Satureja hortensis induces cell death and inhibited cell cycle progression in K562 myelogenous and Jurkat T cell leukemia cell lines

2019 ◽  
Vol 40 (5) ◽  
pp. 459-472
Author(s):  
Morvarid Asadipour ◽  
Zahra Amirghofran
2016 ◽  
Vol 65 (1) ◽  
pp. 82-87 ◽  
Author(s):  
Haihao Wang ◽  
Qiannan Guo ◽  
Peiwen Yang ◽  
Guoxian Long

Adult T-cell leukemia/lymphoma (ATL) is a highly aggressive T-cell malignancy. This study was designed to explore the expression and functional significance of microRNA (miR)-212 in ATL. The expression of miR-212 in human ATL tissues and cell lines were investigated. Gain-of-function experiments were carried out to determine the roles of miR-212 in cell proliferation, tumorigenesis, cell cycle progression, and apoptosis. We also identified and functionally characterized the target genes of miR-212 in ATL cells. Compared with normal lymph node biopsies, lymphoma samples from ATL patients displayed underexpression of miR-212 (p=0.0032). Consistently, miR-212 was downregulated in human ATL cell lines, compared with normal T lymphocytes. Restoration of miR-212 significantly (p<0.05) inhibited ATL cell proliferation and tumorigenesis in mice. Overexpression of miR-212 led to an accumulation of G0/G1-phase cells and a concomitant reduction of S-phase cells. Moreover, enforced expression of miR-212-induced significant apoptosis in ATL cells. CCND3, which encodes a cell cycle regulator cyclin D3, was identified as a direct target of miR-212 in ATL cells. Rescue experiments with a miR-212-resistant variant of CCND3 demonstrated that overexpression of CCND3 restored cell-cycle progression and attenuated apoptotic response in miR-212-overexpressing ATL cells. Taken together, miR-212 exerts growth-suppressive effects in ATL cells largely by targeting CCND3 and may have therapeutic potential in ATL.


2016 ◽  
Vol 107 (9) ◽  
pp. 1302-1314 ◽  
Author(s):  
Shuichiro Okamoto ◽  
Takayuki Tsujioka ◽  
Shin‐ichiro Suemori ◽  
Jun‐ichiro Kida ◽  
Toshinori Kondo ◽  
...  

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1511-1511
Author(s):  
Atsushi Nemoto ◽  
Takeshi Inukai ◽  
Koshi Akahane ◽  
Hiroko Honna- Oshiro ◽  
Kumiko Goi ◽  
...  

