Dasatinib Attenuates Fibrosis in Keloids by Decreasing Senescent Cell Burden

Authors

  • Claudia C. Darmawan Department of Dermatology, Seoul National University College of Medicine, Seoul, Korea; Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Korea
  • Keunyoung Hur Department of Dermatology, Seoul National University College of Medicine, Seoul, Korea; Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Korea; Department of Dermatology, Seoul National University Hospital, Seoul, Korea
  • Novi Kusumaningrum Department of Dermatology, Seoul National University College of Medicine, Seoul, Korea; Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Korea
  • Jin Ho Chung Department of Dermatology, Seoul National University College of Medicine, Seoul, Korea; Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Korea; Department of Dermatology, Seoul National University Hospital, Seoul, Korea
  • Si-Hyung Lee Department of Dermatology, Seoul National University College of Medicine, Seoul, Korea; Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Korea; Department of Dermatology, Seoul National University Hospital, Seoul, Korea
  • Je-Ho Mun Department of Dermatology, Seoul National University College of Medicine, Seoul, Korea; Institute of Human-Environment Interface Biology, Seoul National University, Seoul, Korea; Department of Dermatology, Seoul National University Hospital, Seoul, Korea

DOI:

https://doi.org/10.2340/actadv.v103.4475

Keywords:

cellular senescence, fibrosis, keloid, senotherapeutics, therapy, therapeutics

Abstract

Keloids are skin tumours caused by aberrant growth of dermal fibroblasts. Cellular senescence contributes to aging and various pathological conditions, including cancer, atherosclerosis, and fibrotic diseases. However, the effects of cellular senescence and senolytic drugs on keloids remain largely unknown. This study investigated senescent fibroblasts in keloids and assessed the effects of dasatinib on these cells. Tissues acquired from keloid removal surgery were analysed for senescence-associated β-galactosidase-positive cells, p16 expression, and the effects of dasatinib treatment on keloids. Keloid tissue was xenotransplanted into mice, and the effect of intralesional dasatinib injection on keloid growth was observed. The results showed that the numbers of β-galactosidase-positive and p16-expressing cells were higher in the keloids compared with in the controls. Dasatinib induced selective clearance of senescent cells and decreased procollagen expression in cultured keloid fibroblasts. In this xenotransplant keloid mouse model, intralesional injection of dasatinib reduced gross keloid tissue weight and the expression of both procollagen and p16. In addition, dasatinib-treated keloid fibroblasts conditioned medium reduced procollagen and p16 expression in cultured keloid fibroblasts. In conclusion, these results suggest that an increased number of senescent fibroblasts may play an important role in the pathogenesis of keloids. Therefore, dasatinib could be an alternative treatment for patients with keloids.

Downloads

Download data is not yet available.

References

Lee SS, Yosipovitch G, Chan YH, Goh CL. Pruritus, pain, and small nerve fiber function in keloids: a controlled study. J Am Acad Dermatol 2004; 51: 1002-1006.

https://doi.org/10.1016/j.jaad.2004.07.054 DOI: https://doi.org/10.1016/j.jaad.2004.07.054

Lee HJ, Jang YJ. Recent understandings of biology, prophylaxis and treatment strategies for hypertrophic scars and keloids. Int J Mol Sci 2018; 19: 711.

https://doi.org/10.3390/ijms19030711 DOI: https://doi.org/10.3390/ijms19030711

Childs BG, Gluscevic M, Baker DJ, Laberge RM, Marquess D, Dananberg J, et al. Senescent cells: an emerging target for diseases of ageing. Nat Rev Drug Discov 2017; 16: 718-735.

https://doi.org/10.1038/nrd.2017.116 DOI: https://doi.org/10.1038/nrd.2017.116

Kirkland JL, Tchkonia T, Zhu Y, Niedernhofer LJ, Robbins PD. The clinical potential of senolytic drugs. J Am Geriatr Soc 2017; 65: 2297-2301.

https://doi.org/10.1111/jgs.14969 DOI: https://doi.org/10.1111/jgs.14969

Shah NP, Kantarjian HM, Kim DW, Réa D, Dorlhiac-Llacer PE, Milone JH, et al. Intermittent target inhibition with dasatinib 100 mg once daily preserves efficacy and improves tolerability in imatinib-resistant and -intolerant chronic-phase chronic myeloid leukemia. J Clin Oncol 2008; 26: 3204-3212.

https://doi.org/10.1200/JCO.2007.14.9260 DOI: https://doi.org/10.1200/JCO.2007.14.9260

Kantarjian H, Shah NP, Hochhaus A, Cortes J, Shah S, Ayala M, et al. Dasatinib versus imatinib in newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med 2010; 362: 2260-2270.

https://doi.org/10.1056/NEJMoa1002315 DOI: https://doi.org/10.1056/NEJMoa1002315

Martyanov V, Kim GJ, Hayes W, Du S, Ganguly BJ, Sy O, et al. Novel lung imaging biomarkers and skin gene expression subsetting in dasatinib treatment of systemic sclerosis-associated interstitial lung disease. PLoS One 2017; 12: e0187580.

https://doi.org/10.1371/journal.pone.0187580 DOI: https://doi.org/10.1371/journal.pone.0187580

Montero JC, Seoane S, Ocaña A, Pandiella A. Inhibition of SRC family kinases and receptor tyrosine kinases by dasatinib: possible combinations in solid tumors. Clin Cancer Res 2011; 17: 5546-5552.

https://doi.org/10.1158/1078-0432.CCR-10-2616 DOI: https://doi.org/10.1158/1078-0432.CCR-10-2616

