Taurolidine and povidone-iodine induce different types of cell death in malignant pleural mesothelioma

Lung Cancer ◽  
2007 ◽  
Vol 56 (3) ◽  
pp. 327-336 ◽  
Author(s):  
I. Opitz ◽  
B. Sigrist ◽  
S. Hillinger ◽  
D. Lardinois ◽  
R. Stahel ◽  
...  
2014 ◽  
Vol 25 ◽  
pp. v1 ◽  
Author(s):  
S. Cedres ◽  
S. Ponce Aix ◽  
J. Zugazagoitia ◽  
A.B. Anguita ◽  
I. Sansano ◽  
...  

2017 ◽  
Vol 7 (3) ◽  
pp. e1398874 ◽  
Author(s):  
Chiara Riganti ◽  
Marcello Francesco Lingua ◽  
Iris Chiara Salaroglio ◽  
Chiara Falcomatà ◽  
Luisella Righi ◽  
...  

PLoS ONE ◽  
2015 ◽  
Vol 10 (3) ◽  
pp. e0121071 ◽  
Author(s):  
Susana Cedrés ◽  
Santiago Ponce-Aix ◽  
Jon Zugazagoitia ◽  
Irene Sansano ◽  
Ana Enguita ◽  
...  

Lung Cancer ◽  
2016 ◽  
Vol 96 ◽  
pp. 1-6 ◽  
Author(s):  
S. Cedrés ◽  
S. Ponce-Aix ◽  
N. Pardo-Aranda ◽  
A. Navarro-Mendivil ◽  
A. Martinez-Marti ◽  
...  

Author(s):  
Marika Rossini ◽  
Fernanda Martini ◽  
Elena Torreggiani ◽  
Francesca Fortini ◽  
Giorgio Aquila ◽  
...  

Malignant pleural mesothelioma (MPM) is an aggressive asbestos-related cancer arising from the mesothelial cells lining the pleural cavity. MPM is characterized by a silent clinical progression and a highly resistance to conventional chemo/radio-therapies. MPM patients die in a few months/years from diagnosis. Notch signaling is a well-conserved cell communication system, which regulates many biological processes. In humans, the dysregulation of Notch pathway potentially contributes to cancer onset/progression, including MPM. Metformin is the first-line drug used to treat type 2 diabetes mellitus. Metformin is proven to be an effective antitumor drug in preclinical models of different types of cancer. To date, clinical efficacy is being studied in many clinical trials. In this study, the anti-proliferative effect of metformin on MPM cells and the putative involvement of Notch1 as a mediator of metformin activities, were investigated. MPM cells showed high levels of Notch1 activation compared to normal pleural mesothelial cells. Furthermore, metformin treatment hampered MPM cell proliferation and enhanced the apoptotic process, accompanied by decreased Notch1 activation.


2019 ◽  
Vol 10 (10) ◽  
Author(s):  
Sayantani Sarkar Bhattacharya ◽  
Prabhu Thirusangu ◽  
Ling Jin ◽  
Debarshi Roy ◽  
Deokbeom Jung ◽  
...  

Abstract The metabolic signatures of cancer cells are often associated with elevated glycolysis. Pharmacological (PFK158 treatment) and genetic inhibition of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), a critical control point in the glycolytic pathway, decreases glucose uptake, ATP production, and lactate dehydrogenase activity and arrests malignant pleural mesothelioma (MPM) cells in the G0/G1 phase to induce cell death. To overcome this nutrient stress, inhibition of PFKFB3 activity led to an escalation in endoplasmic reticulum (ER) activity and aggravated ER stress mostly by upregulating BiP and GADD153 expression and activation of the endocytic Rac1-Rab5-Rab7 pathway resulting in a unique form of cell death called “methuosis” in both the sarcomatoid (H28) and epithelioid (EMMeso) cells. Transmission electron microscopy (TEM) analysis showed the formation of nascent macropinocytotic vesicles, which rapidly coalesced to form large vacuoles with compromised lysosomal function. Both immunofluorescence microscopy and co-immunoprecipitation analyses revealed that upon PFKFB3 inhibition, two crucial biomolecules of each pathway, Rac1 and Calnexin interact with each other. Finally, PFK158 alone and in combination with carboplatin-inhibited tumorigenesis of EMMeso xenografts in vivo. Since most cancer cells exhibit an increased glycolytic rate, these results provide evidence for PFK158, in combination with standard chemotherapy, may have a potential in the treatment of MPM.


2009 ◽  
Vol 8 (24) ◽  
pp. 2406-2416 ◽  
Author(s):  
Sharyn I. Katz ◽  
LanLan Zhou ◽  
Grace Chao ◽  
Charles D. Smith ◽  
Thomas Ferrara ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document