Abstract Abstract 1511 Since BCR-ABL plays a central role in cell cycle progression of Philadelphia-chromosome positive (Ph+) leukemia cells and CDK4/6 critically involves in G1-progression of cell cycle, we analyzed sensitivity of Ph+ leukemia cell lines to compounds that act as specific CDK4/6 inhibitors. H3-thymidine uptake assay showed that both PD183812 and CBC219476 significantly inhibited cell growth of Ph+ lymphoid leukemia cell lines (n=9) in comparison with Ph+ myeloid leukemia cell lines (n=7) and Ph- ALL cell lines (n=26). Thus, we next tested the anti-leukemic activity of PD0332991, a potent CDK4/6 inhibitor that is under phase II clinical study for solid tumor patients, and found that 8 of 9 Ph+ lymphoid leukemia cell lines showed extremely higher sensitivity to PD0332991; median IC50 was <25 nM. IC50 of Ph+ lymphoid leukemia cell lines was significantly lower than that of Ph+ myeloid cell lines (200 nM, n=7) and Ph-ALL cell lines (100nM, n=25). PD0332991 effectively dephosphorylated Rb protein (pRb), and subsequently induced G1 arrest on all of Ph+ lymphoid leukemia cell lines. Moreover, PD0332991 gradually induced cell death in 4 Ph+ lymphoid leukemia cell lines. Since CDK4/6 inhibitor acts depending on intact pRb, we analyzed protein and gene expression status of Rb. Of note, all Ph+ lymphoid leukemia cell lines expressed intact pRb except for one cell line that showed relative resistance to PD0332991. In contrast, pRb was almost undetectable in Ph+ myeloid cell lines in spite of comparable level of Rb gene expression, which might be mechanism for resistance to PD0332991. However, most of Ph- ALL cell lines had intact pRb expression in spite of their relative resistance to PD0332991, indicating that Rb status alone did not explain higher PD0332991-sensitivity of Ph+ lymphoid leukemia cell lines. Thus, we assumed that Ph+ lymphoid leukemia cells showed higher PD0332991-sensitivity probably because BCR-ABL regulates CDK4/6 expression for cell cycle progression. To clarify this assumption, we treated Ph+ lymphoid leukemia cell lines with imatinib and performed immunoblot analysis of cell cycle machineries such as CDKs, cyclines, and CDK inhibitors. Of note, CDK4 expression level was frequently downregulated by imatinib in Ph+ lymphoid leukemia cell lines. Moreover, imatinib-induced downregulation of CDK4 in Ph+ lymphoid leukemia cell line was abrogated by the addition of IL-7 and FLT3 ligand, which stimulated cell cycle progression of imatinib-treated Ph+ ALL cell line. LY294002, a PI3K inhibitor, but not U0126, a MAPK inhibitor, and AG490, an inhibitor for JAK/STAT pathway, efficiently downregulated CDK4 expression in Ph+ lymphoid leukemia cell lines. Gene expression level of CDK4 in Ph+ lymphoid leukemia cell lines was downregulated by imatinib, and lactastatin, an inhibitor of protein degradation, partially inhibited imatinib-induced downregulation of CDK4 protein in Ph+ lymphoid leukemia cell lines, indicating that BCR-ABL regulates CDK4 expression both in gene expression level and in protein degradation level. These findings indicated that Ph+ lymphoid leukemia cell lines showed higher sensitivity to PD0332991 since BCR-ABL induces cell cycle progression of Ph+ lymphoid leukemia cells by regulating CDK4 as one of downstream pathways. Accordingly, we tested if PD0332991 shows anti-leukemic activity in Ph+ lymphoid leukemia cells that have a T315I mutation of BCR-ABL. SU/SR is an imatinib-resistant Ph+ ALL cell line with T315I mutation (IC50 for imatinib >10 mM), which was established from SU-Ph2, an imatinib-sensitive Ph+ ALL cell line (IC50 for imatinib <0.1 mM), after long-term culture in the presence of gradually increasing concentration of imatinib. Of note, PD0332991 effectively dephosphorylated pRb and inhibited cell growth of both SU/SR and SU-Ph2. Our findings provide a rationale for efficacy of PD0332991 in the context of anti-leukemic therapy for lymphoid crisis of CML and Ph+ ALL patients even with T315I mutation in BCR-ABL. Disclosures: No relevant conflicts of interest to declare.


Cancers ◽  
2018 ◽  
Vol 10 (10) ◽  
pp. 354 ◽  
Author(s):  
Mouna Sdiri ◽  
Xiangmin Li ◽  
William Du ◽  
Safia El-Bok ◽  
Yi-Zhen Xie ◽  
...  

The extensive applications of Cynomorium species and their rich bioactive secondary metabolites have inspired many pharmacological investigations. Previous research has been conducted to examine the biological activities and numerous interesting pharmaceutical activities have been reported. However, the antitumor activities of these species are unclear. To understand the potential anticancer activity, we screened Cynomorium coccineum and Cynomorium songaricum using three different extracts of each species. In this study, the selected extracts were evaluated for their ability to decrease survival rates of five different cancer cell lines. We compared the cytotoxicity of the three different extracts to the anticancer drug vinblastine and one of the most well-known medicinal mushrooms Amaurederma rude. We found that the water and alcohol extracts of C. coccineum at the very low concentrations possessed very high capacity in decreasing the cancer cells viability with a potential inhibition of tumorigenesis. Based on these primitive data, we subsequently tested the ethanol and the water extracts of C. coccineum, respectively in in vitro and in vivo assays. Cell cycle progression and induction of programmed cell death were investigated at both biological and molecular levels to understand the mechanism of the antitumor inhibitory action of the C. coccineum. The in vitro experiments showed that the treated cancer cells formed fewer and smaller colonies than the untreated cells. Cell cycle progression was inhibited, and the ethanol extract of C. coccineum at a low concentration induced accumulation of cells in the G1 phase. We also found that the C. coccineum’s extracts suppressed viability of two murine cancer cell lines. In the in vivo experiments, we injected mice with murine cancer cell line B16, followed by peritoneal injection of the water extract. The treatment prolonged mouse survival significantly. The tumors grew at a slower rate than the control. Down-regulation of c-myc expression appeared to be associated with these effects. Further investigation showed that treatment with C. coccineum induced the overexpression of the tumor suppressor Foxo3 and other molecules involved in inducing autophagy. These results showed that the C. coccineum extract exerts its antiproliferative activity through the induction of cell death pathway. Thus, the Cynomorium plants appear to be a promising source of new antineoplastic compounds.


