Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
1.
Int J Mol Sci ; 24(5)2023 Feb 24.
Article in English | MEDLINE | ID: mdl-36901935

ABSTRACT

Oral squamous cell carcinoma (OSCC) is the sixth most common type of cancer worldwide. Despite advancement in treatment, advanced-stage OSCC is associated with poor prognosis and high mortality. The present study aimed to investigate the anticancer activities of semilicoisoflavone B (SFB), which is a natural phenolic compound isolated from Glycyrrhiza species. The results revealed that SFB reduces OSCC cell viability by targeting cell cycle and apoptosis. The compound caused cell cycle arrest at the G2/M phase and downregulated the expressions of cell cycle regulators including cyclin A and cyclin-dependent kinase (CDK) 2, 6, and 4. Moreover, SFB induced apoptosis by activating poly-ADP-ribose polymerase (PARP) and caspases 3, 8, and 9. It increased the expressions of pro-apoptotic proteins Bax and Bak, reduced the expressions of anti-apoptotic proteins Bcl-2 and Bcl-xL, and increased the expressions of the death receptor pathway protein Fas cell surface death receptor (FAS), Fas-associated death domain protein (FADD), and TNFR1-associated death domain protein (TRADD). SFB was found to mediate oral cancer cell apoptosis by increasing reactive oxygen species (ROS) production. The treatment of the cells with N-acetyl cysteine (NAC) caused a reduction in pro-apoptotic potential of SFB. Regarding upstream signaling, SFB reduced the phosphorylation of AKT, ERK1/2, p38, and JNK1/2 and suppressed the activation of Ras, Raf, and MEK. The human apoptosis array conducted in the study identified that SFB downregulated survivin expression to induce oral cancer cell apoptosis. Taken together, the study identifies SFB as a potent anticancer agent that might be used clinically to manage human OSCC.


Subject(s)
Carcinoma, Squamous Cell , Mouth Neoplasms , Humans , Apoptosis/drug effects , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Proliferation , Mitogen-Activated Protein Kinase Kinases , Mouth Neoplasms/drug therapy , Mouth Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , ras Proteins/drug effects , ras Proteins/metabolism , Proto-Oncogene Proteins c-raf/drug effects , Proto-Oncogene Proteins c-raf/metabolism
2.
Bioorg Chem ; 130: 106234, 2023 01.
Article in English | MEDLINE | ID: mdl-36375353

ABSTRACT

Ras protein has been considered a fascinating target for anticancer therapy because its malfunction is closely related to cancer. However, Ras has been considered undruggable because of the failure to regulate its malfunction by controlling the Ras activation mechanism. Recently, Lumakras targeting the G12C mutation was approved, and therapeutic interest in Ras for anticancer therapy has been rejuvenated. Here, we present a series of compounds that inhibit Ras via a unique mechanism of action that exploits the relationship between the Wnt/ß-catenin pathway and Ras. KYA1797K (1) binds to axin to stabilize the ß-catenin destruction complex that causes the phosphorylation and subsequent degradation of Ras, similar to canonical ß-catenin regulation. Based on the chemical structure of 1, we performed a structural optimization and identified 3-(2-hydroxyethyl)-5-((6-(4-nitrophenyl)pyridin-2-yl)methylene)thiazolidine-2,4-dione (13d) as the most potent compound. 13d displayed antitumor effects in a colorectal cancer model with enhanced inhibition activity on Ras. The results of this study suggest that the further development of 13d could contribute to the development of Ras inhibitors with novel mechanisms of action.


Subject(s)
Colorectal Neoplasms , beta Catenin , ras Proteins , Humans , Axin Protein/chemistry , Axin Protein/genetics , Axin Protein/metabolism , beta Catenin/chemistry , beta Catenin/drug effects , Colorectal Neoplasms/drug therapy , ras Proteins/drug effects , ras Proteins/metabolism , Wnt Signaling Pathway
3.
Dis Model Mech ; 15(2)2022 02 01.
Article in English | MEDLINE | ID: mdl-35244677

ABSTRACT

Several cancers and rare genetic diseases are caused by dysregulation in the RAS signaling pathway. RAS proteins serve as molecular switches that regulate pathways involved in cellular growth, differentiation and survival. These pathways have been an intense area of investigation for four decades, since the initial identification of somatic RAS mutations linked to human cancers. In the past few years, inhibitors against several RAS effectors, as well as direct inhibitors of the K-RAS mutant G12C, have been developed. This Special Issue in DMM includes original Research articles on RAS-driven cancers and RASopathies. The articles provide insights into mechanisms and biomarkers, and evaluate therapeutic targets. Several articles also present new disease models, whereas others describe technologies or approaches to evaluate the function of RAS in vivo. The collection also includes a series of Review articles on RAS biology and translational aspects of defining and treating RAS-driven diseases. In this Editorial, we summarize this collection and discuss the potential impact of the articles within this evolving area of research. We also identify areas of growth and possible future developments.


