scholarly journals Retama monosperma n-hexane extract induces cell cycle arrest and extrinsic pathway-dependent apoptosis in Jurkat cells

Author(s):  
Lamiae Belayachi ◽  
Clara Aceves-Luquero ◽  
Nawel Merghoub ◽  
Youssef Bakri ◽  
Silvia Fernández de Mattos ◽  
...  
Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4290-4290
Author(s):  
Ina Fabian ◽  
Debby Haite ◽  
Avital Levitov ◽  
Drora Halperin ◽  
Itamar Shalit

Abstract We previously reported that the fluoroquinolone moxifloxacin (MXF) inhibits NF-kB, mitogen-activated protein kinase activation and the synthesis of proinflammatory cytokines in activated human monocytic cells (AAC48:1974,2004). Since MXF acts on topoisomerase II (Topo II) in mammalian cells, we investigated its effect in combination with another Topo II inhibitor, VP-16, on cell proliferation (by the MTT method), cell cycle, caspase-3 activity and proinflammatory cytokine release in THP-1 and Jurkat cells. THP-1 cells were incubated for 24 h with 0.5–3 μg/ml VP-16 in the presence or absence of 5–20 μg/ml MXF. VP-16 induced a dose dependent decrease in cell proliferation. An additional 2.5-and 1.6-fold decrease in cell proliferation was observed upon incubation of the cells with 0.5 or 1 μg/ml VP-16 and 20 μg/ml MXF, respectively (up to 69% inhibition). To further elucidate the mechanism of the antiproliferative activity of MXF, its effect on cell cycle progression was investigated. In control cultures 1%, 45%,18% and 36% of cells were in G0, G1, S and G2/M phases at 24 h, respectively. In contrast, in cultures treated with 1 μg/ml VP-16 and VP-16+ 20 μg/ml MXF, the number of cells in G1 decreased to 5.4 and 6.5%, respectively, while the number of cells in S phase increased to 25.5 and 42%, respectively and the number of cells in G2/M cells increased to 60 and 44%, respectively. These data provide evidence for S-G2/M cell cycle arrest induced by VP-16 and that addition of MXF shifted the S-G2/M arrest more towards the S phase. Since the antiproliferative effects of MXF could also be attributed to apoptotic cell death in addition to cell cycle arrest, we investigated the effect of the drugs on apoptosis. Using the fluorogenic assay for caspse-3 activity, we show that incubation of THP-1 cells for 6 h with 1.5 μg/ml VP-16 resulted in 630±120 unit/50μg protein of caspase-3 activity while the combination of 1.5 μg/ml VP-16 and 20 μg/ml MXF enhanced caspase-3 activity up to 1700±340 units/50μg protein (vs.233±107 in control cells), indicating that MXF synergises with VP-16 in activation of caspase-3. In Jurkat cells, the addition of 0.5 or 1 μg/ml VP-16, did not affect cell proliferation while in the presence of 20 μg/ml MXF and 1 μg/ml VP-16 there was a 62% decrease in cell proliferation (p<0.05). Exposure of Jurkat cells to 3 μg/ml VP-16 alone resulted in 504±114 units/50μg protein of caspase-3 activity and the addition of 20μg/ml MXF enhanced caspase-3 activity up to 1676± 259 units/50μg protein (vs 226±113 units/50μg protein in control cells). We further examined pro-inflammatory cytokine secretion upon stimulation of THP-1 cells with VP-16, MXF or their combination. VP-16 alone at 3 μg/ml increased IL-8 and TNF-α secretion from THP-1 cells by 2.5 and 1.8-fold respectively. Addition of MXF (5–20 μg/ml) inhibited the two cytokines secretion by 72–77% and 58–72%, respectively. The above combined data indicate that MXF, at clinically attainable concentrations, demonstrates pronounced synergistic effect with VP-16 as an anti-proliferative agent mainly by enhancing caspase-3 activity and apoptosis. At the same time MXF inhibits the pro-inflammatory effects conferred by VP-16 in the tumor cells studied. The clinical significance of the above anti-proliferative and anti-inflammatory effects of MXF in combination with VP-16 should be further investigated in animal models.


2019 ◽  
Vol 69 (1) ◽  
pp. 75-86 ◽  
Author(s):  
Affidah Sabran ◽  
Endang Kumolosasi ◽  
Ibrahim Jantan

Abstract Recent studies suggest that annexin A1 (ANXA1) promotes apoptosis in cancerous cells. This study aims to investigate the effects of ANXA1 on apoptosis and cell cycle arrest in K562, Jurkat and U937 cells and peripheral blood mononu-clear cells (PBMC). Cells were treated with ANXA1 and cyclophosphamide prior to flow cytometry analysis for apoptosis and cell cycle arrest induction. At 2.5µM, ANXA1 induced significant apoptosis in K562 (p ≤ 0.001) and U937 (p ≤ 0.05) cells, with EC50 values of 3.6 and 3.8 µM, respectively. In Jurkat cells, induction was not significant (EC50, 17.0 µM). No significant apoptosis induction was observed in PBMC. ANXA1 caused cycle arrest in the G0/G1 phase in K562 and U937 cells with p ≤ 0.001 for both, and (p ≤ 0.01) for Jurkat cells. ANXA1 induced apoptosis and cycle arrest in the G0/G1 phase in K562 and U937 cells, causing only cell cycle arrest in Jurkat cells.


2015 ◽  
Vol 83 (10) ◽  
pp. 4042-4055 ◽  
Author(s):  
Kathleen Boesze-Battaglia ◽  
Lisa P. Walker ◽  
Ali Zekavat ◽  
Mensur Dlakić ◽  
Monika Damek Scuron ◽  
...  

Induction of cell cycle arrest in lymphocytes following exposure to theAggregatibacter actinomycetemcomitanscytolethal distending toxin (Cdt) is dependent upon the integrity of lipid membrane microdomains. Moreover, we have previously demonstrated that the association of Cdt with target cells involves the CdtC subunit which binds to cholesterol via a cholesterol recognition amino acid consensus sequence (CRAC site). In this study, we demonstrate that the active Cdt subunit, CdtB, also is capable of binding to large unilamellar vesicles (LUVs) containing cholesterol. Furthermore, CdtB binding to cholesterol involves a similar CRAC site as that demonstrated for CdtC. Mutation of the CRAC site reduces binding to model membranes as well as toxin binding and CdtB internalization in both Jurkat cells and human macrophages. A concomitant reduction in Cdt-induced toxicity was also noted, indicated by reduced cell cycle arrest and apoptosis in Jurkat cells and a reduction in the proinflammatory response in macrophages (interleukin 1β [IL-1β] and tumor necrosis factor alpha [TNF-α] release). Collectively, these observations indicate that membrane cholesterol serves as an essential ligand for both CdtC and CdtB and, further, that this binding is necessary for both internalization of CdtB and subsequent molecular events leading to intoxication of cells.


2010 ◽  
Vol 152 (3) ◽  
pp. 352-356 ◽  
Author(s):  
Esperanza Martín-Sánchez ◽  
Margarita Sánchez-Beato ◽  
María Elena Rodríguez ◽  
Beatriz Sánchez-Espiridión ◽  
Cristina Gómez-Abad ◽  
...  

Cancers ◽  
2020 ◽  
Vol 12 (12) ◽  
pp. 3845
Author(s):  
Shin Young Park ◽  
Ki Yun Kim ◽  
Do Youn Jun ◽  
Su-Kyeong Hwang ◽  
Young Ho Kim

In vitro antitumor activity of the CDK7 inhibitor BS-181 against human T-ALL Jurkat cells was determined. Treatment of Jurkat clones (JT/Neo) with BS-181 caused cytotoxicity and several apoptotic events, including TRAIL/DR4/DR5 upregulation, c-FLIP down-regulation, BID cleavage, BAK activation, ΔΨm loss, caspase-8/9/3 activation, and PARP cleavage. However, the BCL-2-overexpressing Jurkat clone (JT/BCL-2) abrogated these apoptotic responses. CDK7 catalyzed the activating phosphorylation of CDK1 (Thr161) and CDK2 (Thr160), and CDK-directed retinoblastoma phosphorylation was attenuated in both BS-181-treated Jurkat clones, whereas only JT/BCL-2 cells exhibited G1 cell cycle arrest. The G1-blocker hydroxyurea augmented BS-181-induced apoptosis by enhancing TRAIL/DR4/DR5 upregulation and c-FLIP down-regulation. BS-181-induced FITC–annexin V-positive apoptotic cells were mostly in the sub-G1 and G1 phases. BS-181-induced cytotoxicity and mitochondrial apoptotic events (BAK activation/ΔΨm loss/caspase-9 activation) in Jurkat clones I2.1 (FADD-deficient) and I9.2 (caspase-8-deficient) were significantly lower than in A3 (wild-type). Exogenously added recombinant TRAIL (rTRAIL) markedly synergized BS-181-induced apoptosis in A3 cells but not in normal peripheral T cells. The cotreatment cytotoxicity was significantly reduced by the DR5-blocking antibody but not by the DR4-blocking antibody. These results demonstrated that the BS-181 anti-leukemic activity is attributed to extrinsic TRAIL/DR5-dependent apoptosis preferentially induced in G1-arrested cells, and that BS-181 and rTRAIL in combination may hold promise for T-ALL treatment.


Pharmacology ◽  
2012 ◽  
Vol 89 (5-6) ◽  
pp. 348-356 ◽  
Author(s):  
Jun-Shan Zhu ◽  
Dong-Yun Ouyang ◽  
Zhi-Jian Shi ◽  
Li-Hui Xu ◽  
Yan-Ting Zhang ◽  
...  

2019 ◽  
Vol 7 (4) ◽  
pp. 12-16
Author(s):  
M L Fauzan ◽  
P A Z Hasibuan ◽  
U Harahap

Objective: This study aims to determined phytochemical compounds of simplex and n-hexane extract (nHE) of Vernonia amygdalina Delile Leaves and cell cycle arrest activity against PANC-1 cells. Methods: The leaves of Vernonia amygdalina Delile were dried and extracted with n-hexane, followed by evaporation and freeze-drying. Phytochemicals screening were analyzed with standard procedures. Cytotoxic activity was carried out using MTT assay method. PANC-1 cells were treated with different concentrations of nHE (500 ug/mL, 250 ug/mL, 125 ug/mL, 61.5 ug/mL and 31.25 ug/mL) for 24 hours to obtained IC50 values. The cell cycle inhibition activity of nHE was carried out used flowcytometry method and apoptosis activity used double staining method. Results: Then HE was identified contains steroids/triterpenoids. The IC50 was114.80 ± 1.21 ug/mL. The nHE inhibited cell cycle PANC-1 on M1 phase (67.39%) and it was induced apoptosis process on PANC-1 cells. Conclusions: Our results suggest that extractsn­-hexane of Vernonia amygdalina Delile Leaves had as cancer chemoprevention activities with inhibits cell cycle and spur apoptosis process on PANC-1 cells.   Keywords: 


Sign in / Sign up

Export Citation Format

Share Document