2013 ◽  
Vol 54 (8) ◽  
pp. 1811-1813 ◽  
Author(s):  
Sebastian Schamberger ◽  
Maja Weber ◽  
Claudia Backsch ◽  
Jürgen Sonnemann ◽  
Udo R. Markert

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2055-2055
Author(s):  
Tomohiro Kozako ◽  
Makoto Yoshimitsu ◽  
Naomichi Arima ◽  
Shuhei Soeda ◽  
Shinya Hirata ◽  
...  

Abstract Introduction: Adult T-cell leukemia/lymphoma (ATL) is an aggressive T-cell malignancy that develops after long-term infection with human T-cell leukemia virus (HTLV-1). Despite the recent advances in chemotherapy, allogeneic hematopoietic stem cell transplantation, and supportive care, the prognosis for patients with acute, lymphoma, or unfavorable chronic subtypes (aggressive ATL) is one of the poorest among hematological malignancies; overall survival at 3 years is only 24 % in the more aggressive subtypes of ATL. In case of favourable chronic or smoldering ATL (indolent ATL), watchful waiting until disease progression has been recommended. Some patients with indolent ATL develop infections during this period. Therefore, urgent need for therapy and prophylaxis of ATL are still required. Recently, drug repositioning offers the possibility of reduced time and risk as several stages common to novel drug discovery and development can be bypassed because repositioning candidates have frequently been through several phases of development for their original indication. With the successful clinical introduction of a number of non-cancer drugs for cancer treatment, drug repositioning now became a potent alternative strategy to discover and develop novel anticancer drug candidates from the existing drug space. On the other hand, the transcription factor PPARγ plays various roles in lipid metabolism, immune response, cellular differentiation and apoptosis. Among clinically available angiotensin II receptor blockers, telmisartan is well known for its unique ability to activate PPARγ. Here, we assessed how telmisartan affects ATL cells from patients and leukemia cell lines. Methods and Results: Methods used in this study were carried out in accordance with the approved guidelines by the Committees for Ethical Review of Research involving Human Subjects at Kagoshima University. The subjects were examined by standard serological testing for the presence of HTLV-1 and by haematological/Southern blotting analysis for diagnosis of ATL. The classification of ATL was performed according to the criteria of Shimoyama. Telmisartan reduced cell viability and enhanced apoptotic cells in ex vivo peripheral blood monocytes of acute-type ATL, which has a poor prognosis. Telmisartan-induced apoptosis in cells from ATL patients (acute and chronic-type) and asymptomatic HTLV-1 carriers was significantly increased compared with those from healthy donors. Telmisartan also induced significant growth inhibition and apoptosis (Annexin V+ cells and TUNEL) in leukemia cell lines (HTLV-1-related cell lines: S1T, MT-2; Jurkat and HL60), while other angiotensin II receptor blockers, irbesartan and valsartan, did not induce cell death. In apoptosis, several key events occur in mitochondria, including the release of caspase activators, such as apoptosis-induced factor, and loss of mitochondrial transmembrane potential. Telmisartan induced loss of mitochondrial transmembrane potential and generation of reactive oxygen species, although apoptosis-inducing factor level was stable. Telmisartan also inhibited cell growth via caspase activation (caspase-3, 8 and 9) in the leukemia cells. However, treatment with a caspase inhibitor did not inhibit telmisartan-induced cell death. Interestingly, telmisartan increased the LC3-II-enriched protein fraction, indicating autophagosome accumulation as well as autophagy. Thus, Telmisartan simultaneously caused caspase activation and autophagy. Conclusion: A hypertension medication that has an anti-proliferation effect on leukemia cells is intriguing. These results suggest that telmisartan is highly effective against HTLV-1-infected cells and ATL cells in a caspase-dependent and -independent manner, and its clinical use may suppress the progression from indolent ATL or carrier status to aggressive ATL, and thus improve the prognosis of patients with this fatal disease. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1605-1605
Author(s):  
Xinliang Mao ◽  
Tabitha W. Wood ◽  
Xiaoming Wang ◽  
Johnathan St-Germain ◽  
Michael F. Moran ◽  
...  

