Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Advertisement

npj Aging
  • View all journals
  • Search
  • My Account Login
  • Content Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • RSS feed
  1. nature
  2. npj aging
  3. articles
  4. article

For urgent queries, please contact the Journal Publisher, Aicha Hanna.

Crosstalk between TGF-β and Wnt/β-catenin signaling drives fibrogenic and stem-like phenotypes in senescent MDA-MB-231 breast cancer cells
Download PDF
Download PDF
  • Article
  • Open access
  • Published: 03 January 2026

Crosstalk between TGF-β and Wnt/β-catenin signaling drives fibrogenic and stem-like phenotypes in senescent MDA-MB-231 breast cancer cells

  • Mona El Samarji1 na1,
  • Elissa Alam1 na1,
  • Mariam Dakramanji1 na2,
  • Mariam Bassam1 na2,
  • Jana Santina1 na2,
  • Marc Ayoub1,
  • Alex Aprahamian1 &
  • …
  • Mohamad Rima1 

npj Aging , Article number:  (2026) Cite this article

  • 1804 Accesses

  • 8 Altmetric

  • Metrics details

We are providing an unedited version of this manuscript to give early access to its findings. Before final publication, the manuscript will undergo further editing. Please note there may be errors present which affect the content, and all legal disclaimers apply.

Subjects

  • Cancer
  • Cell biology
  • Molecular biology
  • Stem cells

Abstract

Genotoxic drugs used to treat cancer can trigger senescence, which contributes to chemotherapy resistance and tumor heterogeneity. However, the resulting cellular and molecular alterations following senescence remain poorly characterized. In this study, chemotherapy-induced senescence was triggered by etoposide in MDA-MB-231 breast cancer cells, and their fibrogenic potential, epithelial-to-mesenchymal transition (EMT), and stemness features were examined. In these cells, key mediators of fibrosis were significantly upregulated, suggesting a profibrotic potential involving TGF-β signaling. Etoposide also accentuated the mesenchymal phenotype of MDA-MB-231 cells and increased their motility. Additionally, nuclear β-catenin accumulation and upregulation of its EMT target genes were observed in senescent cells, alongside increased stemness markers, indicating a plastic cellular state involving Wnt/β-catenin signaling. Interestingly, pharmacological inhibition of the TGF-β/Wnt/β-catenin pathways reduced fibrosis, EMT, stemness marker expression, and cell migration, suggesting that these pathways are key regulators of these processes in senescent cells. These findings provide new insights into the molecular mechanisms driving chemotherapy-induced senescence and highlight these pathways as potential targets to alleviate resistance and aggressiveness in breast cancer.

Similar content being viewed by others

Comprehensive molecular interaction map of TGFβ induced epithelial to mesenchymal transition in breast cancer

Article Open access 17 May 2024

Simultaneous screening of overexpressed genes in breast cancer for oncogenic drivers and tumor dependencies

Article Open access 09 June 2024

1H-NMR metabolomics analysis identifies hypoxanthine as a novel metastasis-associated metabolite in breast cancer

Article Open access 02 January 2024

Data availability

The original results presented in the study are included in the article/supplementary data; further inquiries can be directed to the corresponding author.

References

  1. Dimri, G. P. et al. A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc. Natl. Acad. Sci. USA 92, 9363–9367 (1995).

    Google Scholar 

  2. Gorgoulis, V. et al. Cellular senescence: defining a path forward. Cell 179, 813–827 (2019).

    Google Scholar 

  3. Coppé, J.-P., Desprez, P.-Y., Krtolica, A. & Campisi, J. The senescence-associated secretory phenotype: the dark side of tumor suppression. Annu. Rev. Pathol. 5, 99–118 (2010).

    Google Scholar 

  4. Demaria, M. et al. An essential role for senescent cells in optimal wound healing through secretion of PDGF-AA. Dev. Cell 31, 722–733 (2014).

    Google Scholar 

  5. Jun, J.-I. & Lau, L. F. The matricellular protein CCN1 induces fibroblast senescence and restricts fibrosis in cutaneous wound healing. Nat. Cell Biol. 12, 676–685 (2010).

    Google Scholar 

  6. Muñoz-Espín, D. et al. Programmed cell senescence during mammalian embryonic development. Cell 155, 1104–1118 (2013).

    Google Scholar 

  7. Storer, M. et al. Senescence is a developmental mechanism that contributes to embryonic growth and patterning. Cell 155, 1119–1130 (2013).