Subject(s)
Neoplasms , ras Proteins , Antineoplastic Agents/pharmacology , Humans , Mutation/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Protein Processing, Post-Translational , Signal Transduction/genetics , ras Proteins/drug effects , ras Proteins/genetics , ras Proteins/metabolism
4.
Biomolecules ; 11(6)2021 06 16.
Article in English | MEDLINE | ID: mdl-34208655

ABSTRACT

The RTK-RAS-MAPK axis is one of the most extensively studied signaling cascades and is related to the development of both cancers and RASopathies. In the last 30 years, many ideas and approaches have emerged for directly targeting constituent members of this cascade, predominantly in the context of cancer treatment. These approaches are still insufficient due to undesirable drug toxicity, resistance, and low efficacy. Significant advances have been made in understanding the spatiotemporal features of the constituent members of the RTK-RAS-MAPK axis, which are linked and modulated by many accessory proteins. Given that the majority of such modulators are now emerging as attractive therapeutic targets, a very small number of accessory inhibitors have yet to be discovered.


Subject(s)
Mitogen-Activated Protein Kinases/drug effects , Receptor Protein-Tyrosine Kinases/drug effects , ras Proteins/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Humans , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , ras Proteins/metabolism
5.
Aging (Albany NY) ; 13(9): 13333-13348, 2021 05 12.
Article in English | MEDLINE | ID: mdl-33982674

ABSTRACT

This study aimed to explore effects of Sevoflurane on ischemia-reperfusion (I/R) injury after total knee arthroplasty (TKA). To explore potential molecular mechanism, Ras related dexamethasone induced 1 (RASD1), a Protein kinase A (PKA) activator, frequently associated with various models of I/R injury, was also investigated. In vivo mouse models with I/R injury after TKA and in vitro cell models with I/R injury were induced. Contents of creatinine kinase (CK), lactic dehydrogenase (LDH), superoxide dismutase (SOD), and malondialdehyde (MDA), serum levels of inflammatory factors, expression of PKA pathway-related genes and cell proliferation and apoptosis were measured. RASD1 was altered and PKA pathway was inhibited in mice and cells to elucidate the involvement of RASD1 and PKA pathway in Sevoflurane treatment on I/R injury. RASD1 was upregulated in I/R injury after TKA. Sevoflurane treatment or silencing RASD1 reduced RASD1 expression, CK, LDH and MDA contents, inflammation, apoptosis, but increased proliferation, SOD content, cAMP expression, and extents of PKA and cAMP responsive element binding protein (CREB) phosphorylation in skeletal muscle cells of I/R injury. Additionally, PKA pathway activation potentiated the therapeutic effect of Sevoflurane on I/R injury after TKA. Altogether, Sevoflurane treatment confines I/R injury after TKA via RASD1-mediated PKA pathway activation.


Subject(s)
Cell Proliferation/drug effects , Reperfusion Injury/drug therapy , Sevoflurane/pharmacology , ras Proteins/drug effects , Animals , Apoptosis/drug effects , Arthroplasty, Replacement, Knee/methods , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Mice , Protective Agents/pharmacology , Reperfusion Injury/metabolism
6.
Int J Neurosci ; 131(10): 975-983, 2021 Oct.
Article in English | MEDLINE | ID: mdl-32378973

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is the most common aggressive primary cancer occurring in the brain tissue. GBM accounts 16% of primary brain tumors and half of gliomas. Additionally, the incidence of GBM is increases with aging, and reaches the peak at the age of 75 to 84 years. The survival of patients with GBM remains at a low level, only less than 5% patients diagnosed with GBM survive for 5 years. Temozolomide (TMZ) is a DNA alkylating agent and is currently a first line chemotherapeutic treatment for GBM. TMZ combined with radiation therapy has been shown to prolong the overall survival (OS) to 14.6 months compared with 12.1 months for radiation therapy alone. NF-E2-related factor 2 (Nrf2) is a transcription factor that contains seven functional domains. The binding of Keap1 to Nrf2 is a central regulator of the cellular defense mechanism against environmental stresses. METHODS: First, Nrf2 overexpression and inhibition models were constructed in U251 cells using transfection. The percentage of viable cells was detected using the MTT assay. Then, the expression of the HO-1 regulator was detected using qPCR, and the concentrations of oxidative stress related factors were detected using ELISAs. The levels of proteins related to oxidative stress and the Ras/Raf/MEK signaling pathway was detected using western blotting analysis. RESULTS: We initially established Nrf2 inhibition and activation cell models in U251 cells and found that the inhibition of Nrf2 expression decreased the mRNA and protein levels of the anti-oxidative enzymes, as well as the secretion of these enzymes into the cellular microenvironment. These effects might be mediated by the inhibition of Ras/Raf/MEK signaling pathway, leading to the inhibition of cellular proliferation. CONCLUSIONS: Inhibition of Nrf2 expression might enhance the effect of TMZ on the treatment of GBM and might be a new therapeutic strategy.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Glioma/drug therapy , MAP Kinase Kinase Kinases/drug effects , NF-E2-Related Factor 2/drug effects , Temozolomide/pharmacology , raf Kinases/drug effects , ras Proteins/drug effects , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , NF-E2-Related Factor 2/antagonists & inhibitors , Signal Transduction/drug effects , raf Kinases/antagonists & inhibitors , ras Proteins/antagonists & inhibitors
7.
Clin Epigenetics ; 12(1): 76, 2020 06 03.
Article in English | MEDLINE | ID: mdl-32493461

