Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 171
Filter
Add more filters

Publication year range
1.
Mol Biol Rep ; 51(1): 671, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38787503

ABSTRACT

BACKGROUND: TRAIL protein on binding to its cognate death receptors (DR) can induce apoptosis specifically in breast tumor cells sparing normal cells. However, TRAIL also binds to decoy receptors (DCR) thereby inhibiting the apoptotic pathways thus causing TRAIL resistance. Also, one of the barriers due to which TRAIL-based therapy could not become FDA-approved might be because of resistance to therapy. Therefore, in the current study we wanted to explore the role of transcription factors in TRAIL resistance with respect to breast cancer. METHODS: Microarray data from TRAIL-sensitive (TS) and TRAIL-resistant (TR) MDA-MB-231 cells were reanalyzed followed by validation of the candidate genes using quantitative PCR (qPCR), immunoblotting and immunofluorescence technique. Overexpression of the candidate gene was performed in MDA-MB-231 and MCF7 cells followed by cell viability assay and immunoblotting for cleaved caspase-3. Additionally, immunoblotting for DCR2 was carried out. TCGA breast cancer patient survival was used for Kaplan-Meier (KM) plot. RESULTS: Validation of the candidate gene i.e. ELF3 using qPCR and immunoblotting revealed it to be downregulated in TR cells compared to TS cells. ELF3 overexpression in MDA-MB-231 and MCF7 cells caused reversal of TRAIL resistance as observed using cell viability assay and cleaved caspase-3 immunoblotting. ELF3 overexpression also resulted in DCR2 downregulation in the MDA-MB-231 and MCF7 cells. Furthermore, KM analysis found high ELF3 and low DCR2 expression to show better patient survival in the presence of TRAIL. CONCLUSION: Our study shows ELF3 to be an important factor that can influence TRAIL-mediated apoptosis in breast cancer. Also, ELF3 and DCR2 expression status should be taken into consideration while designing strategies for successful TRAIL-based therapy.


Subject(s)
Apoptosis , Breast Neoplasms , DNA-Binding Proteins , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , TNF-Related Apoptosis-Inducing Ligand , Transcription Factors , Humans , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Cell Line, Tumor , Apoptosis/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism , Drug Resistance, Neoplasm/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , MCF-7 Cells , Tumor Necrosis Factor Decoy Receptors/genetics , Tumor Necrosis Factor Decoy Receptors/metabolism , Cell Survival/genetics , Cell Survival/drug effects , Receptors, Tumor Necrosis Factor, Member 10c/genetics , Receptors, Tumor Necrosis Factor, Member 10c/metabolism , Proto-Oncogene Proteins c-ets
2.
BMC Cancer ; 21(1): 76, 2021 Jan 18.
Article in English | MEDLINE | ID: mdl-33461524