    Google Scholar 

  8. Franceschi, C. & Campisi, J. Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J. Gerontol. A Biol. Sci. Med. Sci. 69, S4–S9 (2014).

    Google Scholar 

  9. Mosteiro L. et al. Tissue damage and senescence provide critical signals for cellular reprogramming in vivo. Science. 354 (2016).

  10. Coppé, J.-P. et al. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 6, 2853–2868 (2008).

    Google Scholar 

  11. HAYFLICK, L. & MOORHEAD, P. S. The serial cultivation of human diploid cell strains. Exp. Cell Res. 25, 585–621 (1961).

    Google Scholar 

  12. Campisi, J. The biology of replicative senescence. Eur. J. Cancer 33, 703–709 (1997).

    Google Scholar 

  13. Toussaint, O., Medrano, E. E. & von Zglinicki, T. Cellular and molecular mechanisms of stress-induced premature senescence (SIPS) of human diploid fibroblasts and melanocytes. Exp. Gerontol. 35, 927–945 (2000).

    Google Scholar 

  14. Wyld L. et al. Senescence and cancer: a review of clinical implications of senescence and senotherapies. Cancers. 12 (2020).

  15. Guillon, J. et al. Chemotherapy-induced senescence, an adaptive mechanism driving resistance and tumor heterogeneity. Cell Cycle 18, 2385–2397 (2019).

    Google Scholar 

  16. te Poele, R. H., Okorokov, A. L., Jardine, L., Cummings, J. & Joel, S. P. DNA damage is able to induce senescence in tumor cells in vitro and in vivo. Cancer Res. 62, 1876–1883 (2002).

    Google Scholar 

  17. Chang, B. D. et al. A senescence-like phenotype distinguishes tumor cells that undergo terminal proliferation arrest after exposure to anticancer agents. Cancer Res. 59, 3761–3767 (1999).

    Google Scholar 

  18. Ewald, J. A., Desotelle, J. A., Wilding, G. & Jarrard, D. F. Therapy-induced senescence in cancer. J. Natl. Cancer Inst. 102, 1536–1546 (2010).

    Google Scholar 

  19. Song, K.-X., Wang, J.-X. & Huang, D. Therapy-induced senescent tumor cells in cancer relapse. J. Natl. Cancer Cent. 3, 273–278 (2023).

    Google Scholar 

  20. Dong, Z. et al. Cellular senescence and SASP in tumor progression and therapeutic opportunities. Mol. Cancer 23, 181 (2024).

    Google Scholar 

  21. Childs, B. G., Durik, M., Baker, D. J. & van Deursen, J. M. Cellular senescence in aging and age-related disease: from mechanisms to therapy. Nat. Med. 21, 1424–1435 (2015).

    Google Scholar 

  22. Gewirtz, D. A. Autophagy, senescence and tumor dormancy in cancer therapy. Autophagy 5, 1232–1234 (2009).

    Google Scholar 

  23. De Blander, H., Morel, A.-P., Senaratne, A. P., Ouzounova, M., & Puisieux, A. Cellular plasticity: a route to senescence exit and tumorigenesis. Cancers. 13 (2021).

  24. Jha, S. K. et al. Cellular senescence in lung cancer: Molecular mechanisms and therapeutic interventions. Ageing Res. Rev. 97, 102315 (2024).

    Google Scholar 

  25. Jin, C. et al. Cellular senescence in metastatic prostate cancer: a therapeutic opportunity or challenge (Review). Mol. Med. Rep. 30, 162 (2024).

    Google Scholar 

  26. Childs, B. G., Baker, D. J., Kirkland, J. L., Campisi, J. & van Deursen, J. M. Senescence and apoptosis: dueling or complementary cell fates? EMBO Rep. 15, 1139–1153 (2014).

    Google Scholar 

  27. Khedri, A. et al. Signaling crosstalk of FHIT, p53, and p38 in etoposide-induced apoptosis in MCF-7 cells. J. Cell Biochem. 120, 9125–9137 (2019).

    Google Scholar 

  28. Litwiniec, A., Gackowska, L., Helmin-Basa, A., Zuryń, A. & Grzanka, A. Low-dose etoposide-treatment induces endoreplication and cell death accompanied by cytoskeletal alterations in A549 cells: Does the response involve senescence? The possible role of vimentin. Cancer Cell Int. 13, 9 (2013).

