Survival and Function of Fibroblasts Stored as Stock Cultures at a Non-freezing Temperature

1993 ◽  
Vol 21 (2) ◽  
pp. 206-209
Author(s):  
Anders H. G. Andrén ◽  
Anders P. Wieslander

Cytotoxicity, measured as inhibition of cell growth of cultured cell lines, is a widely used method for testing the safety of biomaterials and chemicals. One major technical disadvantage with this method is the continuous routine maintenance of the cell lines. We decided to investigate the possibility of storing stock cultures of fibroblasts (L-929) in an ordinary refrigerator as a means of reducing the routine workload. Stock cultures of the mouse fibroblast cell line L-929 were prepared in plastic vials with Eagle's minimum essential medium. The vials were stored in a refrigerator at 4–10°C for periods of 7–31 days. The condition of the cells after storage was determined as cell viability, cell growth and the toxic response to acrylamide, measured as cell growth inhibition. We found that the L-929 cell line can be stored for 2–3, weeks with a viabilty > 90% and a cell growth of about 95%, compared to L-929 cells grown and subcultured in the normal manner. The results also show that the toxic response to acrylamide, using refrigerator stored L-929 cells, corresponds to that of control L-929 cells. We concluded that it is possible to store L-929 cells in a refrigerator for periods of up to 3 weeks and still use the cells for in vitro cytotoxic assays.


2018 ◽  
Vol 15 (8) ◽  
pp. 1154-1160
Author(s):  
Edgars Abele ◽  
Ramona Abele ◽  
Lena Golomba ◽  
Ilona Domracheva ◽  
Tatjana Beresneva ◽  
...  

Aims and Objectives: The aim of the research is to obtain and to investigate the cytotoxicity of a novel class of non-toxic oximes – derivatives of N-(benzo[4,5]imidazo[2,1-b]thiazol-3-ylmethoxy)-ω- (hetarylsulfanyl)-alkanamidines. Materials and Methods: To assess the possible toxicity of the compounds, acute oral LD50 was calculated. The calculations were based on tested compounds IC50 values in relation to 3T3 (mouse fibroblast cell line) using NRU assay. Monolayer tumor cell lines HT-1080 (human fibrosarcoma, ATCC® CCL-121™), MH-22A (mouse hepatoma, ECACC code, 96121721) and NIH/3T3 (mouse Swiss Albino embryo fibroblasts, ATCC® CRL-1658™), were cultured in standard medium (Dulbecco`s modified Eagle`s medium) supplemented with 10% fetal bovine serum (“Sigma”). Results: E-Stereoselective synthesis of novel N-(benzo[4,5]imidazo[2,1-b]thiazol-3-ylmethoxy)-ω- (hetarylsulfanyl)alkanamidines as potential cytotoxic agents was carried out in three steps from corresponding thiones. N-(Benzo[4,5]imidazo[2,1-b]thiazol-3-ylmethoxy)-5-(benzothiazol-2-ylsulfanyl)-pentanamidine exhibits high activity in vitro on the monolayer tumour cell lines: MG-22A (mouse hepatoma) and HT-1080 (human fibrosarcoma). Besides this, the above compound exhibits low toxicity on the 3T3 cell line and low estimated acute oral LD50 (LD50 2759 mg/kg). Conclusion: In conclusion, such dramatic decreasing of expected acute toxicity and high cytotoxicity by the introduction of benzo[4,5]imidazo[2,1-b]thiazolyl fragment into N-hydroxy-ω-(hetarylsulfanyl)alkanamidines were demonstrated for the first time (see, for example, toxicity and cytotoxicity of compound 8b and corresponding unsubstituted oxime).



2020 ◽  
Vol 25 (01) ◽  
pp. 10-18
Author(s):  
Fatma Yurt ◽  
Ece Tugba Saka ◽  
Zekeriya Biyiklioglu ◽  
Ayça Tunçel ◽  
Derya Ozel ◽  
...  

