8-Azaguanine-resistant variants of cultured cells of Haplopappus gracilis

1978 ◽  
Vol 56 (21) ◽  
pp. 2660-2665 ◽  
Author(s):  
Robert B. Horsch ◽  
Gary E. Jones

Cultured cell lines of Haplopappus gracilis resistant to the purine analog 8-azaguanine (AG) have been isolated. One such line is 50 times more resistant to the drug than are wild-type cells. This line of AG-resistant cells also exhibits increased resistance to 8-azaadenine, but it responds to kinetin in a manner identical with that of wild-type cells. The resistance phenotype is stable for many generations in the absence of selection. In cultures of wild-type or mutant cells, more than 95% of cells are diploid.

1978 ◽  
Vol 33 (1) ◽  
pp. 157-169
Author(s):  
G. Wolf ◽  
L. Tejmar ◽  
S. Borell ◽  
W. Klietman

SV40-transformed hamster cells were selected for resistance to ethidium bromide (EB). Several cell lines were established, which grew in the presence of up to 250 microgram/ml EB. The EB resistance is genetically stable. The cloned resistant cells show no difference in morphology, with the exception of the mitochondrial ultrastructure, which exhibits condensed cristae formation. The tumorigenicity of these cells in Syrian gold hamsters is considerably reduced. Incorporation of radioactive labelled thymidine into mitochondrial DNA is not influenced by the presence of the drug. Gel electrophoresis with mitochondrial proteins from wild-type and resistant cells reveals significantly different patterns. The mechanism of EB resistance is discussed.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 113-113 ◽  
Author(s):  
Chad C. Bjorklund ◽  
Deborah J. Kuhn ◽  
Jairo A. Matthews ◽  
Michael Wang ◽  
Veerabhadran Baladandayuthapani ◽  
...  

Abstract Abstract 113 Background: Novel drugs such as the immunomodulatory agent lenalidomide have revolutionized the treatment of multiple myeloma, as evidenced by an increasing overall survival for patients with both newly-diagnosed, and relapsed and/or refractory disease. Despite these improvements, myeloma remains incurable, and is still characterized by a trend for increasing chemoresistance at relapse, with a decreasing duration of benefit from each successive line of therapy. By understanding the mechanisms responsible for the emergence of drug resistance, which have so far not been well characterized in the case of lenalidomide, it may be possible to rationally design novel regimens that could either overcome this resistance, or possibly prevent its emergence altogether. Methods: To improve our understanding of the mechanisms responsible for lenalidomide resistance, we developed cell line models of interleukin (IL)-6-dependent (ANBL-6 and KAS-6/1) and –independent (U266 and MM1.S) lenalidomide-resistant multiple myeloma cells. Starting at a concentration that was 1/10 of the IC50 for lenalidomide's anti-proliferative effects in drug-naïve cells, increasing drug concentrations were used until all the cell lines could proliferate and maintain cell membrane integrity in the presence of 10 μM lenalidomide. These cell lines were then used as an in vitro model of lenalidomide-specific drug resistance, and subjected to further characterization, including with gene expression profiling. Results: Resistance to lenalidomide was evidenced by a dramatic, 100-1000-fold increase in the IC50 values of these myeloma cells. In the case of ANBL-6 cells, for example, drug-naïve cells showed an IC50 of 0.14 μM using tetrazolium dye-based viability assays, but this increased to >100 μM in the drug-resistant cells, as was the case in U266 and MM1.S cells. This resistance was a stable phenotype, since removal of lenalidomide for seven to ninety days from cell culture conditions did not re-sensitize them when 10 μM lenalidomide was reintroduced. Gene expression profiling followed by pathway analysis to examine changes at the transcript level between wild-type parental and lenalidomide-resistant cell lines identified the Wnt/β-catenin pathway as the most altered across all cell lines. Increased expression was seen in several members of the low-density-lipoprotein receptor related protein family, including LRP1 and 5; members of the wingless-type MMTV integrations site family, including WNT3 and 4; β-catenin; and downstream Wnt/β-catenin targets such as CD44. Similar changes were detected in primary samples from a patient who developed clinically lenalidomide-refractory disease. Reporter assays revealed an up to 5-fold increase in LEF/TCF-dependent transcription both in drug-naïve cells acutely exposed to lenalidomide, and in their chronically exposed, lenalidomide-resistant clones. Western blotting and flow cytometry confirmed that these lenalidomide-resistant cells had increased expression by 2-20 fold of β-catenin and CD44, as well as other LEF/TCF targets, including Cyclin D1 and c-Myc. Comparable changes occurred after lenalidomide exposure in myeloma cells grown in the context of bone marrow stroma. Notably, lenalidomide-resistant cells showed decreased expression of casein kinase 1 and increased phosphorylation of glycogen synthase kinase 3 at Ser21/9, both of which would reduce the phosphorylation of β-catenin needed for its later proteasome-mediated degradation. Stimulation of the Wnt/β-catenin pathway with recombinant human Wnt3a resulted in resistance to lenalidomide in wild-type, drug-naïve cells, as evidenced by a 10-fold increase in the IC50. Conversely, exposure of lenalidomide-resistant cell lines to quercetin, a known antagonist of the β-catenin/TCF interaction, induced a partial re-sensitization to lenalidomide. Conclusions: These data support the hypothesis that activation of the Wnt/β-catenin pathway represents a mechanism of both acute and chronic resistance to the anti-proliferative effects of lenalidomide in multiple myeloma. Moreover, they support the development of strategies aimed at suppressing Wnt/β-catenin activity to resensitize multiple myeloma to the effects of this immunomodulatory agent in vivo. Disclosures: No relevant conflicts of interest to declare.


