J Pathol Transl Med.  2023 Nov;57(6):305-314. 10.4132/jptm.2023.10.09.

Senescent tumor cells in colorectal cancer are characterized by elevated enzymatic activity of complexes 1 and 2 in oxidative phosphorylation

Affiliations
  • 1Department of Surgery, Ajou University School of Medicine, Suwon, Korea
  • 2Department of Pathology, Ajou University School of Medicine, Suwon, Korea
  • 3Inflamm-Aging Translational Research Center, Ajou University Hospital, Suwon, Korea
  • 4Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Korea
  • 5Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea

Abstract

Background
Cellular senescence is defined as an irreversible cell cycle arrest caused by various internal and external insults. While the metabolic dysfunction of senescent cells in normal tissue is relatively well-established, there is a lack of information regarding the metabolic features of senescent tumor cells.
Methods
Publicly available single-cell RNA-sequencing data from the GSE166555 and GSE178341 datasets were utilized to investigate the metabolic features of senescent tumor cells. To validate the single-cell RNA-sequencing data, we performed senescence-associated β-galactosidase (SA-β-Gal) staining to identify senescent tumor cells in fresh frozen colorectal cancer tissue. We also evaluated nicotinamide adenine dinucleotide dehydrogenase–tetrazolium reductase (NADH-TR) and succinate dehydrogenase (SDH) activity using enzyme histochemical methods and compared the staining with SA-β-Gal staining. MTT assay was performed to reveal the complex 1 activity of the respiratory chain in in-vitro senescence model.
Results
Single-cell RNA-sequencing data revealed an upregulation in the activity of complexes 1 and 2 in oxidative phosphorylation, despite overall mitochondrial dysfunction in senescent tumor cells. Both SA-β-Gal and enzyme histochemical staining using fresh frozen colorectal cancer tissues indicated a high correlation between SA-β-Gal positivity and NADH-TR/SDH staining positivity. MTT assay showed that senescent colorectal cancer cells exhibit higher absorbance in 600 nm wavelength.
Conclusions
Senescent tumor cells exhibit distinct metabolic features, characterized by upregulation of complexes 1 and 2 in the oxidative phosphorylation pathway. NADH-TR and SDH staining represent efficient methods for detecting senescent tumor cells in colorectal cancer.

Keyword

Colorectal neoplasms; Metabolism; Cellular senescence; Oxidative phosphorylation; NADH

Figure

  • Fig. 1. Single-cell RNA-sequencing (scRNA-seq) of human colorectal cancer tissues. (A) Uniform Manifold Approximation and Projection (UMAP) of public scRNA-seq dataset GSE166555 patient #8 who markedly expressed CDKN2A is shown. A total of 274 cells were analyzed. (B) Gene set enrichment analysis (GSEA) of “Fridman Senescence Up” (left panel) and “GOBP Cellular Senescence” gene sets was performed in CDKN2A+ vs. CDKN2A– cancer cells. (C) GSEA of a “GOBP Oxidative Phosphorylation” gene set was performed in CDKN2A+ vs. CDKN2A– cancer cells. (D) GSEA of a “GOBP Mitochondrial Electron Transport NADH to Ubiquinone” gene set was performed in CDKN2A+ vs. CDKN2A– cancer cells. (E) GSEA of a “GOBP Mitochondrial Electron Transport Ubiquinol to Cytochrome c” gene set was performed in CDKN2A+ vs. CDKN2A– cancer cells. (F) GSEA of a “GOBP ATP Synthesis Coupled Electron Transport” gene set was performed in CDKN2A+ vs. CDKN2A– cancer cells. (G) Schematic image of expected electron transport efficiency in senescent and non-senescent tumor cells. respectively. The thickness of red arrows indicates the efficiency of electron flow. ‘padj’ indicates the adjusted p-value. CRC, colorectal cancer; ES, enrichment score; NES, normalized enrichment score.

  • Fig. 2. Nicotinamide adenine dinucleotide dehydrogenase–tetrazolium reductase (NADH-TR) and succinate dehydrogenase (SDH) staining results in the senescence-associated β-galactosidase (SA-β-Gal) high patient. The staining results of SA-β-Gal (A), NADH-TR (B), and SDH staining (C) are shown in the SA-β-Gal high patient.

  • Fig. 3. Nicotinamide adenine dinucleotide dehydrogenase–tetrazolium reductase (NADH-TR) and succinate dehydrogenase (SDH) staining results in the senescence-associated β-galactosidase (SA-β-Gal) low patient. The staining results of SA-β-Gal (A), NADH-TR (B), and SDH staining (C) are shown in the SA-β-Gal low patient.

  • Fig. 4. Scatter plots according to the nicotinamide adenine dinucleotide dehydrogenase–tetrazolium reductase (NADH-TR) (A) and succinate dehydrogenase (SDH) (B) staining results compared with senescence-associated β-galactosidase (SA-β-Gal) staining. p-values are obtained using the hypothesis test for regression slope.


