دورية أكاديمية

NEMO/NF-κB signaling functions as a double-edged sword in PanIN formation versus progression to pancreatic cancer

التفاصيل البيبلوغرافية
العنوان: NEMO/NF-κB signaling functions as a double-edged sword in PanIN formation versus progression to pancreatic cancer
المؤلفون: Miltiadis Tsesmelis, Ulrike F. G. Büttner, Melanie Gerstenlauer, Uta Manfras, Konstantinos Tsesmelis, Ziwei Du, Nadine Sperb, Stephanie Ellen Weissinger, Peter Möller, Thomas F. E. Barth, Harald J. Maier, Lap Kwan Chan, Thomas Wirth
المصدر: Molecular Cancer, Vol 23, Iss 1, Pp 1-20 (2024)
بيانات النشر: BMC, 2024.
سنة النشر: 2024
المجموعة: LCC:Neoplasms. Tumors. Oncology. Including cancer and carcinogens
مصطلحات موضوعية: PDAC, PanINs, NEMO, NF-κB, Senescence, Cerulein, Neoplasms. Tumors. Oncology. Including cancer and carcinogens, RC254-282
الوصف: Abstract Background Pancreatic ductal adenocarcinoma (PDAC) is marked by a dismal survival rate, lacking effective therapeutics due to its aggressive growth, late-stage diagnosis, and chemotherapy resistance. Despite debates on NF-κB targeting for PDAC treatment, no successful approach has emerged. Methods To elucidate the role of NF-κB, we ablated NF-κB essential modulator (NEMO), critical for conventional NF-κB signaling, in the pancreata of mice that develop precancerous lesions (KC mouse model). Secretagogue-induced pancreatitis by cerulein injections was utilized to promote inflammation and accelerate PDAC development. Results NEMO deletion reduced fibrosis and inflammation in young KC mice, resulting in fewer pancreatic intraepithelial neoplasias (PanINs) at later stages. Paradoxically, however, NEMO deletion accelerated the progression of these fewer PanINs to PDAC and reduced median lifespan. Further, analysis of tissue microarrays from human PDAC sections highlighted the correlation between reduced NEMO expression in neoplastic cells and poorer prognosis, supporting our observation in mice. Mechanistically, NEMO deletion impeded oncogene-induced senescence (OIS), which is normally active in low-grade PanINs. This blockage resulted in fewer senescence-associated secretory phenotype (SASP) factors, reducing inflammation. However, blocked OIS fostered replication stress and DNA damage accumulation which accelerated PanIN progression to PDAC. Finally, treatment with the DNA damage-inducing reagent etoposide resulted in elevated cell death in NEMO-ablated PDAC cells compared to their NEMO-competent counterparts, indicative of a synthetic lethality paradigm. Conclusions NEMO exhibited both oncogenic and tumor-suppressive properties during PDAC development. Caution is suggested in therapeutic interventions targeting NF-κB, which may be detrimental during PanIN progression but beneficial post-PDAC development. Graphical Abstract
نوع الوثيقة: article
وصف الملف: electronic resource
اللغة: English
تدمد: 1476-4598
Relation: https://doaj.org/toc/1476-4598
DOI: 10.1186/s12943-024-01989-x
URL الوصول: https://doaj.org/article/d60de3829ea54fce943d4b1ee9fe0c20
رقم الأكسشن: edsdoj.60de3829ea54fce943d4b1ee9fe0c20
قاعدة البيانات: Directory of Open Access Journals
الوصف
تدمد:14764598
DOI:10.1186/s12943-024-01989-x