ABSTRACT

BACKGROUND: Breast cancer is the most frequently diagnosed malignancy among women. However, the role of microRNA (miRNA) expression in breast cancer progression is not fully understood. In this study we examined predictive interactions between differentially expressed miRNAs and mRNAs in breast cancer cell lines representative of the common molecular subtypes. Integrative bioinformatics analysis identified miR-193 and miR-210 as potential regulatory biomarkers of mRNA in breast cancer. Several recent studies have investigated these miRNAs in a broad range of tumors, but the mechanism of their involvement in cancer progression has not previously been investigated. METHODS: The miRNA-mRNA interactions in breast cancer cell lines were identified by parallel expression analysis and miRNA target prediction programs. The expression profiles of mRNA and miRNAs from luminal (MCF-7, MCF-7/AZ and T47D), HER2 (BT20 and SK-BR3) and triple negative subtypes (Hs578T e MDA-MB-231) could be clearly separated by unsupervised analysis using HB4A cell line as a control. Breast cancer miRNA data from TCGA patients were grouped according to molecular subtypes and then used to validate these findings. Expression of miR-193 and miR-210 was investigated by miRNA transient silencing assays using the MCF7, BT20 and MDA-MB-231 cell lines. Functional studies included, xCELLigence system, ApoTox-Glo triplex assay, flow cytometry and transwell inserts were performed to determine cell proliferation, cytotoxicity, apoptosis, migration and invasion, respectively. RESULTS: The most evident effects were associated with cell proliferation after miR-210 silencing in triple negative subtype cell line MDA-MB-231. Using in silico prediction algorithms, TNFRSF10 was identified as one of the potential regulated downstream targets for both miRNAs. The TNFRSF10C and TNFRSF10D mRNA expression inversely correlated with the expression levels of miR-193 and miR210 in breast cell lines and breast cancer patients, respectively. Other potential regulated genes whose expression also inversely correlated with both miRNAs were CCND1, a known mediator on invasion and metastasis, and the tumor suppressor gene RUNX3. CONCLUSIONS: In summary, our findings identify miR-193 and miR-210 as potential regulatory miRNA in different molecular subtypes of breast cancer and suggest that miR-210 may have a specific role in MDA-MB-231 proliferation. Our results highlight important new downstream regulated targets that may serve as promising therapeutic pathways for aggressive breast cancers.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/metabolism , Biomarkers, Tumor/analysis , Breast/pathology , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Computational Biology , Core Binding Factor Alpha 3 Subunit/genetics , Cyclin D1/genetics , Female , GPI-Linked Proteins/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , MicroRNAs/analysis , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Oligonucleotide Array Sequence Analysis , Receptors, Tumor Necrosis Factor, Member 10c/genetics , Tumor Necrosis Factor Decoy Receptors/genetics
3.
Am J Otolaryngol ; 40(6): 102258, 2019.
Article in English | MEDLINE | ID: mdl-31399243

ABSTRACT

OBJECTIVE: Tongue squamous cell carcinoma (TSCC) is one of the most common malignancies in the oral cavity, and its incidence and mortality have been constantly increasing these years. A large number of tumor suppressor genes are involved in the development of the TSCC and it has been reported that the aberrant hypermethylation of tumor suppressor genes may play a key role in the process of the TSCC. In this study, we sought to analyze the association of methylation of DcR1, DcR2, DR4 and DR5 gene promoters and clinical significance in the TSCC to evaluate association between methylation of DcR1, DcR2, DR4 and DR5 gene and Clinical Significance in tongue squamous cell carcinoma. METHODS: Methylation-specific PCR(MSP) was used to analyze the methylation of the promoters of TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) receptors in 45 TSCC cases. Real-Time PCR was used to detect the expression of the DcR1, DcR2, DR4 and DR5 gene. RESULTS: All the four genes (DcR1, DcR2, DR4 and DR5) showed different methylation of promoters in TSCC, while methylation of these promoters in paired adjacent normal tissues were almost undetectable. Patients with high methylation index were diagnosed at younger age when compared with the ones with low methylation index. DcR1 and DR4 hypermethylation was correlated significantly with patients' TNM stage. CONCLUSIONS: Methylation of DcR1, DcR2,DR4 and DR5 promoters are found in TSCC and may associate with its occurrence and development. Taking the reversibility of methylation into account,methylation is a potential targeted therapy of TSCC.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, Tumor Necrosis Factor, Member 10c/metabolism , Tongue Neoplasms/metabolism , Tumor Necrosis Factor Decoy Receptors/metabolism , Adult , Biomarkers/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Female , GPI-Linked Proteins/metabolism , Humans , Male , Methylation , Middle Aged , Neoplasm Staging , Promoter Regions, Genetic , Tongue Neoplasms/genetics , Tongue Neoplasms/pathology
4.
Apoptosis ; 22(7): 920-932, 2017 07.
Article in English | MEDLINE | ID: mdl-28409399