ABSTRACT

BACKGROUND: A long-term opioid use has been associated with hypermethylation of the opioid receptor mu 1 (OPRM1) promoter. Very little is currently known about the early epigenetic response to therapeutic opioids. Here, we examine whether we can detect DNA methylation changes associated with a few days' use of prescribed opioids. Genome-wide DNA methylation was assayed in a cohort of 33 opioid-naïve participants who underwent standard dental surgery followed by opioid self-administration. Saliva samples were collected before surgery (visit 1), and at two postsurgery visits at 2.7 ± 1.5 days (visit 2), and 39 ± 10 days (visit 3) after the discontinuation of opioid analgesics. RESULTS: The perioperative methylome underwent significant changes over the three visits that were primarily due to postoperative inflammatory response and cell heterogeneity. To specifically examine the effect of opioids, we started with a candidate gene approach and evaluated 10 CpGs located in the OPRM1 promoter. There was a significant cross-sectional variability in opioid use, and for participants who self-administered the prescribed drugs, the total dosage ranged from 5-210 morphine milligram equivalent (MME). Participants were categorized by cumulative dosage into three groups: < 25 MME, 25-90 MME, and ≥ 90 MME. Using mixed-effects modeling, 4 CpGs had significant positive associations with opioid dose at two-tailed p value < 0.05, and overall, 9 of the 10 OPRM1 promoter CpGs showed the predicted higher methylation in the higher dose groups relative to the lowest dose group. After adjustment for age, cellular heterogeneity, and past tobacco use, the promoter mean methylation also had positive associations with cumulative MME (regression coefficient = 0.0002, one-tailed p value = 0.02) and duration of opioid use (regression coefficient = 0.003, one-tailed p value = 0.001), but this effect was significant only for visit 3. A preliminary epigenome-wide association study identified a significant CpG in the promoter of the RAS-related signaling gene, RASL10A, that may be predictive of opioid dosage. CONCLUSION: The present study provides evidence that the hypermethylation of the OPRM1 promoter is in response to opioid use and that epigenetic differences in OPRM1 and other sites are associated with a short-term use of therapeutic opioids.


Subject(s)
Analgesics, Opioid/pharmacology , DNA Methylation/drug effects , Opioid-Related Disorders/genetics , Promoter Regions, Genetic/drug effects , Receptors, Opioid, mu/drug effects , Adult , Analgesics, Opioid/administration & dosage , Case-Control Studies , CpG Islands/genetics , Epigenesis, Genetic , Epigenome/drug effects , Epigenome/genetics , Female , Genome-Wide Association Study/methods , Humans , Male , Middle Aged , Opioid-Related Disorders/metabolism , Perioperative Period , Pharmacogenomic Variants/genetics , Promoter Regions, Genetic/genetics , Receptors, Opioid, mu/metabolism , Saliva/metabolism , ras Proteins/drug effects , ras Proteins/genetics
8.
ACS Chem Biol ; 15(6): 1604-1612, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32378881

ABSTRACT

Protein-protein interactions (PPIs) play a critical role in fundamental biological processes. Competitive inhibition of these interfaces requires compounds that can access discontinuous binding epitopes along a large, shallow binding surface area. Conformationally defined protein surface mimics present a viable route to target these interactions. However, the development of minimal protein mimics that engage intracellular targets with high affinity remains a major challenge because mimicry of a portion of the binding interface is often associated with the loss of critical binding interactions. Covalent targeting provides an attractive approach to overcome the loss of noncovalent contacts but have the inherent risk of dominating noncovalent contacts and increasing the likelihood of nonselective binding. Here, we report the iterative design of a proteolytically stable α3ß chimeric helix mimic that covalently targets oncogenic Ras G12C as a model system. We explored several electrophiles to optimize preferential alkylation with the desired C12 on Ras. The designed lead peptide modulates nucleotide exchange, inhibits activation of the Ras-mediated signaling cascade, and is selectively toxic toward mutant Ras G12C cancer cells. The relatively high frequency of acquired cysteines as missense mutations in cancer and other diseases suggests that covalent peptides may offer an untapped therapeutic approach for targeting aberrant protein interactions.


Subject(s)
Drug Delivery Systems , Drug Design , Peptidomimetics/pharmacology , ras Proteins/drug effects , Biophysical Phenomena , Cell Line, Tumor , Humans , Ligands , Peptidomimetics/chemistry , Protein Conformation , Protein Interaction Maps , Proteolysis , Signal Transduction
9.
Folia Neuropathol ; 58(1): 45-56, 2020.
Article in English | MEDLINE | ID: mdl-32337957