1992 ◽  
Vol 75 (3) ◽  
pp. 474-480 ◽  
Author(s):  
Hamed K Abbas ◽  
Chester J Mirocha ◽  
W Thomas Shier ◽  
Roland Gunther

Abstract Rats and cultured cells were treated with purified wortmannin toxin, crude fungal extracts, and partially purified preparations for periods of 5-96 h. A single dose of the toxic fractions containing the equivalent of 2.5 g fungus-infested rice resulted in death, hematuria, and hemorrhage in the urinary bladder, Intestine, stomach, heart, and thymus of rats. Purified wortmannin produced the same effects at 4 mg/kg. Fractions that were toxic in vivo also had cytotoxic properties with various cultured cell lines at concentrations ranging from 1.2 to >50 mg equivalents of fungus-infested rice extracted/mL Purified wortmannin was cytotoxic at 50 μg/mL.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1927-1927
Author(s):  
Anna C. Cunningham ◽  
Charles M. Mansbach ◽  
Alan E. Mast

Abstract Tissue factor pathway inhibitor (TFPI) is the major endogenous inhibitor of tissue factor initiated blood coagulation and a is key regulator of the development of intravascular thrombosis. TFPI indirectly binds to the endothelial surface through tight association with a GPI-anchored co-receptor. The location of recombinant TFPI expression in mammalian cells varies depending on the cell line used. In cell lines that do not produce endogenous TFPI, CHO and HEK293 cells, recombinant TFPI is secreted into the culture media. However, in a cell line that produces endogenous surface TFPI, EaHy926 cells, recombinant TFPI is expressed on the cell surface. These data suggest that TFPI is expressed on the surface of cells that produce the GPI-anchored co-receptor and is secreted by cells that do not. To further investigate the function of the co-receptor in TFPI cellular trafficking we developed aerolysin resistant ECV304 and EaHy926 cells lines. Both of these cell lines produce endogenous, surface associated TFPI. The cell lines were mutated with ethyl methanesulfonate and selected with aerolysin. Mutant cells lacking surface GPI-anchored proteins are resistant to the toxic effects of aerolysin and survive. The morphology and growth rate of the two aerolysin resistant cell lines are identical to that of the wild-type cells. They were first characterized to rule out the presence of a mutation that could directly alter cellular metabolism of TFPI. Sequencing of TFPI cDNA indicates that no mutations are present in the TFPI exons. Analysis of mRNA by real time PCR demonstrates that the aerolysin resistant cells make similar amounts of TFPI mRNA as their wild-type counterparts. Thus, transcription and translation of TFPI appear identical to the wild-type cells. In addition, the two independently derived cell lines have very similar phenotypes, as described below, indicating that the aerolysin resistant cell lines have a defect in GPI-anchor biosynthesis but not additional random mutations that could alter cellular processing of TFPI. Characterization of protein expression by flow cytometry indicates that the aerolysin resistant cell lines