Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Int J Mol Sci ; 25(3)2024 Jan 24.
Article in English | MEDLINE | ID: mdl-38338709

ABSTRACT

Oncogenic Ras proteins are known to present multiple conformational states, as reported by the great variety of crystallographic structures. The GTP-bound states are grouped into two main states: the "inactive" state 1 and the "active" state 2. Recent reports on H-Ras have shown that state 2 exhibits two substates, directly related to the orientation of Tyr32: toward the GTP-bound pocket and outwards. In this paper, we show that N-Ras exhibits another substate of state 2, related to a third orientation of Tyr32, toward Ala18 and parallel to the GTP-bound pocket. We also show that this substate is highly sampled in the G12V mutation of N-Ras and barely present in its wild-type form, and that the G12V mutation prohibits the sampling of the GTPase-activating protein (GAP) binding substate, rendering this mutation oncogenic. Furthermore, using molecular dynamics simulations, we explore the importance of the membrane on N-Ras' conformational state dynamics and its strong influence on Ras protein stability. Moreover, the membrane has a significant influence on the conformational (sub)states sampling of Ras. This, in turn, is of crucial importance in the activation/deactivation cycle of Ras, due to the binding of guanine nucleotide exchange factor proteins (GEFs)/GTPase-activating proteins (GAPs).


Subject(s)
Guanine Nucleotide Exchange Factors , Point Mutation , Proto-Oncogene Proteins p21(ras) , Guanine Nucleotide Exchange Factors/genetics , Guanosine Triphosphate/metabolism , Mutation , ras Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/physiology , Molecular Dynamics Simulation
2.
Curr Issues Mol Biol ; 45(3): 2505-2520, 2023 Mar 17.
Article in English | MEDLINE | ID: mdl-36975534

ABSTRACT

The development of K-Ras independence may explain the failure of targeted therapy for pancreatic cancer (PC). In this paper, active N as well as K-Ras was shown in all human cell lines tested. In a cell line dependent on mutant K-Ras, it was shown that depleting K-Ras reduced total Ras activity, while cell lines described as independent had no significant decline in total Ras activity. The knockdown of N-Ras showed it had an important role in controlling the relative level of oxidative metabolism, but only K-Ras depletion caused a decrease in G2 cyclins. Proteasome inhibition reversed this, and other targets of APC/c were also decreased by K-Ras depletion. K-Ras depletion did not cause an increase in ubiquitinated G2 cyclins but instead caused exit from the G2 phase to slow relative to completion of the S-phase, suggesting that the mutant K-Ras may inhibit APC/c prior to anaphase and stabilise G2 cyclins independently of this. We propose that, during tumorigenesis, cancer cells expressing wild-type N-Ras protein are selected because the protein protects cancer cells from the deleterious effects of the cell cycle-independent induction of cyclins by mutant K-Ras. Mutation independence results when N-Ras activity becomes adequate to drive cell division, even in cells where K-Ras is inhibited.