    Google Scholar 

  29. Hassibi, S., Baker, J., Barnes, P. & Donnelly, L. Generating senescent airway epithelial cell populations using low-concentration doxorubicin or etoposide. In: Airway cell biology and immunopathology. PA3687, (European Respiratory Society, 2021).

  30. Nagano, T. et al. Identification of cellular senescence-specific genes by comparative transcriptomics. Sci. Rep. 6, 31758 (2016).

    Google Scholar 

  31. Hande, K. R. Etoposide: four decades of development of a topoisomerase II inhibitor. Eur. J. Cancer 34, 1514–1521 (1998).

    Google Scholar 

  32. Milczarek, M. The premature senescence in breast cancer treatment strategy. Cancers 12, 1815 (2020).

    Google Scholar 

  33. Santarosa, M. et al. Premature senescence is a major response to DNA cross-linking agents in BRCA1-defective cells: implication for tailored treatments of BRCA1 mutation carriers. Mol. Cancer Ther. 8, 844–854 (2009).

    Google Scholar 

  34. Wu, D., Pepowski, B., Takahashi, S. & Kron, S. J. A cmap-enabled gene expression signature-matching approach identifies small-molecule inducers of accelerated cell senescence. BMC Genomics 20, 290 (2019).

    Google Scholar 

  35. Saleh, T. et al. Clearance of therapy-induced senescent tumor cells by the senolytic ABT-263 via interference with BCL-XL -BAX interaction. Mol. Oncol. 14, 2504–2519 (2020).

    Google Scholar 

  36. Ren, L.-L. et al. TGF-β as a master regulator of aging-associated tissue fibrosis. Aging Dis. 14, 1633–1650 (2023).

    Google Scholar 

  37. Kim, S. W., Kim, S. J., Langley, R. R. & Fidler, I. J. Modulation of the cancer cell transcriptome by culture media formulations and cell density. Int. J. Oncol. 46, 2067–2075 (2015).

    Google Scholar 

  38. Walles, S. A. S., Zhou, R. & Liliemark, E. DNA damage induced by etoposide; a comparison of two different methods for determination of strand breaks in DNA. Cancer Lett. 105, 153–159 (1996).

    Google Scholar 

  39. O’Reilly, S., Tsou, P.-S. & Varga, J. Senescence and tissue fibrosis: opportunities for therapeutic targeting. Trends Mol. Med. 30, 1113–1125 (2024).

    Google Scholar 

  40. Bonner, J. C. Regulation of PDGF and its receptors in fibrotic diseases. Cytokine Growth Factor Rev. 15, 255–273 (2004).

    Google Scholar 

  41. Abou Zaghla, H. M. A., El Sebai, A. A., Ahmed, O. A., Ahmed, A. F. & Saab, A. A. R. Growth differentiation factor 15: an emerging diagnostic biomarker of liver fibrosis in chronic hepatitis C patients. Egypt. Liver J. 11, 6 (2021).

    Google Scholar 

  42. Wan, Y. & Fu, J. GDF15 as a key disease target and biomarker: linking chronic lung diseases and ageing. Mol. Cell Biochem 479, 453–466 (2024).

    Google Scholar 

  43. Radwanska A. et al. Increased expression and accumulation of GDF15 in IPF extracellular matrix contribute to fibrosis. JCI Insight. 7 (2022).

  44. Takenouchi, Y., Kitakaze, K., Tsuboi, K. & Okamoto, Y. Growth differentiation factor 15 facilitates lung fibrosis by activating macrophages and fibroblasts. Exp. Cell Res. 391, 112010 (2020).

    Google Scholar 

  45. Emmerson P. J., Duffin K. L., Chintharlapalli S., Wu X. GDF15 and growth control. Front. Physiol. 9 (2018).

  46. Pence B. D. Growth differentiation Factor-15 in immunity and aging. Front. Aging. 3 (2022).

  47. Kurundkar A. R. et al. The matricellular protein CCN1 enhances TGF-β1/SMAD3-dependent profibrotic signaling in fibroblasts and contributes to fibrogenic responses to lung injury. FASEB J. 30, 2135–2150 (2016).