do not express GPI-anchored proteins (CD59, uPAR) or TFPI on the cell surface but do have wild-type surface expression of transmembrane proteins (CD9, tissue factor). Interestingly, instead of being secreted, western blot analysis of cellular lysates indicates that TFPI is degraded within the aerolysin resistant cells in a manner similar to that observed for GPI-anchored proteins. Intracellular degradation of TFPI is prevented by brefeldin A indicating that degradation takes place in a post endoplasmic reticulum compartment. Pepstatin A, but not MG-132, also prevents degradation, indicating that degradation is lysosomal rather than proteosomal. It appears that binding of TFPI to its co-receptor occurs early in cellular processing, likely within the endoplasmic reticulum. Cellular trafficking of TFPI is controlled by its co-receptor, which has not yet been identified. The co-receptor directs TFPI to the cell surface in wild-type endothelial cells or to be degraded in aerolysin resistant cells. In the absence if its co-receptor TFPI is secreted. Therefore, regulation of co-receptor expression provides a mechanism for the production of cell associated TFPI, as occurs in endothelial cells, versus soluble TFPI, as may occur in megakaryocytes/platelets.


2010 ◽  
Vol 298 (4) ◽  
pp. F935-F940 ◽  
Author(s):  
Janet D. Klein ◽  
Mitsi A. Blount ◽  
Otto Fröhlich ◽  
Chad E. Denson ◽  
Xiaoxiao Tan ◽  
...  

Vasopressin is the primary hormone regulating urine-concentrating ability. Vasopressin phosphorylates the UT-A1 urea transporter in rat inner medullary collecting ducts (IMCDs). To assess the effect of UT-A1 phosphorylation at S486, we developed a phospho-specific antibody to S486-UT-A1 using an 11 amino acid peptide antigen starting from amino acid 482 that bracketed S486 in roughly the center of the sequence. We also developed two stably transfected mIMCD3 cell lines: one expressing wild-type UT-A1 and one expressing a mutated form of UT-A1, S486A/S499A, that is unresponsive to protein kinase A. Forskolin stimulates urea flux in the wild-type UT-A1-mIMCD3 cells but not in the S486A/S499A-UT-A1-mIMCD3 cells. The phospho-S486-UT-A1 antibody identified UT-A1 protein in the wild-type UT-A1-mIMCD3 cells but not in the S486A/S499A-UT-A1-mIMCD3 cells. In rat IMCDs, forskolin increased the abundance of phospho-S486-UT-A1 (measured using the phospho-S486 antibody) and of total UT-A1 phosphorylation (measured by 32P incorporation). Forskolin also increased the plasma membrane accumulation of phospho-S486-UT-A1 in rat IMCD suspensions, as measured by biotinylation. In rats treated with vasopressin in vivo, the majority of the phospho-S486-UT-A1 appears in the apical plasma membrane. In summary, we developed stably transfected mIMCD3 cell lines expressing UT-A1 and an S486-UT-A1 phospho-specific antibody. We confirmed that vasopressin increases UT-A1 accumulation in the apical plasma membrane and showed that vasopressin phosphorylates UT-A1 at S486 in rat IMCDs and that the S486-phospho-UT-A1 form is primarily detected in the apical plasma membrane.