3.
Int J Mol Sci ; 24(4)2023 Feb 12.
Article in English | MEDLINE | ID: mdl-36835085

ABSTRACT

Epigenetic mechanisms such as microRNA (miRNA) deregulation seem to exert a central role in breast cancer initiation and progression. Therefore, targeting epigenetics deregulation may be an effective strategy for preventing and halting carcinogenesis. Studies have revealed the significant role of naturally occurring polyphenolic compounds derived from fermented blueberry fruits in cancer chemoprevention by modulation of cancer stem cell development through the epigenetic mechanism and regulation of cellular signaling pathways. In this study, we first investigated the phytochemical changes during the blueberry fermentation process. Fermentation favored the release of oligomers and bioactive compounds such as protocatechuic acid (PCA), gallic acid, and catechol. Next, we investigated the chemopreventive potentials of a polyphenolic mixture containing PCA, gallic acid, and catechin found in fermented blueberry juice in a breast cancer model by measuring miRNA expression and the signaling pathways involved in breast cancer stemness and invasion. To this end, 4T1 and MDA-MB-231 cell lines were treated with different doses of the polyphenolic mixture for 24 h. Additionally, female Balb/c mice were fed with this mixture for five weeks; two weeks before and three weeks after receiving 4T1 cells. Mammosphere formation was assayed in both cell lines and the single-cell suspension obtained from the tumor. Lung metastases were counted by isolating 6-thioguanine-resistant cells present in the lungs. In addition, we conducted RT-qPCR and Western blot analysis to validate the expression of targeted miRNAs and proteins, respectively. We found a significant reduction in mammosphere formation in both cell lines treated with the mixture and in tumoral primary cells isolated from mice treated with the polyphenolic compound. The number of colony-forming units of 4T1 cells in the lungs was significantly lower in the treatment group compared to the control group. miR-145 expression significantly increased in the tumor samples of mice treated with the polyphenolic mixture compared to the control group. Furthermore, a significant increase in FOXO1 levels was noted in both cell lines treated with the mixture. Overall, our results show that phenolic compounds found in fermented blueberry delay the formation of tumor-initiating cells in vitro and in vivo and reduce the spread of metastatic cells. The protective mechanisms seem to be related, at least partly, to the epigenetic modulation of mir-145 and its signaling pathways.


Subject(s)
Blueberry Plants , Breast Neoplasms , MicroRNAs , Polyphenols , Animals , Female , Mice , Blueberry Plants/chemistry , Cell Line, Tumor , Cell Proliferation , Chemoprevention , Fermentation , Gallic Acid/pharmacology , Gene Expression Regulation, Neoplastic , MicroRNAs/drug effects , MicroRNAs/metabolism , Polyphenols/pharmacology , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/metabolism , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism
4.
J Cancer Res Ther ; 18(4): 1052-1060, 2022.
Article in English | MEDLINE | ID: mdl-36149161

ABSTRACT

Breast cancer is the leading invasive cancer in women globally. This study aimed at evaluating the anti-apoptotic activity of p-Coumaric acid (PCA) on MCF-7 breast cancer cell line. Experiments were conducted in which the MCF-7 cell line was treated with PCA. which showed decreased cell viability, increased lactate dehydrogenase activity, and caspase-3 activation. The results were evaluated with real-time polymerase chain reaction which revealed that PCA reduced the amount of H-Ras and K-Ras transcript in MCF-7 breast cancer cells. In the presence of PCA there was a significant increase in the levels of mRNA gene Bax and late apoptotic cells which was dose dependent. It also retarded the relative expression of antiapoptotic gene, Bcl2 in treated cells. The results suggest that PCA exhibits anti-cancer properties against MCF-7 cells. PCA inhibited the growth of MCF7 cell. The optimum concentration of PCA was 75-150 mM. PCA can inhibit the growth of MCF-7 cells by reducing Ras expression and inducing cell apoptosis. Our results suggest that PCA could prove valuable in the search for possible inhibitors of Ras oncogene functionality and gain further support for its potential utilization in the treatment of patients with breast cancer. PCA is safe and could complement current treatments employed for the disease.


Subject(s)
Breast Neoplasms , Apoptosis/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Caspase 3/metabolism , Cell Proliferation/genetics , Female , Gene Expression , Genes, ras , Humans , Lactate Dehydrogenases/genetics , MCF-7 Cells , RNA, Messenger/metabolism , bcl-2-Associated X Protein/genetics
5.
Front Cell Dev Biol ; 10: 876000, 2022.
Article in English | MEDLINE | ID: mdl-35419362

ABSTRACT

[This corrects the article DOI: 10.3389/fcell.2020.00836.].

6.
Asian Pac J Cancer Prev ; 23(1): 125-130, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-35092380

ABSTRACT

OBJECTIVE: Acute myeloid leukemia is caused by the clonal proliferation of undifferentiated myeloid hematopoietic precursors. AML prognosis is highly involved in the treatment response and is determined by mutations in several genes such as N-RAS. This study aims to identify the distribution of common N-RAS mutations (codons 12, 13, and 61) in AML patients using the HRM method and confirm this method's efficiency for mutation detection by comparing its results with the sequencing data as the Gold standard method. METHODS: Peripheral blood samples were taken from 50 newly diagnosed AML patients. Mononuclear cells were isolated from samples, and DNA was extracted. Then, mutation detection was investigated using the HRM method. Efficacy of the HRM method in mutation detection was determined in comparison with direct sequencing. RESULTS: N-RAS mutations were detected in 7 of the 50 samples (14%). Most of the mutations were found in codon 12 (57.14%), and 28.57% and 14.28% of mutations were in codons 61 and 13, respectively. There was no statistically significant association between patients' demographic data and HRM results. CONCLUSION: According to mutation detection results and the HRM results confirmation with the sequencing method, this method can be introduced as an efficient, low-cost, and fast method for detecting common mutations.


Subject(s)
DNA Mutational Analysis/methods , Genes, ras/genetics , Leukemia, Myeloid, Acute/genetics , Nucleic Acid Denaturation , Adult , Codon , Female , Humans , Leukemia, Myeloid, Acute/blood , Leukocytes, Mononuclear , Male , Middle Aged , Mutation
7.
Molecules ; 26(14)2021 Jul 17.
Article in English | MEDLINE | ID: mdl-34299605

ABSTRACT

Scientific evidence supports the early deregulation of epigenetic profiles during breast carcinogenesis. Research shows that cellular transformation, carcinogenesis, and stemness maintenance are regulated by epigenetic-specific changes that involve microRNAs (miRNAs). Dietary bioactive compounds such as blueberry polyphenols may modulate susceptibility to breast cancer by the modulation of CSC survival and self-renewal pathways through the epigenetic mechanism, including the regulation of miRNA expression. Therefore, the current study aimed to assay the effect of polyphenol enriched blueberry preparation (PEBP) or non-fermented blueberry juice (NBJ) on the modulation of miRNA signature and the target proteins associated with different clinical-pathological characteristics of breast cancer such as stemness, invasion, and chemoresistance using breast cancer cell lines. To this end, 4T1 and MB-MDM-231 cell lines were exposed to NBJ or PEBP for 24 h. miRNA profiling was performed in breast cancer cell cultures, and RT-qPCR was undertaken to assay the expression of target miRNA. The expression of target proteins was examined by Western blotting. Profiling of miRNA revealed that several miRNAs associated with different clinical-pathological characteristics were differentially expressed in cells treated with PEBP. The validation study showed significant downregulation of oncogenic miR-210 expression in both 4T1 and MDA-MB-231 cells exposed to PEBP. In addition, expression of tumor suppressor miR-145 was significantly increased in both cell lines treated with PEBP. Western blot analysis showed a significant increase in the relative expression of FOXO1 in 4T1 and MDA-MB-231 cells exposed to PEBP and in MDA-MB-231 cells exposed to NBJ. Furthermore, a significant decrease was observed in the relative expression of N-RAS in 4T1 and MDA-MB-231 cells exposed to PEBP and in MDA-MB-231 cells exposed to NBJ. Our data indicate a potential chemoprevention role of PEBP through the modulation of miRNA expression, particularly miR-210 and miR-145, and protection against breast cancer development and progression. Thus, PEBP may represent a source for novel chemopreventative agents against breast cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/drug therapy , Forkhead Box Protein O1/genetics , MicroRNAs/genetics , Neoplastic Stem Cells/drug effects , Polyphenols/pharmacology , Animals , Antineoplastic Agents, Phytogenic/chemistry , Blueberry Plants/chemistry , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Polyphenols/chemistry
8.
Int J Mol Sci ; 22(12)2021 Jun 17.
Article in English | MEDLINE | ID: mdl-34204435

ABSTRACT

The central protein in the oncogenic circuitry is the Ras GTPase that has been under intense scrutiny for the last four decades. From its discovery as a viral oncogene and its non-oncogenic contribution to crucial cellular functioning, an elaborate genetic, structural, and functional map of Ras is being created for its therapeutic targeting. Despite decades of research, there still exist lacunae in our understanding of Ras. The complexity of the Ras functioning is further exemplified by the fact that the three canonical Ras genes encode for four protein isoforms (H-Ras, K-Ras4A, K-Ras4B, and N-Ras). Contrary to the initial assessment that the H-, K-, and N-Ras isoforms are functionally similar, emerging data are uncovering crucial differences between them. These Ras isoforms exhibit not only cell-type and context-dependent functions but also activator and effector specificities on activation by the same receptor. Preferential localization of H-, K-, and N-Ras in different microdomains of the plasma membrane and cellular organelles like Golgi, endoplasmic reticulum, mitochondria, and endosome adds a new dimension to isoform-specific signaling and diverse functions. Herein, we review isoform-specific properties of Ras GTPase and highlight the importance of considering these towards generating effective isoform-specific therapies in the future.


Subject(s)
Proto-Oncogene Proteins p21(ras)/genetics , Biomarkers , Biomarkers, Tumor , Gene Expression Regulation , Humans , Mutation , Protein Isoforms , Proto-Oncogene Proteins p21(ras)/chemistry , Proto-Oncogene Proteins p21(ras)/metabolism , Research , Signal Transduction , Structure-Activity Relationship , Translational Research, Biomedical , ras Proteins/metabolism
9.
Oncol Rep ; 45(1): 390-400, 2021 01.
Article in English | MEDLINE | ID: mdl-33200225

ABSTRACT

The present study aimed to analyze the compensatory signaling pathways induced by forkhead domain inhibitor­6 (FDI­6), which is a forkhead box protein M1 (FOXM1) inhibitor, in ovarian cancer cells and evaluate the effectiveness of simultaneous inhibition of FOXM1 and the compensatory signaling pathway in decreasing the survival of ovarian cancer cells. The present study identified the proteins involved in the compensatory mechanism activated by FDI­6 in HeyA8 ovarian cancer cells using western blot analysis and a reverse­phase protein array. In addition, a cell viability assay was performed to determine the effects of FDI­6 and the compensatory signaling pathway on cancer cell viability. All experiments were performed in three­dimensional cell cultures. The present study observed that FDI­6 stimulated the upregulation of N­Ras, phosphoprotein kinase Cδ (p­PKCδ) (S664) and HER3 in HeyA8 cells. Tipifarnib as an N­Ras inhibitor, rottlerin as a p­PKCδ (S664) inhibitor and sapitinib as a HER3 inhibitor were selected. The combination of FDI­6 with tipifarnib attenuated the upregulation of N­Ras induced by FDI­6 and the combination of FDI­6 with sapitinib also attenuated HER3 downstream signaling pathway in HeyA8 cells, as shown by on western blot analysis. Rottlerin downregulated p­PKCδ (S664) by inhibiting the activity of a Src­related tyrosine kinase that transfers a phosphate group to PKCδ. Compared with FDI­6 alone, the addition of tipifarnib or rottlerin to FDI­6 was significantly more effective in reducing the growth of HeyA8 cells. However, the combination of FDI­6 and sapitinib did not induce a significant decrease in survival of HeyA8 cells. In conclusion, the addition of tipifarnib or rottlerin to inhibit N­Ras or p­PKCδ (S664), respectively, inhibited the compensatory signaling pathway response induced by FDI­6 in HeyA8 cells. These inhibitors increased the efficacy of FDI­6, which inhibits FOXM1, in reducing ovarian cancer cell viability.


Subject(s)
Forkhead Box Protein M1/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Pyridines/pharmacology , Thiophenes/pharmacology , Acetophenones/pharmacology , Benzopyrans/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Female , Forkhead Box Protein M1/analysis , GTP Phosphohydrolases/metabolism , Humans , Membrane Proteins/metabolism , Ovarian Neoplasms/pathology , Quinazolines/pharmacology , Quinolones/pharmacology , Signal Transduction/drug effects
10.
Ecotoxicol Environ Saf ; 208: 111453, 2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33068984

ABSTRACT

Trichloroethylene (TCE), an important volatile organic solvent, causes a series of toxic damage to human. Conventional genetic mechanisms cannot fully explain its toxicity and carcinogenicity, indicative of the possible involvement of epigenetic mechanisms. Our study was intended to investigate the epigenetic toxicity and underlying mechanisms of TCE. Data showed that 0.3 mM TCE treatment for 24 h increased the growth of L-02 cells transiently. In contrast, subacute exposure to TCE inhibited cell growth and induced the genomic DNA hypomethylation and histone hyperacetylation. Further studies have revealed the TCE-induced DNA hypomethylation in the promoter regions of tumor-related genes, N-Ras, c-Jun, c-Myc, c-Fos and IGF-II, promoting their protein levels in a time-dependent manner. These results reveal there is a negative relationship existing between DNA hypomethylation and protein expression in tumor-related gene after TCE exposure under specific epigenetic microenvironment, serving as early biomarkers for TCE-associated diseases.


Subject(s)
Epigenesis, Genetic/physiology , Solvents/toxicity , Trichloroethylene/toxicity , Cell Line , Cell Proliferation/drug effects , DNA/metabolism , DNA Methylation/drug effects , Gene Expression/drug effects , Hepatocytes/metabolism , Histones/metabolism , Humans , Neoplasms , Tumor Microenvironment/drug effects
11.
Front Cell Dev Biol ; 8: 836, 2020.
Article in English | MEDLINE | ID: mdl-32984331

ABSTRACT

Malignant melanoma is one of the most common and dangerous skin cancers with a high rate of death every year. Furthermore, N-RAS and B-RAF mutations in melanoma cells increase the difficulties for clinical treatment in patients. Therefore, development of effective and universal drugs against melanoma is urgently needed. Here we demonstrate that baicalein and baicalin, the active components of the Chinese traditional medicinal plant Scutellaria baicalensis Georgi, can significantly inhibit melanoma cell growth and proliferation, suppress tumor cell colony formation and migration, as well as induce apoptosis and senescence in melanoma cells. The anti-tumor effects mediated by baicalein and baicalin are independent of N-RAS and B-RAF mutation statuses in melanoma cells. Mechanistically, we identify that the suppression of baicalein and baicalin on melanoma cells is due to inhibition of tumor cell glucose uptake and metabolism by affecting the mTOR-HIF-1α signaling pathway. In addition, we demonstrated that baicalein and baicalin can suppress tumorigenesis and tumor growth in vivo in the melanoma model. These studies clearly indicate that baicalein and baicalin can control tumor growth and development metabolically and have great potential as novel and universal drugs for melanoma therapy.

12.
Methods Mol Biol ; 2164: 129-143, 2020.
Article in English | MEDLINE | ID: mdl-32607890

ABSTRACT

Transgenic mouse are reliable, convenient models for studying human hepatocellular carcinoma (HCC). The development of a synthetically engineered Sleeping Beauty (SB) transposon system further enables the viral-free, efficient delivery of desired oncogenes to mouse tissues. Here, we describe an SB transposon-based approach to induce HCC in mice by expressing a hyperactive form of N-RAS, N-RASG12V, while silencing the endogenous Trp53 gene via hydrodynamic tail vein injection, a method to rapidly deliver naked plasmids to mouse liver.


Subject(s)
Carcinoma, Hepatocellular/genetics , DNA Transposable Elements/genetics , Liver Neoplasms/genetics , Animals , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Female , Gene Transfer Techniques , Genetic Therapy/methods , Hydrodynamics , Liver/pathology , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plasmids/genetics , Tail
13.
Genes Dis ; 6(4): 378-384, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31832517

ABSTRACT

Ras gene mutation has been observed in more than 30% of cancers, and 90% of pancreatic, lung and colon cancers. Ras proteins (K-Ras, H-Ras, N-Ras) act as molecular switches which are activated by binding to GTP. They play a role in the cascade of cell process control (proliferation and cell division). In the inactive state, transforming GTP to GDP leads to the activation of GTpase in Ras gene. However, the mutation in Ras leads to the loss of internal GTPase activity and permanent activation of the protein. The activated Ras can promote the cell death or stop cell growth, which are facilitated by Ras-association domain family. Various studies have been conducted to determine the importance of losing RASSF proteins in Ras-induced tumors. This paper examines the role of Ras and RASSF proteins. In general, RASSF proteins can be used as a suitable means for targeting a large group of Ras-induced tumors.

14.
Mar Drugs ; 17(7)2019 Jul 19.
Article in English | MEDLINE | ID: mdl-31331053

ABSTRACT

Low molecular weight fucoidan extract (LMF), prepared by an abalone glycosidase digestion of a crude fucoidan extracted from Cladosiphon novae-caledoniae Kylin, exhibits various biological activities, including anticancer effect. Various cancers express programmed cell death-ligand 1 (PD-L1), which is known to play a significant role in evasion of the host immune surveillance system. PD-L1 is also expressed in many types of normal cells for self-protection. Previous research has revealed that selective inhibition of PD-L1 expressed in cancer cells is critical for successful cancer eradication. In the present study, we analyzed whether LMF could regulate PD-L1 expression in HT1080 fibrosarcoma cells. Our results demonstrated that LMF suppressed PD-L1/PD-L2 expression and the growth of HT1080 cancer cells and had no effect on the growth of normal TIG-1 cells. Thus, LMF differentially regulates PD-L1 expression in normal and cancer cells and could serve as an alternative complementary agent for treatment of cancers with high PD-L1 expression.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Fibrosarcoma/drug therapy , Phaeophyceae/chemistry , Plant Extracts/pharmacology , Polysaccharides/pharmacology , Apoptosis/drug effects , B7-H1 Antigen/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Fibrosarcoma/pathology , Humans , Molecular Weight , Plant Extracts/chemistry , Plant Extracts/therapeutic use , Polysaccharides/chemistry , Polysaccharides/therapeutic use
15.
Int J Hematol ; 110(2): 213-227, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31129802

ABSTRACT

A MEK1/2 inhibitor, binimetinib is promising as a therapeutic agent for malignant melanoma with N-RAS mutation. We examined in vitro effects of binimetinib on 10 human myeloid/lymphoid leukemia cell lines, and found that three of five cell lines with N-RAS mutation and one of five without N-RAS mutation were responsive to treatment with binimetinib. Binimetinib inhibited cell growth mainly by inducing G1 arrest and this action mechanism was assisted by gene set enrichment analysis. To identify signaling pathways associated with binimetinib response, we examined the status of MAP kinase/ERK and PI3Kinase/Akt pathways. The basal levels of phosphorylated ERK and Akt varied between the cell lines, and the amounts of phosphorylated ERK and Akt appeared to be reciprocal of each other. Interestingly, most of the binimetinib-resistant cell lines revealed strong Akt phosphorylation compared with binimetinib-sensitive ones. The effect of binimetinib may not be predicted by the presence/absence of N-RAS mutation, but rather by Akt phosphorylation status. Moreover, combination of binimetinib with a PI3K/Akt inhibitor showed additive growth-suppressive effects. These results suggest that binimetinib shows potential anti-leukemic effects and the basal level of phosphorylated Akt might serve as a biomarker predictive of therapeutic effect.


Subject(s)
Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Genes, ras , Leukemia/pathology , Oncogene Protein p21(ras)/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Aminopyridines/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , DNA, Neoplasm/genetics , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , G1 Phase/drug effects , Gene Expression Regulation, Leukemic/drug effects , Humans , Leukemia/genetics , MAP Kinase Signaling System/drug effects , Morpholines/pharmacology , Mutation , Neoplasm Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
16.
Medicina (Kaunas) ; 55(4)2019 Mar 31.
Article in English | MEDLINE | ID: mdl-30935124

ABSTRACT

Background and objectives: Cancer represents the miscommunication between and within the body cells. The mutations of the oncogenes encoding the MAPK pathways play an important role in the development of tumoral diseases. The mutations of KRAS and BRAF oncogenes are involved in colorectal cancer and melanoma, while the NRAS mutations are associated with melanoma. Thiazolidine-2,4-dione is a versatile scaffold in medicinal chemistry and a useful tool in the development of new antitumoral compounds. The aim of our study was to predict the pharmacokinetic/pharmacodynamic properties, the drug-likeness and lead-likeness of two series of synthetic 5-arylidene(chromenyl-methylene)-thiazolidinediones, the molecular docking on the oncoproteins K-Ras, N-Ras and B-Raf, and to investigate the cytotoxicity of the compounds, in order to select the best structural profile for potential anticancer agents. Materials and Methods: In our paper we studied the cytotoxicity of two series of thiazolidine-2,4-dione derivatives, their ADME-Tox properties and the molecular docking on a mutant protein of K-Ras, two isoforms of N-Ras and an isoform of B-Raf with 16 mutations. Results: The heterocyclic compounds strongly interact with K-Ras and N-Ras right after their posttranslational processing and/or compete with GDP for the nucleotide-binding site of the two GTPases. They are less active against the GDP-bound states of the two targets. All derivatives have a similar binding pattern in the active site of B-Raf. Conclusions: The data obtained encourage the further investigation of the 5-arylidene(chromenyl-methylene)-thiazolidinediones as potential new agents against the oncoproteins K-Ras, N-Ras and B-Raf.


Subject(s)
Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Melanoma/drug therapy , Oncogene Protein p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Skin Neoplasms/drug therapy , Thiazolidinediones/chemistry , Thiazolidinediones/therapeutic use , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Drug Discovery , GTP Phosphohydrolases/chemistry , Guanosine Diphosphate/chemistry , Humans , Melanoma, Experimental/drug therapy , Mice , Molecular Docking Simulation/methods , Mutation , Oncogene Protein p21(ras)/genetics , Protein Binding , Protein Structure, Secondary , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Thiazolidinediones/chemical synthesis
17.
Small GTPases ; 10(1): 20-25, 2019 01.
Article in English | MEDLINE | ID: mdl-28129035

ABSTRACT

Ras is the best-studied member of the superfamily of small GTPases because of its role in cancer. Ras proteins transmit signals for proliferation, differentiation and survival. Three RAS genes encode 4 isoforms. All Ras isoforms have long been considered membrane bound, a localization required for function. Our recent study revealed that N-Ras differs from all other isoforms in being largely cytosolic even following modification with a prenyl lipid. Endogenous, cytosolic N-Ras chromatographed in both high and low molecular weight pools, a pattern that required prenylation, suggesting prenyl-dependent interaction with other proteins. VPS35, a coat protein of the retromer, was shown to interact with prenylated N-Ras in the cytosol. Silencing VPS35 results in partial N-Ras mislocalization on vesicular and tubulovesicular structures, reduced GTP-loading of Ras proteins, and inhibited proliferation and MAPK signaling in an oncogenic N-Ras-driven tumor cell line. Our data revealed a novel regulator of N-Ras trafficking and signaling.


Subject(s)
Cytosol/metabolism , Vesicular Transport Proteins/metabolism , ras Proteins/metabolism , Humans , Protein Prenylation , Protein Transport , Signal Transduction
18.
Semin Cancer Biol ; 54: 114-120, 2019 02.
Article in English | MEDLINE | ID: mdl-29307569

ABSTRACT

Membrane-anchored oncogenic KRas can dimerize, form nanoclusters, and signal through the MAPK (Raf/MEK/ERK) and PI3Kα/Akt/mTOR. Both pathways are needed in KRAS-driven proliferation. Here we ask: Is oncogenic KRas nanoclustering (or dimerization) essential for all KRas signaling pathways? Raf kinase domain dimerization, thus MAPK activation, requires KRas nanoclusters. By contrast, the PI3Kα heterodimer acts as a monomeric unit; thus, does PI3Kα activation and PI3Kα/Akt/mTOR signaling require nanoclustering? Further, calmodulin binds only to oncogenic KRas4B. Here we ask: Does calmodulin downregulate KRas4B cancer development as suggested early on, or promote it? We also ask: Why is oncogenic KRas4B the most abundant isoform? Does wild-type Ras indeed inhibit its oncogenic variants as data appeared to suggest? And related to the last question, why is wild-type KRas a more potent inhibitor of its oncogenic form than wild-type NRas of its oncogenic form? Resolving these cardinal questions, and others, such as how exactly does RASSF5 (NORE1A) act as tumor suppressor, and why Ras isoforms tend to occur in distinct cancer types are crucial for effective pharmacology. In this review, we take a nanoclustering/dimerization-centric outlook and show that many questions can be explained by simply considering Ras nanoclustering.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing , Animals , Apoptosis Regulatory Proteins , Calmodulin/metabolism , Cell Membrane/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Humans , Monomeric GTP-Binding Proteins/metabolism , Protein Binding , Protein Isoforms , Protein Multimerization/drug effects , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction/drug effects
19.
J Cell Mol Med ; 22(4): 2117-2130, 2018 04.
Article in English | MEDLINE | ID: mdl-29377576

ABSTRACT

Acute myeloid leukaemia (AML) comprises a range of disparate genetic subtypes, involving complex gene mutations and specific molecular alterations. Post-translational modifications of specific proteins influence their translocation, stability, aggregation and even leading disease progression. Therapies that target to post-translational modification of specific proteins in cancer cells represent a novel treatment strategy. Non-homogenous subcellular distribution of PLSCR1 is involved in the primary AML cell differentiation. However, the nuclear translocation mechanism of PLSCR1 remains poorly understood. Here, we leveraged the observation that nuclear translocation of PLSCR1 could be induced during wogonoside treatment in some primary AML cells, despite their genetic heterogeneity that contributed to the depalmitoylation of PLSCR1 via acyl protein thioesterase 1 (APT-1), an enzyme catalysing protein depalmitoylation. Besides, we found a similar phenomenon on another AML-related protein, N-RAS. Wogonoside inhibited the palmitoylation of small GTPase N-RAS and enhanced its trafficking into Golgi complex, leading to the inactivation of N-RAS/RAF1 pathway in some primary AML cells. Taken together, our findings provide new insight into the mechanism of wogonoside-induced nuclear translocation of PLSCR1 and illuminate the influence of N-RAS depalmitoylation on its Golgi trafficking and RAF1 signalling inactivation in AML.


Subject(s)
Flavanones/pharmacology , GTP Phosphohydrolases/metabolism , Glucosides/pharmacology , Leukemia, Myeloid, Acute/metabolism , Lipoylation , Membrane Proteins/metabolism , Phospholipid Transfer Proteins/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Humans , Leukemia, Myeloid, Acute/pathology , Lipoylation/drug effects , Protein Transport/drug effects , Thiolester Hydrolases/metabolism , Tumor Cells, Cultured
20.
J Gastrointest Cancer ; 49(2): 124-131, 2018 Jun.
Article in English | MEDLINE | ID: mdl-28044264

ABSTRACT

BACKGROUND: K-RAS and recently N-RAS gene mutation testing are mandatory requirements prior to anti-epidermal growth factor receptor (EGFR) monoclonal antibody treatment of metastatic CRC. Mutation prevalence and distribution in Indonesian colorectal cancer (CRC) are not known. METHODS: Combined methods of PCR high-resolution melt (HRM), restriction fragment length polymorphism (RFLP), and direct DNA sequencing were used to genotype exons 2, 3, and 4 of both K-RAS and N-RAS genes for routine clinical testing of CRC patients. Descriptive analytical review of 595 consecutive CRC patients (years 2013 to 2016) was performed to find associations between gene mutations and clinicopathologic features. RESULTS: This retrospective study revealed overall K-RAS gene mutation in exon 2 (codon 12 and 13) rates being 34.9%. Women (42.5%), stages I and II (43.4%), and well and moderate differentiations (37.7%) had higher frequency of K-RAS exon 2 mutations than men (29%, p = 0.006), stages (III and IV 31.9%, p = 0.05), and poor differentiation (11.8%, p = 0.002), respectively. At later period (2015-2016), 121 of 595 patients were genotyped for the remaining exons 3 and 4 of K-RAS as well as exons 2, 3, and 4 of N-RAS mutations resulting in overall RAS mutation prevalence of 41%. Mucinous histology had highest frequency of N-RAS mutation. CONCLUSIONS: Combination of PCR HRM with either RFLP or direct DNA sequencing was useful to detect K-RAS exon 2 and extended RAS mutations, respectively. Frequency of all RAS mutations in stage IV Indonesian (41%) was similar among Asians (41-49%), which tend to be lower than western (55%) CRC.


Subject(s)
Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Adolescent , Adult , Aged , Cohort Studies , Colorectal Neoplasms/epidemiology , Female , Humans , Indonesia/epidemiology , Male , Middle Aged , Proto-Oncogene Proteins p21(ras)/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...