scholarly journals Effects of short-term infusion of lipid emulsions on pro-inflammatory cytokines and lymphocyte apoptosis in septic and non-septic rats

2011 ◽  
Vol 106 (1) ◽  
pp. 27-32 ◽  
Author(s):  
Patrick Scheiermann ◽  
Juliane Ott ◽  
Sandra Hoegl ◽  
Matthias Hecker ◽  
Christian Hofstetter ◽  
...  

Long-term administration of PUFA is known to modulate immune functions and apoptotic pathways depending on the respective amount of n-6 and n-3 fatty acids (FA). Data on short-term effects on apoptotic pathways are rare. Apoptosis of splenic lymphocytes is the hallmark of detrimental sepsis. Therefore, we aimed to compare the immediate effects of parenterally administered n-6-enriched soyabean oil (SO)- and n-3-enriched fish oil (FO)-based lipid emulsions after laparotomy (LAP; sham procedure) and after induction of acute, severe sepsis by caecal ligation and incision. After 390 min of observation time, plasma was analysed for IL-1β, IL-6 and NEFA. Apoptosis in splenic lymphocytes was quantified by Annexin-V expression. After LAP, infusion of both FO and SO did not change cytokine concentrations. Sepsis increased both cytokines. FO but not SO further augmented the rise. After LAP, SO increased NEFA, and both lipid emulsions reduced free arachidonic acid (AA). Sepsis resulted in a dramatic decrease in NEFA and AA. The drop in NEFA and AA was prevented by both SO and FO. In addition, FO resulted in an increased concentration of n-3 FA under both conditions. Infusion of both lipid emulsions induced apoptosis in splenic lymphocytes after LAP. Sepsis-induced apoptosis was not further enhanced by FO or SO. The present study shows that short-term administration of FO as opposed to SO caused pro-inflammatory effects during sepsis. Moreover, short-term administration of both SO and FO suffices to induce apoptosis in splenic lymphocytes. Finally, SO and FO do not further enhance sepsis-induced splenic apoptosis.

1991 ◽  
Vol 150 (4) ◽  
pp. 238-241 ◽  
Author(s):  
M. Maghnie ◽  
A. Valtorta ◽  
A. Moretta ◽  
D. Larizza ◽  
M. A. Girani ◽  
...  

2019 ◽  
Vol 39 (5) ◽  
Author(s):  
Dongxu Zhang ◽  
Houxian Liu ◽  
Binbin Yang ◽  
Jiasheng Hu ◽  
Yue Cheng

Abstract The present study aims to evaluate the anticancer effect of L-securinine on androgen-independent prostate cancer (AIPC) DU145 cells. L-securinine (2.5, 5, and 10 μM) treatment for 24, 48 and 72 h displayed strong growth inhibitory effect on DU145 cells in a concentration and time-dependent fashion but has less toxicity toward normal androgen-dependent LNCaP cells. Hoechst 332582 staining of DU145 cells and Annexin V-FITC/ PI dual-labeling followed by flow cytometry assay identified that this growth inhibition by L-securinine would be due to the induction of apoptosis. Moreover Transwell assay revealed that L-securinine significantly inhibited the cell migration/invasion ability of DU145 cells. Furthermore, results of western blotting showed that the involvement of mitochondrial apoptotic pathway in the L-securinine-induced apoptosis of DU145 cell, as evidenced by an increase in the protein expression of Bax, cleaved caspase-9, cleaved caspase-3, cytosolic cytochrome c, and cleaved PARP, together with a unchanged cleaved caspase-8 and decreased Bcl-2 protein expression. Also, L-securinine-induced antimetastatic activity in DU145 cells was associated with decreased protein expression of MMP-2 and MMP-9 and concurrent reduction of VEGF. In addition, further studies revealed that L-securinine may inhibit the protein expression of AGTR1, p-MEK1/2, p-ERK1/2, p-STAT3, PAX2, and p-PAX2, while the expression of ERK1/2, MEK1/2, and STAT3 protein retains intact. These findings suggest that L-securinine may be a promising chemopreventive agent against AIPC.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1364-1364
Author(s):  
Paolo Lunghi ◽  
Laura Mazzera ◽  
Vittorio Rizzoli ◽  
Antonio Bonati

Abstract We recently reported that PD184352 (PD) (Pfizer), a highly selective inhibitor of MEK1 phosphorylation and activation, sensitizes primary acute myelogenous leukemia (AML) to ATO-induced apoptosis via p73, a p53 paralogue, and Bad pro-apoptotic pathways activation (Blood107: 4549–4554, 2006). We also demonstrated that high doses of ATO (2μM) induced p53 accumulation (more than two-fold increase compared with control) in 11 out of 21 patients (52%), indicating a possible contribution of p53 pathway in apoptosis induction of dual treated blasts. TP53 mutations are quite rare in AML (5–10%) and MDM2 (murine double minute 2), its principal negative regulator, has been found to be frequently overexpressed in AML, a process that can actively enhance tumorigenic potential and resistance to apoptosis. The aim of this study was to investigate whether Nutlin-3, a potent and selective small-molecule MDM2 antagonist, can potentiate the apoptotic effect of the PD plus ATO combination in AML cells that retain a functional p53. Apoptosis was evaluated, after 24 and 48 hours of treatment, by measurement of sub-G1 DNA content, annexin V binding and mitochondrial transmembrane potential assays. We first analyzed the pharmacologic interactions between Nutlin-3, PD and ATO (0, 0.25, 0.5, 1, 2, 5, or 10 μM) using a fixed-ratio (1:1:1) experimental design in OCI-AML-3 cell line that has wild-type p53. We found that the three-drugs combination showed cytotoxic synergism stronger than PD plus ATO combination indicating that the inhibition of the p53-MDM2 interaction can positively influence the pro-apoptotic efficacy of dual-treated (PD plus ATO) cells: the averaged Combination Index (CI) values calculated from the ED50 (50% effective dose), ED75 and ED90, in Nutlin-3 plus PD plus ATO versus PD plus ATO treated cells were 0.36± 0.03 and 0.66± 0.02 respectively (CI values equal to 0.85 or less indicate synergism). In order to investigate the molecular effectors involved in Nutlin-3-PD-ATO or PD-ATO-induced apoptosis we first studied the kinetics (2h, 12h, 24h and 48h) of p53, p73 and phospho-Bad at Ser112 in OCI-AML-3. In the absence of Nutlin-3 ATO, even at high doses, did not promote a p53 accumulation whereas modulated the expression of the p73 gene by inducing the pro-apoptotic and anti-proliferative TAp73 and the anti-apoptotic and pro-proliferative ΔNp73 isoforms, thereby failing to elevate the TA/ΔNp73 ratio. Conversely, treatment with PD reduced the level of ΔNp73 and blunted the ATO-mediated up-regulation of ΔNp73 thus causing an increase in the TA/ΔNp73 ratio of dual-treated cells. In the presence of Nutlin-3, p53 accumulated and the triple combination of Nutlin-3, PD and ATO enhanced the loss of mitochondrial membrane potential occurred in PD plus ATO treatment. Furthermore, pretreatment with MEK1 inhibitor strongly increased the expression of dephosphorylated Bad and blunted the ATO-mediated phosphorylation of Bad at Ser112, thereby activating its pro-apoptotic functions. These findings suggest that the pro-apoptotic p73 and Bad pathways, both involved in PD plus ATO efficacy, can be potentiated by the rescue of p53 pathway in AML cells that possess a functional p53 pathway.


2009 ◽  
Vol 21 (1) ◽  
pp. 222 ◽  
Author(s):  
D. Kalo ◽  
Z. Roth

Programmed cell death through the sphingomyelin pathway has been suggested to underlie the mechanism by which heat shock disturbs oocyte developmental competence. Serial experiments were performed to characterize the role of the sphingolipid ceramide in heat-shock-induced apoptosis, and to determine whether ceramide formation could be regulated. In all experiments, bovine cumulus–oocyte complexes (COC) were aspirated from ovaries collected at the abattoir. Cumulus–oocyte complexes were matured (22 h, 38.5°C, 5% CO2), in vitro fertilized (18 h, 38.5°C, 5% CO2), and cultured for 8 days (KSOM; 38.5°C, 5% CO2, 5% O2). In the first experiment, COC were matured at 38.5°C (n = 673) or 41°C (heat shock; n = 718). Exposure of COC to heat shock during maturation reduced cleavage (92.3 v. 84.2%; P < 0.05) and blastocyst formation rates (24.1 v. 14.6%; P < 0.05) relative to the control groups. In the second experiment, COC (n = 563) were exposed to either long-term (22 h) or short-term (0.5 to 3.5 h) heat shock during maturation. An Annexin V-FITC assay (Bender) was used to identify the turnover of phosphatidylserine, one of the features of early-stage apoptosis. In addition, a subgroup of matured oocytes (n = 384) were denuded and fixed in 2% paraformaldehyde, followed by immunofluorescent staining using anti-ceramide (Alexis) to detect ceramide levels and counterstaining with Hoechst (Sigma). Immunofluorescent staining showed no difference in endogenous ceramide levels between groups. Similarly, annexin expression did not differ between groups at the end of the maturation but was higher (P < 0.05) in oocytes exposed to short-term heat shock than in those subjected to long-term heat shock, suggesting that early features of apoptosis should be examined at a specific time of heat exposure. Another experiment was performed to examine the effect of ceramide on oocyte developmental competence. Cumulus–oocyte complexes (n = 1185) were matured with or without 10, 30, and 50 μm C2-ceramide (Sigma). An Annexin V-FITC assay was used with a subgroup of oocytes (n = 137) that were matured with or without 50 μm C2-ceramide for 2 h. To examine the major pathway of ceramide generation, heat-shocked COC were matured with or without 5 μm fumonisin B1 (FB1; Sigma), a specific inhibitor of dihydroceramide synthase (i.e. de novo formation) and ceramide synthase (i.e. salvage pathway), or with 5 μm desipramine hydrochloride (DH; Sigma), a specific inhibitor of the acid sphingomyelinase (i.e. hydrolysis). Results revealed that maturation in 50 μm C2-ceramide increased (P < 0.05) annexin expression in association with reducing cleavage rate and blastocyst formation (P < 0.05). Although not significant, maturation with FB1 moderated the heat-shock effect on oocyte developmental competence. Similarly, 5 μm DH increased the blastocyst formation rate for heat-shocked oocytes from a level of 17% to the control level (22.5%). In summary, ceramide appears to play an important role in heat-shock-induced apoptosis because blocking ceramide formation alleviated its deleterious effect. Research was supported in part by USDA Grant 2007-35203 and BARD Grant 3986-07


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1885-1885
Author(s):  
Yoshitaka Kikukawa ◽  
Hiroaki Mitsuya ◽  
Hiroyuki Hata

Abstract Abstract 1885 Poster Board I-908 Treatment of MGUS by hesperetin, potentially delaying/blocking the conversion of MGUS to symptomatic myeloma Yoshitaka Kikukawa, Hiroaki Mitsuya, Hiroyuki Hata. Department of Hematology, Kumamoto Unversity Hospital INTRODUCTION: Although prognosis of multiple myeloma has recently been improved by novel therapeutic regimens, all currently available therapuetics target symptomatic myeloma only. We saw an MGUS patient, experiencing a gradual and continuous decrease of M-protein (IgA-k) from 2,080 mg/dl to 878 mg/dl over 3 years (Fig. 1). History examiantion revealed that the decrease of M-protein initiated when the patient started taking a dietary supplement containing hesperetin-7-glucoside, which is a glycosylated form of hesperetin designated to achive an approximately 104-fold increase of bioavailability comparing to aglycon structure (Hayashibara Biochemical Laboratories, Inc.). Because this compound is eventually metabolized to hesperetin, a possible anti-myeloma effect of hesperetin was examined in vitro. METHODS: Myeloma cells were obtained from myeloma patients and purified with CD138 magnetic beads. Anti-myeloma effects of test compounds on purified myeloma cells or myeloma cell lines were evaluated by WST-8 assay. Apoptosis of myeloma cells was quantified either by annexin V staining or the propidium iodide method, followed by flowcytomerty analysis or by morphological analysis of cytospin slides. Mitochondrial membrane potential was quantified using JC-1 staining Kit (Cayman Chemical Co.) and flow cytometry. Caspase activation and total ubiqutinated protein were analysed by western blotting. Proteasomal chymotrypsin-like activity was measured using 20S Proteasome Assay Kit (Enzo Life Sciences). RESULTS: Hesperetin showed inhibitory effects in a dose-dependent manner on the growth of 4 myeloma cell lines and freshly isolated myeloma cells. Two myeloma cell ines, RPMI8226 and 12PE, were utilized as representative cell lines for further analysis. Hesperetin induced annexin V/PI positive cells, morphological fragmentation of the nucleus of myeloma cells, and activation of caspase-3, 8 and 9, at a concentration around 500 microM, which is clinically achievable with peroral administration of glycosylated hesperetin, suggesting that the observed anti-myeloma effects by hesperitine through apoptotic pathways. Further analysis revealed that hesperetin disrupted mitochondrial membrane potential which leads to release of cytochrome c from mitochondria to cytoplasm as assessed by western blot analysis. Caspase-8 and 9 inhibitors(Z-IETD-fmk and Z-LEHD-fmk, respectively)did not inhibit the hesperetine-induced apoptosis, although they completely inhibited anti-Fas antibody-induced apoptosis, suggesting that the hesperitine-induced apoptosis is not dependent on death receptor signaling. A pan-caspase inhibitor, Z-VAD-fmk, completely blocked the hesperetin-induced apoptosis in 12PE cells, but only partially in RPMI8226 cells, suggesting that hesperetin also mediated caspase-independent apoptosis in RPMI8226 cells. Moreover, western blot showed that hesperetin treatment induced an accumlation of poly-ubiquitine proteins. Analysis of proteasome activity revealed that hesperetin at 500 microM exerted a moderate inhibition of proteasome activity with 72.6% of the inhibition exerted by bortezomib at 40 nM. CONCLUTIONS: This is the first report showing anti-myeloma effect of hesperetin. It showed anti-myeloma effects via caspase-dependent and independent apoptotic pathways and proteasome inhibition activity. Given a fact that hesperitin has been approved by Japanese Goverment as a safe supplementary compound, and the present MGUS case had been taking hesperitin over >3 years without any adverse events, this compound should be well torelated over a long period. If a delay of conversion from either MGUS or asymptomatic myeloma to symptomatic myeloma could be achieved, prognosis of MM could be improve. Based on these findings, an open-label, pilot clinical trial to test the efficacy of hespertin, enroling asymptomatic myeloma patients, has currently been underway (Approved by IRB of Kumamoto Univerisity Hospital). Disclosures: No relevant conflicts of interest to declare.


2001 ◽  
Vol 120 (5) ◽  
pp. A314-A314
Author(s):  
K HADERSLEV ◽  
P JEPPESEN ◽  
B HARTMANN ◽  
J THULESEN ◽  
J GRAFF ◽  
...  

2019 ◽  
Vol 18 (10) ◽  
pp. 1448-1456 ◽  
Author(s):  
Bahareh Movafegh ◽  
Razieh Jalal ◽  
Zobeideh Mohammadi ◽  
Seyyede A. Aldaghi

Objective: Cell resistance to doxorubicin and its toxicity to healthy tissue reduce its efficiency. The use of cell-penetrating peptides as drug delivery system along with doxorubicin is a strategy to reduce its side effects. In this study, the influence of poly-L-arginine on doxorubicin cytotoxicity, its cellular uptake and doxorubicin-induced apoptosis on human prostate cancer DU145 cells are assessed. Methods: The cytotoxicity of doxorubicin and poly-L-arginine, alone and in combination, in DU145 cells was evaluated at different exposure times using MTT assay. The influence of poly-L-arginine on doxorubicin delivery into cells was evaluated by fluorescence microscopy and ultraviolet spectroscopy. DAPI and ethidium bromide- acridine orange stainings, flow cytometry using annexin V/propidium iodide, western blot analysis with anti-p21 antibody and caspase-3 activity were used to examine the influence of poly-L-arginine on doxorubicininduced cell death. Results: Poly-L-arginine had no cytotoxicity at low concentrations and short exposure times. Poly-L-arginine increased the cytotoxic effect of doxorubicin in DU145 cells in a time-dependent manner. But no significant reduction was found in HFF cell viability. Poly-L-arginine seems to facilitate doxorubicin uptake and increase its intracellular concentration. 24h combined treatment of cells with doxorubicin (0.5 µM) and poly-L-arginine (1 µg ml-1) caused a small increase in doxorubicin-induced apoptosis and significantly elevated necrosis in DU145 cells as compared to each agent alone. Conclusion: Our results indicate that poly-L-arginine at lowest and highest concentrations act as proliferationinducing and antiproliferative agents, respectively. Between these concentrations, poly-L-arginine increases the cellular uptake of doxorubicin and its cytotoxicity through induction of necrosis.


2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Saba Sameri ◽  
Chiman Mohammadi ◽  
Mehrnaz Mehrabani ◽  
Rezvan Najafi

Abstract Background Silibinin, as a chemopreventive agent, has shown anti-cancer efficacy against different types of cancers. In the present study, we investigated the anti-cancer activities of silibinin on CT26 mouse colon cell line. Methods CT26 cells were treated with different concentrations of silibinin. To examine the cytotoxic effect of silibinin on proliferation, apoptosis, autophagy, angiogenesis, and migration, MTT, colony-forming assay, Annexin V/PI flow cytometry, RT-qPCR, and Scratch assay were used. Results Silibinin was found to significantly reduce CT26 cells survival. Furthermore, silibinin strongly induced apoptosis and autophagy by up-regulating the expression of Bax, Caspase-3, Atg5, Atg7 and BECN1 and down-regulating Bcl-2. Silibinin considerably down-regulated the expression of COX-2, HIF-1α, VEGF, Ang-2, and Ang-4 as well as the expression of MMP-2, MMP-9, CCR-2 and CXCR-4. Conclusions The present study revealed that silibinin shows anticancer activities by targeting proliferation, cell survival, angiogenesis, and migration of CT26 cells.


Author(s):  
Amber M. Tavener ◽  
Megan C. Phelps ◽  
Richard L. Daniels

AbstractGlioblastoma (GBM) is a lethal astrocyte-derived tumor that is currently treated with a multi-modal approach of surgical resection, radiotherapy, and temozolomide-based chemotherapy. Alternatives to current therapies are urgently needed as its prognosis remains poor. Anthracyclines are a class of compounds that show great potential as GBM chemotherapeutic agents and are widely used to treat solid tumors outside the central nervous system. Here we investigate the cytotoxic effects of doxorubicin and other anthracyclines on GL261 glioma tumor cells in anticipation of novel anthracycline-based CNS therapies. Three methods were used to quantify dose-dependent effects of anthracyclines on adherent GL261 tumor cells, a murine cell-based model of GBM. MTT assays quantified anthracycline effects on cell viability, comet assays examined doxorubicin genotoxicity, and flow cytometry with Annexin V/PI staining characterized doxorubicin-induced apoptosis and necrosis. Dose-dependent reductions in GL261 cell viability were found in cells treated with doxorubicin (EC50 = 4.9 μM), epirubicin (EC50 = 5.9 μM), and idarubicin (EC50 = 4.4 μM). Comet assays showed DNA damage following doxorubicin treatments, peaking at concentrations of 1.0 μM and declining after 25 μM. Lastly, flow cytometric analysis of doxorubicin-treated cells showed dose-dependent induction of apoptosis (EC50 = 5.2 μM). Together, these results characterized the cytotoxic effects of anthracyclines on GL261 glioma cells. We found dose-dependent apoptotic induction; however at high concentrations we find that cell death is likely necrotic. Our results support the continued exploration of anthracyclines as compounds with significant potential for improved GBM treatments.


Sign in / Sign up

Export Citation Format

Share Document