2009 ◽  
Vol 27 (15_suppl) ◽  
pp. e22103-e22103
Author(s):  
H. Otani ◽  
M. Jida ◽  
M. Takaoka ◽  
T. Kubo ◽  
T. Hayashi ◽  
...  

e22103 Background: Mutations in the epidermal growth factor receptor (EGFR) gene is the predictive factor for sensitivity of EGFR tyrosine kinase inhibitors (TKIs) in non-small cell lung cancer. Focal adhesion kinase (FAK) that is the downstream molecule of EGFR has been reported to be highly expressed in NSCLC suggesting novel therapeutic target of NSCLC. TAE226, dual inhibitor for FAK and insulin like growth factor-I receptor (IGF-IR), have been developed as anticancer reagent. In this study, we examined the effect of TAE226 on NSCLC from the view point of EGFR mutation status. Methods: We used NSCLC cell lines consisting of 4 EGFR mutant cell lines (PC9, H3255, HCC827, H1975) and 3 EGFR wild type cell lines (H1819, H1299, A549). We also used PC9 derived resistant cell line (RPC9). Antiproliferative effect of TAE226 on NSCLC cell lines was examined with MTS assay. The status of EGFR related molecules including its downstream signal pathway was investigated by western blotting analysis. The effect of TAE226 on xenograft mouse models was also examined. Results: TAE226 was effective on NSCLC cell lines with EGFR mutation including T790M mutation, compared to those with EGFR wild type. The value of IC50 (μmol/L) for PC-9, H3255, HCC827, H1975, RPC-9 and H1819, H1299, A549 was 0.16, 0.12, 0.086, 0.17, 0.31 and 4.7, 2.8, 1.4, respectively. Western blotting assay showed that TAE226 preferentially inhibited phosphor-EGFR and its downstream signaling mediators. We could confirm the anticancer effect of TAE226 on EGFR mutant cells was confirmed in xenograft mouse models. Conclusions: We indicated that TAE226 showed antitumor effect on EGFR mutant cell lines even T790M mutant cells. Further study is necessary to understand the mechanism of TAE226 effect on EGFR mutant cell lines. Our results suggest that TAE226 will be expected as the novel strategy for NSCLC. No significant financial relationships to disclose.


2015 ◽  
Vol 8 (1) ◽  
pp. 22-24
Author(s):  
Tahere Kashkolinejad-Koohi ◽  
Iraj Saadat ◽  
Mostafa Saadat

Abstract Teicoplanin is a glycopeptide antibiotic with a wide variation in human serum half-life. It is also a valuable alternative of vancomycin. There is however no study on its effect on cultured cells. The aim of the present study was to test the effect of teicoplanin on cultured cell lines CHO, Jurkat E6.1 and MCF-7. The cultured cells were exposed to teicoplanin at final concentrations of 0–11000 μg/ml for 24 hours. To determine cell viability, the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) test was performed. At low concentrations of teicoplanin the numbers of cultured cells (due to cell proliferation) were increased in the three cell lines examined. The maximum cell proliferation rates were observed at concentrations of 1000, 400, and 200 μg/ml of teicoplanin for CHO, MCF-7 and Jurkat cell lines, respectively. Cell toxicity was observed at final concentrations over 2000, 6000, and 400 μg/ml of teicoplanin for CHO, MCF-7 and Jurkat cell lines, respectively. A dose-dependent manner of cell toxicity was observed. Our present findings indicated that teicoplanin at clinically used concentrations induced cell proliferation. It should therefore be used cautiously, particularly in children, pregnant women and patients with cancer.


Cancers ◽  
2020 ◽  
Vol 12 (2) ◽  
pp. 517 ◽  
Author(s):  
Karuna Mittal ◽  
Da Hoon Choi ◽  
Guanhao Wei ◽  
Jaspreet Kaur ◽  
Sergey Klimov ◽  
...  

