Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

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.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2020 Sep 1;80(17):3606-3619.
doi: 10.1158/0008-5472.CAN-20-0108. Epub 2020 Jul 8.

Cellular Senescence Promotes Skin Carcinogenesis through p38MAPK and p44/42MAPK Signaling

Affiliations

Cellular Senescence Promotes Skin Carcinogenesis through p38MAPK and p44/42MAPK Signaling

Fatouma Alimirah et al. Cancer Res. .

Abstract

Cellular senescence entails an irreversible growth arrest that evolved in part to prevent cancer. Paradoxically, senescent cells secrete proinflammatory and growth-stimulatory molecules, termed the senescence-associated secretory phenotype (SASP), which is correlated with cancer cell proliferation in culture and xenograft models. However, at what tumor stage and how senescence and the SASP act on endogenous tumor growth in vivo is unknown. To understand the role of senescence in cancer etiology, we subjected p16-3MR transgenic mice, which permit the identification and selective elimination of senescent cells in vivo, to the well-established two-step protocol of squamous cell skin carcinoma, in which tumorigenesis is initiated by a carcinogen 7,12-dimethylbenz[α]anthracene, and then promoted by 12-O-tetradecanoyl-phorbol-13-acetate (TPA). We show that TPA promotes skin carcinogenesis by inducing senescence and a SASP. Systemic induction of senescence in nontumor-bearing p16-3MR mice using a chemotherapy followed by the two-step carcinogenesis protocol potentiated the conversion of benign papillomas to carcinomas by elevating p38MAPK and MAPK/ERK signaling. Ablation of senescent cells reduced p38MAPK and MAPK/ERK signaling, thereby preventing the progression of benign papillomas to carcinomas. Thus, we show for the first time that senescent cells are tumor promoters, not tumor initiators, and that they stimulate skin carcinogenesis by elevating p38MAPK and MAPK/ERK signaling. These findings pave the way for developing novel therapeutics against senescence-fueled cancers. SIGNIFICANCE: These findings identify chemotherapy-induced senescence as a culprit behind tumor promotion, suggesting that elimination of senescent cells after chemotherapy may reduce occurrence of second cancers decades later. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/17/3606/F1.large.jpg.

PubMed Disclaimer

Conflict of interest statement

Conflict of Interests

JC is a co-founder of Unity Biotechnology and MD owns equity in Unity Biotechnology. All other authors declare no competing financial interests.

