Your browser doesn't support javascript.
loading
PD-L1 intrinsically promotes the proliferation of breast cancer cells through the SKP2-p27/p21 axis.
Elfoly, Marwa; Mirza, Jumanah Y; Alaiya, Ayodele; Al-Hazzani, Amal A; Tulbah, Asma; Al-Alwan, Monther; Ghebeh, Hazem.
Afiliação
  • Elfoly M; Cell Therapy and Immunobiology Department, King Faisal Specialist Hospital & Research Centre, Riyadh, 11211, Kingdom of Saudi Arabia.
  • Mirza JY; Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia.
  • Alaiya A; Cell Therapy and Immunobiology Department, King Faisal Specialist Hospital & Research Centre, Riyadh, 11211, Kingdom of Saudi Arabia.
  • Al-Hazzani AA; Cell Therapy and Immunobiology Department, King Faisal Specialist Hospital & Research Centre, Riyadh, 11211, Kingdom of Saudi Arabia.
  • Tulbah A; Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia.
  • Al-Alwan M; Department of Pathology, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia.
  • Ghebeh H; Cell Therapy and Immunobiology Department, King Faisal Specialist Hospital & Research Centre, Riyadh, 11211, Kingdom of Saudi Arabia.
Cancer Cell Int ; 24(1): 161, 2024 May 09.
Article em En | MEDLINE | ID: mdl-38725021
ABSTRACT

BACKGROUND:

PD-L1 intrinsically promotes tumor progression through multiple mechanisms, which potentially leads to resistance to anti-PD-1/PD-L1 therapies. The intrinsic effect of PD-L1 on breast cancer (BC) cell proliferation has not been fully elucidated.

METHODS:

we used proteomics, gene expression knockdown (KD), quantitative immunofluorescence (qIF), western blots, functional assays including colony-forming assay (CFA) and real-time cell analyzer (RTCA), and in vivo data using immunohistochemistry in breast cancer patients.

RESULTS:

PD-L1 promoted BC cell proliferation by accelerating cell cycle entry at the G1-to-S phase transition. Global proteomic analysis of the differentially expressed nuclear proteins indicated the involvement of several proliferation-related molecules, including p21CIP1/WAF1. Western blotting and qIF demonstrated the higher expression of SKP2 and the lower expression of p21CIP1/WAF1 and p27Kip1 in PD-L1 expressing (PD-L1pos) cells as compared to PD-L1 KD (PD-L1KD) cells. Xenograft-derived cells and the TCGA BC dataset confirmed this relationship in vivo. Functionally, CFA and RTCA demonstrated the central role of SKP2 in promoting PD-L1-mediated proliferation. Finally, immunohistochemistry in 74 breast cancer patients confirmed PD-L1 and SKP-p21/p27 axis relationship, as it showed a highly statistically significant correlation between SKP2 and PD-L1 expression (p < 0.001), and both correlated significantly with the proliferation marker Ki-67 (p < 0.001). On the other hand, there was a statistically significant inverse relationship between PD-L1 and p21CIP1/WAF1 expression (p = 0.005). Importantly, double negativity for p21CIP1/WAF1 and p27Kip1 correlated significantly with PD-L1 (p < 0.001), SKP2 (p = 0.002), and Ki-67 (p = 0.002).

CONCLUSIONS:

we have demonstrated the role of the SKP2-p27/p21 axis in intrinsic PD-L1-enhanced cell cycle progression. Inhibitors of SKP2 expression can alleviate resistance to ICPIs.
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2024 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Idioma: En Ano de publicação: 2024 Tipo de documento: Article