Zhu Y, Tchkonia T, Pirtskhalava T, Gower AC, Ding H, Giorgadze N, et al. The Achilles's hill of senescent cells: from transcriptome to senolytic drugs. Aging Cell 2015; 14: 644-658.

https://doi.org/10.1111/acel.12344 DOI: https://doi.org/10.1111/acel.12344

Roos CM, Zhang B, Palmer AK, Ogrodnik MB, Pirtskhalava T, Thalji NM, et al. Chronic senolytic treatment alleviates established vasomotor dysfunction in aged or atherosclerotic mice. Aging Cell 2016; 15; 973-977.

https://doi.org/10.1111/acel.12458 DOI: https://doi.org/10.1111/acel.12458

Martyanov V, Whitfield ML, Varga J. Senescence signature in skin biopsies from systemic sclerosis patients treated with senolytic therapy: potential predictor of clinical response? Arthritis Rheumatol 2019; 71: 1766-1767.

https://doi.org/10.1002/art.40934 DOI: https://doi.org/10.1002/art.40934

Justice JN, Nambiar AM, Tchkonia T, LeBrasseur NK, Pascual R, Hashmi SK, et al. Senolytics in idiopathic pulmonary fibrosis: results from a first-in-human, open-label, pilot study. EBioMedicine 2019; 40: 554-563.

https://doi.org/10.1016/j.ebiom.2018.12.052 DOI: https://doi.org/10.1016/j.ebiom.2018.12.052

Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative CT method. Nat Protoc 2008; 3: 1101-1108.

https://doi.org/10.1038/nprot.2008.73 DOI: https://doi.org/10.1038/nprot.2008.73

Darmawan CC, Montenegro SE, Jo G, Kusumaningrum N, Lee SH, Chung JH, et al. Adiponectin-based peptide (ADP355) inhibits transforming growth factor-β1-induced fibrosis in keloids. Int J Mol Sci 2020; 21: 2833.

https://doi.org/10.3390/ijms21082833 DOI: https://doi.org/10.3390/ijms21082833

Schafer MJ, Haak AJ, Tschumperlin DJ, LeBrasseur NK. Targeting senescent cells in fibrosis: pathology, paradox, and practical considerations. Curr Rheumatol Rep 2018; 20: 3.

https://doi.org/10.1007/s11926-018-0712-x DOI: https://doi.org/10.1007/s11926-018-0712-x

Limandjaja GC, Belien JM, Scheper RJ, Niessen FB, Gibbs S. Hypertrophic and keloid scars fail to progress from the CD34- /α-smooth muscle actin (α-SMA)+ immature scar phenotype and show gradient differences in α-SMA and p16 expression. Br J Dermatol 2020; 182: 974-986.

https://doi.org/10.1111/bjd.18219 DOI: https://doi.org/10.1111/bjd.18219

Varmeh S, Egia A, McGrouther D, Tahan SR, Bayat A, Pandolfi PP. Cellular senescence as a possible mechanism for halting progression of keloid lesions. Genes Cancer 2011; 2: 1061-1066.

https://doi.org/10.1177/1947601912440877 DOI: https://doi.org/10.1177/1947601912440877

Lee JU, Cheong HS, Shim EY, Bae DJ, Chang HS, Uh ST, et al. Gene profile of fibroblasts identify relation of CCL8 with idiopathic pulmonary fibrosis. Respir Res 2017; 18: 3.

https://doi.org/10.1186/s12931-016-0493-6 DOI: https://doi.org/10.1186/s12931-016-0493-6

Li BH, He FP, Yang X, Chen YW, Fan JG. Steatosis induced CCL5 contributes to early-stage liver fibrosis in nonalcoholic fatty liver disease progress. Transl Res 2017; 180: 103-117.e4.

https://doi.org/10.1016/j.trsl.2016.08.006 DOI: https://doi.org/10.1016/j.trsl.2016.08.006

Ong VH, Evans LA, Shiwen X, Fisher IB, Rajkumar V, Abraham DJ, et al. Monocyte chemoattractant protein 3 as a mediator of fibrosis: overexpression in systemic sclerosis and the type 1 tight-skin mouse. Arthritis Rheum 2003; 48: 1979-1991.

https://doi.org/10.1002/art.11164 DOI: https://doi.org/10.1002/art.11164

Yanaba K, Komura K, Kodera M, Matsushita T, Hasegawa M, Takehara K, et al. Serum levels of monocyte chemotactic protein-3/CCL7 are raised in patients with systemic sclerosis: association with extent of skin sclerosis and severity of pulmonary fibrosis. Ann Rheum Dis 2006; 65: 124-126.

https://doi.org/10.1136/ard.2005.040782 DOI: https://doi.org/10.1136/ard.2005.040782

Zhang C, Hu X, Qi F, Luo J, Li X. Identification of CD2, CCL5 and CCR5 as potential therapeutic target genes for renal interstitial fibrosis. Ann Transl Med 2019; 7: 454.

https://doi.org/10.21037/atm.2019.08.62 DOI: https://doi.org/10.21037/atm.2019.08.62

Xiao M, Chen W, Wang C, Wu Y, Zhu S, Zeng C, et al. Senescence and cell death in chronic liver injury: roles and mechanisms underlying hepatocarcinogenesis. Oncotarget 2017; 9: 8772-8784.

https://doi.org/10.18632/oncotarget.23622 DOI: https://doi.org/10.18632/oncotarget.23622

Published

2023-04-06

How to Cite

Darmawan, C. C., Hur, K., Kusumaningrum, N., Chung, J. H., Lee, S.-H., & Mun, J.-H. (2023). Dasatinib Attenuates Fibrosis in Keloids by Decreasing Senescent Cell Burden. Acta Dermato-Venereologica, 103, adv4475. https://doi.org/10.2340/actadv.v103.4475