In this study, two SiPcs have been selected and the photodynamic therapy potentials were evaluated of the Pcs. Synthesis of Axially 2-decyn-1-oxy disubstituted Es-SiPc-2 was newly synthesized by the reaction of SiPcCl2 with 2-decyn-1-ol in the presence of NaH in toluene. Furthermore, their nuclear imaging potentials were evaluated in human colon adenocarcinoma (HT-29) and human lung fibroblast cell (WI-38) cell lines. The uptake results have indicated that Es-SiPc labeled with [Formula: see text]I radionuclide ([Formula: see text]I-Es-SiPc) was approximately 2-fold higher in the HT-29 cell line than the WI-38 cell line. In other words, the target/non-target tissue ratio is defined as two in the HT-29/WI-38 cell lines. Besides, the uptake values of [Formula: see text]I-Es-SiPc were found to be higher than [Formula: see text]I-Es-SiPc-2. [Formula: see text]I-Es-SiPc and [Formula: see text]I-Es-SiPc-2 are promising for imaging or treating colon adenocarcinoma. In vitrophotodynamic therapy (PDT) studies have shown that both compounds are suitable and can be used in this field. Also, Es-SiPc has been shown to have higher phototoxicity than Es-SiPc-2.



Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1653-1653
Author(s):  
Silvia Locatelli ◽  
Arianna Giacomini ◽  
Anna Guidetti ◽  
Loredana Cleris ◽  
Michele Magni ◽  
...  

Abstract Abstract 1653 Introduction: A significant proportion of Hodgkin lymphoma (HL) patients refractory to first-line chemotherapy or relapsing after autologous transplantation are not cured with currently available treatments and require new treatments. The PI3K/AKT and RAF/MEK/ERK pathways are constitutively activated in the majority of HL. These pathways can be targeted using the AKT inhibitor perifosine (Æterna Zentaris GmBH, Germany, EU), and the RAF/MEK/ERK inhibitor sorafenib (Nexavar®, Bayer, Germany, EU). We hypothesized that perifosine in combination with sorafenib might have a therapeutic activity in HL by overcoming the cytoprotective and anti-apoptotic effects of PI3K/Akt and RAF/MEK/ERK pathways. Since preclinical evidence supporting the anti-lymphoma effects of the perifosine/sorafenib combination are still lacking, the present study aimed at investigating in vitro and in vivo the activity and mechanism(s) of action of this two-drug combination. METHODS: Three HL cell lines (HD-MyZ, L-540 and HDLM-2) were used to investigate the effects of perifosine and sorafenib using in vitro assays analyzing cell growth, cell cycle distribution, gene expression profiling (GEP), and apoptosis. Western blotting (WB) experiments were performed to determine whether the two-drug combination affected MAPK and PI3K/AKT pathways as well as apoptosis. Additionally, the antitumor efficacy and mechanism of action of perifosine/sorafenib combination were investigated in vivo in nonobese diabetic/severe combined immune-deficient (NOD/SCID) mice. RESULTS: While perifosine and sorafenib as single agents exerted a limited activity against HL cells, exposure of HD-MyZ and L-540 cell lines, but not HDLM-2 cells, to perifosine/sorafenib combination resulted in synergistic cell growth inhibition (40% to 80%) and cell cycle arrest. Upon perifosine/sorafenib exposure, L-540 cell line showed significant levels of apoptosis (up to 70%, P ≤.0001) associated with severe mitochondrial dysfunction (cytochrome c, apoptosis-inducing factor release and marked conformational change of Bax accompanied by membrane translocation). Apoptosis induced by perifosine/sorafenib combination did not result in processing of caspase-8, -9, -3, or cleavage of PARP, and was not reversed by the pan-caspase inhibitor Z-VADfmk, supporting a caspase-independent mechanism of apoptosis. In responsive cell lines, WB analysis showed that anti-proliferative events were associated with dephosphorylation of MAPK and PI3K/Akt pathways. GEP analysis of HD-MyZ and L-540 cell lines, but not HDLM-2 cells indicated that perifosine/sorafenib treatment induced upregulation of genes involved in amino acid metabolism and downregulation of genes regulating cell cycle, DNA replication and cell death. In addition, in responsive cell lines, perifosine/sorafenib combination strikingly induced the expression of tribbles homologues 3 (TRIB3) both in vitro and in vivo. Silencing of TRIB3 prevented cell growth reduction induced by perifosine/sorafenib treatment. In vivo, the combined perifosine/sorafenib treatment significantly increased the median survival of NOD/SCID mice xenografted with HD-MyZ cell line as compared to controls (81 vs 45 days, P ≤.0001) as well as mice receiving perifosine alone (49 days, P ≤.03) or sorafenib alone (54 days, P ≤.007). In mice bearing subcutaneous nodules generated by HD-MyZ and L-540 cell lines but not HDLM-2 cell line, perifosine/sorafenib treatment induced significantly increased levels of apoptosis (2- to 2.5-fold, P ≤.0001) and necrosis (2- to 8-fold, P ≤.0001), as compared to controls or treatment with single agents. CONCLUSIONS: Perifosine/sorafenib combination resulted in potent anti-HL activity both in vitro and in vivo. These results warrant clinical evaluation in HL patients. Disclosures: No relevant conflicts of interest to declare.





Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4496-4496
Author(s):  
Dario Ferrero ◽  
Graziella Bellone ◽  
Valentina Giai ◽  
Anna Carbone ◽  
Patrizia Scaravaglio ◽  
...  

Abstract In vitro growth of acute myelogenous leukemia blasts as an established cell line is an uncommon event and molecular mechanisms allowing permanent in vitro proliferation have not been defined. In particular, few acute monoblastic leukemia (AMoL) cell lines have been described. Here we report a new AMoL cell line expressing a combination of growth factors and growth factor receptor that might be involved in an autocrine stimulation loop. AMoL (M5a according to F.A.B) was diagnosed in November 2004 in a 74 old woman. Caryotypic analysis revealed a combination of 46,XX, 47,XX,+8, 47,XX,+8,6q− caryotypes. A complete remission was achieved by an age- adapted FLAG-Ida regimen but caryotype analysis still revealed about 50% mitosis with 47,XX,+8 and in February 2005 the patient presented an isolated cutaneous relapse. In spite of treatment with low dose cytarabine, etoposide and retinoids, hematologic progression occurred in May 2005. Further treatments with vinblastine, hydroxyurea and gentuzumab-ozogamycine were only temporary effective and in June 2005 the patient died, with hyperleukocytosis, extensive cutaneous and possibly CNS involvement. A 10 ml peripheral blood sample was taken, after informed consent, at the time of the last progression and mononuclear cells were collected. Part the cells were cryopreserved and part cultured in RPMI medium + 15% fetal bovine serum (FBS). Cultured cells survived without significant expansion for 4 weeks, then progressive growth started. After a few passage the cell line (named PI-BR) has been established, with a doubling time of approximately 48 hours. During early passages cell growth was density dependent, impaired at concentrations lower than 3 ×10e4/ml. Morphology and cytochemical stains were in agreement to M5a F.A.B. subtype. Immunophenotypic analysis revealed an antigenic expression comparable to that of patient’s blasts: CD13++, CD33++, HLA-DR ++, CD64++, CD117 +, CD15+/−, CD34−, CD19−, CD9−. Caryotypic analysis was performed on patient’s cells cryopreserved when culture was initiated: only 4 mitosis were found: 2 with normal 46,XX, 2 with 47,XX,+8 caryotype. Caryotypic analysis, repeated on PI-BR cell line at passage 9, revealed a mixture of mitosis with 47,XX,+8, 47,XX,+ ring, 48,XX,+8,+ ring, normal 46,XX and hypodiploid (42–44 chromosome) caryotype. Therefore, although some chromosome abnormalities were acquired during in vitro culture, they do not seem responsible for the capability of continuous in vitro growth, since about 55% of cells at passage 9 maintained the same caryotypes of patient’s cells in vivo. Real time PCR revealed expression of genes codifying for some monocytic cytokines (IL6, IL8, IL10, IL18, TGF-beta1), Vascular Endotelium Growth Factor (VEGF)-A, VEGF-D and its receptor Flt-4. Immunoistochemistry confirmed the synthesis of Flt-4, VEGF-A and VEGF-D. VEGF-D was also detected by ELISA assay in cell line supernatant (14.7 pg/ml) at early passages. VEGF A and C and VEGF receptors have been reported to be expressed in some AML cell lines and in some fresh AML cell samples, displaying anti-apoptotic and proliferative activity. In the case of PI-BR cell line, synthesis of VEGF-D and of its receptor Flt-4 may have established an autocrine loop involved in continuous cell growth.



Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4063-4063 ◽  
Author(s):  
Shiva Bamezai ◽  
Jing He ◽  
Deniz Sahin ◽  
Fabian Mohr ◽  
Fabio Ciccarone ◽  
...  

Abstract DNA methylation patterns are highly deregulated in human acute myeloid leukemia (AML) cases and stratify AML patient samples into different subgroup. AML1-ETO is the most commonly occurring fusion gene in AML and these AML cases exhibit an aberrant and distinct methylation pattern. So far, the underlying mechanisms for this are only poorly understood. The TET1 dioxygenase has recently emerged as an important epigenetic modifier: by catalyzing the conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) TET1 plays an important role in active demethylation, thereby regulating a variety of biological processes. It was linked to tumorigenesis based on the observation that its expression is frequently deregulated in solid cancer. However, the role of TET1 in AML1-ETO+ (AE+)human AML cases is yet unexplored. Using quantitative real time (qRT)- PCR we now show that AE+ AML is characterized by high and aberrant expression of TET1: the gene was significantly higher expressed in the majority of AE+ patients (n=7, p<0.01) compared to other AML subtypes such as inv(16) (n=11), PML-RARα+ (n=31), cytogenetically normal (CN)-AML patients (n=33) and CD34+ normal BM cells (n=4). This observation was consistent with published cDNA microarray data on large patient cohorts (Haferlach et al., JCO 2010, p<0.008 t-test, p<0.01 Anova) and recently published transcriptome data (TCGA) of AML patients. In contrast to TET1, TET2 and TET3 did not show significant higher expression in AE+ patients compared to other AML subtypes. In line with patient data, TET1 was highest expressed in the AE+ AML cell line KASUMI-1 and SKNO-1 compared to other AML cell lines (p<0.05 and n=3). Compared to normal CD34+ and myeloid (CD33+, CD15+ and CD14+) cells (n=3), TET1 was 10-fold and 16-fold higher expressed in AE+ patient samples (n= 7). Aberrant expression of TET1 in AE+ leukemic cells was associated with hypomethylation of its promoter and enrichment for H3K4me3 euchromatic marks at its promoter as determined by LC/MS and ChIP-qPCR respectively. Knockdown (KD) of TET1 mRNA using two short hairpin RNAs (shRNAs) in AE+ AML cell lines impaired their cell growth and clonogenicity by over 50% in vitro (n=3 and p<0.01). shRNA mediated depletion of TET1 did not impact the cell growth and clonogenicity of the TET1 negative cell line RAJI, ruling out off target effects of the shRNAs (n=3). In mice, KD of Tet1 in leukemic bone marrow cells expressing the truncated leukemogenic AML1-ETO9a (AE9a) fusion, dramatically inhibited cell growth (>60% compared to scrambled, n=3, p<0.01), clonogenicity (>50-70% reduction in primary CFCs, p<0.01, n=3) and importantly delayed onset of leukemia in vivo (median survival 35 days for scr vs 80 days for shRNA mice, n=4/arm, p<0.03). Tet1-knock-out c-kit+ hematopoietic stem and progenitor cells (HSPCs) transduced with AE9a showed reduced primary colony formation and impaired serial replanting capacity in vitro compared to AE9a transduced Tet1-wild-type HSPCs (>50% and >70%, respectively; p<0.001, n=3). Global analysis of 5hmC and 5mC levels using hMeDIP/MeDIP-seq performed on TET1 depleted KASUMI-1 cells revealed lower global 5hmC levels and increase in 5mC as compared to cells transduced with scrambled control (n=2). 3324 promoter regions lost 5hmC and gained 5mC upon TET1 depletion (-5kTSS, Fold enrichment cut off <2-fold, q-value<1e5). Recent studies have shown that PARP activity induces TET1 expression by regulating its promoter epigenetically. We could show that aberrant TET1 expression could be antagonized by the PARP inhibitor olaparib in AE+ leukemic cell lines. Furthermore, olaparib treatment decreased 5hmC levels and reduced cell growth and clonogenicity of human AE+ cell lines and of the murine AE9a+ leukemic cell line in vitro (n=3, p<0.01). In conclusion, our data indicates that aberrant TET1 expression contributes to the growth of AE+ AML by maintaining the 5-hydroxymethylome and that the PARP inhibitor olaparib can at least partially antagonize the oncogenic effect of TET in AML. Disclosures Mulaw: NuGEN: Honoraria. Buske:Celltrion, Inc.: Consultancy, Honoraria.



2021 ◽  
Vol 11 ◽  
Author(s):  
Fangqin Yu ◽  
Runsheng Ma ◽  
Chenguang Liu ◽  
Lele Zhang ◽  
Kaixiang Feng ◽  
...  

BackgroundThyroid cancer is one of the most common endocrine malignancies worldwide, and papillary thyroid cancer (PTC) is the most common pathologic type of thyroid cancer. SQSTM1/p62 activity mediates different biological functions. This study aimed to investigate the effect of SQSTM1/p62, a multifunctional receptor, on biological function and autophagy characteristics in the human PTC cell line TPC-1.MethodsA total of 105 primary PTC samples and matched adjacent normal thyroid tissue samples were obtained to evaluate the expression of p62 in clinical patients. A similar p62 expression pattern was found in PTC cell lines and normal human thyroid follicular epithelial cells. To evaluate the effect of SQSTM1/p62 on TPC-1 cells, we constructed the p62 knockout cell line p62-KO-TPC-1. Cell proliferation, cell cycle, and cell apoptosis were analyzed by colony formation tests, Cell Counting Kit-8 (CCK-8) assays and flow cytometry in vitro. TPC-1 and p62-KO-TPC-1 human PTC cell lines in the logarithmic growth phase were subcutaneously implanted into BALB/c nude mice to verify their proliferation effect in vivo. Furthermore, western blotting and immunohistochemistry (IHC) were used to detect the expression of AKT/AMPK/mTOR signaling pathway-related proteins.ResultsOverall, p62 expression was higher in tumor tissues than in normal tissues in 73 of 105 PTC patients (69.5%). The expression level of p62 in the PTC cell line was higher than that in the normal thyroid cell line. Our data indicated that in vitro, p62 deficiency could decrease the number of colonies, inhibit cell growth and the cell cycle, and induce apoptosis. Tumor xenograft experiments in BALB/c nude mice corroborated these findings. Moreover, the molecular mechanism was explored by western blotting, and we found that the AMPK/AKT/mTOR pathway was involved.ConclusionsThe results indicate that p62 might mediate cell autophagy and apoptosis in TPC-1 cells via the AMPK/AKT/mTOR pathway and could be used as a potential therapeutic approach for PTC.



Molecules ◽  
2018 ◽  
Vol 23 (8) ◽  
pp. 2067 ◽  
Author(s):  
Krzysztof Słomiak ◽  
Andrzej Łazarenkow ◽  
Lilianna Chęcińska ◽  
Joachim Kusz ◽  
Justyn Ochocki ◽  
...  

