scholarly journals Cytotoxic and Antiproliferative Effects of Diarylheptanoids Isolated from Curcuma comosa Rhizomes on Leukaemic Cells

Molecules ◽  
2020 ◽  
Vol 25 (22) ◽  
pp. 5476
Author(s):  
Natsima Viriyaadhammaa ◽  
Aroonchai Saiai ◽  
Waranya Neimkhum ◽  
Wariya Nirachonkul ◽  
Wantida Chaiyana ◽  
...  

Curcuma comosa belongs to the Zingiberaceae family. In this study, two natural compounds were isolated from C. comosa, and their structures were determined using nuclear magnetic resonance. The isolated compounds were identified as 7-(3,4-dihydroxyphenyl)-5-hydroxy-1-phenyl-(1E)-1-heptene (1) and trans-1,7-diphenyl-5-hydroxy-1-heptene (2). Compound 1 showed the strongest cytotoxicity effect against HL-60 cells, while its antioxidant and anti-inflammatory properties were stronger than those of compound 2. Compound 1 proved to be a potent antioxidant, compared to ascorbic acid. Neither compounds had any effect on red blood cell haemolysis. Furthermore, compound 1 significantly decreased Wilms’ tumour 1 protein expression and cell proliferation in KG-1a cells. Compound 1 decreased the WT1 protein levels in a time- and dose- dependent manner. Compound 1 suppressed cell cycle at the S phase. In conclusion, compound 1 has a promising chemotherapeutic potential against leukaemia.

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4713-4713
Author(s):  
Zhen Cai ◽  
Wenjun Wu Master

Abstract Myelodysplastic syndromes (MDS) represent a heterogenous group of clonal stem cell disorders with qualitative and quantitative abnormalities of blood cells and a high probability of evolving to acute leukemia. Intensive induction chemotherapy in order to reduce the malignant clone and reconstruct normal hematopoiesis is a classic therapy of MDS, especially high risk MDS. Topotecan (TPT), a semisynthetic water-soluble derivative of camptothecin, is a potent inhibitor of DNA topoisomerase I and has been extensively studied in hematologic malignances. However, little is known about how TPT acts against neoplastic cells. The aim of this study is to evaluate apoptotic effect of TPT on the MDS cell line MUTZ-1 and its associated changes in the expression of inhibitors of apoptosis protein (IAPs). The effect of TPT on MUTZ-1 growth was determined by using MTT assay. Characteristics associated with apoptosis induced by TPT were evaluated by transmission electron microscope, DNA gel electrophoresis and flow cytometry (FCM). Cell cycle shift were observed by FCM. Semi-quantitative RT-PCR was used to evaluate the mRNA expression of members of IAP gene family, including survivin, XIAP, Bcl-2, Bax, cIAP1 andcIAP2. The potential of mitochondrial membrane potential (MMP) was determined by using JC-1 probe. The results demonstrated that TPT significantly inhibited MUTZ-1 cell growth in a time- and dose-dependent manner with IC50 of 5.011 mmol/L, 1.297mmol/L and 0.483mmol/L at 24h, 48h and 72h respectively. Morphological features associated with TPT-induced apoptosis observed by transmission electron microscopy included cytoplasmic and nuclear shrinkage, karyorrhexis, nuclear convolution, chromatin condensation and margination, cytoplasmic vacuolization, and membrane-bound apoptotic bodies. An ambiguous DNA ladder was observed following treatment with 5mmol/L TPT for 24h, and a typical DNA ladder was observed with 10mmol/L TPT for 24h. The apoptotic rates were 11.69±0.51%, 34.07±1.73%, and 48.59±2.01%, respectively, after 24h culture with TPT as 1, 5, 10 μmol/L, significantly higher than that of the control (3.47%±0.3%; F=31.642, P<0.01). The percentage of MUTZ-1 cells in G2/M phase of the cell cycle decreased while in S and G0/G1 phase increased after treatment with 1mmol/L TPT for 24h,. The majority of the cells were arrested in S phase. After 24h culture with TPT at1, 5, and 10μmol/L, the mRNA levels of survivin, XIAP, cIAP1 and cIAP2 were decreased (P<0.01). This down-regulation was negatively correlated with TPT-induced apoptotic rates(P<0.05). There was no significant change in the Bax and Bcl-2 mRNA levels after TPT treatment (P>0.05). After 24h culture with 1μmol/L TPT, the MMP of TPT treated cells decreased significantly(P<0.01). Together, we conclude TPT can inhibit the growth and induce apoptosis of MUTZ-1 cells in a time- and dose- dependent manner. TPT can also induce the cell cycle changes, with the majoritoy of cells being arrested in S phase. The TPT-induced apoptosis in MUTZ-1 cells is associated with down-regulation of suvivin, XIAP, cIAP1and cIAP2 mRNA expresison. As well, MMP may be play a important role in the apoptotic process of MUTZ-1 cells induced by TPT.