Reference

References

1. Hayflick L, Moorhead PS. The serial cultivation of human diploid cell strains. Exp Cell Res. 1961; 25:585–621.
Article
2. Park SS, Choi YW, Kim JH, Kim HS, Park TJ. Senescent tumor cells: an overlooked adversary in the battle against cancer. Exp Mol Med. 2021; 53:1834–41.
Article
3. Munoz-Espin D, Serrano M. Cellular senescence: from physiology to pathology. Nat Rev Mol Cell Biol. 2014; 15:482–96.
Article
4. Bartkova J, Rezaei N, Liontos M, et al. Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature. 2006; 444:633–7.
Article
5. Kim YH, Choi YW, Lee J, Soh EY, Kim JH, Park TJ. Senescent tumor cells lead the collective invasion in thyroid cancer. Nat Commun. 2017; 8:15208.
Article
6. Salam R, Saliou A, Bielle F, et al. Cellular senescence in malignant cells promotes tumor progression in mouse and patient Glioblastoma. Nat Commun. 2023; 14:441.
Article
7. Choi YW, Kim YH, Oh SY, et al. Senescent tumor cells build a cytokine shield in colorectal cancer. Adv Sci (Weinh). 2021; 8:2002497.
Article
8. Park SS, Lee YK, Park SH, et al. p15(INK4B) is an alternative marker of senescent tumor cells in colorectal cancer. Heliyon. 2023; 9:e13170.
Article
9. Wiley CD, Campisi J. The metabolic roots of senescence: mechanisms and opportunities for intervention. Nat Metab. 2021; 3:1290–301.
Article
10. Wiley CD, Campisi J. From ancient pathways to aging cells-connecting metabolism and cellular senescence. Cell Metab. 2016; 23:1013–21.
Article
11. Faget DV, Ren Q, Stewart SA. Unmasking senescence: context-dependent effects of SASP in cancer. Nat Rev Cancer. 2019; 19:439–53.
Article
12. Miwa S, Kashyap S, Chini E, von Zglinicki T. Mitochondrial dysfunction in cell senescence and aging. J Clin Invest. 2022; 132:e158447.
Article
13. James EL, Michalek RD, Pitiyage GN, et al. Senescent human fibroblasts show increased glycolysis and redox homeostasis with extracellular metabolomes that overlap with those of irreparable DNA damage, aging, and disease. J Proteome Res. 2015; 14:1854–71.
Article
14. Ashton TM, McKenna WG, Kunz-Schughart LA, Higgins GS. Oxidative phosphorylation as an emerging target in cancer therapy. Clin Cancer Res. 2018; 24:2482–90.
Article
15. Kumar PR, Moore JA, Bowles KM, Rushworth SA, Moncrieff MD. Mitochondrial oxidative phosphorylation in cutaneous melanoma. Br J Cancer. 2021; 124:115–23.
Article
16. LeBleu VS, O’Connell JT, Gonzalez Herrera KN, et al. PGC-1alpha mediates mitochondrial biogenesis and oxidative phosphorylation in cancer cells to promote metastasis. Nat Cell Biol. 2014; 16:992–1003.
Article
17. Vellinga TT, Borovski T, de Boer VC, et al. SIRT1/PGC1alpha-dependent increase in oxidative phosphorylation supports chemotherapy resistance of colon cancer. Clin Cancer Res. 2015; 21:2870–9.
18. Wang Y, Nasiri AR, Damsky WE, et al. Uncoupling hepatic oxidative phosphorylation reduces tumor growth in two murine models of colon cancer. Cell Rep. 2018; 24:47–55.
Article
19. Uhlitz F, Bischoff P, Peidli S, et al. Mitogen-activated protein kinase activity drives cell trajectories in colorectal cancer. EMBO Mol Med. 2021; 13:e14123.
Article
20. Pelka K, Hofree M, Chen JH, et al. Spatially organized multicellular immune hubs in human colorectal cancer. Cell. 2021; 184:4734–52.
21. Yoon YS, Byun HO, Cho H, Kim BK, Yoon G. Complex II defect via down-regulation of iron-sulfur subunit induces mitochondrial dysfunction and cell cycle delay in iron chelation-induced senescence-associated growth arrest. J Biol Chem. 2003; 278:51577–86.
Article
22. Yoon YS, Lee JH, Hwang SC, Choi KS, Yoon G. TGF beta1 induces prolonged mitochondrial ROS generation through decreased complex IV activity with senescent arrest in Mv1Lu cells. Oncogene. 2005; 24:1895–903.
Article
23. Moiseeva O, Bourdeau V, Roux A, Deschenes-Simard X, Ferbeyre G. Mitochondrial dysfunction contributes to oncogene-induced senescence. Mol Cell Biol. 2009; 29:4495–507.
Article
