [ PMC free article ] [ PubMed ] [ Google Scholar ]
36. Kang TW, Yevsa T, Woller N, et al. Senescence surveillance of pre–malignant hepatocytes limits liver cancer development . Nature . 2011; 479 :547‐551. [ PubMed ] [ Google Scholar ]
37. Demaria M, Ohtani N, Youssef SA, et al. An essential role for senescent cells in optimal wound healing through secretion of PDGF‐AA . Dev Cell . 2014; 31 :722‐733. [ PMC free article ] [ PubMed ] [ Google Scholar ]
38. Schnabl B, Purbeck CA, Choi YH, Hagedorn CH, Brenner D. Replicative senescence of activated human hepatic stellate cells is accompanied by a pronounced inflammatory but less fibrogenic phenotype . Hepatology . 2003; 37 :653‐664. [ PubMed ] [ Google Scholar ]
39. Krizhanovsky V, Yon M, Dickins RA, et al. Senescence of activated stellate cells limits liver fibrosis . Cell . 2008; 134 :657‐667. [ PMC free article ] [ PubMed ] [ Google Scholar ]
40. Storer M, Mas A, Robert‐Moreno A, et al. Senescence is a developmental mechanism that contributes to embryonic growth and patterning . Cell . 2013; 155 :1119‐1130. [ PubMed ] [ Google Scholar ]
41. Munoz‐Espin D, Canamero M, Maraver A, et al. Programmed cell senescence during mammalian embryonic development . Cell . 2013; 155 :1104‐1118. [ PubMed ] [ Google Scholar ]
42. He S, Sharpless NE. Senescence in health and disease . Cell . 2017; 169 :1000‐1011. [ PMC free article ] [ PubMed ] [ Google Scholar ]
43. Ovadya Y, Landsberger T, Leins H, et al. Impaired immune surveillance accelerates accumulation of senescent cells and aging . Nat Commun . 2018; 9 :5435. [ PMC free article ] [ PubMed ] [ Google Scholar ]
44. Freund A, Orjalo AV, Desprez PY, Campisi J. Inflammatory networks during cellular senescence: causes and consequences . Trends Mol Med . 2010; 16 :238‐246. [ PMC free article ] [ PubMed ] [ Google Scholar ]
45. Yoshimoto S, Loo TM, Atarashi K, et al. Obesity‐induced gut microbial metabolite promotes liver cancer through senescence secretome . Nature . 2013; 499 :97‐101. [ PubMed ] [ Google Scholar ]
46. Faget DV, Ren Q, Stewart SA. Unmasking senescence: context‐dependent effects of SASP in cancer . Nat Rev Cancer . 2019; 19 :439‐453. [ PubMed ] [ Google Scholar ]
47. Wang T, Notta F, Navab R, et al. Senescent carcinoma‐associated fibroblasts upregulate IL8 to enhance prometastatic phenotypes . Mol Cancer Res . 2017; 15 :3‐14. [ PubMed ] [ Google Scholar ]
48. Loo TM, Kamachi F, Watanabe Y, et al. Gut microbiota promotes obesity‐associated liver cancer through PGE2‐mediated suppression of antitumor immunity . Cancer Discov . 2017; 7 :522‐538. [ PubMed ] [ Google Scholar ]
49. Chien Y, Scuoppo C, Wang X, et al. Control of the senescence‐associated secretory phenotype by NF‐kappaB promotes senescence and enhances chemosensitivity . Genes Dev . 2011; 25 :2125‐2136. [ PMC free article ] [ PubMed ] [ Google Scholar ]
50. Orjalo AV, Bhaumik D, Gengler BK, Scott GK, Campisi J. Cell surface‐bound IL‐1alpha is an upstream regulator of the senescence‐associated IL‐6/IL‐8 cytokine network . Proc Natl Acad Sci USA . 2009; 106 :17031‐17036. [ PMC free article ] [ PubMed ] [ Google Scholar ]
51. Takahashi A, Imai Y, Yamakoshi K, et al. DNA damage signaling triggers degradation of histone methyltransferases through APC/C(Cdh1) in senescent cells . Mol Cell . 2012; 45 :123‐131. [ PubMed ] [ Google Scholar ]
52. Chen H, Ruiz PD, McKimpson WM, Novikov L, Kitsis RN, Gamble MJ. MacroH2A1 and ATM play opposing roles in paracrine senescence and the senescence‐associated secretory phenotype . Mol Cell . 2015; 59 :719‐731. [ PMC free article ] [ PubMed ] [ Google Scholar ]
53. Guerrero A, Gil J. HMGB2 holds the key to the senescence‐associated secretory phenotype . J Cell Biol . 2016; 215 :297‐299. [ PMC free article ] [ PubMed ] [ Google Scholar ]
54. Capell BC, Drake AM, Zhu J, et al. MLL1 is essential for the senescence‐associated secretory phenotype . Genes Dev . 2016; 30 :321‐336. [ PMC free article ] [ PubMed ] [ Google Scholar ]
55. Tasdemir N, Banito A, Roe JS, et al. BRD4 connects enhancer remodeling to senescence immune surveillance . Cancer Discov . 2016; 6 :612‐629. [ PMC free article ] [ PubMed ] [ Google Scholar ]
56. Laberge RM, Sun Y, Orjalo AV, et al. MTOR regulates the pro–tumorigenic senescence‐associated secretory phenotype by promoting IL1A translation . Nat Cell Biol . 2015; 17 :1049‐1061. [ PMC free article ] [ PubMed ] [ Google Scholar ]
57. Herranz N, Gallage S, Mellone M, et al. mTOR regulates MAPKAPK2 translation to control the senescence‐associated secretory phenotype . Nat Cell Biol . 2015; 17 :1205‐1217. [ PMC free article ] [ PubMed ] [ Google Scholar ]
58. Nacarelli T, Lau L, Fukumoto T, et al. NAD(+) metabolism governs the proinflammatory senescence‐associated secretome . Nat Cell Biol . 2019; 21 :397‐407. [ PMC free article ] [ PubMed ] [ Google Scholar ]
59. Davalos AR, Kawahara M, Malhotra GK, et al. p53‐dependent release of Alarmin HMGB1 is a central mediator of senescent phenotypes . J Cell Biol . 2013; 201 :613‐629. [ PMC free article ] [ PubMed ] [ Google Scholar ]
60. Hari P, Millar FR, Tarrats N, et al. The innate immune sensor Toll‐like receptor 2 controls the senescence‐associated secretory phenotype . Sci Adv . 2019; 5 :eaaw0254. [ PMC free article ] [ PubMed ] [ Google Scholar ]
61. Kawasaki T, Kawai T. Toll‐like receptor signaling pathways . Front Immunol . 2014; 5 :461. [ PMC free article ] [ PubMed ] [ Google Scholar ]
62. Gluck S, Guey B, Gulen MF, et al. Innate immune sensing of cytosolic chromatin fragments through cGAS promotes senescence . Nat Cell Biol . 2017; 19 :1061‐1070. [ PMC free article ] [ PubMed ] [ Google Scholar ]
63. Yang H, Wang H, Ren J, Chen Q, Chen ZJ. cGAS is essential for cellular senescence . Proc Natl Acad Sci USA . 2017; 114 :E4612‐E4620. [ PMC free article ] [ PubMed ] [ Google Scholar ]
64. Li T, Chen ZJ. The cGAS‐cGAMP‐STING pathway connects DNA damage to inflammation, senescence, and cancer . J Exp Med . 2018; 215 :1287‐1299. [ PMC free article ] [ PubMed ] [ Google Scholar ]
65. Ishikawa H, Barber GN. STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling . Nature . 2008; 455 :674‐678. [ PMC free article ] [ PubMed ] [ Google Scholar ]
66. Ishikawa H, Ma Z, Barber GN. STING regulates intracellular DNA‐mediated, type I interferon‐dependent innate immunity . Nature . 2009; 461 :788‐792. [ PMC free article ] [ PubMed ] [ Google Scholar ]
67. Sun L, Wu J, Du F, Chen X, Chen ZJ. Cyclic GMP‐AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway . Science . 2013; 339 :786‐791. [ PMC free article ] [ PubMed ] [ Google Scholar ]
68. Dou Z, Ghosh K, Vizioli MG, et al. Cytoplasmic chromatin triggers inflammation in senescence and cancer . Nature . 2017; 550 :402‐406. [ PMC free article ] [ PubMed ] [ Google Scholar ]
69. Fenech M, Kirsch‐Volders M, Natarajan AT, et al. Molecular mechanisms of micronucleus, nucleoplasmic bridge and nuclear bud formation in mammalian and human cells . Mutagenesis . 2011; 26 :125‐132. [ PubMed ] [ Google Scholar ]
70. Sun B, Sundström KB, Chew JJ, et al. Dengue virus activates cGAS through the release of mitochondrial DNA . Sci Rep . 2017; 7 :3594. [ PMC free article ] [ PubMed ] [ Google Scholar ]
71. Stetson DB, Ko JS, Heidmann T, Medzhitov R. Trex1 prevents cell‐intrinsic initiation of autoimmunity . Cell . 2008; 134 :587‐598. [ PMC free article ] [ PubMed ] [ Google Scholar ]
72. Lan YY, Londono D, Bouley R, Rooney MS, Hacohen N. Dnase2a deficiency uncovers lysosomal clearance of damaged nuclear DNA via autophagy . Cell Rep . 2014; 9 :180‐192. [ PMC free article ] [ PubMed ] [ Google Scholar ]
73. Takahashi A, Loo TM, Okada R, et al. Downregulation of cytoplasmic DNases is implicated in cytoplasmic DNA accumulation and SASP in senescent cells . Nat Commun . 2018; 9 :1249. [ PMC free article ] [ PubMed ] [ Google Scholar ]
74. De Cecco M, Ito T, Petrashen AP, et al. L1 drives IFN in senescent cells and promotes age‐associated inflammation . Nature . 2019; 566 :73‐78. [ PMC free article ] [ PubMed ] [ Google Scholar ]
75. Beck CR, Collier P, Macfarlane C, et al. LINE‐1 retrotransposition activity in human genomes . Cell . 2010; 141 :1159‐1170. [ PMC free article ] [ PubMed ] [ Google Scholar ]
76. Takahashi A, Okada R, Nagao K, et al. Exosomes maintain cellular homeostasis by excreting harmful DNA from cells . Nat Commun . 2017; 8 :15287. [ PMC free article ] [ PubMed ] [ Google Scholar ]
77. Takasugi M, Okada R, Takahashi A, Virya Chen D, Watanabe S, Hara E. Small extracellular vesicles secreted from senescent cells promote cancer cell proliferation through EphA2 . Nat Commun . 2017; 8 :15729. [ PMC free article ] [ PubMed ] [ Google Scholar ]
78. Ritchie C, Cordova AF, Hess GT, Bassik MC, Li L. SLC19A1 Is an Importer of the Immunotransmitter cGAMP . Mol Cell . 2019; 75 ( 2 ):372‐381.e375. [ PMC free article ] [ PubMed ] [ Google Scholar ]
79. Luteijn RD, Zaver SA, Gowen BG, et al. SLC19A1 transports immunoreactive cyclic dinucleotides . Nature . 2019; 573 :434‐438. [ PMC free article ] [ PubMed ] [ Google Scholar ]
80. Kitajima S, Ivanova E, Guo S, et al. Suppression of STING Associated with LKB1 loss in KRAS‐driven lung cancer . Cancer Discov . 2019; 9 :34‐45. [ PMC free article ] [ PubMed ] [ Google Scholar ]
81. Childs BG, Gluscevic M, Baker DJ, et al. Senescent cells: an emerging target for diseases of ageing . Nat Rev Drug Discov . 2017; 16 :718‐735. [ PMC free article ] [ PubMed ] [ Google Scholar ]
82. Motwani M, Pesiridis S, Fitzgerald KA. DNA sensing by the cGAS‐STING pathway in health and disease . Nat Rev Genet . 2019; 20 ( 11 ):657‐674. [ PubMed ] [ Google Scholar ]

Articles from Cancer Science are provided here courtesy of Wiley