  48. Ihn, H. Pathogenesis of fibrosis: role of TGF-β and CTGF. Curr. Opin. Rheumatol. 14, 681–685 (2002).

    Google Scholar 

  49. Chung, J. Y.-F. et al. TGF-β signaling: from tissue fibrosis to tumor microenvironment. Int. J. Mol. Sci. 22, 7575 (2021).

    Google Scholar 

  50. Cao, Z.-Q., Wang, Z. & Leng, P. Aberrant N-cadherin expression in cancer. Biomedicine Pharmacother. 118, 109320 (2019).

    Google Scholar 

  51. WANG, M. et al. N-cadherin promotes epithelial-mesenchymal transition and cancer stem cell-like traits via ErbB signaling in prostate cancer cells. Int. J. Oncol. 48, 595–606 (2016).

    Google Scholar 

  52. Ferber, E. C. et al. A role for the cleaved cytoplasmic domain of E-cadherin in the nucleus. J. Biol. Chem. 283, 12691–12700 (2008).

    Google Scholar 

  53. Xu, R. et al. Arf6 regulates EGF-induced internalization of E-cadherin in breast cancer cells. Cancer Cell Int. 15, 11 (2015).

    Google Scholar 

  54. Adhikary A. et al. Inhibition of epithelial to mesenchymal transition by E-cadherin up-regulation via repression of slug transcription and inhibition of E-cadherin degradation: dual role of scaffold/matrix attachment region-binding protein 1 (SMAR1) in breast cancer cells. J. Biol. Chem. 289, 25431–25444 (2014).

  55. Tian, X. et al. E-cadherin/β-catenin complex and the epithelial barrier. J. Biomed. Biotechnol. 2011, 567305 (2011).

    Google Scholar 

  56. Song P. et al. Wnt/β-catenin signaling pathway in carcinogenesis and cancer therapy. J. Hematol. Oncol. 17, 46 (2024).

  57. Suh, Y. et al. Claudin-1 induces epithelial–mesenchymal transition through activation of the c-Abl-ERK signaling pathway in human liver cells. Oncogene 32, 4873–4882 (2013).

    Google Scholar 

  58. Ke B., Fan C., Yang L., Fang X. Matrix metalloproteinases-7 and kidney fibrosis. Front. Physiol. 8 (2017).

  59. Yin, S., Cheryan, V. T., Xu, L., Rishi, A. K. & Reddy, K. B. Myc mediates cancer stem-like cells and EMT changes in triple negative breast cancers cells. PLoS One 12, e0183578 (2017).

    Google Scholar 

  60. Migault, M., Sapkota, S. & Bracken, C. P. Transcriptional and post-transcriptional control of epithelial-mesenchymal plasticity: why so many regulators? Cell Mol. Life Sci. 79, 182 (2022).

    Google Scholar 

  61. Perez-Oquendo M., Gibbons D. L. Regulation of ZEB1 function and molecular associations in tumor progression and metastasis. Cancers.14 (2022).

  62. Glackin CA. Nanoparticle delivery of TWIST small interfering RNA and anticancer drugs: a therapeutic approach for combating cancer. In p. 83–101. (2018).

  63. Sledge, G. W. Etoposide in the management of metastatic breast cancer. Cancer 67, 266–270 (1991).

    Google Scholar 

  64. Nichols, C. R. Role of etoposide in treatment of breast cancer. Semin Oncol. 19, 67–71 (1992).

    Google Scholar 

  65. Benjamin, C. W., Hiebsch, R. R. & Jones, D. A. Caspase activation in MCF7 cells responding to etoposide treatment. Mol. Pharm. 53, 446–450 (1998).

    Google Scholar 

  66. Alpsoy, A., Yasa, S. & Gündüz, U. Etoposide resistance in MCF-7 breast cancer cell line is marked by multiple mechanisms. Biomed. Pharmacother. 68, 351–355 (2014).

    Google Scholar 

  67. Gordon, R. R. & Nelson, P. S. Cellular senescence and cancer chemotherapy resistance. Drug Resistance Updates 15, 123–131 (2012).

    Google Scholar 

  68. Campisi, J. Aging, cellular senescence, and cancer. Annu. Rev. Physiol. 75, 685–705 (2013).

    Google Scholar 

  69. Muñoz D. P. et al. Targetable mechanisms driving immunoevasion of persistent senescent cells link chemotherapy-resistant cancer to aging. JCI Insight. 5 (2019).

  70. Baker, D. J. et al. Naturally occurring p16Ink4a-positive cells shorten healthy lifespan. Nature 530, 184–189 (2016).

    Google Scholar 

  71. Demaria, M. et al. Cellular Senescence Promotes Adverse Effects of Chemotherapy and Cancer Relapse. Cancer Discov. 7, 165–176 (2017).

    Google Scholar 

  72. Saleh T. et al. Expression of therapy-induced senescence markers in breast cancer samples upon incomplete response to neoadjuvant chemotherapy. Biosci. Rep. 41 (2021).

  73. Jackson, J. G. et al. p53-mediated senescence impairs the apoptotic response to chemotherapy and clinical outcome in breast cancer. Cancer Cell 21, 793–806 (2012).

    Google Scholar 

  74. Elmore, L. W. et al. Adriamycin-induced senescence in breast tumor cells involves functional p53 and telomere dysfunction. J. Biol. Chem. 277, 35509–35515 (2002).

    Google Scholar 

  75. Inao, T. et al. Different sensitivities of senescent breast cancer cells to immune cell-mediated cytotoxicity. Cancer Sci. 110, 2690–2699 (2019).

    Google Scholar 

  76. Huun, J., Lønning, P. E. & Knappskog, S. Effects of concomitant inactivation of p53 and pRb on response to doxorubicin treatment in breast cancer cell lines. Cell Death Discov. 3, 17026 (2017).

    Google Scholar 

  77. Jackson, J. G. & Pereira-Smith, O. M. Primary and compensatory roles for RB family members at cell cycle gene promoters that are deacetylated and downregulated in doxorubicin-induced senescence of breast cancer cells. Mol. Cell Biol. 26, 2501–2510 (2006).

    Google Scholar 

  78. Bojko A., Czarnecka-Herok J., Charzynska A., Dabrowski M., Sikora E. Diversity of the senescence phenotype of cancer cells treated with chemotherapeutic agents. Cells. 8 (2019).

  79. Gomes, L. R. et al. ATR mediates cisplatin resistance in 3D-cultured breast cancer cells via translesion DNA synthesis modulation. Cell Death Dis. 10, 459 (2019).

    Google Scholar 

  80. Ago, T. & Sadoshima, J. GDF15, a Cardioprotective TGF-β Superfamily Protein. Circ. Res 98, 294–297 (2006).

    Google Scholar 

  81. Kok, H. M., Falke, L. L., Goldschmeding, R. & Nguyen, T. Q. Targeting CTGF, EGF and PDGF pathways to prevent progression of kidney disease. Nat. Rev. Nephrol. 10, 700–711 (2014).

    Google Scholar 

  82. Siddiqui, J. A. et al. Pathophysiological role of growth differentiation factor 15 (GDF15) in obesity, cancer, and cachexia. Cytokine Growth Factor Rev. 64, 71–83 (2022).

    Google Scholar 

  83. Zhao, X., Chen, J., Sun, H., Zhang, Y. & Zou, D. New insights into fibrosis from the ECM degradation perspective: the macrophage-MMP-ECM interaction. Cell Biosci. 12, 117 (2022).

    Google Scholar 

  84. Herrera, A. et al. Endothelial cell activation on 3D-matrices derived from PDGF-BB-stimulated fibroblasts is mediated by Snail1. Oncogenesis 7, 76 (2018).

    Google Scholar 

  85. Throckmorton, D. C., Brogden, A. P., Min, B., Rasmussen, H. & Kashgarian, M. PDGF and TGF-β mediate collagen production by mesangial cells exposed to advanced glycosylation end products. Kidney Int 48, 111–117 (1995).

    Google Scholar 

  86. Grotendorst, G. R. Connective tissue growth factor: a mediator of TGF-β action on fibroblasts. Cytokine Growth Factor Rev. 8, 171–179 (1997).

    Google Scholar 

  87. Massagué, J. & Sheppard, D. TGF-β signaling in health and disease. Cell 186, 4007–4037 (2023).

    Google Scholar 

  88. Massagué, J. TGFβ in cancer. Cell 134, 215–230 (2008).

    Google Scholar 

  89. Villarejo, A., Cortés-Cabrera, Á, Molina-Ortíz, P., Portillo, F. & Cano, A. Differential role of Snail1 and Snail2 Zinc fingers in E-cadherin repression and epithelial to mesenchymal transition. J. Biol. Chem. 289, 930–941 (2014).