24. Yagi T, Matsuno-Yagi A. The proton-translocating NADH-quinone oxidoreductase in the respiratory chain: the secret unlocked. Biochemistry. 2003; 42:2266–74.
Article
25. Kumar P, Nagarajan A, Uchil PD. Analysis of cell viability by the MTT assay. Cold Spring Harb Protoc. 2018 Jun 1 [Epub]. https://doi.org/10.1101/pdb.prot095505.
Article
26. Surin AM, Sharipov RR, Krasil’nikova IA, et al. Disruption of functional activity of mitochondria during MTT assay of viability of cultured neurons. Biochemistry (Mosc). 2017; 82:737–49.
Article
27. Deberardinis RJ, Sayed N, Ditsworth D, Thompson CB. Brick by brick: metabolism and tumor cell growth. Curr Opin Genet Dev. 2008; 18:54–61.
Article
28. Liberti MV, Locasale JW. The Warburg effect: how does it benefit cancer cells? Trends Biochem Sci. 2016; 41:211–8.
Article
29. Warburg O. On respiratory impairment in cancer cells. Science. 1956; 124:269–70.
Article
30. Zheng J. Energy metabolism of cancer: Glycolysis versus oxidative phosphorylation (Review). Oncol Lett. 2012; 4:1151–7.
Article
31. Vasan K, Werner M, Chandel NS. Mitochondrial metabolism as a target for cancer therapy. Cell Metab. 2020; 32:341–52.
Article
32. Porporato PE, Filigheddu N, Pedro JM, Kroemer G, Galluzzi L. Mitochondrial metabolism and cancer. Cell Res. 2018; 28:265–80.
Article
33. Galluzzi L, Kepp O, Vander Heiden MG, Kroemer G. Metabolic targets for cancer therapy. Nat Rev Drug Discov. 2013; 12:829–46.
Article
34. Allison KE, Coomber BL, Bridle BW. Metabolic reprogramming in the tumour microenvironment: a hallmark shared by cancer cells and T lymphocytes. Immunology. 2017; 152:175–84.
Article
35. Chapman J, Fielder E, Passos JF. Mitochondrial dysfunction and cell senescence: deciphering a complex relationship. FEBS Lett. 2019; 593:1566–79.
Article
36. Sahin E, Colla S, Liesa M, et al. Telomere dysfunction induces metabolic and mitochondrial compromise. Nature. 2011; 470:359–65.
Article
37. Wiley CD, Velarde MC, Lecot P, et al. Mitochondrial dysfunction induces senescence with a distinct secretory phenotype. Cell Metab. 2016; 23:303–14.
Article
38. Martini H, Passos JF. Cellular senescence: all roads lead to mitochondria. FEBS J. 2023; 290:1186–202.
Article
39. Nelson G, Kucheryavenko O, Wordsworth J, von Zglinicki T. The senescent bystander effect is caused by ROS-activated NF-kappaB signalling. Mech Ageing Dev. 2018; 170:30–6.
40. Joyce NC, Oskarsson B, Jin LW. Muscle biopsy evaluation in neuromuscular disorders. Phys Med Rehabil Clin N Am. 2012; 23:609–31.
Article
41. Zhao Z, Mei Y, Wang Z, He W. The effect of oxidative phosphorylation on cancer drug resistance. Cancers (Basel). 2022; 15:62.
Article
42. Collado M, Blasco MA, Serrano M. Cellular senescence in cancer and aging. Cell. 2007; 130:223–33.
Article
43. Haferkamp S, Tran SL, Becker TM, Scurr LL, Kefford RF, Rizos H. The relative contributions of the p53 and pRb pathways in oncogene-induced melanocyte senescence. Aging (Albany NY). 2009; 1:542–56.
Article
44. Heinzel PA, Balaram P, Bernard HU. Mutations and polymorphisms in the p53, p21 and p16 genes in oral carcinomas of Indian betel quid chewers. Int J Cancer. 1996; 68:420–3.
45. Soto Martinez JL, Cabrera Morales CM, Serrano Ortega S, Lopez-Nevot MA. Mutation and homozygous deletion analyses of genes that control the G1/S transition of the cell cycle in skin melanoma: p53, p21, p16 and p15. Clin Transl Oncol. 2005; 7:156–64.
Article
46. Sadaie M, Salama R, Carroll T, et al. Redistribution of the Lamin B1 genomic binding profile affects rearrangement of heterochromatic domains and SAHF formation during senescence. Genes Dev. 2013; 27:1800–8.
Article
47. Itahana K, Campisi J, Dimri GP. Methods to detect biomarkers of cellular senescence: the senescence-associated beta-galactosidase assay. Methods Mol Biol. 2007; 371:21–31.
48. Krishna DR, Sperker B, Fritz P, Klotz U. Does pH 6 beta-galactosidase activity indicate cell senescence? Mech Ageing Dev. 1999; 109:113–23.
Full Text Links
  • JPTM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr