Cytotoxic Effect of Alcohol on Liver Cells and Fibroblasts in Vitro

1974 ◽  
Vol 19 (3) ◽  
pp. 125-127 ◽  
Author(s):  
F. Walker ◽  
W. Elmslie ◽  
R. A. Fraser ◽  
P. E. Snape ◽  
G. C. M. Watt

Cultures of liver cells (Chang) and of fibroblasts (3T6) were exposed to media containing a range of concentrations of ethanol. At the end of a standard period of time the number of viable cells was determined. As the concentration of ethanol increased over 0.25 per cent v/v liver cell survival decreased progressively. Fibroblast survival decreased progressively at ethanol concentrations over 0.50 per cent v/v. This indicates that ethanol is more toxic to liver cells than to fibroblasts. Certain alcoholic beverages, particularly brandy, were found to be more toxic than pure ethanol.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 47-47
Author(s):  
Jessie-F Fecteau ◽  
Ila Bharati ◽  
Morgan O'Hayre ◽  
Tracy Handel ◽  
Thomas J. Kipps ◽  
...  

Abstract Abstract 47 Chronic Lymphocytic Leukemia (CLL) is characterized by an accumulation of mature monoclonal B cells in the blood, secondary lymphoid tissue, and marrow. Despite their accumulation in vivo, CLL cells undergo spontaneous apoptosis in vitro unless rescued by extrinsic factors derived from the leukemia-cell microenvironment. Monocyte-derived Nurse-Like Cells (NLCs) and Marrow Stromal Cells (MSCs), representing the leukemic microenvironment, have been show to sustain CLL cell survival and more importantly to protect CLL cells from drug-induced apoptosis in vitro and possibly in vivo. Such protective niches are thought to prevent current therapies from achieving complete remission in patients. Investigating the mechanism(s) by which cells from the microenvironment promote CLL cell survival, particularly the signaling pathways triggered, will allow for the identification of new therapeutic targets aiming to disrupt these protective interactions. NLCs and MSCs have been shown to produce the chemokine SDF-1 (CXCL12), which can enhance CLL cell survival. We recently found that ZAP-70+ aggressive CLL cells responded by an increased survival to this chemokine, compared to ZAP-70- indolent CLL cells, and that this response was accompanied by the activation of the ERK pathway. Attempting to abrogate this survival pathway, we found that sorafenib (BAY 43–9006, Nexavar) a multi-kinase inhibitor targeting among others Raf kinases and thereby the RAF/MEK/ERK pathway, strongly reduced CLL cell viability in a time and dose dependent manner. A regimen of one single dose of 10uM of sorafenib significantly reduced CLL cell viability to 18+/−10% cells after 48hrs compared to vehicle control (DMSO; 100%; n=5). The daily addition of 1uM sorafenib also significantly decreased CLL cell viability, leading to 31+/−21% and 11+/−5% viable cells after 6 and 7 days respectively, compared to DMSO (n=5). More importantly, our results show that sorafenib induces CLL cell death in the presence of NLCs and MSCs. A single dose of sorafenib (10uM) rapidly decreased the fraction of viable CLL cells overtime, passing from 40+/−16% after 1 day to 10+/−3% after 4 days (n=4) in the context of NLCs and to 25+/−3% after 2 days and 14+/−3% after 4 days in the presence of MSCs, when compared to vehicle control (>80%; n=4). In the presence of NLCs, the 1uM daily regimen also uncovered an increased sensitivity of ZAP-70+ CLL cells to this drug, reducing in 6 days their viability to 13+/−2% (n=4), which approximately half the fraction of viable cells remaining in the ZAP-70- group (40+/−16%; n=7). We next studied sorafenib-mediated cytotoxicity by investigating its impact on the expression of pro-survival molecules. We found that Mcl-1, Bcl-2 and Bcl-xL protein expression was reduced in CLL cells compared to vehicle control, when stimulated with CXCL12 (n=3). In the presence of NLCs and MSCs, only Mcl-1 expression was downregulated, which was also associated with a reduction of the active form of the transcription factor CREB, involved in Mcl-1 expression. Because Mcl-1 expression can be regulated by ERK and AKT pathways, we next investigated if they were abrogated by sorafenib. We indeed found that MEK, ERK, and AKT activation were reduced by this inhibitor compared to vehicle control (n=3). We therefore propose that the cytotoxic effect of sorafenib on CLL cells is due to its interference with at least these two major survival pathways. Since sorafenib caused apoptosis of CLL cells in context of the microenvironment, we reasoned that it might also cause apoptosis of chemotherapy resistant CLL cells. To test this hypothesis, we studied cells from fludarabine-refractory patients. In the presence of NLCs, a single dose of 10uM sorafenib induced a significant reduction in CLL cell viability after 2 days: only 4+/−2% viable cells remained compared to 78+/−12% for the vehicle control (n=4). A comparable observation was made in the presence of MSCs: sorafenib potently induced apoptosis, leaving 12+/−3% live cells after 2 days, compared to vehicle control (71+/−16%; n=4). These results are very promising as they suggest that sorafenib could be an effective novel therapeutic for CLL, affecting the viability of the leukemic cells even in protective niches. Since sorafenib has been approved by the FDA in 2007 for the treatment of advanced hepatocellular carcinoma, a pilot study is currently being planned at UCSD to evaluate the potential of this drug in CLL in vivo. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3330-3330 ◽  
Author(s):  
Kyle Crassini ◽  
William S Stevenson ◽  
Stephen P. Mulligan ◽  
Oliver Giles Best

Abstract BACKGROUND Chronic Lymphocytic Leukemia (CLL) is characterised by the clonal expansion of apoptosis resistant B-lymphocytes. However, in vitro and in the absence of pro-survival factors primary CLL cells undergo spontaneous apoptosis. B-cell receptor (BCR) signalling plays a major role in the survival and proliferation of CLL cells, which is highlighted by the clinical efficacy of the Btk and PI3-kinase inhibitors, ibrutinib and idelalisib. Mitogen activated protein kinase (MAPK) is an important mediator of signals downstream of both Btk and PI3-kinase but few studies have shown that inhibitors of MEK1/2, a critical component in the MAPK pathway, have any potential benefit for therapy of CLL. METHODS We sought to investigate the potential of the MEK1/2 inhibitor MEK162 against CLL cells in vitro. To mimic the tonic BCR stimulation experienced in vivo, primary CLL cells were stimulated using an immobilised antibody to IgM or were treated with PMA, a less specific B-cell activator which promotes protein kinase C-dependent MAPK-ERK1/2 signaling. Sensitivity to MEK162 and effects on MAPK-ERK1/2 pathway activity were assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and western blot analyses respectively. RESULTS MEK162 treatment of CLL cells cultured in media alone resulted in a modest but significant (P < 0.01) reduction in cell viability; 20µM MEK162 reduced the proportion of viable cells remaining after 48 h to 77.40 +/- 7.81 % relative to vehicle-treated controls. In contrast, BCR stimulation through IgM ligation promoted cell survival 3.2 +/- 1.01 fold and sensitised CLL cells to MEK162; 20µM MEK162 reduced the proportion of viable cells by 56.28 +/- 2.37 % (P < 0.001 relative to control). A similar effect was observed in response to PMA stimulation; cell viability increased 1.78 +/- 0.15 fold and was reduced by 59.55 +/- 10.33 % (P < 0.001 relative to control) following treatment with 20µM MEK162 (Figure 1). At concentrations > 0.05 µM MEK162 was significantly (P < 0.05) more effective against CLL cells stimulated with either anti-IgM or PMA than against cells cultured in media alone. By western blotting we observed low levels of MAPK-ERK1/2 activity in cells cultured in media alone, which we suggest may contribute to the spontaneous apoptosis of these cells and the low degree of sensitivity to MEK162 under these conditions. We confirmed that stimulation with either IgM or PMA results in activation of MAPK-ERK1/2 and show that this response can be effectively blocked using MEK162. The effects of anti-IgM and PMA on cell survival and response to MEK162 were independent of ZAP-70 expression or ATM/TP53 functional status. CONCLUSIONS Our data illustrate the important role of MAPK-ERK1/2 activity in BCR-mediated CLL cell survival and suggest that MEK162 may have potential for CLL therapy. These data highlight the importance of employing appropriate culture conditions in order to make accurate assessments concerning the efficacy of novel agents for the treatment of CLL. Figure 1. Stimulation with IgM or PMA sensitises B-CLL cells to MEK1/2 inhibition by MEK162. Figure 1. Stimulation with IgM or PMA sensitises B-CLL cells to MEK1/2 inhibition by MEK162. Disclosures Mulligan: Roche: Consultancy, Honoraria, Research Funding, Speakers Bureau; Sanofi Aventis: Research Funding; Janssen: Consultancy, Honoraria, Speakers Bureau; Celgene: Consultancy, Honoraria.


2019 ◽  
Vol 2019 ◽  
pp. 1-13 ◽  
Author(s):  
Catherine A. Lombard ◽  
Gwenaëlle Sana ◽  
Joël LeMaoult ◽  
Mehdi Najar ◽  
Joachim Ravau ◽  
...  

One of the main challenges in liver cell therapy (LCT) is the induction of a tolerogenic microenvironment to promote graft acceptance in the recipient. Little is known about the immunomodulatory potential of the hepatic cells used in liver cell therapy. In this work, we wanted to evaluate the immunosuppressive properties of human hepatocytes and adult-derived human liver stem/progenitor cells (ADHLSCs), as well as the potential involvement of the immunomodulatory molecule HLA-G. We demonstrated that both cell types were capable of inhibiting the proliferative response of PBMCs to an allogenic stimulus and that the immune inhibitory potential of ADHLSCs, although lower than that of hepatocytes, increased after hepatogenic differentiation. We demonstrated that liver cells express HLA-G and that the immune inhibition pattern was clearly associated to its expression. Interestingly, HLA-G expression increased after the third step of differentiation, wherein oncostatin M (OSM) was added. A 48 hr treatment with OSM was sufficient to induce HLA-G expression in ADHLSCs and result in immune inhibition. Surprisingly, blocking HLA-G partially reversed the immune inhibition mediated by hepatocytes and differentiated ADHLSCs, but not that of undifferentiated ADHLSCs, suggesting that additional immune inhibitory mechanisms may be used by these cells. In conclusion, we demonstrated that both hepatocytes and ADHLSCs present immunomodulatory properties mediated, at least in part, through HLA-G, which can be upregulated following hepatogenic differentiation or liver cell pretreatment with OSM. These observations open up new perspectives for the induction of tolerance following LCT and for potential therapeutic applications of these liver cells.


2015 ◽  
Vol 43 (1) ◽  
pp. 41-46 ◽  
Author(s):  
Milan S. STANKOVIĆ ◽  
Tatjana Lj. MITROVIĆ ◽  
Ivana Z. MATIĆ ◽  
Marina D. TOPUZOVIĆ ◽  
Slaviša M. STAMENKOVIĆ

