Your browser doesn't support javascript.
loading
Cistanche phenylethanoid glycosides induce apoptosis and pyroptosis in T-cell lymphoma.
Tang, Ying; Zhao, Fangxin; Zhang, Xuan; Niu, Yan; Liu, Xiulan; Bu, Renqiqige; Ma, Yunlong; Wu, Geyemuri; Li, Beibei; Yang, Hongxin; Wu, Jianqiang.
Afiliação
  • Tang Y; School of Life Sciences, Inner Mongolia University Hohhot, Inner Mongolia, China.
  • Zhao F; College of Basic Medicine, Inner Mongolia Medical University Hohhot, Inner Mongolia, China.
  • Zhang X; School of Life Sciences, Inner Mongolia University Hohhot, Inner Mongolia, China.
  • Niu Y; College of Basic Medicine, Inner Mongolia Medical University Hohhot, Inner Mongolia, China.
  • Liu X; College of Basic Medicine, Inner Mongolia Medical University Hohhot, Inner Mongolia, China.
  • Bu R; College of Basic Medicine, Inner Mongolia Medical University Hohhot, Inner Mongolia, China.
  • Ma Y; College of Basic Medicine, Inner Mongolia Medical University Hohhot, Inner Mongolia, China.
  • Wu G; College of Basic Medicine, Inner Mongolia Medical University Hohhot, Inner Mongolia, China.
  • Li B; School of Life Sciences, Inner Mongolia Agricultural University Hohhot, Inner Mongolia, China.
  • Yang H; College of Basic Medicine, Inner Mongolia Medical University Hohhot, Inner Mongolia, China.
  • Wu J; College of Basic Medicine, Inner Mongolia Medical University Hohhot, Inner Mongolia, China.
Am J Cancer Res ; 14(3): 1338-1352, 2024.
Article em En | MEDLINE | ID: mdl-38590417
ABSTRACT
Cistanche deserticola, known for its extensive history in Traditional Chinese Medicine (TCM), is valued for its therapeutic properties. Recent studies have identified its anticancer capabilities, yet the mechanisms underlying these properties remain to be fully elucidated. In this study, we determined that a mixture of four cistanche-derived phenylethanoid glycosides (CPhGs), echinacoside, acteoside, 2-acetylacteoside, and cistanoside A, which are among the main bioactive compounds in C. deserticola, eliminated T-cell lymphoma (TCL) cells by inducing apoptosis and pyroptosis in vitro and attenuated tumor growth in vivo in a xenograft mouse model. At the molecular level, these CPhGs elevated P53 by inhibiting the SIRT2-MDM2/P300 and PI3K/AKT carcinogenic axes and activating PTEN-Bax tumor-suppressing signaling. Moreover, CPhGs activated noncanonical and alternative pathways to trigger pyroptosis. Interestingly, CPhGs did not activate canonical NLRP3-caspase-1 pyroptotic signaling pathway; instead, CPhGs suppressed the inflammasome factor NLRP3 and the maturation of IL-1ß. Treatment with a caspase-1/4 inhibitor and silencing of Gasdermin D (GSDMD) or Gasdermin E (GSDME) partially rescued CPhG-induced cell death. Conversely, forced expression of NLRP3 restored cell proliferation. In summary, our results indicate that CPhGs modulate multiple signaling pathways to achieve their anticancer properties and perform dual roles in pyroptosis and NLRP3-driven proliferation. This study offers experimental support for the potential application of CPhGs in the treatment of TCL.
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Revista: Am J Cancer Res Ano de publicação: 2024 Tipo de documento: Article País de afiliação: China

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Revista: Am J Cancer Res Ano de publicação: 2024 Tipo de documento: Article País de afiliação: China