2019 ◽  
Vol 14 (2) ◽  
pp. 187-197 ◽  
Author(s):  
Chuanjun Qu ◽  
Weiwei Zhu ◽  
Kaijie Dong ◽  
Zhaohai Pan ◽  
Ying Chen ◽  
...  

Background:A recent patent has been issued for hydroxysafflor yellow A (HSYA) as a drug to prevent blood circulation disorders. Hydroxysafflor yellow B (HSYB), an isomer of HSYA with antioxidative effects, has been isolated from the florets of Carthamus tinctorius. The effects of HSYB on the proliferation of cancer cells and its mechanism of action have not been investigated.Objective:The aims of this study were to investigate the anti-cancer effects and the molecular mechanism of HSYB for breast cancer MCF-7 cells.Methods:MTT assays and colony formation assays were used to assess the survival and proliferation of MCF-7 cells, respectively. Hoechst 33258 and flow cytometry were used to measure cell apoptosis and flow cytometry to determine effects on the cell cycle. Western blots were used to measure protein levels.Results:Treatment with HSYB reduced survival and proliferation of human breast cancer MCF-7 cells in a dose-dependent manner. Furthermore, HSYB arrested the MCF-7 cell cycle at the S phase and downregulated cyclin D1, cyclin E, and CDK2. Compared with a control group, HSYB suppressed the protein levels of p-PI3K, PI3K, AKT, and p-AKT in MCF-7 cells. In addition, HSYB decreased the levels of Bcl- 2, increased the levels of Bax, cleaved caspase-3 and caspase-9, and subsequently induced MCF-7 cell apoptosis.Conclusion:These data demonstrate that HSYB arrests the MCF-7 cell cycle at the S phase and induces cell apoptosis. Patent US20170246228 indicates that HSYB can be potentially used for the prevention and treatment of human breast cancer.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 4616-4616
Author(s):  
Azhar R. Hussain ◽  
Naif A. Al-Jomah ◽  
Mehar Sultana ◽  
Manugaran S. Pulicat ◽  
Khawla S. Al-Kuraya ◽  
...  

Abstract Proteosome inhibition is a novel approach for treating malignancy and has been approved for clinical use. The proteosome is the primary proteolytic mechanism in eukaryotic cells and inhibition of its catalytic activity initiates a cascade of events affecting cell cycle and apoptotic activities. These activities ultimately lead to cell cycle arrest and apoptosis in malignant cells however, the normal counterpart of these cells are spared. In this study, we used a panel of primary effusion lymphoma cell lines (BC1, BC3, BCBL1 and HBL6) to study the effects of proteosome inhibitor, MG132 on cell proliferation and apoptosis. Our data showed that proteosome inhibitor MG132 decreased cell viability as well as induced apoptosis in a dose dependent manner ranging from 0.5–10μM. Furthermore, treatment with 2.5μM MG132 for 24hours induced 41% apoptosis in BC1, 51% in BC3, 41% in BCBL1 and 48% in HBL6 cell lines as detected by annexinV/PI dual staining. S-phase kinase-associated protein 2 (skp-2) is a proto-oncogene and over expressed in various types of tumors. We sought to determine the role of Skp-2 following proteosome inhibition in PELs. MG132 treatment of PEL cell lines resulted in down-regulation of SKP-2 protein in a dose dependent manner whereas the expression of p-27 was up-regulated demonstrating an inverse relationship between these two proteins. Furthermore, MG132 treatment of PELs led to conformational changes in Bax protein and translocation to the mitochondria leading to the loss of mitochondrial membrane potential with subsequent release of cytochrome c from mitochondria into cytosol. Cytochrome c release caused activation of caspase-3 followed by polyadenosin-5′-diphosphate-ribose polymerase (PARP) cleavage. In addition, proteosome inhibitor treatment also caused down-regulation of inhibitor of apoptosis protein, XIAP. Taken together, our findings show that proteosome inhibition causes down-regulation of skp-2, up-regulation of p-27, inhibition of proliferation as well as caspase-dependent apoptosis in primary effusion lymphoma cells suggesting a role of proteosome inhibitors in the treatment of these aggressive cancers.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Yuan Wang ◽  
Huazhang Wu ◽  
Nannan Dong ◽  
Xu Su ◽  
Mingxiu Duan ◽  
...  