Figures

Figure 1:
Figure 1:. TPA induces senescence and a SASP in human keratinocytes and mouse skin
(A-C) Human keratinocytes were treated with DMSO (control) or TPA (10 nM) for 48 h. 12 d later, the cells were stained for SA-ß-gal (A-B) and assessed for cell proliferation (C). N=3 independent experiments. (D) Total RNA was isolated from human keratinocytes and analyzed for p16, p21 and SASP mRNAs normalized to actin. N=3 independent experiments, presented as means ±SD; **p<0.01, ***p<0.001, ****p<0.0001. (E) Protein lysates from human keratinocytes treated with DMSO or TPA (5 nM or 10 nM) were evaluated for Lmnb1 and p16 levels by immunoblotting. (F) Total RNA was isolated from mouse skin topically treated with either acetone or TPA for 2 mos. One month after the last treatments, the skin was analyzed for the indicated mRNAs, normalized to actin and tubulin. N=5 for the acetone group, N=7 for the TPA group. Shown are means ±SEM; *p<0.05, **p<0.01 (Student t-test). (G-H) Frozen tissue sections of acetone- or TPA-treated skin were stained for SA-ß-gal (blue) and nuclei (red). (G) Representative images are shown (4x magnification), and arrows denote SA-ß-gal positive areas. (H) Percent of SA-ß-gal positive surface area of skin compared to unstained skin. N=5 for acetone group, N=7 for TPA group. Shown are means ±SEM; *p<0.05 (Student t-test).
Figure 2:
Figure 2:. TPA-induced senescence (pre-treatment) stimulates skin tumor growth
(A) Schematic of treated p16–3MR mice. (B) Representative images of tumor- bearing mice, 24 wks after DMBA treatment. (C) Tumors of Acetone + PBS, Acetone + GCV, TPA + PBS, TPA + GCV groups were monitored once a wk; mean tumor volume over 24 wks is shown. Shown are means ±SEM; **p<0.01, ***p<0.001, ****p<0.0001 (two-way ANOVA, Tukey’s test for multiple comparisons was used post-analyses). (D) Luminescence of tumor-bearing skin in arbitrary units (A.U.) units 24 wks after DMBA treatment. N = 8–10 per group. Shown are means ±SEM; *p<0.05 (one-way ANOVA, Sidak’s multiple comparisons test was used post-analyses). (E) Representative images of H&E staining of skin (top panels) and tumors (middle panels) (200 μm and 50 μm respectively), and Ki-67 staining of tumors (bottom panels) (50 μm). (F) Percentage of Ki-67-positive cells in tumors from the 4 treatment groups. Shown are means ±SEM; *p<0.05 (one-way ANOVA, Sidak’s multiple comparisons test was used post-analyses).
Figure 3:
Figure 3:. DOXO induces senescence and a SASP in human keratinocytes and mouse skin
(A-C) Human keratinocytes were treated with DMSO or DOXO (250 nM) for 24 h. 10 d later, the cells were stained for SA-ß-gal (A-B) and assessed for cell proliferation (C). N=3 independent experiments. Shown are means ±SD, *p<0.5, ***p<0.001, (Student t test). (D) Total RNA was isolated from the indicated cells and analyzed for p16, p21 and SASP mRNAs, normalized to actin. N=3 independent experiments. Shown are means ±SD; **p<0.01, ***p<0.001. (E) Protein lysates from human keratinocytes were evaluated for p16 and LMNB1 by immunoblotting. (F) Total RNA was isolated from the skin of PBS- or DOXO-treated mice and analyzed for the indicated mRNAs, normalized to actin and tubulin. N=6 per each treatment group. Shown are means ±SEM; *p<0.05, **p<0.01 (Student t-test). (G-H) Representative image (G) and quantification in arbitrary units (A.U.) of whole body luminescence of p16–3MR mice one mo after PBS or DOXO treatments. N=6 per each treatment group. Shown are means ±SEM; *p<0.05 (Student t test).
Figure 4:
Figure 4:. DOXO-induced senescence fuels skin tumor growth