ABSTRACT

Trauma is the main cause of death for people aged 1-45, and among them, traumatic brain injury (TBI) is the major condition, which causes over 50,000 deaths each year and costs over 80 billion per year. Tetrahydroxystilbene glucoside (TSG) is the active ingredient of polygonum multiflorum, a traditional Chinese herbal medicine, which presented multiple pharmacological effects, including antioxidative, anti-inflammatory, reducing blood fat and neuroprotection effects. However, the effect of TSG in promoting the recovery of the nerve system after TBI is not fully understood. PARP1 is a key enzyme in repair of the damage in DNA, which is activated by binding to DNA breaks, initiating both single-strand and double-strand DNA break repair. And we thought that overexpression of TSG might enhance the effect of TSG in TBI treatment. In this study, we firstly detected the oxidative stress response related molecules in serum samples of TBI patients and a TBI mice model, and found that oxidative stress response was activated after TBI, and TSG would reduce this effect. We further noticed that inflammation related molecules presented a similar trend with oxidative stress response related molecules. These results indicated that inflammatory response and oxidative stress processes were both activated after TBI, and reduced after TSG treatment. We further detected that the apoptosis related proteins and anti-oxidative proteins were increased after TSG treatment, and these effects were enlarged after overexpression of PARP1. We further noticed that these effects might be mediated by inhibition of the Ras/JNK signalling pathway. Thus, we thought overexpression of PARP1 might enhance the therapeutic effect of TSG in TBI treatment.


Subject(s)
Brain Injuries, Traumatic/pathology , Glucosides/pharmacology , MAP Kinase Signaling System/drug effects , Poly (ADP-Ribose) Polymerase-1/metabolism , Stilbenes/pharmacology , ras Proteins/drug effects , Adult , Animals , Brain Injuries, Traumatic/metabolism , Humans , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Oxidative Stress/drug effects , Oxidative Stress/physiology , Reactive Oxygen Species/blood , ras Proteins/metabolism
10.
Org Biomol Chem ; 18(16): 3069-3081, 2020 04 29.
Article in English | MEDLINE | ID: mdl-32101243

ABSTRACT

The use of selective covalent inhibitors with low binding affinity and high reactivity with the target enzyme is a promising way to solve a long-standing problem of the "undruggable" RAS-like proteins. Specifically, compounds of the ARS family that prevent the activation of the GDP-bound G12C mutant of Kirsten RAS (KRAS) are in the focus of recent experimental research. We report the first computational characterization of the entire reaction mechanism of the covalent binding of ARS-853 to the KRASG12C·GDP complex. The application of molecular dynamics, molecular docking and quantum mechanics/molecular mechanics approaches allowed us to model the inhibitor binding to the protein and the chemical reaction of ARS-853 with Cys12 in the enzyme binding site. We estimated a full set of kinetic constants and carried out numerical kinetic analysis of the process. Thus, we were able to compare directly the physicochemical parameters of the reaction obtained in silico and the macroscopic parameters observed in experimental studies. From our computational results, we explain the observed unusual dependence of the rate constant of covalent complex formation, kobs, on the ARS concentration. The latter depends both on the non-covalent binding step with the equilibrium constant, Ki, and on the rate constant of covalent adduct formation, kinact. The calculated ratio kinact/Ki = 213 M-1 s-1 reproduces the corresponding experimental value of 250 ± 30 M-1 s-1 for the interaction of ARS-853 with KRASG12C. Electron density analysis in the reactive region demonstrates that covalent bond formation occurs efficiently according to the Michael addition mechanism, which assumes the activation of the C[double bond, length as m-dash]C bond of ARS-853 by a water molecule and Lys16 in the binding site of KRASG12C. We also refine the kinact and Ki constants of the ARS-107 compound, which shares common features with ARS-853, and show that the decrease in the kinact/Ki ratio in the case of ARS-107 is explained by changes in both Ki and kinact constants.


Subject(s)
Azetidines/metabolism , Piperazines/metabolism , ras Proteins/antagonists & inhibitors , Azetidines/pharmacology , Binding Sites , Guanosine Diphosphate/metabolism , Humans , Kinetics , Molecular Docking Simulation , Mutation , Piperazines/pharmacology , Proof of Concept Study , Protein Binding , ras Proteins/drug effects , ras Proteins/genetics , ras Proteins/metabolism
11.
BMC Anesthesiol ; 20(1): 43, 2020 02 21.
Article in English | MEDLINE | ID: mdl-32085741

ABSTRACT

BACKGROUND: Substantial clinical and preclinical evidence have indicated the association between amide-linked local anesthesia and the long-term outcomes of cancer patients. However, the potential effects of local anesthesia on cancer recurrence are inconclusive and the underlying mechanisms remain poorly understood. METHODS: We systematically examined the effects of three commonly used local anesthetics in melanoma cells and analyzed the underlying mechanisms focusing on small GTPases. RESULTS: Ropivacaine and lidocaine but not bupivacaine inhibited migration and proliferation, and induced apoptosis in melanoma cells. In addition, ropivacaine and lidocaine but not bupivacaine significantly augmented the in vitro efficacy of vemurafenib (a B-Raf inhibitor for melanoma with BRAF V600E mutation) and dacarbazine (a chemotherapeutic drug). Mechanistically, ropivacaine but not bupivacaine decreased the activities of Ras superfamily members with the dominant inhibitory effects on RhoA and Ras, independent of sodium channel blockade. Rescue studies using constitutively active Ras and Rho activator calpeptin demonstrated that ropivacaine inhibited migration mainly through RhoA whereas growth and survival were mainly inhibited through Ras in melanoma cells. We further detected a global reduction of downstream signaling of Ras and RhoA in ropivacaine-treated melanoma cells. CONCLUSION: Our study is the first to demonstrate the anti-melanoma activity of ropivacaine and lidocaine but not bupivacaine, via targeting small GTPases. Our findings provide preclinical evidence on how amide-linked local anesthetics could affect melanoma patients.