    Google Scholar 

  90. Loh, C.-Y. et al. The E-Cadherin and N-cadherin switch in epithelial-to-mesenchymal transition: signaling, therapeutic implications, and challenges. Cells 8, 1118 (2019).

    Google Scholar 

  91. Pal, M., Bhattacharya, S., Kalyan, G. & Hazra, S. Cadherin profiling for therapeutic interventions in Epithelial Mesenchymal Transition (EMT) and tumorigenesis. Exp. Cell Res 368, 137–146 (2018).

    Google Scholar 

  92. Anzai, E. et al. FOXA1 Induces E-Cadherin Expression at the Protein Level via Suppression of Slug in Epithelial Breast Cancer Cells. Biol. Pharm. Bull. 40, 1483–1489 (2017).

    Google Scholar 

  93. Eslami Amirabadi, H. et al. Characterizing the invasion of different breast cancer cell lines with distinct E-cadherin status in 3D using a microfluidic system. Biomed. Microdevices 21, 101 (2019).

    Google Scholar 

  94. Mbalaviele, G. et al. E-cadherin expression in human breast cancer cells suppresses the development of osteolytic bone metastases in an experimental metastasis model. Cancer Res 56, 4063–4070 (1996).

    Google Scholar 

  95. Marambaud, P. et al. A presenilin-1/gamma-secretase cleavage releases the E-cadherin intracellular domain and regulates disassembly of adherens junctions. EMBO J. 21, 1948–1956 (2002).

    Google Scholar 

  96. David, J. M. & Rajasekaran, A. K. Dishonorable discharge: the oncogenic roles of cleaved E-cadherin fragments. Cancer Res 72, 2917–2923 (2012).

    Google Scholar 

  97. Zuo, J. et al. Activation of EGFR promotes squamous carcinoma SCC10A cell migration and invasion via inducing EMT-like phenotype change and MMP-9-mediated degradation of E-cadherin. J. Cell Biochem. 112, 2508–2517 (2011).

    Google Scholar 

  98. Yusuf, M. et al. N-Cadherin Expression with Metastasis of Neck Lymph Nodes in Patients with Nasopharyngeal Carcinoma. Int J. Gen. Med 16, 1029–1037 (2023).

    Google Scholar 

  99. Rodriguez, F. J., Lewis-Tuffin, L. J. & Anastasiadis, P. Z. E-cadherin’s dark side: possible role in tumor progression. Biochim. Biophys. Acta 1826, 23–31 (2012).

    Google Scholar 

  100. Shoval, I., Ludwig, A. & Kalcheim, C. Antagonistic roles of full-length N-cadherin and its soluble BMP cleavage product in neural crest delamination. Development 134, 491–501 (2007).

    Google Scholar 

  101. Zhao, Z. et al. Epithelial-mesenchymal transition in cancer: Role of the IL-8/IL-8R axis. Oncol. Lett. 13, 4577–4584 (2017).

    Google Scholar 

  102. Du, W. et al. From cell membrane to the nucleus: an emerging role of E-cadherin in gene transcriptional regulation. J. Cell Mol. Med 18, 1712–1719 (2014).

    Google Scholar 

  103. Xiang W. et al. N-cadherin cleavage: A critical function that induces diabetic retinopathy fibrosis via regulation of β-catenin translocation. FASEB J. 37. (2023).

  104. Williams, H., Johnson, J. L., Jackson, C. L., White, S. J. & George, S. J. MMP-7 mediates cleavage of N-cadherin and promotes smooth muscle cell apoptosis. Cardiovasc. Res. 87, 137–146 (2010).

  105. Orsulic, S., Huber, O., Aberle, H., Arnold, S. & Kemler, R. E-cadherin binding prevents β-catenin nuclear localization and β-catenin/LEF-1-mediated transactivation. J. Cell Sci. 112, 1237–1245 (1999).

    Google Scholar 

  106. Turano M. et al. Characterisation of mesenchymal colon tumour-derived cells in tumourspheres as a model for colorectal cancer progression. Int. J. Oncol. (2018).

  107. Xu, J., Lamouille, S. & Derynck, R. TGF-β-induced epithelial to mesenchymal transition. Cell Res. 19, 156–172 (2009).