The hydrazine and hydrazide derivatives of benzo-γ-pyrones with fluorine substituents remain an unexplored group of chemical compounds. This preliminary study reports the synthesis, structural assessment, initial microbiological screening and biological testing of the synthesized compounds on cell lines using the XTT-assay. A series of 10 novel hydrazine and hydrazide derivatives of 3-formylchromone were synthesized and their structures determined. Structural assessment consisted of elemental analysis, IR, 1H-NMR, 13C-NMR, MS and crystallographic studies. Antimicrobial activity was tested on standard strains representing different groups of microorganisms. The tested compounds were found to inhibit microbial growth. Concentrations of 0.01–1250 µmol/L were found to influence cell proliferation, demonstrating antiproliferative and stimulation of proliferation against two cell lines: the L929 cell line (mouse fibroblast cell line) and the EA.hy926 cell line (the human umbilical vein, somatic cell hybrid).



2001 ◽  
Vol 169 (2) ◽  
pp. 309-320 ◽  
Author(s):  
CM Pariante ◽  
BD Pearce ◽  
TL Pisell ◽  
C Su ◽  
AH Miller

RU40555 is a recently available glucocorticoid receptor (GR) antagonist that differs from RU486 by a methyl radical. We have used the mouse fibroblast cell line L929 to study the in vitro effects of RU40555 on GR translocation and function and on the membrane steroid hormones transporter. The results showed that: 1) RU40555 competed for the binding of labelled dexamethasone (Dex) with a K(i) of 2.4 nM; 2) both RU40555 and RU486 were equally potent inhibitors of Dex-induced GR-mediated gene transcription; 3) maximum GR translocation induced by micromolar concentrations of Dex and the GR antagonists was approximately 30-55% loss in the cytoplasmic GR and approximately 40-90% increase in the nuclear GR (assessed by GR immunostaining in cytoplasm and nucleus and western blots of immunoprecipitated GR protein in cytosolic and nuclear fractions) and was similar for the two antagonists; 4) at nanomolar concentrations, RU40555 and RU486 induced more GR translocation than Dex (assessed by [(3)H]Dex binding and western blot of immunoreactive GR in the same cytosolic homogenates); 5) blocking the steroids membrane transporter with verapamil (100 microM) in the presence of Dex (10 nM) increased GR translocation to levels similar to those induced by RU40555 (10 nM) and RU486 (10 nM) alone; 6) verapamil did not affect GR translocation in the presence of RU40555 or RU486. These data demonstrate similar quantitative effects on GR translocation by RU486 and the new GR antagonist, RU40555. Moreover, RU40555, like RU486, is an effective GR antagonist. Finally, there is no evidence that the intracellular concentrations of RU40555 or RU486 are regulated by the steroids membrane transporter in L929 cells.



2017 ◽  
Vol 63 (1) ◽  
pp. 141-145
Author(s):  
Yuliya Khochenkova ◽  
Eliso Solomko ◽  
Oksana Ryabaya ◽  
Yevgeniya Stepanova ◽  
Dmitriy Khochenkov

The discovery for effective combinations of anticancer drugs for treatment for breast cancer is the actual problem in the experimental chemotherapy. In this paper we conducted a study of antitumor effect of the combination of sunitinib and bortezomib against MDA-MB-231 and SKBR-3 breast cancer cell lines in vitro. We found that bortezomib in non-toxic concentrations can potentiate the antitumor activity of sunitinib. MDA-MB-231 cell line has showed great sensitivity to the combination of bortezomib and sunitinib in vitro. Bortezomib and sunitinib caused reduced expression of receptor tyrosine kinases VEGFR1, VEGFR2, PDGFRa, PDGFRß and c-Kit on HER2- and HER2+ breast cancer cell lines



Sign in / Sign up

Export Citation Format

Share Document