Human papillomavirus-negative (HPV-neg) oropharyngeal squamous cell carcinomas (OPSCCs) are associated with poorer overall survival (OS) compared with HPV-positive (HPV-pos) OPSCCs. The major obstacle in improving outcomes of HPV-neg patients is the lack of robust biomarkers and therapeutic targets. Herein, we investigated the role of centrosome amplification (CA) as a prognostic biomarker in HPV-neg OPSCCs. A quantitative evaluation of CA in clinical specimens of OPSCC revealed that (a) HPV-neg OPSCCs exhibit higher CA compared with HPV-pos OPSCCs, and (b) CA was associated with poor OS, even after adjusting for potentially confounding clinicopathologic variables. Contrastingly, CA was higher in HPV-pos cultured cell lines compared to HPV-neg ones. This divergence in CA phenotypes between clinical specimens and cultured cells can therefore be attributed to an inaccurate recapitulation of the in vivo tumor microenvironment in the cultured cell lines, namely a hypoxic environment. The exposure of HPV-neg OPSCC cultured cells to hypoxia or stabilizing HIF-1α genetically increased CA. Both the 26-gene hypoxia signature as well as the overexpression of HIF-1α positively correlated with increased CA in HPV-neg OPSCCs. In addition, we showed that HIF-1α upregulation is associated with the downregulation of miR-34a, increase in CA and expression of cyclin- D1. Our findings demonstrate that the evaluation of CA may aid in therapeutic decision-making, and CA can serve as a promising therapeutic target for HPV-neg OPSCC patients.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2683-2683 ◽  
Author(s):  
Weiguo Zhang ◽  
Gautam Borthakur ◽  
Chen Gao ◽  
Ye Chen ◽  
Yong S. Lan ◽  
...  

Abstract Internal tandem duplication (ITD) or point mutation of Fms-like tyrosine kinase 3 (FLT3) and N/KRAS mutations in patients with acute myeloid leukemia (AML) lead to aberrant activation of FLT3 and/or RAS–mitogen-activated protein kinase (MAPK) pathways and are associated with poor prognosis (Kottaridis et al, Leuk Lymphoma Vol. 44:905, 2003; Thiede et al, Blood Vol. 99;4326, 2002). Therapy with inhibitors targeting these pathways individually may at best result in short lasting responses in the appropriate mutational context (Borthakur et al, Haematologica Vol. 96:62, 2011; Cortes et al, Blood Vol. 114:636a, 2009). Persistent activation of MEK/ERK signaling pathway is seen in cells resistant to FLT3-ITD inhibitor sorafenib that harbor acquired point mutations of FLT3 in tyrosine kinase domains in addition to ITD mutation (Moore et al, Leukemia Vol. 26:1462, 2012). E6201 is a MEK1/FLT3 dual inhibitor with inhibitory activity in low nanomolar concentrations against both targets. We tested E6201 against AML cells including FLT3-inhibitor resistant cells, AML patient samples and investigated its efficacy in murine AML model. E6201 inhibited cell growth and induced apoptosis in AML cells with FLT3 ITD mutations (including sorafenib-resistant cells harboring ITD plus N676D/Y842C point mutations) at nanomolar levels, and showed 600 to 1000-fold more selective activity against cells with FLT3-ITD mutations than those with FLT3-WT (IC50s 0.003µM, 0.005 and 0.002µM, respectively, in Ba/F3-ITD and FLT3-ITD mutant MOLM13 and MV4-11cells compared to 3.18µM in Ba/F3-FLT3-WT cells). In addition, OCI/AML3 cells [FLT3 and RAS wild-type(WT)], which have high basal p-ERK level and are resistant to most of chemotherapeutic drugs, were sensitive to E6201 (IC50 = 0.037µM). Consistent with its MEK1 inhibitory activity, E6201 was more active against NRAS mutation carrying OCI/AML3 and MV4-11 cells than their NRAS-WT isogenic cells. E6201-induced apoptosis appears to be p53 dependent as p53-wild-type OCI/AML3 and MOLM13 cells were significantly more sensitive compared to their paired p53-knockdown cells. EC50 of E6201 was at sub-micromolar levels in all 5 FLT3-ITD mutant primary AML samples, which included one with FLT3-ITD/RAS dual mutation. NOG mice bearing xenografts of MOLM13-Luc-GFP (FLT3-ITD mutated) cells were treated with E6201 i.v. starting on day 5 after leukemia cell injection until day 21 on a twice-per-week schedule. Bioluminescence imaging revealed that the tumor burden (mean luminescence) was reduced (3.1 x 106 and 2.7 x 106vs. 5.6 x 106 Photons/sec, p< .01 in 20mg/kg and 40mg/kg groups compared with vehicle group)(Fig 1) and histologically leukemia cells infiltrations were profoundly reduced in the bone marrows, spleens, livers and lungs on Day 9 after first drug treatment. Additionally, the median survival was modestly extended from 16 days of vehicle-treated mice to 18 days of E6201-treated mice (P <0.01). Mechanistically, E6201 significantly suppressed p-FLT3 and p-ERK in all tested FLT3 mutant AML cell lines and p53 wild-type OCI/AML3 cells. In addition, decrease of Bcl-xL and Mcl-1 levels and increase of cleaved-caspase-3 was observed in all FLT3 mutant cell lines after treatment with E6201 for 24 hours. In conclusion, E6201 is active against AML cells with FLT3 and/or RAS mutation. A clinical trial is in development in FLT3 and/or RAS mutant AML. Disclosures: Borthakur: Eisai, Inc.: Research Funding. Nomoto:Eisai, Inc.: Employment. Zhao:Eisai, Inc.: Employment.