    Google Scholar 

  108. Wang, Y., Shi, J., Chai, K., Ying, X. & Zhou, B. P. The role of snail in EMT and tumorigenesis. Curr. Cancer Drug Targets 13, 963–972 (2013).

    Google Scholar 

  109. Dou, Z. & Berger, S. L. Senescence elicits stemness: a surprising mechanism for cancer relapse. Cell Metab. 27, 710–711 (2018).

    Google Scholar 

  110. Kim, B. N. et al. TGF-β induced EMT and stemness characteristics are associated with epigenetic regulation in lung cancer. Sci. Rep. 10, 10597 (2020).

    Google Scholar 

  111. Tsubakihara, Y. et al. TGFβ selects for pro-stemness over pro-invasive phenotypes during cancer cell epithelial–mesenchymal transition. Mol. Oncol. 16, 2330–2354 (2022).

    Google Scholar 

  112. Bellomo, C., Caja, L. & Moustakas, A. Transforming growth factor β as regulator of cancer stemness and metastasis. Br. J. Cancer 115, 761–769 (2016).

    Google Scholar 

  113. Meng, X., Nikolic-Paterson, D. J. & Lan, H. Y. TGF-β: the master regulator of fibrosis. Nat. Rev. Nephrol. 12, 325–338 (2016).

    Google Scholar 

  114. Hu, H.-H., Cao, G., Wu, X.-Q., Vaziri, N. D. & Zhao, Y.-Y. Wnt signaling pathway in aging-related tissue fibrosis and therapies. Ageing Res. Rev. 60, 101063 (2020).

    Google Scholar 

  115. Lam, A. P. & Gottardi, C. J. β-catenin signaling: a novel mediator of fibrosis and potential therapeutic target. Curr. Opin. Rheumatol. 23, 562–567 (2011).

    Google Scholar 

  116. Leung, R. W. H. & Lee, T. K. W. Wnt/β-catenin signaling as a driver of stemness and metabolic reprogramming in hepatocellular carcinoma. Cancers 14, 5468 (2022).

    Google Scholar 

  117. Katoh, M. & Katoh, M. WNT signaling and cancer stemness. Essays Biochem. 66, 319–331 (2022).

    Google Scholar 

  118. Kessler, M. et al. The Notch and Wnt pathways regulate stemness and differentiation in human fallopian tube organoids. Nat. Commun. 6, 8989 (2015).

    Google Scholar 

  119. Akhmetshina, A. et al. Activation of canonical Wnt signalling is required for TGF-β-mediated fibrosis. Nat. Commun. 3, 735 (2012).

    Google Scholar 

  120. Shao, S. et al. Lysyl hydroxylase 3 increases collagen deposition and promotes pulmonary fibrosis by activating TGFβ1/Smad3 and Wnt/β-catenin pathways. Arch. Med. Sci. 16, 436–445 (2020).

    Google Scholar 

  121. Działo, E., Tkacz, K. & Błyszczuk, P. Crosstalk between TGF-β and WNT signalling pathways during cardiac fibrogenesis. Acta Biochim. Pol. 65, 341–349 (2018).

    Google Scholar 

  122. Sun, M. et al. Role of the Wnt signaling pathway in the complex microenvironment of breast cancer and prospects for therapeutic potential (Review). Int. J. Oncol. 66, 1–22 (2025).

    Google Scholar 

  123. Sheikh, K. et al. Exploring TGF-β signaling in cancer progression: prospects and therapeutic strategies. Onco Targets Ther. 18, 233–262 (2025).

    Google Scholar 

  124. Kadota, T. et al. Human bronchial epithelial cell-derived extracellular vesicle therapy for pulmonary fibrosis via inhibition of TGF-β-WNT crosstalk. J Extracell Vesicles. 10. 2021;

  125. McCloy, R. A. et al. Partial inhibition of Cdk1 in G 2 phase overrides the SAC and decouples mitotic events. Cell Cycle 13, 1400–1412 (2014).

    Google Scholar 

  126. Count Nuclear Foci - ImageJ. https://microscopy.duke.edu/guides/count-nuclear-foci-ImageJ.

  127. Rima, M. et al. The β<inf>4</inf>subunit of the voltage-gated calcium channel (Cacnb4) regulates the rate of cell proliferation in Chinese Hamster Ovary cells. Int. J. Biochem. Cell Biol. 89 (2017).