(A) A schematic of the PBS/DOXO and skin carcinogenesis regimens of p16–3MR mice. TPA treatment lasted 16 wks. (B) Quantification of whole body luminescence of p16–3MR mice 10 d after PBS/DOXO treatments and one wk after DMBA initiation. Quantification is in arbitrary units (A.U.). Shown are means ±SEM; *p<0.05 (one-way ANOVA, Sidak’s multiple comparisons test was used post-analyses). (C) Total RNA was isolated from the skin of the 4 treatments groups and analyzed for p16 normalized to actin and tubulin (PBS + PBS: n=10, PBS + GCV: n=8, DOXO + PBS: n=9, DOXO + GCV: n=8). Shown are means ±SEM; *p<0.05, **p<0.01 (one-way ANOVA, Sidak’s multiple comparisons test was used post-analyses). (D) Representative images of tumor-bearing mice 21 wks after DMBA treatment and 5 wks after TPA treatment in PBS + PBS, PBS + GCV, DOXO + PBS, DOXO + GCV groups. (E) The mean tumor volume of the same mice is shown over 21 wks. Shown are means ±SEM; *p<0.05, **p<0.01, ****p<0.0001 (two way ANOVA, Tukey’s test for multiple comparisons was used post-analyses). (F-G) Frozen skin sections of (PBS + PBS: n=10, PBS + GCV: n=10, DOXO + PBS: n=14, DOXO + GCV: n=14) treatment groups were stained for SA-ß-gal (blue) and nuclei (red). (F) Representative images are shown (50 μm), and arrows denote SA-ß-gal positive areas. (G) Percent of SA-ß-gal positive surface area of skin compared to non-stained skin. Shown are means ±SEM; *p<0.5, **p<0.01, ***p<0. 001 (one-way ANOVA, Sidak’s multiple comparisons test was used post-analyses).
Figure 5:
Figure 5:. Eliminating DOXO-induced senescent cells prevents malignant tumor development
Representative images of H&E staining of the skin (top panels) and tumors (bottom panels) in PBS + PBS, PBS + GCV, DOXO + PBS, DOXO + GCV treated-groups (200 μm). (B-D) In the same 4 groups (PBS + PBS: n =9, PBS + GCV: n=7, DOXO + PBS: n=11, DOXO + GCV: n=7), representative IHC images for Ki-67 (B), vimentin (C) and CD-31 (D) (50 μm) with the corresponding quantification of percent positive cells in the right panels. Shown are means ±SEM; *p<0.05, **p<0.01, ***p<0. 001 (one-way ANOVA, Sidak’s multiple comparisons test was used post-analyses).
Figure 6:
Figure 6:. The SASP drives skin carcinogenesis
(A-D) Representative images and quantification of tumorspheres of A-431 cells embedded in reduced growth factor Matrigel incubated for 7 d with conditioned media from non-senescent (quiescent) or senescent HCA2 fibroblasts with or without SB203580 (A-B) or anti-IL1α (C-D). (A and C) Tumorspheres are shown stained with DAPI (blue) and EdU (green). (B and D) Quantification of percentage EdU-positive cells. N=3 independent experiments. Shown are mean ±SEM; **p<0.01, ****p<0.0001 (one-way ANOVA, Sidak’s multiple comparisons test was used post-analyses). (E) Representative tumor and adjacent skin luminescence images of PBS- or DOXO-treated mice, 21 wks after DMBA treatment and 5 wks after the last TPA treatment. Luminescent skin and tumors are labeled on the images as (S) and (T), respectively. (F) Luminescence quantification of tumors and adjacent skin shown in (E) in arbitrary units (A.U.). N= 8 per each treatment group. Shown are mean ±SEM; ***p<0.001 (Student t test). (G) Luciferase activity of untreated skin compared to DOXO-tumor, protein lysates were normalized to total protein content. N= 4–5 for skin and DOXO tumors respectively. Shown are mean ±SEM; *p<0.05 (Student t test). (H) Total RNA was isolated from skin adjacent to tumors from mice in (E-F) and analyzed for the indicated mRNAs, normalized to actin and tubulin. N= 8 per treatment group. Shown are mean ±SEM; *p<0.5, **p<0.001 (Student t test).
Figure 7:
Figure 7:. Phospho-p38 and phospho-p44/42 MAPK signaling is increased in tumors from DOXO-treated mice
(A-B) Representative IHC images of tumors from PBS + PBS: n =9, PBS + GCV: n=7, DOXO + PBS: n=11, DOXO + GCV: n=7, stained with p-p38 (A) or p-p44/42 (B) with the corresponding quantification of percent positive cells in the right panels. Shown are means ±SEM; *p<0.05, **p<0.01, (one-way ANOVA, Sidak’s multiple comparisons test was used post-analyses). (C-D) Representative IHC images of normal human skin and hSCC lesions (grades I-III) stained with p-p38 (C) and p-p44/42 (D). (E) Working model showing that DOXO induces senescence and a SASP in keratinocytes and fibroblasts, creating a microenvironment permissive for tumor growth and invasion. Once tumors are formed, senescent cells within the tumors also reinforce tumor growth by elevating p-p38 and p-p44/42 signaling.

