scholarly journals Thymoquinone and Difluoromethylornithine (DFMO) Synergistically Induce Apoptosis of Human Acute T Lymphoblastic Leukemia Jurkat Cells Through the Modulation of Epigenetic Pathways

2020 ◽  
Vol 19 ◽  
pp. 153303382094748
Author(s):  
Mahmoud Alhosin ◽  
Syed Shoeb I. Razvi ◽  
Ryan A. Sheikh ◽  
Jalaluddin A. Khan ◽  
Mazin A. Zamzami ◽  
...  

Thymoquinone (TQ), a natural anticancer agent exerts cytotoxic effects on several tumors by targeting multiple pathways, including apoptosis. Difluoromethylornithine (DFMO), an irreversible inhibitor of the ornithine decarboxylase (ODC) enzyme, has shown promising inhibitory activities in many cancers including leukemia by decreasing the biosynthesis of the intracellular polyamines. The present study aimed to investigate the combinatorial cytotoxic effects of TQ and DFMO on human acute T lymphoblastic leukemia Jurkat cells and to determine the underlying mechanisms. Here, we show that the combination of DFMO and TQ significantly reduced cell viability and resulted in significant synergistic effects on apoptosis when compared to either DFMO or TQ alone. RNA-sequencing showed that many key epigenetic players including Ubiquitin-like containing PHD and Ring finger 1 (UHRF1) and its 2 partners DNA methyltransferase 1 (DNMT1) and histone deacetylase 1 (HDAC1) were down-regulated in DFMO-treated Jurkat cells. The combination of DFMO and TQ dramatically decreased the expression of UHRF1, DNMT1 and HDAC1 genes compared to either DFMO or TQ alone. UHRF1 knockdown led to a decrease in Jurkat cell viability. In conclusion, these results suggest that the combination of DFMO and TQ could be a promising new strategy for the treatment of human acute T lymphoblastic leukemia by targeting the epigenetic code.

Genes ◽  
2021 ◽  
Vol 12 (5) ◽  
pp. 622
Author(s):  
Omeima Abdullah ◽  
Ziad Omran ◽  
Salman Hosawi ◽  
Ali Hamiche ◽  
Christian Bronner ◽  
...  

Silencing of tumor suppressor genes (TSGs) through epigenetic mechanisms, mainly via abnormal promoter DNA methylation, is considered a main mechanism of tumorigenesis. The abnormal DNA methylation profiles are transmitted from the cancer mother cell to the daughter cells through the involvement of a macromolecular complex in which the ubiquitin-like containing plant homeodomain (PHD), and an interesting new gene (RING) finger domains 1 (UHRF1), play the role of conductor. Indeed, UHRF1 interacts with epigenetic writers, such as DNA methyltransferase 1 (DNMT1), histone methyltransferase G9a, erasers like histone deacetylase 1 (HDAC1), and functions as a hub protein. Thus, targeting UHRF1 and/or its partners is a promising strategy for epigenetic cancer therapy. The natural compound thymoquinone (TQ) exhibits anticancer activities by targeting several cellular signaling pathways, including those involving UHRF1. In this review, we highlight TQ as a potential multitarget single epidrug that functions by targeting the UHRF1/DNMT1/HDAC1/G9a complex. We also speculate on the possibility that TQ might specifically target UHRF1, with subsequent regulatory effects on other partners.


Genes ◽  
2021 ◽  
Vol 13 (1) ◽  
pp. 42
Author(s):  
Omeima Abdullah ◽  
Mahmoud Alhosin

HAUSP (herpes virus-associated ubiquitin-specific protease), also known as Ubiquitin Specific Protease 7, plays critical roles in cellular processes, such as chromatin biology and epigenetics, through the regulation of different signaling pathways. HAUSP is a main partner of the “Epigenetic Code Replication Machinery,” ECREM, a large protein complex that includes several epigenetic players, such as the ubiquitin-like containing plant homeodomain (PHD) and an interesting new gene (RING), finger domains 1 (UHRF1), as well as DNA methyltransferase 1 (DNMT1), histone deacetylase 1 (HDAC1), histone methyltransferase G9a, and histone acetyltransferase TIP60. Due to its deubiquitinase activity and its ability to team up through direct interactions with several epigenetic regulators, mainly UHRF1, DNMT1, TIP60, the histone lysine methyltransferase EZH2, and the lysine-specific histone demethylase LSD1, HAUSP positions itself at the top of the regulatory hierarchies involved in epigenetic silencing of tumor suppressor genes in cancer. This review highlights the increasing role of HAUSP as an epigenetic master regulator that governs a set of epigenetic players involved in both the maintenance of DNA methylation and histone post-translational modifications.


2021 ◽  
Vol 12 (5) ◽  
Author(s):  
Weilin Sun ◽  
Gang Ma ◽  
Li Zhang ◽  
Pengliang Wang ◽  
Nannan Zhang ◽  
...  

AbstractADAMTS9 belongs to the ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) protein family, and its expression is frequently silenced due to promoter hypermethylation in various human cancers. However, the underlying mechanisms remain largely unknown. In this study, we investigated the inhibitory effects of ADAMTS9 on gastric cancer (GC) cells. We initially examined ADAMTS9 protein level in 135 GC and adjacent normal tissue pairs, showing that ADAMTS9 was strikingly decreased in the malignant specimens and patients with low ADAMTS9 expression exhibited more malignant phenotypes and poorer outcome. ADAMTS9 expression was restored in AGS and BGC-823 cells, which then markedly suppressed cellular viability and motility in vitro and in vivo. As ADAMTS9 was enriched in the nuclei of gastric mucosal cells, RNA-sequencing experiment showed that ADAMTS9 significantly altered gene expression profile in BGC-823 cells. Additionally, DNA methyltransferase 3α (DNMT3A) was identified to be responsible for the hypermethylation of ADAMTS9 promoter, and this methyltransferase was ubiquitinated by ring finger protein 180 (RNF180) and then subject to proteasome-mediated degradation. In conclusion, we uncovered RNF180/DNMT3A/ADAMTS9 axis in GC cells and showed how the signaling pathway affected GC cells.


2022 ◽  
Vol 12 (4) ◽  
pp. 763-769
Author(s):  
Liang Yu ◽  
Sheng Zhang ◽  
Wei He

microRNA-136 can inhibit the proliferating activity of malignant cells and also participate in chemotherapy resistance of colorectal cancer via modulating HDAC1. This study assessed miR-136’s effect on NSCLC cell proliferation and underlying mechanisms. Tumor tissues and paracancerous tissues from NSCLC patients were collected to measure miR-136 and HDAC1 level. Cells were transfected with miR-136-mimics, miR-136-inhibitors or miR-136 mimics+HDAC1-OE followed by analysis of cell viability and apoptosis by CCK-8 method and flow cytometry, phosphorylation of Jak2/STAT3 by western blot. miR-136 was significantly downregulated in tumor tissues and NSCLC cells, accompanied by upregulated HDAC1. miR-136 overexpression suppressed HDAC1 expression, retarded phosphorylation and activation of Jak2/STAT3 signaling, reduced NSCLC cell viability and enhanced apoptosis. In addition, co-transfection of miR-136-mimics and HDAC1-OE reversed the inhibitory effects of miR-136 on NSCLC cells. In conclusion, miR-136 is reduced and HDAC1 is increased in NSCLC and miR-136 overexpression inhibited NSCLC cell proliferation and increased apoptosis possibly through regulating HDAC1/Jak2/STAT3 signal pathway, indicating that miR-136 might be a novel target for the treatment of NSCLC.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3020-3020
Author(s):  
Frank C. Passero ◽  
Ravi Dashnamoorthy ◽  
Afshin Beheshti ◽  
J. Tyson McDonald ◽  
Andrew M Evens

Abstract Background: Activation of NF-kB pathways are a hallmark feature of TCL and HL, making proteasome inhibition an attractive therapeutic target. Previous studies have demonstrated prominent in vivo efficacy of ixazomib, an oral proteasome inhibitor for both TCL and HL. Among the common significantly regulated genes identified via systems biology approach include upregulation of genes encoding for ubiquitin proteasome subunits (Ravi et al. Cancer Res. 2016). Further combination studies with HDACi belinostat were synergistic in Jurkat, HH and L428 cell lines, and NRF2 was discovered as mediator of proteasome gene expression. We hypothesized that HDACi abrogates NRF2 mediated proteasome recovery leading to synergistic effects on cell viability in combination with ixazomib. Methods: Global transcriptome analysis was performed on RNA isolated from multiple cell lines include TCL (Jurkat) and HL (L540, L428) treated with ixazomib and control, as well as Jurkat cell lines treated with single agent ixazomib, belinostat and combination. Significant genes were determined by applying a one-way ANOVA with an adjusted Bonferroni correction for a false discovery rate (FDR) < 0.05. Further pathway analysis from significant genes was performed by using a fold change greater than ±1.2 comparing all samples to each other and observing pathway relationships using Ingenuity Pathway Analysis. Gene Set Enrichment Analysis was performed with FDR <0.05 for functional analysis. Proteasome-Glo cell based assay was used to evaluate caspase-, chymotrypsin, and trypsin-like activity. Proteasome activity was measured at 24 and 72 hours after Jurkat cell lines were treated with control, ixazomib or belinostat as single agents, and in combination. SiRNA knockdown experiments were performed in Jurkat cell line with NRF2 and non-targeting (NT) SiRNA transfection. Real-time quantitative PCR (qPCR) for proteasome subunit and NRF2 genes was performed on RNA isolated from treated cells. Results: Transcriptome analysis revealed upregulation of proteasome genes in ixazomib treated cell lines Jurkat, L540 and L428 at 24 hours. In Jurkat TCL, ixazomib caused decreased caspase-like and chymotrypsin-like proteasome activity at 24 hours that was followed by recovery of these activities at 72 hours. The combination of ixazomib and belinostat significantly decreased proteasome activity for chymotrypsin-like, caspase-like and trypsin-like activity at 72 hours compared to single agent ixazomib or belinostat. In Jurkat cells, NRF2 was identified as a transcriptional regulator involved in proteasome gene regulation, showing upregulation of proteasomal genes and NRF2 with ixazomib single agent, downregulation with belinostat single agent and in combination with ixazomib. These results were confirmed with qPCR for NRF2 and proteasome genes in Jurkat and L428. SiRNA knockdown for NRF2 in Jurkat cells resulted in decreased cell viability, NRF2 and proteasome gene expression compared with NT SiRNA following ixazomib treatment. Conclusions: Treatment with single agent ixazomib induced prominent proteasome gene expression in all TCL and HL cell lines. In Jurkat, recovery of chymotrypsin and caspase-like proteasome activity occurred by 72 hours suggesting that transcriptional changes induced by proteasome inhibition contributed to proteasome function recovery. Combination therapy with belinostat resulted in downregulation of proteasome genes in Jurkat and L428 and prevented functional recovery of the proteasome observed in Jurkat. Our results suggest that targeting the proteasome itself with ixazomib and preventing the induced recovery of proteasome genes with belinostat contributes to synergistic effects observed on proteasome function and cell viability in TCL and HL. Further studies with CRISPR/Cas to confirm the effect of NRF2 on proteasome gene and functional recovery in the context of proteasome inhibition are ongoing and will be reported. Disclosures Evens: Takeda: Other: Advisory board.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3701-3701
Author(s):  
Hideki Goto ◽  
Rachel Wilkie Grantham ◽  
Motoaki Mizuuchi ◽  
Tetsuji Yamaguchi ◽  
Takanori Teshima ◽  
...  

Abstract Apoptosis-based therapies have been used as a powerful weapon against hematologic malignancies (Reed JC et al, Blood 2005 15; 408-418). One strategy for attacking cancer is to overcome its resistance to apoptosis via specific targeting molecules. In this study, we are focusing on protein tyrosine kinase 7 (PTK7) as a new target for cancer therapy. PTK7, also known as CCK4, is a member of transmembrane tyrosine kinase family proteins that has been implicated in cell migration, polarity and apoptosis. PTK7 is overexpressed in acute T-cell lymphoblastic leukemia cells, and a recent report indicated that PTK7 expression was associated with poor prognosis. However, the mechanism by which PTK7 elicits its anti-apoptotic effects and directly promotes tumor proliferation has not yet been clarified. In addition, no data has been reported regarding the relationship between doxorubicin resistance and PTK7 expression. In this study, we investigated the role of PTK7 in chemotherapy resistance. Two different shRNA lentiviruses targeting PTK7 were stably infected in Jurkat cells (Jurkat tet-shPTK7#1 and #2), and gene knockdown assays against PTK7 were performed. In the presence of doxycycline, we observed a decrease in the viability of PTK7 knockdown cells following stimulation with αFAS-Ab (Jurkat tet-shPTK7#1: 28.6% ± 5.2%, #2: 8.7% ± 0.3%) compared to control cells (considered to be 100%). Similar results of decreased cell viability were also obtained with TRAIL stimulation (Jurkat tet-shPTK7#1: 22.5% ± 5.9%, #2: 14.7% ± 9.8%) compared to control cells (considered to be 100%). Furthermore, we observed a decrease in the viability of PTK7 knockdown cells following treatment with Doxorubicin compared to control cells treated with scramble siRNA. The reduction of cell viability in PTK7 knockdown cells could not be attributed to PTK7 knockdown itself since annexin-V and PI staining showed limited apoptotic changes in Jurkat tet-shPTK7 cells. These changes were also not the result of Fas receptor (CD95) and Trail receptor (DR4, DR5) expression up-regulation as flow cytometric analysis revealed no differences in the expression rates of Fas receptor and Trail receptor between scramble control cells and PTK7 knockdown cells. Very interestingly, PTK7 knockdown had no effect on viability when non-cancer cell lines were stimulated with Fas, TRAIL and Doxorubicin, suggesting that the synergistic effect of PTK7 knockdown and TNF family cytokine treatment is cancer specific. Next, to identify interaction partners of the PTK7 cytoplasmic domain (PTK7-cyto), we performed a yeast two-hybrid assay, and showed that FADD-DD was the only molecule found to bind to PTK7-cyto in both LEU2 reporter and LacZ reporter assays. This protein interaction was further verified by co-immunoprecipitation assays. Finally, to demonstrate the activation of Caspase-8 and PARP, which is induced by TNF-family death receptors, immunoblotting was performed. The shRNA-mediated silencing of PTK7 in Jurkat cells indeed increased Fas- and TRAIL-induced processing of caspase-8 and PARP compared to control RNA. These data suggest that PTK7 appears to be important for a proximal step in Fas and TRAIL signaling, which is consistent with the selective sensitization of tumor cells to the extrinsic apoptosis pathway. Together, our findings reveal an unexpected role for PTK7 in terms of death receptor-mediated apoptosis, suggesting that PTK7 could provide a new target for cancer therapy. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2001 ◽  
Vol 97 (7) ◽  
pp. 1999-2007 ◽  
Author(s):  
Yasuhiko Kano ◽  
Miyuki Akutsu ◽  
Saburo Tsunoda ◽  
Hiroyuki Mano ◽  
Yuko Sato ◽  
...  

Abstract The BCR/ABL tyrosine kinase has been implicated in the pathogenesis of chronic myelogenous leukemia (CML) and Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). STI571 is a novel anticancer agent that selectively inhibits the BCR/ABL tyrosine kinase. The cytotoxic effects of STI571 were studied in combination with antileukemic agents against Ph+leukemia cell lines, KU812, K-562, TCC-S, and TCC-Y. The cells were exposed to STI571 and to other agents simultaneously for 5 or 7 days. Cell growth inhibition was determined by MTT assay. The cytotoxic effects in combinations at the inhibitory concentration of 80% level were evaluated by the isobologram. STI571 produced synergistic effects with recombinant and natural α-interferons in 2 of 3 and 3 of 3 cell lines, respectively. STI571 produced additive effects with hydroxyurea, cytarabine, homoharringtonine, doxorubicin, and etoposide in all 4 cell lines. STI571 with 4-hydroperoxy-cyclophosphamide, methotrexate, or vincristine produced additive, antagonistic, and synergistic effects in 3 of 4 cell lines, respectively. These findings suggest that the simultaneous administration of STI571 with other agents except methotrexate would be advantageous for cytotoxic effects against Ph+ leukemias. Among them, the simultaneous administration of STI571 and α-interferons or vincristine would be highly effective against Ph+ leukemias and these combinations would be worthy of clinical trials. In contrast, the simultaneous administration of STI571 with methotrexate would have little therapeutic efficacy. Although there are gaps between in vitro studies and clinical trials, the present findings provide useful information for the establishment of clinical protocols involving STI571.


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii94-ii94
Author(s):  
Fatemeh Jamali ◽  
Paul Daniel ◽  
Kevin Petrecca ◽  
Bassam Abdulkarim ◽  
Siham Sabri

Abstract Glioblastoma (GBM), the most common and advanced malignant primary brain tumor in adults remains incurable. GBM tumor recurrence occurs within a short time reflecting the failure to eradicate chemo-radioresistant GBM stem cells (GSCs). GSCs alter the redox system including Thioredoxin (Trx) and glutathione (GSH) systems to counteract increased reactive oxygen species (ROS). We previously showed the role of O6-methylguanine-DNA methyltransferase (MGMT) in response to PRIMAMET, a drug that targets the TP53 tumor suppressor gene and decrease Thioredoxin reductase1 (TRxR1) levels and further identified a positive relationship between MGMT and TrxR1 in established GBM cell lines isogenic for MGMT. Auranofin (Au), an orally available ROS-inducing FDA-approved drug is a potent irreversible inhibitor of TrxR1. The effect of Au on GSCs and the potential relationship between TrXR1, MGMT and p53 specifically in GSCs remain unknown. We hypothesized that increased ROS levels in GSCs might affect their drug resistant phenotype. we investigated the cytotoxic effects of Au on GSCs with known MGMT and P53 status and the mechanisms underlying these effects. Our results suggest that Au exerts strong cytotoxic effects in GSCs within a micromolar range. These effects were associated with increased ROS levels, decreased TrXR1, MGMT, phosphorylation of ERk1/2, activation of p53 and increased apoptosis, we also used a knockdown strategy to assess the role of p53 and showed that increased sensitivity to Au for p53-knockdown. We showed the role of ROS in response to Au using ROS inducer L-Buthionine Sulfoxamine (L-BSO), a GSH inhibitor, and ROS scavenger N-Acetylcysteine (NAc). L-BSO combined with Au drastically decreased the IC50 within a nanomolar range suggesting the crucial role of both Trx and GSH systems in GSCs redox balance. Increased ROS with concomitant GSH depletion using L-BSO might circumvent drug-induced oxidative stress and overcome drug resistance in MGMT-positive and negative GSCs


2020 ◽  
Vol 20 (4) ◽  
pp. 307-317
Author(s):  
Yuan Yang ◽  
Jin Huang ◽  
Jianzhong Li ◽  
Huansheng Yang ◽  
Yulong Yin

Background: Butyric acid (BT), a short-chain fatty acid, is the preferred colonocyte energy source. The effects of BT on the differentiation, proliferation, and apoptosis of small intestinal epithelial cells of piglets and its underlying mechanisms have not been fully elucidated. Methods: In this study, it was found that 0.2-0.4 mM BT promoted the differentiation of procine jejunal epithelial (IPEC-J2) cells. BT at 0.5 mM or higher concentrations significantly impaired cell viability in a dose- and time-dependent manner. In addition, BT at high concentrations inhibited the IPEC-J2 cell proliferation and induced cell cycle arrest in the G2/M phase. Results: Our results demonstrated that BT triggered IPEC-J2 cell apoptosis via the caspase8-caspase3 pathway accompanied by excess reactive oxygen species (ROS) and TNF-α production. BT at high concentrations inhibited cell autophagy associated with increased lysosome formation. It was found that BT-reduced IPEC-J2 cell viability could be attenuated by p38 MAPK inhibitor SB202190. Moreover, SB202190 attenuated BT-increased p38 MAPK target DDIT3 mRNA level and V-ATPase mRNA level that were responsible for normal acidic lysosomes. Conclusion: In conclusion, 1) at 0.2-0.4 mM, BT promotes the differentiation of IPEC-J2 cells; 2) BT at 0.5 mM or higher concentrations induces cell apoptosis via the p38 MAPK pathway; 3) BT inhibits cells autophagy and promotes lysosome formation at high concentrations.


2020 ◽  
Vol 20 (16) ◽  
pp. 1966-1980
Author(s):  
Jaleh Varshosaz ◽  
Saeedeh Fardshouraki ◽  
Mina Mirian ◽  
Leila Safaeian ◽  
Setareh Jandaghian ◽  
...  

Background: Using imatinib, a tyrosine kinase inhibitor drug used in lymphoblastic leukemia, has always had limitations due to its cardiotoxicity and hepatotoxicity side effects. The objective of this study is to develop a target-oriented drug carrier to minimize these adverse effects by the controlled release of the drug. Methods: KIT-5 nanoparticles were functionalized with 3-aminopropyltriethoxysilane and conjugated to rituximab as the targeting agent for the CD20 positive receptors of the B-cells. Then they were loaded with imatinib and their physical properties were characterized. The cell cytotoxicity of the nanoparticles was studied by MTT assay in Ramos (CD20 positive) and Jurkat cell lines (CD20 negative) and their cellular uptake was shown by fluorescence microscope. Wistar rats received an intraperitoneal injection of 50 mg/kg of the free drug or targeted nanoparticles for 21 days. Then the level of aspartate Aminotransferase (AST), alanine Aminotransferase (ALT), Alkaline Phosphatase (ALP) and Lactate Dehydrogenase (LDH) were measured in serum of animals. The cardiotoxicity and hepatotoxicity of the drug were also studied by hematoxylin and eosin staining of the tissues. Results: The targeted nanoparticles of imatinib showed to be more cytotoxic to Ramos cells rather than Jurkat cells. The results of the biochemical analysis displayed a significant reduction in AST, ALT, ALP, and LDH levels in animals treated with targeted nanoparticles, compared to the free drug group. By comparison with the free imatinib, histopathological results represented less cardiotoxicity and hepatotoxicity in the animals, which received the drug through the current designed delivery system. Conclusion: The obtained results confirmed that the rituximab targeted KIT-5 nanoparticles are promising in the controlled release of imatinib and could decrease its cardiotoxicity and hepatotoxicity side effects.


Sign in / Sign up

Export Citation Format

Share Document