Abstract D-cyclins are universally dysregulated in multiple myeloma and frequently over-expressed in acute leukemia. Therefore, to better understand the regulation of the D-cyclins and identify leads for novel therapeutic agents for the treatment of hematologic malignancies, we conducted a high throughput screen of 50,000 novel chemical compounds to identify inhibitors of D-cyclin transactivation. From this screen, we identified a chromene-based compound 8-ethoxy-2-(4-fluorophenyl)-3-nitro-2H-chromene (pichromene). In secondary assays, pichromene reduced expression of cyclins D1, D2, and D3 in myeloma and leukemia cell lines at low micromolar concentrations. Furthermore, pichromene arrested the cells in the G0/G1 phase of the cell cycle. In myeloma and leukemia cell lines, pichromene decreased levels of phospho-AKT, but did not alter levels of total AKT. PI3 kinases regulate AKT phosphorylation that, in turn, regulate D-cyclin expression and cell cycle progression. Therefore, we evaluated the effects of pichromene on the enzymatic activity of PI3 kinases. In cell-free enzymatic assays, pichromene inhibited the enzymatic activity of all four isoforms of the PI3 kinase, PI3Kalpha, beta, delta and gamma, with similar efficacy. In contrast it did not markedly inhibit the enzymatic activities of unrelated kinases AKT 1, 2 or 3, PDK 1 or 2, or GSK3β or 3α at concentrations up to 300 μM in a similar cell-free assay. However, in intact cells, due to its inhibition of PI3 kinases, pichomene inhibited AKT activity as noted above. As inhibitors of PI3 kinases are pro-apoptotic and may have anti-cancer activity, we evaluated the effects of pichromene on the viability of leukemia and myeloma cells. Leukemia and myeloma cell lines were treated with increasing concentrations of pichromene and cell viability was measured after 72 hours by an MTS assay. Pichromene induced cell death in 9/10 leukemia and 9/10 myeloma cell lines with an ED50 &lt; 10 μM. In contrast, it was less cytotoxic to primary normal hematopoietic cells obtained from volunteer donors of stem cells for allotransplant. Apoptosis was confirmed by Annexin V staining. Cell death was associated with caspase activation as demonstrated by the cleavage of caspase-3 and PARP through immunoblotting. Interestingly, U266 was the one myeloma cell line that was resistant to pichromene, and lacked detectable basal levels of phospho-AKT by immunoblotting. Given the effects of pichromene on malignant cells, we evaluated the efficacy of this compound in a leukemia xenograft mouse model. K562 cells were implanted subcutaneously into sublethally irradiated NOD/SCID mice. Mice were then treated with pichromene (50 mg/kg/day) or buffer control by oral gavage. Pichromene decreased tumor weight and volume by more than 35% as early as 8 days after treatment. No evidence of weight loss or gross organ toxicity was observed even when mice were treated with up to 500mg/kg/day of pichromene by oral gavage or intraperitoneally. Thus, in summary, we have identified a novel pan-inhibitor of PI3 kinases that displays preclinical efficacy in myeloma and leukemia.


Oncogene ◽  
2001 ◽  
Vol 20 (17) ◽  
pp. 2055-2067 ◽  
Author(s):  
Ritsuko Iwanaga ◽  
Kiyoshi Ohtani ◽  
Takeshi Hayashi ◽  
Masataka Nakamura

2000 ◽  
Vol 64 (5) ◽  
pp. 315-322 ◽  
Author(s):  
Yasuaki Yamada ◽  
Kazuyuki Sugahara ◽  
Kazuto Tsuruda ◽  
Kazuo Nohda ◽  
Naoki Mori ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document