关键词: 3D spheroids Budesonide Cell proliferation Glucocorticoids Metabolic reprogramming Pancreatic cancer

Mesh : Humans Budesonide / pharmacology therapeutic use Mice Pancreatic Neoplasms / drug therapy metabolism pathology Energy Metabolism / drug effects Cell Proliferation / drug effects Animals Cell Line, Tumor Carcinoma, Pancreatic Ductal / drug therapy metabolism pathology Xenograft Model Antitumor Assays Cell Movement / drug effects

来  源:   DOI:10.1186/s13046-024-03072-1   PDF(Pubmed)

Abstract:
BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is the most lethal cancer with an aggressive metastatic phenotype and very poor clinical prognosis. Interestingly, a lower occurrence of PDAC has been described in individuals with severe and long-standing asthma. Here we explored the potential link between PDAC and the glucocorticoid (GC) budesonide, a first-line therapy to treat asthma.
METHODS: We tested the effect of budesonide and the classical GCs on the morphology, proliferation, migration and invasiveness of patient-derived PDAC cells and pancreatic cancer cell lines, using 2D and 3D cultures in vitro. Furthermore, a xenograft model was used to investigate the effect of budesonide on PDAC tumor growth in vivo. Finally, we combined genome-wide transcriptome analysis with genetic and pharmacological approaches to explore the mechanisms underlying budesonide activities in the different environmental conditions.
RESULTS: We found that in 2D culture settings, high micromolar concentrations of budesonide reduced the mesenchymal invasive/migrating features of PDAC cells, without affecting proliferation or survival. This activity was specific and independent of the Glucocorticoid Receptor (GR). Conversely, in a more physiological 3D environment, low nanomolar concentrations of budesonide strongly reduced PDAC cell proliferation in a GR-dependent manner. Accordingly, we found that budesonide reduced PDAC tumor growth in vivo. Mechanistically, we demonstrated that the 3D environment drives the cells towards a general metabolic reprogramming involving protein, lipid, and energy metabolism (e.g., increased glycolysis dependency). This metabolic change sensitizes PDAC cells to the anti-proliferative effect of budesonide, which instead induces opposite changes (e.g., increased mitochondrial oxidative phosphorylation). Finally, we provide evidence that budesonide inhibits PDAC growth, at least in part, through the tumor suppressor CDKN1C/p57Kip2.
CONCLUSIONS: Collectively, our study reveals that the microenvironment influences the susceptibility of PDAC cells to GCs and provides unprecedented evidence for the anti-proliferative activity of budesonide on PDAC cells in 3D conditions, in vitro and in vivo. Our findings may explain, at least in part, the reason for the lower occurrence of pancreatic cancer in asthmatic patients and suggest a potential suitability of budesonide for clinical trials as a therapeutic approach to fight pancreatic cancer.
摘要:
背景:胰腺导管腺癌(PDAC)是最致命的癌症,具有侵袭性转移表型和非常差的临床预后。有趣的是,据报道,重度和长期哮喘患者的PDAC发生率较低.在这里,我们探讨了PDAC和糖皮质激素(GC)布地奈德之间的潜在联系,治疗哮喘的一线疗法。
方法:我们测试了布地奈德和经典GCs对形态学的影响,扩散,患者来源的PDAC细胞和胰腺癌细胞系的迁移和侵袭,使用2D和3D体外培养。此外,使用异种移植模型研究布地奈德对体内PDAC肿瘤生长的影响.最后,我们将全基因组转录组分析与遗传和药理学方法相结合,以探索不同环境条件下布地奈德活性的潜在机制.
结果:我们发现在2D文化设置中,高微摩尔浓度的布地奈德降低了PDAC细胞的间充质侵袭/迁移特征,不影响增殖或存活。该活性是特异性的并且不依赖于糖皮质激素受体(GR)。相反,在更生理的3D环境中,低纳摩尔浓度的布地奈德以GR依赖性方式显著降低PDAC细胞增殖.因此,我们发现布地奈德可降低体内PDAC肿瘤的生长.机械上,我们证明了3D环境驱动细胞向涉及蛋白质的一般代谢重编程,脂质,和能量代谢(例如,糖酵解依赖性增加)。这种代谢变化使PDAC细胞对布地奈德的抗增殖作用敏感,相反,这会引起相反的变化(例如,线粒体氧化磷酸化增加)。最后,我们提供布地奈德抑制PDAC生长的证据,至少在某种程度上,通过肿瘤抑制因子CDKN1C/p57Kip2。
结论:总的来说,我们的研究表明,微环境影响PDAC细胞对GC的敏感性,并为布地奈德在3D条件下对PDAC细胞的抗增殖活性提供了前所未有的证据,在体外和体内。我们的发现可以解释,至少在某种程度上,在哮喘患者中胰腺癌发病率较低的原因,并提示布地奈德可能适合作为抗胰腺癌的治疗方法进行临床试验。
公众号