Similar articles

Cited by

  • Cellular senescence exacerbates features of aging in the eyes.
    Kitazawa K, Numa K, Patel SK, King CD, Matsumoto A, Sotozono C, Desprez PY, Schilling B, Campisi J. Kitazawa K, et al. Aging Biol. 2023;1(1):20230014. doi: 10.59368/agingbio.20230014. Epub 2023 Sep 15. Aging Biol. 2023. PMID: 39239324 Free PMC article.
  • NLRP1 inflammasome promotes senescence and senescence-associated secretory phenotype.
    Muela-Zarzuela I, Suarez-Rivero JM, Gallardo-Orihuela A, Wang C, Izawa K, de Gregorio-Procopio M, Couillin I, Ryffel B, Kitaura J, Sanz A, von Zglinicki T, Mbalaviele G, Cordero MD. Muela-Zarzuela I, et al. Inflamm Res. 2024 Aug;73(8):1253-1266. doi: 10.1007/s00011-024-01892-7. Epub 2024 Jun 21. Inflamm Res. 2024. PMID: 38907167 Free PMC article.
  • Therapy-induced senescence through the redox lens.
    Robert M, Kennedy BK, Crasta KC. Robert M, et al. Redox Biol. 2024 Aug;74:103228. doi: 10.1016/j.redox.2024.103228. Epub 2024 Jun 6. Redox Biol. 2024. PMID: 38865902 Free PMC article. Review.
  • SenNet recommendations for detecting senescent cells in different tissues.
    Suryadevara V, Hudgins AD, Rajesh A, Pappalardo A, Karpova A, Dey AK, Hertzel A, Agudelo A, Rocha A, Soygur B, Schilling B, Carver CM, Aguayo-Mazzucato C, Baker DJ, Bernlohr DA, Jurk D, Mangarova DB, Quardokus EM, Enninga EAL, Schmidt EL, Chen F, Duncan FE, Cambuli F, Kaur G, Kuchel GA, Lee G, Daldrup-Link HE, Martini H, Phatnani H, Al-Naggar IM, Rahman I, Nie J, Passos JF, Silverstein JC, Campisi J, Wang J, Iwasaki K, Barbosa K, Metis K, Nernekli K, Niedernhofer LJ, Ding L, Wang L, Adams LC, Ruiyang L, Doolittle ML, Teneche MG, Schafer MJ, Xu M, Hajipour M, Boroumand M, Basisty N, Sloan N, Slavov N, Kuksenko O, Robson P, Gomez PT, Vasilikos P, Adams PD, Carapeto P, Zhu Q, Ramasamy R, Perez-Lorenzo R, Fan R, Dong R, Montgomery RR, Shaikh S, Vickovic S, Yin S, Kang S, Suvakov S, Khosla S, Garovic VD, Menon V, Xu Y, Song Y, Suh Y, Dou Z, Neretti N. Suryadevara V, et al. Nat Rev Mol Cell Biol. 2024 Jun 3. doi: 10.1038/s41580-024-00738-8. Online ahead of print. Nat Rev Mol Cell Biol. 2024. PMID: 38831121 Review.
  • Senescent characteristics of human corneal endothelial cells upon ultraviolet-A exposure.
    Numa K, Patel SK, Zhang ZA, Burton JB, Matsumoto A, Hughes JB, Sotozono C, Schilling B, Desprez PY, Campisi J, Kitazawa K. Numa K, et al. Aging (Albany NY). 2024 Apr 26;16(8):6673-6693. doi: 10.18632/aging.205761. Epub 2024 Apr 26. Aging (Albany NY). 2024. PMID: 38683123 Free PMC article.

References

    1. Rodier F, Campisi J. Four faces of cellular senescence. J Cell Biol 2011;192:547–56. - PMC - PubMed
    1. Wiley CD, Velarde MC, Lecot P, Liu S, Sarnoski EA, Freund A et al. Mitochondrial Dysfunction Induces Senescence with a Distinct Secretory Phenotype. Cell Metab 2016;23:303–14. - PMC - PubMed
    1. Harley CB, Futcher AB, Greider CW. Telomeres shorten during aging of human fibroblasts. Nature 1990;345:458–60. - PubMed
    1. Serrano M, Lin AW, McCurrach ME, Beach D, Lowe SW. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 1997;88:593–602. - PubMed
    1. Zhang R, Adams PD. Heterochromatin and its relationship to cell senescence and cancer therapy. Cell Cycle 2007;6:784–9. - PubMed

Publication types