The cytotoxicity of seven Teucrium species, a long time ago used as a food spices, for beverages and teas preparing, as well as therapeutics for digestive and respiratory diseases, were examined against human cervix adenocarcinoma HeLa, human melanoma Fem-x, human chronic myelogenous leukemia K562 and human breast adenocarcinoma MDA-MB-361 cells. MTT assay was used for determination of target cell survival. The most prominent cytotoxic effect was observed against K562 cells, especially by T. scordioides, T. montanum and T. botrys. All Teucrium extracts showed good cytotoxic activity on HeLa cells, but very low cytotoxic effect on MDA-MB-361 cells. In addition, the cytotoxic activities of T. scordioides and T. montanum extract were tested on healthy resting and phytohaemagglutinin-stimulated peripheral blood mononuclear cells (PHA-stimulated PBMC). T. scordioides and T. montanum extracts at concentration of 200 µg/ml reduced the resting PBMC and PHA-stimulated PBMC survival up to 10% and 20%, while the reduction of K562 cell survival at the same concentration of extracts was 94% and 97%, respectively. These results point to selectivity in their antitumor actions. Teucrium species can be regarded as promising candidates for natural plant sources of effective biological compounds as a supplements in the food industry, as well as for therapeutic use.


2012 ◽  
Vol 92 (1) ◽  
pp. 126-130 ◽  
Author(s):  
Joan Oliva ◽  
Jin Zhong ◽  
Virgil S. Buslon ◽  
Samuel W. French
Keyword(s):  

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 670-670 ◽  
Author(s):  
Reina Improgo ◽  
Grace Tiao ◽  
Adam Kiezun ◽  
Yaoyu Wang ◽  
Lillian Werner ◽  
...  

Abstract Constitutive activation of the NF-κB pathway is a critical feature of hematologic malignancies and is associated with increased lymphocyte proliferation and survival. Various mechanisms leading to altered NF-κB activation have been described in lymphomas, including activating mutations in and upregulation of NF-κB pathway genes. We have previously reported whole-exome sequencing results from a cohort of 160 CLL patients showing mutations in several NF-κB pathway genes (as defined in the Ingenuity Pathway Analysis database), including MYD88 (n=12), RIPK1 (n=4), NRAS (n=4), KRAS (n=3), CARD11 (n=1), IRAK4 (n=1), PIK3CA (n=1) and TRAF3 (n=1). Here, we describe functional approaches to evaluate the biological significance of NF-κB pathway mutations. Using the CellTiter-Glo assay in serum-free conditions, we assessed the relationship between NF-κB pathway mutation status and survival of CLL cells in vitro. We found that CLL cells with no NF-κB mutations exhibited spontaneous apoptosis in vitro, with 3.3 - 39.5% viable cells remaining after 48 hours in culture (n=4 patient samples; n=5 replicates per patient). Similarly, cells harboring the MYD88 L265P mutation yielded 0.6 - 23.1% viable cells after 48 hours (n=5 patient samples; n=5 replicates per patient). In contrast, cells harboring RIPK1 Q375*, RIPK1 K559R, KRAS Q61H, CARD11 E756K, IRAK4 K400E, and PIK3CA I143V displayed apoptotic resistance, with 48.6 - 132.5% viable cells remaining after 48 hours (1 patient sample per mutation; n=5 replicates per patient). Similar findings were observed in the context of B-cell receptor activation via IgM stimulation, with IgM stimulation generally enhancing CLL cell viability (mean = 24% increase after 48 hours). In the case of RIPK1 Q375*, however, no further increase in viability was observed, suggesting that this truncating mutation may obviate the need for external survival signals. To identify NF-κB mutations that might associate with susceptibility to different therapies, we examined the relationship between NF-κB pathway mutation status and sensitivity to the NF-κB inhibitor, SN50. Treatment with SN50 for 48 hours resulted in loss of viability in cells that were negative for NF-κB pathway mutations (43.3 - 98.9% decrease in viability with 5 μM SN50; n=4 patient samples; n=5 replicates per patient). Similarly, cells harboring the MYD88 L265P mutation responded to SN50 treatment (43.1 - 97.0% decrease in viability; n=5 patient samples; n=5 replicates per patient). In contrast, cells harboring RIPK1 Q375*, RIPK1 K559R, CARD11 E756K, and PIK3CA I143V were more resistant to SN50 treatment (18.6% increase - 38.9% decrease in viability; 1 patient sample per mutation; n=5 replicates per patient). These results suggest that specific NF-κB pathway mutations confer resistance to NF-κB inhibition. The above results prompted the question of whether NF-κB pathway mutations may also confer resistance to the BTK inhibitor, ibrutinib, which has previously been shown to block NF-κB pathway activation by inhibiting the phosphorylation of p65 and preventing its nuclear translocation. To address this question, we treated cells with 2.5 μM ibrutinib for 48 hours. Ibrutinib treatment led to a loss in viability in cells that had no NF-κB pathway mutations (26.3 - 82.9% decrease in viability; n=4 patient samples; n=5 replicates per patient). Cells harboring MYD88 L265P mutations also appeared to be susceptible to ibrutinib (25.8 - 82.9% decrease in viability; n=5 patient samples; n=5 replicates per patient). Notably, cells harboring the RIPK1 Q375* and KRAS Q61H mutations appeared to be more resistant to treatment (7.0% decrease and 2.0% increase in viability, respectively; 1 patient sample per mutation; n=5 replicates per patient). Furthermore, RIPK1 Q375* and KRAS Q61H cells remained resistant to ibrutinib in the context of BCR activation. Though our study utilized a limited number of patient samples representing a variety of mutations, the results are suggestive that specific NF-κB pathway mutations are functional and may influence intrinsic CLL cell survival, responsiveness to IgM stimulation, and sensitivity to drug treatment. Identification of specific mutations that confer resistance to ibrutinib is of particular clinical interest for predicting response and understanding drug resistance. Disclosures: Brown: Novartis: Consultancy; Vertex: Consultancy; Sanofi Aventis: Consultancy; Onyx: Consultancy; Emergent: Consultancy; Celgene: Consultancy, Research Funding; Genentech: Consultancy; Pharmacyclics: Consultancy; Genzyme: Research Funding.


2007 ◽  
Vol 18 (3) ◽  
pp. 179-184 ◽  
Author(s):  
Esther Rieko Takamori ◽  
Eduardo Aleixo Figueira ◽  
Rumio Taga ◽  
Mari Cleide Sogayar ◽  
José Mauro Granjeiro

Treatment of bovine bone with peroxides and chaotropic agents aims to obtain an acellular bone matrix that is able to maintain the collagen-apatite complex and a higher mechanical resistance, a mixed biomaterial hereby named mixed bovine bone (MBB). The purpose of this study was to evaluate the cytocompatibility of MBB and cell-MBB interaction. Cell morphology, number of viable cells, ability to reduce methyltetrazolium and to incorporate neutral red upon exposure to different concentrations of the hydrosoluble extract of MBB were assessed in Balb-c 3T3 cells according to ISO 10993-5 standard. The interaction between cells and MBB surface was evaluated by scanning electron microscopy. The water-soluble MBB extracts were cytotoxic and led to cell death possibly due to its effect on mitochondrial function and membrane permeability. Cells plated directly onto the MBB did not survive, although after dialysis and material conditioning in DMEM + 10% FCS, the cells adhered and proliferated onto the material. It may be concluded that, in vitro, water-soluble MBB extracts were cytotoxic. Nevertheless, MBB cytotoxic effect was reverted by dialysis resulting in a material that is suitable for cell based-therapy in the bioengineering field.


2014 ◽  
Author(s):  
K. Aprile von Hohenstaufen ◽  
I. Puoti ◽  
M. Meloni ◽  
B. De Servi

BackgroundThe acute myeloid leukemia (AML) cell line OCI-AML3, carrying both NPM1 mutation A and the heterozygous DNMT3A R882C mutation, represents the model for in vitro studies on AML with mutated NPM11. AML with mutated NPM1 harbours a hypo-methylated profile distinct from those of the other AML subtypes2. This characteristic is probably related to the inhibitory effect of the mutant DNMT3A on the wild type protein3. S-adenosylmethionine (SAM) is a universal methyl donor acting as a coenzyme of DNMT3A. There are growing evidences of the antineoplastic effect of SAM in vitro and in murine models of gastric cancer, colon cancer and hepatocellular carcinoma, where SAM induces the downregulation of several oncogenes4-10. Moreover SAM upregulates the expression of DNMT enzymes in lung cancer cells11. In our knowledge there are no published data exploring the effect of SAM on the growth of OCI-AML3 cells and its ability to modulate DNMT3A activity in this cell line.Study design and methodsThe present data have been generated between August 2013 and April 2014 at the VITROSCREEN facilities in Milan–ITALY. We used a 3-(4,5-dimethylthiazol-2-yl)-2,5-dephenyl tetrazolium bromide (MTT) assay to assess the cytotoxic effect of SAM iodide (Sigma-Aldrich) on OCI-AML3 cells (DSMZ Leibniz Institut). We analyzed then the ability of SAM to induce apoptosis by Tali Image-Based Cytometer (green Annexin V – Alexa Fluor 488 for apoptotic cells, red propidium and green Annexin V-Alexa Fluor 488 for necrotic cells).ResultsThe MTT assays were performed after having treated the OCI-AML3 cells with various concentrations of the indicated drug for 24 hours. We observed no significant effects on cells viability using 0.5μM, 10 μM and 100 μM of SAM (data not shown). In contrast, a dose dependent cytotoxic effect of SAM on OCI-AML3 cells was evident for concentrations equal or superior to 500 μM, with an IC50 of 500 μM (Figure 1). Since a Cmax of 211(SD 94)μM after single intravenous infusion of SAM was previously reported in healthy voluntarees12, we decided to investigate the cytotoxic effect of SAM for concentrations close to 211 μM using the MTT test. A significant dose dependent reduction of the cells viability was observed with SAM 200μM (62,74% viable cells) and SAM 300μM (53.32% viable cells), (Figure 2). The Apoptosis assay after 24 hours of treatment with SAM showed no differences in the percentages of apoptotic cells between the OCI-AML3 cells treated with SAM 300-500-2500 μM and the untreated cells (data not shown). After 72 hours, only a minimal effect on the amount of apoptotic cells was obtained, while a clear dose dependent increase in the proportion of dead cells was noted (Figure 3), confirming the results of the aforementioned MTT tests.ConclusionsSAM showed remarkable in vitro cytotoxic activity on OCI-AML3 cells at concentrations similar to those achievable in humans after intravenous administration. SAM is not able to induce apoptosis of OCI-AML3 cells in vitro after 72 hours of treatment. However, the increase in the amount of dead cells after SAM treatment may be due to mechanisms other than apoptosis. In order to verify if the observed cytotoxicity was mediated by the enzymatic activity of DNMT3A, we planned to repeat the cytotoxicity assays after DNMT3A silencing. The in vivo antineoplastic effect of SAM could be assessed in NOD/SCID mice engrafted with OCI-AML3 cells.Authors contributionKAvH wrote the study rationale, designed the study, interpreted the data and wrote the article; IP revised the article, MM and BDS planned and interpreted the experiments and BDS performed the experiments.


Author(s):  
Robert R. Cardell

Hypophysectomy of the rat renders this animal deficient in the hormones of the anterior pituitary gland, thus causing many primary and secondary hormonal effects on basic liver functions. Biochemical studies of these alterations in the rat liver cell are quite extensive; however, relatively few morphological observations on such cells have been recorded. Because the available biochemical information was derived mostly from disrupted and fractionated liver cells, it seemed desirable to examine the problem with the techniques of electron microscopy in order to see what changes are apparent in the intact liver cell after hypophysectomy. Accordingly, liver cells from rats which had been hypophysectomized 5-120 days before sacrifice were studied. Sham-operated rats served as controls and both hypophysectomized and control rats were fasted 15 hours before sacrifice.


2011 ◽  
Vol 49 (01) ◽  
Author(s):  
SA Hoffmann ◽  
M Lübberstedt ◽  
U Müller-Vieira ◽  
D Knobeloch ◽  
A Nüssler ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document