AbstractSulforaphane (SFN) extracted from broccoli sprout has previously been investigated for its potential properties in cancers, however, the underlying mechanisms of the anticancer activity of SFN remain not fully understood. In the present study, we investigate the effects of SFN on cell proliferation, cell cycle, cell apoptosis, and also the expression of several cell cycle and apoptosis-related genes by MTT assay, flow cytometry and western blot analysis in gastric cancer (GC) cells. The results showed that SFN could impair the colony-forming ability in BGC-823 and MGC-803 cell lines compared with the control. In addition, SFN significantly suppressed cell proliferation by arresting the cell cycle at the S phase and enhancing cell apoptosis in GC cells in a dose-dependent manner. Western blot results showed that SFN treatment significantly increased the expression levels of p53, p21 and decreased CDK2 expression, which directly regulated the S phase transition. The Bax and cleaved-caspase-3 genes involved in apoptosis executive functions were significantly increased in a dose-dependent manner in BGC-823 and MGC-803 cells. These results suggested that SFN-induced S phase cell cycle arrest and apoptosis through p53-dependent manner in GC cells, which suggested that SFN has a potential therapeutic application in the treatment and prevention of GC.


2013 ◽  
Vol 41 (03) ◽  
pp. 665-682 ◽  
Author(s):  
Sansan Chen ◽  
Xinming Qu ◽  
Pei Wan ◽  
Qing Wen Li ◽  
Ziyi Wang ◽  
...  

Norcantharidin (NCTD) is currently used for anticancer therapy but the exact mechanism of action remains unknown. Pre-replicative complexes (pre-RCs) are essential for cell DNA replication and highly related to malignant proliferation. Here, we examined the inhibitory effect of NCTD on pre-RC components in HepG2 cells. We showed that NCTD induced degradation of Cdc6 and Mcm2 in a dose-dependent manner. Under 100 μM NCTD concentration, about 70% of Cdc6 and 50% of Mcm2 were degraded. In addition, the nuclear translocation of Mcm6 was inhibited by NCTD. Further studies aiming at G1 synchronous cells showed that, NCTD reduced the chromatin-bound Cdc6, Mcm2 and Mcm6. Moreover, the cells were blocked from entering the S phase and accumulated at the G1 phase when released synchronously into the cell cycle. Consistently, the DNA replication was inhibited by NCTD. Finally, the combination NCTD with Cdc6 depletion lead to more severe cytotoxicity (88%) than NCTD (52%) and Cdc6 depletion (39%) alone. A synergic cytotoxicity was observed between Cdc6 depletion and NCTD. In conclusion, our results demonstrate that NCTD inhibits pre-RC assembly; subsequently blocks the G1 to S transition; and inhibits DNA replication in HepG2 cells. Pre-RCs are an intriguing target for cancer therapy, which merits further investigations for anticancer development.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2254-2254 ◽  
Author(s):  
Yoko Tabe ◽  
Masako Harada ◽  
Yuka Miyamae ◽  
Saiko Kazuno ◽  
Tsutomu Fujimura ◽  
...  