Subject(s)
Anesthetics, Local/pharmacology , Melanoma/metabolism , Sodium Channel Blockers/metabolism , ras Proteins/drug effects , rhoA GTP-Binding Protein/drug effects , Bupivacaine/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Humans , In Vitro Techniques , Lidocaine/pharmacology , Melanoma/drug therapy , Ropivacaine/pharmacology , Signal Transduction/drug effects , ras Proteins/metabolism , rhoA GTP-Binding Protein/metabolism
12.
Life Sci ; 239: 116916, 2019 Dec 15.
Article in English | MEDLINE | ID: mdl-31626792

ABSTRACT

AIMS: To investigate the effects of sevoflurane on proliferation, cell cycle, apoptosis, autophagy, invasion and epithelial-mesenchymal transition of colon cancer cell line SW480, and to explore its possible mechanism. MATERIALS AND METHODS: SW480 and SW620 cells were treated with a mixture of 95% O2+5% CO2 containing different concentrations of sevoflurane (1.7% SAV, 3.4% SAV and 5.1% SAV) for 6 h. Meanwhile, we performed a rescue experiment by treating cells with the ERK pathway activator LM22B-10 prior to treatment of cells with 5.1% sevoflurane。 KEY FINDINGS: High concentration (5.1%) of sevoflurane significantly inhibited the proliferation and invasion of cells, causing G0/G1 phase arrest and promoted apoptosis and autophagy. 5.1% sevoflurane can participate in the regulation of EMT by regulating the expression of E-cadherin, Vimentin and N-cadherin proteins. LM22B-10 promoted proliferation and invasion of cancer cells and inhibited apoptosis and autophagy, while 5.1% sevoflurane could reverse the effect of LM22B-10 on the biological characteristics of cells. Sevoflurane can significantly inhibit tumor growth in SW480 cells transplanted nude mice. Moreover, 5.1% sevoflurane significantly increased the expression of p-Raf, p-MEK1/2, and p-ERK1/2 in SW480 cells and tumor tissues without affecting p-JNK and p-p38 proteins, meanwhile, 5.1% sevoflurane can inhibit the activation of ERK signaling pathway by LM22B-10 in vitro and in vivo. SIGNIFICANCE: Sevoflurane can inhibit the proliferation and invasion of colon cancer cells, induce apoptosis and autophagy, and participate in the regulation of epithelial-mesenchymal transition, which may be related to its inhibition of the ERK signaling pathway.


Subject(s)
Colonic Neoplasms/metabolism , Sevoflurane/metabolism , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cadherins/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , China , Colonic Neoplasms/physiopathology , Epithelial-Mesenchymal Transition/drug effects , Humans , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Sevoflurane/pharmacology , Signal Transduction/drug effects , raf Kinases/drug effects , ras Proteins/drug effects
13.
Respir Res ; 20(1): 194, 2019 Aug 23.
Article in English | MEDLINE | ID: mdl-31443651

ABSTRACT

BACKGROUND: The aim of this study was to investigate the effects and mechanisms of ectonucleoside triphosphate phosphohydrolase-7 (ENTPD7) on lung cancer cells. METHODS: The expression characteristics of ENTPD7 and its effect on the survival of lung cancer patients were analyzed by referring to The Cancer Genome Atlas (TCGA). Streptavidin-peroxidase (SP) staining was performed to detect the ENTPD7 protein in tumor tissues and adjacent tissues. Plasmid transfection technology was also applied to silence ENTPD7 gene. Crystal violet staining and flow cytometry were performed to determine cell proliferation and apoptosis. Tumor-bearing nude mice model was established to investigate the effect of sh-ENTPD7 on tumors. RESULTS: The results showed that patients with low levels of ENTPD7 had higher survival rates. ENTPD7 was up-regulated in lung cancer tissues and cells. Down-regulation of the expression of ENTPD7 inhibited proliferation but promoted apoptosis of lung cancer cell. Silencing ENTPD7 also inhibited the expression levels of Ras and Raf proteins and the phosphorylation of mitogen-activated protein kinase (MEK) and extracellular signal-regulated kinase (ERK). Tumor-bearing nude mice experiments showed that silencing ENTPD7 had an inhibitory effect on lung cancer cells. CONCLUSIONS: ENTPD7 was overexpressed in lung cancer cells. Down-regulating ENTPD7 could inhibit lung cancer cell proliferation and promote apoptosis via inhibiting the Ras/Raf/MEK/ERK pathway.


Subject(s)
Apyrase/antagonists & inhibitors , Apyrase/genetics , Lung Neoplasms/therapy , Signal Transduction/drug effects , Adult , Aged , Animals , Apoptosis , Biomarkers , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Lung Neoplasms/genetics , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Mitogen-Activated Protein Kinases/drug effects , Plasmids , Survival Analysis , raf Kinases/antagonists & inhibitors , raf Kinases/genetics , ras Proteins/drug effects , ras Proteins/genetics
14.
Cancer Cell ; 34(4): 611-625.e7, 2018 10 08.
Article in English | MEDLINE | ID: mdl-30300582

ABSTRACT

Targeting KRAS mutant tumors through inhibition of individual downstream pathways has had limited clinical success. Here we report that RAF inhibitors exhibit little efficacy in KRAS mutant tumors. In combination drug screens, MEK and PI3K inhibitors synergized with pan-RAF inhibitors through an RAS-GTP-dependent mechanism. Broad cell line profiling with RAF/MEK inhibitor combinations revealed synergistic efficacy in KRAS mutant and wild-type tumors, with KRASG13D mutants exhibiting greater synergy versus KRASG12 mutant tumors. Mechanistic studies demonstrate that MEK inhibition induced RAS-GTP levels, RAF dimerization and RAF kinase activity resulting in MEK phosphorylation in synergistic tumor lines regardless of KRAS status. Taken together, our studies uncover a strategy to rewire KRAS mutant tumors to confer sensitivity to RAF kinase inhibition.


Subject(s)
Phosphatidylinositol 3-Kinases/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins p21(ras)/drug effects , Cell Line, Tumor , Guanosine Triphosphate/metabolism , Humans , Mutation/drug effects , Mutation/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins B-raf/drug effects , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , ras Proteins/drug effects , ras Proteins/genetics
15.
World Neurosurg ; 120: e380-e391, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30144594

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is the most common and fatal human brain tumor, with the worst prognosis. The aberrant microenvironment, enhanced by the activation of proangiogenic mediators such as hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor (VEGF), and their downstream effectors, sustain GBM malignancy. Proangiogenic signaling represents an attractive chemotherapeutic target. Recent evidence suggests a therapeutic benefit from aspirin (acetylsalicylic acid, or ASA) intake in reducing risk and cancer progression. METHODS: In the present study, human primary GBM-endothelial cells (ECs) were used to ascertain whether ASA could inhibit angiogenesis and improve cell sensitivity to drugs. The impact of ASA was observed by measuring cell viability, tube-like structure formation, migration, VEGF production, and proliferative, proangiogenic, and apoptotic modulators expression, such as HIF-1α/VEGF/vascular endothelial growth factor receptor/(VEGFR)-1/VEGFR-2, Ras/mitogen-activated protein kinase kinase/extracellular signal-regulated kinase, phosphoinositide 3-kinase/AKT signaling axis, and Bcl-2-associated X protein/B-cell lymphoma 2 (BCL-2) ratio. Furthermore, we evaluated the effect of ASA alone or in combination with temozolomide (TMZ), bevacizumab (BEV), and sunitinib (SUN). RESULTS: Our data reported that ASA affected GBM-EC viability, tube-like structure formation, cell migration, and VEGF releasing in a dose-dependent manner and that combined treatments with TMZ, BEV, and SUN synergized to counteract proangiogenic cell ability. mRNA expression analysis displayed a marked effect of ASA in reducing VEGF, VEGFR-1, HIF-1α, RAS, mitogen-activated protein kinase kinase, AKT, and BCL-2, as well a combined anticancer effect of ASA together with TMZ, BEV, and SUN. Levels of HIF-1α, VEGFR-2, Bcl-2-associated X protein, and BCL-2 protein expression confirmed a positive trend. CONCLUSIONS: ASA and antiangiogenic therapies showed synergetic anticancer efficacy in human primary GBM-ECs. Thus, the combination of conventional chemotherapy with ASA may offer a new strategy to counteract tumor malignancy.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents/pharmacology , Aspirin/pharmacology , Bevacizumab/pharmacology , Brain Neoplasms/blood supply , Endothelial Cells/drug effects , Glioblastoma/blood supply , Sunitinib/pharmacology , Temozolomide/pharmacology , Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Blotting, Western , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Survival/drug effects , Drug Synergism , Endothelial Cells/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mitogen-Activated Protein Kinase Kinases/drug effects , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-1/drug effects , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/drug effects , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , bcl-2-Associated X Protein/drug effects , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , ras Proteins/drug effects , ras Proteins/genetics , ras Proteins/metabolism
16.
Tumour Biol ; 39(10): 1010428317734947, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28990465

ABSTRACT

Recently, statins have been demonstrated to improve cancer-related mortality or prognosis in patients of various cancers. However, the details of the apoptosis-inducing mechanisms remain unknown. This study showed that the induction of apoptosis by statins in hematopoietic tumor cells is mediated by mitochondrial apoptotic signaling pathways, which are activated by the suppression of mevalonate or geranylgeranyl pyrophosphate biosynthesis. In addition, statins decreased the levels of phosphorylated extracellular signal-regulated kinase 1/2 and mammalian target of rapamycin through suppressing Ras prenylation. Furthermore, inhibition of extracellular signal-regulated kinase 1/2 and mammalian target of rapamycin by statins induced Bim expression via inhibition of Bim phosphorylation and ubiquitination and cell-cycle arrest at G1 phase via enhancement of p27 expression. Moreover, combined treatment of U0126, a mitogen-activated protein kinase kinase 1/2 inhibitor, and rapamycin, a mammalian target of rapamycin inhibitor, induced Bim and p27 expressions. The present results suggested that statins induce apoptosis by decreasing the mitochondrial transmembrane potential, increasing the activation of caspase-9 and caspase-3, enhancing Bim expression, and inducing cell-cycle arrest at G1 phase through inhibition of Ras/extracellular signal-regulated kinase and Ras/mammalian target of rapamycin pathways. Therefore, our findings support the use of statins as potential anticancer agents or concomitant drugs of adjuvant therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Hematologic Neoplasms/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Signal Transduction/drug effects , Bcl-2-Like Protein 11/drug effects , Bcl-2-Like Protein 11/metabolism , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Immunoprecipitation , Proliferating Cell Nuclear Antigen/drug effects , Proliferating Cell Nuclear Antigen/metabolism , ras Proteins/drug effects , ras Proteins/metabolism
17.
Cell Host Microbe ; 21(5): 611-618.e5, 2017 May 10.
Article in English | MEDLINE | ID: mdl-28494242

ABSTRACT

Neutrophils are the first line of defense against bacterial infections, and the generation of reactive oxygen species is a key part of their arsenal. Pathogens use detoxification systems to avoid the bactericidal effects of reactive oxygen species. Here we demonstrate that the Gram-negative pathogen Pseudomonas aeruginosa is susceptible to reactive oxygen species but actively blocks the reactive oxygen species burst using two type III secreted effector proteins, ExoS and ExoT. ExoS ADP-ribosylates Ras and prevents it from interacting with and activating phosphoinositol-3-kinase (PI3K), which is required to stimulate the phagocytic NADPH-oxidase that generates reactive oxygen species. ExoT also affects PI3K signaling via its ADP-ribosyltransferase activity but does not act directly on Ras. A non-ribosylatable version of Ras restores reactive oxygen species production and results in increased bacterial killing. These findings demonstrate that subversion of the host innate immune response requires ExoS-mediated ADP-ribosylation of Ras in neutrophils.


Subject(s)
ADP Ribose Transferases/antagonists & inhibitors , Bacterial Toxins/antagonists & inhibitors , Neutrophils/immunology , Neutrophils/metabolism , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/metabolism , Reactive Oxygen Species/metabolism , ras Proteins/drug effects , ADP Ribose Transferases/metabolism , ADP-Ribosylation/drug effects , Animals , Bacterial Toxins/immunology , Colony Count, Microbial , Epithelium/pathology , Eye/pathology , Female , GTPase-Activating Proteins/antagonists & inhibitors , Humans , Immunity, Innate , Mice, Inbred C57BL , NADPH Oxidases/metabolism , Neutrophils/enzymology , Phagocytosis , Phosphatidylinositol 3-Kinases/metabolism , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/pathogenicity , Signal Transduction/drug effects , Survival Analysis , Type III Secretion Systems/drug effects , ras Proteins/metabolism
18.
Oncotarget ; 8(13): 21754-21769, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28423516

ABSTRACT

Here we showed that the addition of the COX-2 inhibitor celecoxib improved the antitumor efficacy in colorectal cancer (CRC) of the monoclonal anti-EGFR antibody cetuximab. The addition of celecoxib augmented the efficacy of cetuximab to inhibit cell proliferation and to induce apoptosis in CRC cells. Moreover, the combination of celecoxib and cetuximab was more effective than either treatment alone in reducing the tumor volume in a mouse xenograft model. The combined treatment enhanced the inhibition of EGFR signaling and altered the subcellular distribution of ß-catenin. Moreover, knockdown of FOXM1 showed that this transcription factor participates in this enhanced antitumoral response. Besides, the combined treatment decreased ß-catenin/FOXM1 interaction and reduced the cancer stem cell subpopulation in CRC cells, as indicated their diminished capacity to form colonospheres. Notably, the inmunodetection of FOXM1 in the nuclei of tumor cells in human colorectal adenocarcinomas was significantly associated with response of patients to cetuximab. In summary, our study shows that the addition of celecoxib enhances the antitumor efficacy of cetuximab in CRC due to impairment of EGFR-RAS-FOXM1-ß-catenin signaling axis. Results also support that FOXM1 could be a predictive marker of response of mCRC patients to cetuximab therapy.


Subject(s)
Adenocarcinoma/pathology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Celecoxib/pharmacology , Cetuximab/pharmacology , Colorectal Neoplasms/pathology , Signal Transduction/drug effects , Animals , Blotting, Western , Drug Synergism , ErbB Receptors/drug effects , ErbB Receptors/metabolism , Fluorescent Antibody Technique , Forkhead Box Protein M1/drug effects , Forkhead Box Protein M1/metabolism , Humans , Immunohistochemistry , Mice , Mice, Inbred NOD , Mice, SCID , Microscopy, Confocal , Xenograft Model Antitumor Assays , beta Catenin/drug effects , beta Catenin/metabolism , ras Proteins/drug effects , ras Proteins/metabolism
19.
Asia Pac J Clin Oncol ; 13(2): e132-e137, 2017 Apr.
Article in English | MEDLINE | ID: mdl-26245851

ABSTRACT

AIM: Cetuximab improves the prognosis for wild-type KRAS metastatic colorectal cancer (MCRC). We evaluated the safety and efficacy of cetuximab in combination with irinotecan in Japanese patients with wild-type KRAS MCRC refractory to irinotecan, oxaliplatin and fluoropyrimidines. METHODS: Cetuximab was administered initially at a dose of 400 mg/m2 , followed by weekly infusions at 250 mg/m2 . Irinotecan was administered every 2 weeks at 150 mg/m2 . Primary endpoint was the incidence of grade 3/4 adverse events; secondary endpoints included overall survival (OS), progression-free survival (PFS), response rate (RR), time to treatment failure (TTF), and TTF for irinotecan. RESULTS: Thirty-four patients were enrolled. Grade 3 or 4 toxicities were leucopenia (11.8%), neutropenia (23.5%), anemia (11.8%), fatigue (2.9%), anorexia (2.9%), diarrhea (14.7%) and hypomagnesemia (5.9%). Skin toxicities were as follows (any grade/grade 3): acne (94.2/8.8%), rash (55.9/0%), nail changes (75.5/8.8%) and hand-foot syndrome (55.9/5.9%). Median PFS was 6.0 months (95%CI; 4.7-7.4). Median OS was 12.9 months (95%CI; 10.0-15.9). RR was 26.4%. Median TTF was 5.1 months and median TTF for irinotecan was 5.0 months (95%CI; 4.3-5.6). CONCLUSION: Cetuximab with irinotecan therapy was well tolerated in Japanese patients with wild-type KRAS colorectal cancer refractory to irinotecan, oxaliplatin and fluoropyrimidine, thus demonstrating the feasibility of their usage.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/analogs & derivatives , Cetuximab/therapeutic use , Colorectal Neoplasms/drug therapy , ras Proteins/drug effects , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Asian People , Camptothecin/administration & dosage , Camptothecin/pharmacology , Camptothecin/therapeutic use , Cetuximab/administration & dosage , Cetuximab/pharmacology , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Female , Humans , Irinotecan , Male , Middle Aged , Prognosis , Survival Rate , Treatment Outcome
20.
Mutagenesis ; 32(1): 77-90, 2017 01.
Article in English | MEDLINE | ID: mdl-28011749

ABSTRACT

We used the marine bivalve (Mytilus galloprovincialis) to assess a range of biological or biomarker responses following exposure to a model-engineered nanoparticle, C60 fullerene, either alone or in combination with a model polycyclic aromatic hydrocarbon, benzo(α)pyrene [B(α)P]. An integrated biomarker approach was used that included: (i) determination of 'clearance rates' (a physiological indicator at individual level), (ii) histopathological alterations (at tissue level), (iii) DNA strand breaks using the comet assay (at cellular level) and (iv) transcriptional alterations of p53 (anti-oncogene) and ras (oncogene) determined by real-time quantitative polymerase chain reaction (at the molecular/genetic level). In addition, total glutathione in the digestive gland was measured as a proxy for oxidative stress. Here, we report that mussels showed no significant changes in 'clearance rates' after 1 day exposure, however significant increases in 'clearance rates' were found following exposure for 3 days. Histopathology on selected organs (i.e. gills, digestive glands, adductor muscles and mantles) showed increased occurrence of abnormalities in all tissues types, although not all the exposed organisms showed these abnormalities. Significantly, increased levels of DNA strand breaks were found after exposure for 3-days in most individuals tested. In addition, a significant induction for p53 and ras expression was observed in a tissue and chemical-specific pattern, although large amounts of inter-individual variability, compared with other biomarkers, were clearly apparent. Overall, biological responses at different levels showed variable sensitivity, with DNA strand breaks and gene expression alterations exhibiting higher sensitivities. Furthermore, the observed genotoxic responses were reversible after a recovery period, suggesting the ability of mussels to cope with the toxicants C60 and/or B(α)P under our experimental conditions. Overall, in this comprehensive study, we have demonstrated mussels as a suitable model marine invertebrate species to study the potential detrimental effects induced by possible genotoxicants and toxicants, either alone or in combinations at different levels of biological organisation (i.e. molecular to individual levels).


Subject(s)
Bivalvia/drug effects , DNA Damage , Fullerenes/toxicity , Tumor Suppressor Protein p53/drug effects , ras Proteins/drug effects , Animals , Benzo(a)pyrene/pharmacology , Benzo(a)pyrene/toxicity , Bivalvia/genetics , Bivalvia/metabolism , Comet Assay , DNA/drug effects , Fullerenes/pharmacology , Gene Expression Regulation , Glutathione/analysis , Glutathione/drug effects , Models, Animal , Organ Specificity , Oxidative Stress/drug effects , Tumor Suppressor Protein p53/genetics , ras Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...