  128. Foty, R. A simple hanging drop cell culture protocol for generation of 3D spheroids. J. Vis. Exp. (2011).

Download references

Acknowledgements

This research was funded by the Lebanese American University (LAU) under grant number PIRF-I0087 and supported by intramural funds from the Department of Biological Sciences (LAU) to secure space, equipment, and consumables.

Author information

Author notes
  1. These authors contributed equally: Mona El Samarji, Elissa Alam.

  2. These authors jointly supervised this work: Mariam Dakramanji, Mariam Bassam, Jana Santina.

Authors and Affiliations

  1. Department of Biological Sciences, Lebanese American University, Byblos, Lebanon

    Mona El Samarji, Elissa Alam, Mariam Dakramanji, Mariam Bassam, Jana Santina, Marc Ayoub, Alex Aprahamian & Mohamad Rima

Authors
  1. Mona El Samarji
    View author publications

    Search author on:PubMed Google Scholar

  2. Elissa Alam
    View author publications

    Search author on:PubMed Google Scholar

  3. Mariam Dakramanji
    View author publications

    Search author on:PubMed Google Scholar

  4. Mariam Bassam
    View author publications

    Search author on:PubMed Google Scholar

  5. Jana Santina
    View author publications

    Search author on:PubMed Google Scholar

  6. Marc Ayoub
    View author publications

    Search author on:PubMed Google Scholar

  7. Alex Aprahamian
    View author publications

    Search author on:PubMed Google Scholar

  8. Mohamad Rima
    View author publications

    Search author on:PubMed Google Scholar

Contributions

M.R. designed the study and was responsible for funding acquisition, methodology, project administration and supervision. M.E.S., E.A., M.D., J.S., M.B., M.A., A.A., and M.R. performed and analyzed the experiments. M.R. prepared the figures and wrote the manuscript with input from all the authors. All authors have read and agreed to the published version of the manuscript.

Corresponding author

Correspondence to Mohamad Rima.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

El Samarji, M., Alam, E., Dakramanji, M. et al. Crosstalk between TGF-β and Wnt/β-catenin signaling drives fibrogenic and stem-like phenotypes in senescent MDA-MB-231 breast cancer cells. npj Aging (2026). https://doi.org/10.1038/s41514-025-00322-0

Download citation

  • Received: 17 August 2025

  • Accepted: 16 December 2025

  • Published: 03 January 2026

  • DOI: https://doi.org/10.1038/s41514-025-00322-0

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

Download PDF

Advertisement

Explore content

  • Research articles
  • Reviews & Analysis
  • News & Comment
  • Collections
  • Follow us on Twitter
  • Sign up for alerts
  • RSS feed

About the journal

  • Aims & Scope
  • Journal Information
  • Content types
  • About the Editors
  • Contact
  • Open Access
  • Calls for Papers
  • Article Processing Charges
  • Editorial policies
  • Journal Metrics
  • About the Partner

Publish with us

  • For Authors and Referees
  • Language editing services
  • Open access funding
  • Submit manuscript

Search

Advanced search

Quick links

  • Explore articles by subject
  • Find a job
  • Guide to authors
  • Editorial policies

npj Aging (npj Aging)

ISSN 2731-6068 (online)

nature.com sitemap

About Nature Portfolio

  • About us
  • Press releases
  • Press office
  • Contact us

Discover content

  • Journals A-Z
  • Articles by subject
  • protocols.io
  • Nature Index

Publishing policies

  • Nature portfolio policies
  • Open access

Author & Researcher services

  • Reprints & permissions
  • Research data
  • Language editing
  • Scientific editing
  • Nature Masterclasses
  • Research Solutions

Libraries & institutions

  • Librarian service & tools
  • Librarian portal
  • Open research
  • Recommend to library

Advertising & partnerships

  • Advertising
  • Partnerships & Services
  • Media kits
  • Branded content

Professional development

  • Nature Awards
  • Nature Careers
  • Nature Conferences

Regional websites

  • Nature Africa
  • Nature China
  • Nature India
  • Nature Japan
  • Nature Middle East
  • Privacy Policy
  • Use of cookies
  • Legal notice
  • Accessibility statement
  • Terms & Conditions
  • Your US state privacy rights
Springer Nature

© 2026 Springer Nature Limited

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer