14 research outputs found

    Viridiflorol Induces Anti-Neoplastic Effects on Breast, Lung, and Brain Cancer Cells Through Apoptosis

    Get PDF
    All active natural molecules are not fully exploited as therapeutic agents, causing delays in the advancement of anticancer drug discovery. Viridiflorol is a natural volatile element that may work as anti-cancer compound. We tested the anticancer properties of viridiflorol at different concentrations ranging from 0.03 to 300 Ī¼M in vitro on three cancer cells including breast (MCF-7), lung (A549) and brain (Daoy). The cancer cells responses were documented after treatment using MTT and Annexin V assays. Viridiflorol showed cytotoxic effects against all tested cell lines, reducing cell viability in a concentration-dependent manner with variable IC50 values. Daoy and A549 cell lines were more sensitive to viridiflorol when compared with temozolomide and doxorubicin, respectively. Viridiflorol demonstrated the highest anticancer activity against the Daoy cells with an estimated IC50 of 0.1 ĀµM followed by MCF-7 at 10 ĀµM, and A549 at 30 ĀµM. In addition, upon exposure to concentrations ranging from 30 ĀµM to 300 ĀµM of viridiflorol, early and late apoptotic cell death was induced in a concentration dependent manner in Daoy (55.8%-72.1%), MCF-7 (36.2%-72.7%) and A459 (35%-98.9%) cell lines, respectively. In conclusion, viridiflorol demonstrates cytotoxic and apoptotic ability in three different cancer cell lines (brain, breast and lung)

    The loss of microglia activities facilitates glaucoma progression in association with CYP1B1 gene mutation (p.Gly61Glu).

    No full text
    BackgroundGlaucoma represents the second main cause of irreversible loss of eyesight worldwide. Progression of the disease is due to changes around the optic nerve, eye structure and optic nerve environment. Focusing on primary congenital glaucoma, which is not completely understood, we report an evaluation of an untested mutation (c.182G>A, p.Gly61Glu) within the CYP1B1 gene in the context of microglia, astrocytes and mesenchymal stem cells. We investigated the behaviours of these cells, which are needed to maintain eye homeostasis, in response to the CYP1B1 mutation.Methods and resultsCRISPR technology was used to edit normal CYP1B1 genes within normal astrocytes, microglia and stem cells in vitro. Increased metabolic activities were found in microglia and astrocytes 24 hours after CYP1B1 manipulation. However, these activities dropped by 40% after 72 hrs. In addition, the nicotinamide adenine dinucleotide phosphate (NADP)/NADPH reducing equivalent process decreased by 50% on average after 72 hrs of manipulation. The cytokines measured in mutated microglia showed progressive activation leading to apoptosis, which was confirmed with annexin-V. The cytokines evaluated in mutant astrocytes were abnormal in comparison to those in the control.ConclusionsThe results suggest a progressive inflammation that was induced by mutations (p.Gly61Glu) on CYP1B1. Furthermore, the mutations enhanced the microglia's loss of activity. We are the first to show the direct impact of the mutation on microglia. This progressive inflammation might be responsible for primary congenital glaucoma complications, which could be avoided via an anti-inflammatory regimen. This finding also reveals that progressive inflammation affects recovery failure after surgeries to relieve glaucoma. Moreover, microglia are important for the survival of ganglion cells, along with the clearing of pathogens and inflammation. The reduction of their activities may jeopardise homeostasis within the optic nerve environment and complicate the protection of optic nerve components (such as retinal ganglion and glial cells)

    miR-100 decreases tumor size.

    No full text
    <div><p>(A) Treatment of 22T GBM cells with a miR-100 expressing vector (V<sub>miR-100</sub>) increased miR-100 levels by 3-fold compared to control vector (V<sub>cont</sub>) treated cells. Statistics: a is p < 0.05 compared to astrocyte control and b is p < 0.05 compared to V<sub>cont</sub>. </p> <p>(B) The size of the xenograft (black arrow) is larger in the V<sub>cont</sub>. group compared to V<sub>miR-100</sub> group when brains were harvested at death or when animals were moribund post tumor implantation. Dorsal surfaces of brains and arrows to the xenografts are shown in both images. (C) A1-B1, brain section of animal from panel (B) stained with haematoxylin and eosin (H&E). A2-B2, tissue sections were immuno-labeled with SMRT/NCOR2 (brown staining). In xenografts, more SMRT/NCOR2 was seen in control (V<sub>cont</sub>) brain than V<sub>miR-100</sub> brain. A3-B3, No primary antibody control (D) Quantitation of the sections from C shows more SMRT/NCOR2 positive cells in the V<sub>cont</sub>. Group over the V<sub>miR-100</sub> group. (E) A1-B1, brain section of animal from panel (B) stained with H&E. A2-B2, tissue sections were immuno-stained with Ki-67 (brown staining). More Ki-67 staining was seen in control (V<sub>cont</sub>) brain than V<sub>miR-100</sub> brain.(F) Quantitation of Ki-67 positive microscopic fields show more staining in control brains than miR-100 one. A3-B3, tissue sections were immuno-stained with no Ki-67 primary antibody. (Ł€Ł€Ł€) Scale bar, 50 um.</p></div

    microRNA-100 Targets SMRT/NCOR2, Reduces Proliferation, and Improves Survival in Glioblastoma Animal Models

    Get PDF
    <div><p>Glioblastoma (GBM) is the most frequently diagnosed malignant human glioma, and current median patient survival is less than two years despite maximal surgery followed by temozolomide chemoradiation therapies. Novel microRNA-related therapies are now being developed for cancers such as GBM. Differential microRNA expression profiling revealed that miR-100 expression is down-regulated in GBM compared to normal controls. We report that miR-100 expression reduces GBM tumorigenicity. In vitro, four GBM lines (U87, U251, 22T, and 33T) demonstrated reduced proliferation 24 hours after transient miR100 overexpression via transfection. miR-100 triggered cell death an average 70% more than scrambled miR controls 24 hours after transient transfection (p < 0.01). miR-100 targeted inhibition of the ā€œsilencing mediator of retinoid or thyroid hormone receptor-2ā€ (SMRT/NCOR2) gene was confirmed via reporter assays. Ki67 proliferation index was decreased 40% in tumor xenografts generated from stable miR-100 transfected GBM lines versus controls (p < 0.01). Furthermore, treatment of tumor xenografts with a single pre-mir-100 injection (60 pmol) significantly extended survival of mice bearing intracranial GBM xenografts 25% more than scrambled controls (p < 0.01; n=8). These studies establish miR-100ā€™s effect on tumor GBM growth, and suggest clinical potential for microRNA-related GBM therapy.</p> </div

    miR-100 decreases GBM proliferation and targets SMRT/NCOR2.

    No full text
    <p>(A) Quantitative PCR showed native miR-100 expression in multiple GBM tumor lines relative to normal control (astrocyte extracted from non-tumor brain). All GBM lines showed significantly lower miR-100 expression. (B) miR-100 overexpression decreased GBM cell growth compared to control miR transfected group. Control scrambled miRs did not affect GBM cell growth compared to miR-100. (C) miR-100 overexpression decreased GBM proliferation up to 45% compared to control miR group. (D) Transient luciferase reporter assay with either wild-type or mutated SMRT/NCOR2 3ā€™UTR vectors. Both vectors were co-transfected with pre-miR-100 or control miRs. The normal bar represents transient luciferase reporter expression with no further treatment. (E) Western blot quantitation of SMRT/NCOR2 protein level after miR-100 expression. SMRT/NCOR2 level was reduced by transfection with either pre-miR-100 or SMRT/NCOR2 siRNAs (siSMRT/NCOR2) compared to control miR transfected group. (N=3; p Ė‚ 0.05). (F) EdU proliferation assay shows miR-100 overexpression along with SMRT overexpression (pSMRT) prevented proliferation inhibition reported in panel (C). </p

    SMRT rescued tumor cells and avoids decreased cell death.

    No full text
    <p>(A) Treatment with both pre-miR-100 and SMRT/NCOR2 (pSMRT/NCOR2) overexpression prevented avoided induced cell death as shown by TUNEL staining compared to pre-miR-100 alone. The control cells were treated with control miR (Ł€Ł€Ł€) Scale bar, 50 um. (B) Quantitative analysis show marked decrease of TUNEL-positive cells in GBMs transfected with both SMRT/NCOR2 and pre-miR-100 similar to the level of control miR treated group.</p

    miR-100 induces cell death

    No full text
    <p>(A) Treatment with either pre-miR-100 or SMRT/NCOR2 siRNA (siSMRT/NCOR2) induced cell death as shown by TUNEL staining. The control cells were treated with control miR (Ł€Ł€Ł€) Scale bar, 200 um. (B) Quantitative analysis show marked increase of TUNEL-positive cells in GBMs transfected with either siSMRT/NCOR2 or pre-miR-100 compared control miR treated group. </p

    Deficiency in the Treatment Description of mTOR Inhibitor Resistance in Medulloblastoma, a Systematic Review

    No full text
    Medulloblastoma is a common fatal pediatric brain tumor. More treatment options are required to prolong survival and decrease disability. mTOR proteins play an essential role in the disease pathogenesis, and are an essential target for therapy. Three generations of mTOR inhibitors have been developed and are clinically used for immunosuppression and chemotherapy for multiple cancers. Only a few mTOR inhibitors have been investigated for the treatment of medulloblastoma and other pediatric tumors. The first-generation mTOR, sirolimus, temsirolimus, and everolimus, went through phase I clinical trials. The second-generation mTOR, AZD8055 and sapanisertib, suppressed medulloblastoma cell growth; however, limited studies have investigated possible resistance pathways. No clinical trials have been found to treat medulloblastoma using third-generation mTOR inhibitors. This systematic review highlights the mechanisms of resistance of mTOR inhibitors in medulloblastoma and includes IDO1, T cells, Mnk2, and eIF4E, as they prolong malignant cell survival. The findings promote the importance of combination therapy in medulloblastoma due to its highly resistant nature

    ONECUT2 regulates proliferation and apoptosis in glioblastoma cell lines

    No full text
    Glioblastoma multiforme (GBM) is a highly aggressive brain tumor associated with a high mortality rate, with an average survival time of less than two years. GBM treatment faces significant challenges due to its infiltrative nature, genetic diversity, protection by the bloodā€“brain barrier (BBB), drug resistance, and post-treatment side effects. Transcription factors (TFs) play a crucial role in regulating gene expression during cancer initiation and progression. This study aimed to investigate the impact of altering the function of ONECUT-2 (OC-2) in GBM cells, focusing on metabolic activity, proliferation, cell cycle, and apoptosis. To confirm the successful reduction of OC-2 expression in U251 and U87 cells compared to the control cells (wild types; WT), quantitative real-time polymerase chain reaction (qPCR) was performed. The downregulation of OC-2 resulted in a significant decrease in metabolic activity (MTT) of U251 cells by 47Ā % (PĀ =Ā 0.0056) and U87 cells by 36.4Ā % (PĀ =Ā 0.0003) compared to WT cells. In U251 cells, OC-2 downregulation caused cell cycle arrest in both the G0/G1 phase (13.6Ā %) and the S/G2 phase (52.52Ā %) compared to WT cells. Similarly, in U87 cells, downregulation of OC-2 led to cell cycle arrest in the G0/G1 phase (90.5Ā %) and the G2 phase (76.61Ā %) compared to WT cells. Furthermore, OC-2 downregulation significantly affected early-stage apoptosis in U251 cells (95.46Ā %, PĀ =Ā 0.0005) and U87 cells (19.64Ā %, PĀ =Ā 0.0004). Conversely, upregulation of OC-2 significantly increased the metabolic activity (MTT) of U251 cells by 94Ā % (PĀ =Ā 0.0067) and U87 cells by 58Ā % (PĀ =Ā 0.0028) compared to WT cells. These findings highlight the essential role of OC-2 in regulating the progression of GBM cells. Consequently, OC-2 represents a potential therapeutic target for inhibiting GBM. Overall, this study demonstrates that OC-2 plays a significant role in cell progression, suggesting its potential as a therapeutic target for GBM treatment

    The role of TAOK3 in cancer progression and development as a prognostic marker: A pan-cancer analysis study

    No full text
    The protein kinase TAOK3, belongs to the MAP kinase family, is one of three closely related members, namely TAOK1, TAOK2, and TAOK3. We performed a pan-cancer investigation of TAOK3 across different cancer types, including uterine carcinosarcoma, adenocarcinoma of the stomach and pancreas, and endometrial carcinoma of the uterus, to better understand TAOK3ā€²s role in cancer. In at least 16 types of cancer, our findings indicate that TAOK3 expression levels differ considerably between normal and tumor tissues. In addition, our study is the first to identify the oncogenic role of TAOK3 locus S331 and S471 in renal clear cell carcinoma, Glioblastoma Multiforme, hepatocellular carcinoma, Lung adenocarcinoma, and Pancreatic adenocarcinoma, indicating their involvement in cancer progression. In addition, our data analysis indicates that copy number variation is the most prevalent form of mutation in the TAOK3 gene, and that there is a negative correlation between TAOK3 mRNA and DNA promoter methylation. Moreover, our analysis suggests that TAOK3 may serve as a prognostic marker for several kinds of cancer, including Colon adenocarcinoma, renal clear cell carcinoma, Lower Grade Glioma, Lung adenocarcinoma, Mesothelioma, and hepatocellular carcinoma. In addition, our research on signature cancer genes has uncovered a positive association between TAOK3 and SMAD2, SMAD4, and RNF168 in most of the malignancies we have examined. TAOK3 is also correlated with the frequency of mutations and microsatellite instability in four types of cancer. Numerous immune-related genes are closely associated with TAOK3 levels in numerous malignancies. TAOK3 expression is positively correlated with immune infiltrates, which include activated CD4 T cells, CD8 T cells, and type 2T helper cells. Our pan-cancer analysis of TAOK3 provides vital insight into its potential role across a variety of cancer types
    corecore