2009 ◽  
Vol 27 (15_suppl) ◽  
pp. e22064-e22064
Author(s):  
T. Kubo ◽  
N. Kobayashi ◽  
M. Jida ◽  
T. Hayashi ◽  
H. Yamamoto ◽  
...  

e22064 Background: It has been reported that NSCLC cell lines with activating mutations of EGFRgene were significantly sensitive to Heat Shock Protein 90 (Hsp 90 ) ihibitors regardless of the secondary T790M mutation. Methods: We examined the antitumor effect of 17-dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG), the orally active Hsp90 inhibitor, on the growth of NSCLC cell lines in vitro and in vivo. Results: In MTS assay, the IC50 values for 3 cell lines with EGFR mutation (PC-9, RPC-9, and NCI-H1975) ranged from 0.15 to 0.28 μM. RPC-9 is a gefitinib-resistant lung cancer cell line with the secondary T790M mutation established from PC-9. By contrast, those of 7 EGFR-wild type cell lines ranged from 1.63 to 28.5 μM. Western blot analysis revealed that mutant EGFRs were more readily depleted than wild-type EGFRs after the treatment of 17-DMAG. Furthermore, expression of phospho-Akt, phospho-MAPK, cdk4, and cyclin D1 was depleted after exposure to low concentrations of 17-DMAG in EGFR-mutant cells. Celeaved PARP expression confirmed apoptosis in response to 17-DMAG in EGFR-mutant cells. In animal models, 17-DMAG significantly reduced the growth of EGFR-mutant cell lines PC-9 and RPC-9 compared with controls and induced degradation of mutant EGFR. In contrast, 17-DMAG did not inhibit the growth of EGFR wild type cell lines compared with controls. Conclusions: These results suggested that 17-DMAG could become a novel therapeutic agent for patients with lung tumors expressing mutant EGFR and having developed clinical resistance to gefitinib. No significant financial relationships to disclose.


Sign in / Sign up

Export Citation Format

Share Document