Abstract Mantle cell lymphoma (MCL), which overexpresses cyclin-D1 through an alteration in the t(11;14)(q13;q32) chromosomal region, is associated with brief disease-free and overall survival durations characteristic of aggressive B-cell lymphomas. Bruton tyrosine kinase (BTK) has been identified as a key component of the B-cell antigen receptor (BCR) signaling pathway and is implicated in the pathogenesis of certain B-cell malignancies. Phase III clinical trials of BKT inhibitor ibrutinib in MCL patients have demonstrated clinical responses characterized by mobilization of tissue-resident MCL cells into the peripheral blood. However, since the time to maximum response with ibrutinib is relatively long and patients may become resistant to BTK inhibition, combination regimens that accelerate time to remission and increase depth of remission are of considerable interest. We hypothesized that combinations of ibrutinib with proapoptotic drugs that function independently of BCR signaling could yield synergistic anti-lymphoma interactions. Thus we investigated the antitumor effects and molecular mechanisms of simultaneous treatment with ibrutinib and selexinor, an oral selective inhibitor of nuclear export (SINE)(KPT-330, Karyopharm), or ABT-199, a selective Bcl-2 inhibitor. SINE agents exhibit antiproliferative and proapoptotic activities against MCL cells via inhibition of nuclear export of tumor suppresor proteins, transcription factors and oncogenic mRNAs and repression of ribosomal biogenesis (Tabe et al. ASH 2013). Selinexor showed promising anti tumor activity in agrresive lymphoma as part of ongoing Phase 1 study (ASCO 2014). ABT-199 has promising proapoptotic activity in relapsed/refractory CLL and NHL without inducing thrombocytopenia. In this study, we utilized four MCL cell lines: MINO, Z138, Jeko-1, and JVM2. Inhibition of BTK activity by ibrutinib resulted in reduction of cell proliferation in a dose-dependent manner with G0/G1 cell cycle arrest but no apoptosis induction (IC50 at 48 hrs by MTT: 5.4 mM for MINO, 3.5 mM for Z138, 0.5 mM for Jeko-1, 3.1 mM for JVM2). Western blot analysis demonstrated ibrutinib-induced downregulation of phospho-(p-)BTK, p-Akt, mTORC1 substrates p-S6K and p-4EBP1, and cyclin D1 expression. Single-agent selinexor induced cell growth inhibition with G0/G1 cell cycle arrest in a dose-dependent manner (IC50 ranging from 10 nM to 130 nM). The ibrutinib/selinexor combination resulted in further decrease of p-4EBP1 and cyclin D1 expression and downregulation of p-Rb, c-Myc, and Mcl-1, which translated into synergistic reduction of cell proliferation in three of the four tested cell lines (combination index [CI]: 0.4 for MINO, 0.2 for Jeko-1, 0.2 for JVM2). ABT-199 inhibited cell proliferation with apoptosis induction in MINO and Z138 cells (IC50: 1.5 nM for MINO, 17.5 nM for Z138), which synergistically enhanced the antiproliferative effects of ibrutinib (CI: 0.6 for MINO, 0.8 for Z138) with striking reductions of p-4EBP1, cyclin D1, p-Rb, and c-Myc expression along with induction of Bax and cleaved caspase-3. To investigate the molecular modifications of the cellular pathway network in response to BTK blockade by ibrutinib alone or in combination with selinexor or ABT-199, we employed the proteomic technology of isobaric tags for relative and absolute quantitation (iTRAQ). In MINO cells, iTRAQ identified 1,401 unique proteins. Ibrutinib induced downregulation of isoform BTK (p=0.02) and the cell cycle initiation of mitosis pathway (p=0.003) with decreases of ribosomal proteins and elongation factors. Combination with selinexor upregulated the apoptosis and oxidative stress–associated pathways with increases of cytochrome c, voltage-dependent anion channels, HSP10, and histone H1, all of which function as dynamic initiators of mitochondria-mediated apoptosis (p < 0.05). ABT-199 by itself induced upregulation of the apoptosis and oxidative stress–associated pathways (p < 0.001), and these effects were significantly enhanced by combination with ibrutinib. Taken together, our findings suggest that treatment with combinations of ibrutinib and selinexor or ABT-199 exerts synergistic antiproliferative effects through inhibition of mTOR signaling, downregulation of ribosomal biosynthesis, and induction of mitochondria-mediated apoptosis. These combinations warrant further evaluation in clinical trials in MCL patients. Disclosures Andreeff: Karyopharm: Research Funding.


2015 ◽  
Vol 2015 ◽  
pp. 1-10 ◽  
Author(s):  
Yong-sen Jia ◽  
Xue-qin Hu ◽  
Hegyi Gabriella ◽  
Li-juan Qin ◽  
Nora Meggyeshazi

Objective. The purpose of the study was to elucidate the molecular mechanism of tenacissoside H (TDH) inhibiting esophageal carcinoma infiltration and proliferation.Methods.In vitro, EC9706 cells were treated with TDH. Cells proliferation and cell cycle were assayed. PI3K and NF-κB mRNAs expression were determined by real time PCR.In vivo, model of nude mice with tumor was established. Mice were treated with TDH. Inhibition ratio of tumor volume was calculated. PCNA expression was examined. Protein expression in PI3K/Akt-NF-κB signaling pathway was determined.Results.In vitro, TDH significantly inhibited cells proliferation in a time-and-dose-dependent manner. TDH arrested the cell cycle in S phase and significantly inhibited PI3K and NF-κB mRNA expression, compared with blank controlled group (P<0.05). In vivo, TDH strongly inhibits tumor growth and volume. PCNA expression was significantly decreased after treatment of TDH. TDH downregulated proteins expression in PI3K/Akt-NF-κB transduction cascade (P<0.05).Conclusion. TDH inhibited esophageal carcinoma infiltration and proliferation bothin vitroandin vivo. The anticancer activity has relation to arresting the cell cycle at the S phase, inhibited the PCNA expression of transplanted tumors in nude mice, and regulated the protein expression in the PI3K/Akt-NF-κB transduction cascade.


2019 ◽  
Vol 19 (4) ◽  
pp. 557-566 ◽  
Author(s):  
Nerella S. Goud ◽  
Mahammad S. Ghouse ◽  
Jatoth Vishnu ◽  
Jakkula Pranay ◽  
Ravi Alvala ◽  
...  

Background: Human Galectin-1, a protein of lectin family showing affinity towards β-galactosides has emerged as a critical regulator of tumor progression and metastasis, by modulating diverse biological events including homotypic cell aggregation, migration, apoptosis, angiogenesis and immune escape. Therefore, galectin-1 inhibitors might represent novel therapeutic agents for cancer. Methods: A new series of heterocyclic imines linked coumarin-thiazole hybrids (6a-6r) was synthesized and evaluated for its cytotoxic potential against a panel of six human cancer cell lines namely, lung (A549), prostate (DU-145), breast (MCF-7 & MDA-MB-231), colon (HCT-15 & HT-29) using MTT assay. Characteristic apoptotic assays like DAPI staining, cell cycle, annexin V and Mitochondrial membrane potential studies were performed for the most active compound. Furthermore, Gal-1 inhibition was confirmed by ELISA and fluorescence spectroscopy. Results: Among all, compound 6g 3-(2-(2-(pyridin-2-ylmethylene) hydrazineyl) thiazol-4-yl)-2H-chromen-2- one exhibited promising growth inhibition against HCT-15 colorectal cancer cells with an IC50 value of 1.28 ± 0.14 µM. The characteristic apoptotic morphological features like chromatin condensation, membrane blebbing and apoptotic body formation were clearly observed with compound 6g on HCT-15 cells using DAPI staining studies. Further, annexin V-FITC/PI assay confirmed effective early apoptosis induction by treatment with compound 6g. Loss of mitochondrial membrane potential and enhanced ROS generation were confirmed with JC-1 and DCFDA staining method, respectively by treatment with compound 6g, suggesting a possible mechanism for inducing apoptosis. Moreover, flow cytometric analysis revealed that compound 6g blocked G0/G1 phase of the cell cycle in a dose-dependent manner. Compound 6g effectively reduced the levels of Gal-1 protein in a dose-dependent manner. The binding constant (Ka) of 6g with Gal-1 was calculated from the intercept value which was observed as 1.9 x 107 M-1 by Fluorescence spectroscopy. Molecular docking studies showed strong interactions of compound 6g with Gal-1 protein. Conclusion: Our studies demonstrate the anticancer potential and Gal-1 inhibition of heterocyclic imines linked coumarin-thiazole hybrids.


Nutrients ◽  
2021 ◽  
Vol 13 (7) ◽  
pp. 2178
Author(s):  
Fabio Morandi ◽  
Veronica Bensa ◽  
Enzo Calarco ◽  
Fabio Pastorino ◽  
Patrizia Perri ◽  
...  

Neuroblastoma (NB) is the most common extra-cranial solid tumor of pediatric age. The prognosis for high-risk NB patients remains poor, and new treatment strategies are desirable. The olive leaf extract (OLE) is constituted by phenolic compounds, whose health beneficial effects were reported. Here, the anti-tumor effects of OLE were investigated in vitro on a panel of NB cell lines in terms of (i) reduction of cell viability; (ii) inhibition of cell proliferation through cell cycle arrest; (iii) induction of apoptosis; and (iv) inhibition of cell migration. Furthermore, cytotoxicity experiments, by combining OLE with the chemotherapeutic topotecan, were also performed. OLE reduced the cell viability of NB cells in a time- and dose-dependent manner in 2D and 3D models. NB cells exposed to OLE underwent inhibition of cell proliferation, which was characterized by an arrest of the cell cycle progression in G0/G1 phase and by the accumulation of cells in the sub-G0 phase, which is peculiar of apoptotic death. This was confirmed by a dose-dependent increase of Annexin V+ cells (peculiar of apoptosis) and upregulation of caspases 3 and 7 protein levels. Moreover, OLE inhibited the migration of NB cells. Finally, the anti-tumor efficacy of the chemotherapeutic topotecan, in terms of cell viability reduction, was greatly enhanced by its combination with OLE. In conclusion, OLE has anti-tumor activity against NB by inhibiting cell proliferation and migration and by inducing apoptosis.


1993 ◽  
Vol 13 (3) ◽  
pp. 1610-1618 ◽  
Author(s):  
J E Slansky ◽  
Y Li ◽  
W G Kaelin ◽  
P J Farnham

Enhanced expression of genes involved in nucleotide biosynthesis, such as dihydrofolate reductase (DHFR), is a hallmark of entrance into the DNA synthesis (S) phase of the mammalian cell cycle. To investigate the regulated expression of the DHFR gene, we stimulated serum-starved NIH 3T3 cells to synchronously reenter the cell cycle. Our previous results show that a cis-acting element at the site of DHFR transcription initiation is necessary for serum regulation. Recently, this element has been demonstrated to bind the cloned transcription factor E2F. In this study, we focused on the role of E2F in the growth regulation of DHFR. We demonstrated that a single E2F site, in the absence or presence of other promoter elements, was sufficient for growth-regulated promoter activity. Next, we showed that the increase in DHFR mRNA at the G1/S-phase boundary required protein synthesis, raising the possibility that a protein(s) lacking in serum-starved cells is required for DHFR transcription. We found that, similar to DHFR mRNA expression, levels of murine E2F1 mRNA were low in serum-starved cells and increased at the G1/S-phase boundary in a protein synthesis-dependent manner. Furthermore, in a cotransfection experiment, expression of human E2F1 stimulated the DHFR promoter 22-fold in serum-starved cells. We suggest that E2F1 may be the key protein required for DHFR transcription that is absent in serum-starved cells. Expression of E2F also abolished the serum-stimulated regulation of the DHFR promoter and resulted in transcription patterns similar to those seen with expression of the adenoviral oncoprotein E1A. In summary, we provide evidence for the importance of E2F in the growth regulation of DHFR and suggest that alterations in the levels of E2F may have severe consequences in the control of cellular proliferation.


Sign in / Sign up

Export Citation Format

Share Document