Faculty Opinions recommendation of Deoxyribonucleic acid profiling analysis of 40 human thyroid cancer cell lines reveals cross-contamination resulting in cell line redundancy and misidentification.

Author(s):  
Barry Nelkin
2020 ◽  
Vol 4 (Supplement_1) ◽  
Author(s):  
Neel Rajan ◽  
Luis Bautista ◽  
Amy Adik ◽  
Matthew David Ringel

Abstract Cancer cells display lineage and cancer-specific transcriptional control of expression of oncogenic driver genes, particularly those regulated through super-enhancers. Compounds that target CDK7 and 9, key regulators of RNA polymerase II (RNAPII)-mediated gene transcription, selectively reduce transcription of “superenhanced” oncogenes and are in clinical trials. We previously identified that medullary cancer cell lines are highly sensitive to CDK7 inhibition and that the RET-gene is super-enhanced. In the present study, we sought to determine sensitivities of a panel of follicular cell-derived thyroid cancer cell lines to the CDK7 inhibitor, THZ1 and CDK9 inhibitor, AZD4573. We selected 8 independently confirmed thyroid cancer cell lines (TPC1, FTC133, BCPAP, SW1376; K1, THJ16, C643, and 8505C) from a variety of histological subtypes with different drive mutations and performed WST assays with increasing concentration of THZ1 or AZD4573 to determine IC50 for each cell line. Western blots were performed in parallel for target validation. IC50 values ranged from 5-100 nM for both compounds for all cell lines. All cell lines showed inhibition of CDK 7 phosphorylation (Ser 5) of RNAPII with retained CDK7 levels with THZ1 and loss of CDK9 RNAPII phosphorylation (Ser 2) with both compounds. However, treatment with either agent unexpectedly caused a reduction of total RNAPII protein levels. qRT-PCR did not reveal reduced mRNA levels in TPC1 cells with THZ1 treatment while, Bortezomib (proteasome inhibitor) co-treatment with THZ1 rescued RNAPII protein. These results are consistent with THZ1-induced proteasome degradation of RNAPII. BRAF protein levels also decreased in the hemizygous BRAF V600E-mutated cell line (8505C) but not the BRAF WT cell line (TPC1). qRT-PCR of both cell lines treated THZ-1 showed stable BRAF gene expression; further mechanistic studies are ongoing. In summary, human thyroid cancer cell lines are sensitive to inhibition of CDK7 and CDK9 likely through several mechanisms not all directly attributable to RNAPII inhibition.


2019 ◽  
Vol 25 (10) ◽  
pp. 3141-3151 ◽  
Author(s):  
Iñigo Landa ◽  
Nikita Pozdeyev ◽  
Christopher Korch ◽  
Laura A. Marlow ◽  
Robert C. Smallridge ◽  
...  

Thyroid ◽  
2013 ◽  
Vol 23 (3) ◽  
pp. 317-328 ◽  
Author(s):  
Geneviève Dom ◽  
Vanessa Chico Galdo ◽  
Maxime Tarabichi ◽  
Gil Tomás ◽  
Aline Hébrant ◽  
...  

2019 ◽  
Vol 25 (22) ◽  
pp. 6883-6884
Author(s):  
Iñigo Landa ◽  
Nikita Pozdeyev ◽  
Jeffrey A. Knauf ◽  
Bryan R. Haugen ◽  
James A. Fagin ◽  
...  

2001 ◽  
Vol 169 (2) ◽  
pp. 417-424 ◽  
Author(s):  
M Iitaka ◽  
S Kakinuma ◽  
S Fujimaki ◽  
I Oosuga ◽  
T Fujita ◽  
...  

Zinc at concentrations of 150, microM or higher induced necrosis as well as apoptosis in thyroid cancer cell lines. Necrosis was induced by zinc in a dose-dependent manner, whereas apoptosis did not increase at higher concentrations of zinc. The expression of the antiapoptotic protein phosphorylated Bad was markedly increased, whereas the expression of the proapoptotic proteins Bax and Bad decreased following Zn(2+) exposure. Zn(2+) induced rapid degradation of IkappaB, and an increase in the binding of nuclear transcription factor-kappaB (NF-kappaB). These observations indicate that antiapoptotic pathways were activated in thyroid cancer cells following exposure to Zn(2+). This may be a self-defence mechanism against apoptosis and may underlie the general resistance of thyroid cancer cells to apoptotic stimuli. Zinc may be a potential cytotoxic agent for the treatment of thyroid cancer.


2012 ◽  
Vol 214 (2) ◽  
pp. 207-216 ◽  
Author(s):  
Joanna Klubo-Gwiezdzinska ◽  
Kirk Jensen ◽  
Andrew Bauer ◽  
Aneeta Patel ◽  
John Costello ◽  
...  

The translocator protein (TSPO), formerly known as a peripheral benzodiazepine receptor, exerts pro-apoptotic function via regulation of mitochondrial membrane potential. We examined TSPO expression in human thyroid tumors (25 follicular adenomas (FA), 15 follicular cancers (FC), and 70 papillary cancers (PC)). The role of TSPO in the regulation of cell growth, migration, and apoptosis was examined in thyroid cancer cell lines after TSPO knockdown with siRNA and after treatment with TSPO antagonist (PK11195). Compared with normal thyroid, the level of TSPO expression was increased in FA, FC, and PC in 24, 26.6, and 55.7% of cases respectively. Thyroid cancer cell lines demonstrated variable levels of TSPO expression, without specific association with thyroid oncogene mutations. Treatment with inhibitors of PI3K/AKT or MEK/ERK signaling was not associated with changes in TSPO expression. Treatment with histone deacetylase inhibitor (valproic acid) increased TSPO expression in TSPO-deficient cell lines (FTC236 cells). TSPO gene silencing or treatment with PK11195 did not affect thyroid cancer cell growth and migration but prevented depolarization of mitochondrial membranes induced by oxidative stress. Induction of TSPO expression by valproic acid was associated with increased sensitivity of FTC236 to oxidative stress-inducible apoptosis. Overall, we showed that TSPO expression is frequently increased in PC. In vitro data suggested the role of epigenetic mechanism(s) in the regulation of TSPO in thyroid cells. Implication of TSPO in the thyroid cancer cell response to oxidative stress suggested its potential role in the regulation of thyroid cancer cell response to treatment with radioiodine and warrants further investigation.


2009 ◽  
Vol 27 (15_suppl) ◽  
pp. e14608-e14608
Author(s):  
T. Behlendorf ◽  
W. Voigt ◽  
T. Mueller ◽  
K. Jordan ◽  
D. Arnold ◽  
...  

e14608 Background: Recent data suggest that aberrant activation of PIK3/AKT-pathway and mTOR are involved in the development of thyroid cancer, particularly of anaplastic (ATC) and follicular (FTC) subtype. Therefore, mTOR could be a potential treatment target in thyroid cancer. Methods: To asses the potential role of mTOR as target for the treatment of thyroid cancer, two human ATC cell lines SW1736 and 8505C, the papillary thyroid cancer (PTC) cell line BCPAP and the FTC cell line FTC133 were exposed for 96h to increasing concentrations of the mTOR inhibitor RAD001 (Everolimus, kindly provided by Novartis, Switzerland). For combination experiments 10 nM of RAD001 were combined with increasing concentrations of either doxorubicin (DOX) or cisplatin (CDDP) continuously. Cytotoxicity was measured using the sulforhodamine B assay. IC50-values were calculated with Sigma Plot (Jandel Scientific) and drug interaction was determined by the model of Drewinko. Results: The observed IC50-values of RAD001 were 1nM (FTC133), 10 nM (BCPAP), 1000 nM (SW1736) and 9400 nM (8505C). In contrast to the pronounced differences in sensitivity as assessed on the basis of IC50, a growth inhibitory effect ≥ 25 % was seen in all cell lines at a concentration of 1 nM of RAD001. IC50 for CDDP ranged from 1,3–4,8 μM and for DOX from 8–40 nM. Combination of 10 nM RAD001 with either DOX or CDDP resulted in additive drug interaction with the exception in cell line 8505C where significant synergy was found for the combination with CDDP. Conclusion: RAD001 exerted interesting preclinical activity in two differentiated thyroid cancer cell lines. Mainly additive drug interaction in thyroid cancer cell lines was observed for combinations with CDDP and DOX. Mechanistic investigations are underway and will be presented at the meeting. At least for differentiated thyroid cancer mTOR-inhibition appeared promising, further evaluation in thyroid cancer seems warranted. No significant financial relationships to disclose.


Sign in / Sign up

Export Citation Format

Share Document