ABSTRACT

Breast cancer is one of the most lethal malignancies for women. Retinoic acid (RA) and double-stranded RNA (dsRNA) are considered signaling molecules with potential anticancer activity. RA, co-administered with the dsRNA mimic polyinosinic-polycytidylic acid (poly(I:C)), synergizes to induce a TRAIL (Tumor-Necrosis-Factor Related Apoptosis-Inducing Ligand)- dependent apoptotic program in breast cancer cells. Here, we report that RA/poly(I:C) co-treatment, synergically, induce the activation of Interferon Regulatory Factor-3 (IRF3) in breast cancer cells. IRF3 activation is mediated by a member of the pathogen recognition receptors, Toll-like receptor-3 (TLR3), since its depletion abrogates IRF3 activation by RA/poly(I:C) co-treatment. Besides induction of TRAIL, apoptosis induced by RA/poly(I:C) correlates with the increased expression of pro-apoptotic TRAIL receptors, TRAIL-R1/2, and the inhibition of the antagonistic receptors TRAIL-R3/4. IRF3 plays an important role in RA/poly(I:C)-induced apoptosis since IRF3 depletion suppresses caspase-8 and caspase-3 activation, TRAIL expression upregulation and apoptosis. Interestingly, RA/poly(I:C) combination synergizes to induce a bioactive autocrine/paracrine loop of type-I Interferons (IFNs) which is ultimately responsible for TRAIL and TRAIL-R1/2 expression upregulation, while inhibition of TRAIL-R3/4 expression is type-I IFN-independent. Our results highlight the importance of IRF3 and type-I IFNs signaling for the pro-apoptotic effects induced by RA and synthetic dsRNA in breast cancer cells.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Interferon Regulatory Factor-3/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , Apoptosis/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , GPI-Linked Proteins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interferon Type I/genetics , RNA, Double-Stranded/administration & dosage , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, Tumor Necrosis Factor, Member 10c/genetics , Toll-Like Receptor 3/genetics , Tretinoin/administration & dosage
5.
BMC Cancer ; 17(1): 280, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28420351

ABSTRACT

BACKGROUND: Tumor epithelial cells (TEpCs) and spindle-shaped stromal cells, not associated with the vasculature, of patients with early breast cancer express osteoprotegerin (OPG), tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), receptor activator of nuclear factor kappa B ligand, stromal cell derived factor-1, interleukin-6, macrophage colony stimulating factor, chemokine (C-C motif) ligand-2 (CCL-2) and their receptors at significantly higher levels compared with non-neoplastic breast tissues. We evaluated the clinicopathological significance of these ligands and receptors in TEpC and spindle-shaped stromal cells, not associated with the vasculature, to determine their impact on prognosis of patients with early-stage breast cancer. METHODS: We conducted immunohistochemical analyses of protein expression in primary tumors of patients with early breast cancer and analyzed their association with standard prognostic parameters and clinical outcomes, including local relapse, metastatic recurrence, disease-free survival (DFS), metastasis-free survival (MFS), and overall survival (OS). RESULTS: Elevated levels of TRAIL-R3 and chemokine (C-C motif) receptor 2 (CCR-2) in TEpCs and OPG and CCL-2 in stromal cells were significantly associated with a higher risk of metastasis (p = 0.032, p = 0.003, p = 0.038, and p = 0.049; respectively). Moreover, high expression of TRAIL-R3 and CCR-2 in TEpCs was associated with shorter DFS, MFS, and OS. High TRAIL-R3 expression in TEpCs was an independent prognostic factor for DFS and OS, and high CCR-2 expression in these cells was an independent prognostic factor for MFS. CONCLUSIONS: High levels of TRAIL-R3 and CCR-2 expression in TEpCs identified patients with early breast cancer with poor outcomes.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/pathology , Epithelial Cells/metabolism , Receptors, CCR2/biosynthesis , Receptors, Tumor Necrosis Factor, Member 10c/biosynthesis , Adult , Aged , Aged, 80 and over , Breast Neoplasms/mortality , Epithelial Cells/pathology , Female , GPI-Linked Proteins/analysis , GPI-Linked Proteins/biosynthesis , Humans , Immunohistochemistry , Middle Aged , Prognosis , Receptors, CCR2/analysis , Receptors, Tumor Necrosis Factor, Member 10c/analysis , Retrospective Studies
6.
Neoplasma ; 64(2): 182-191, 2017.
Article in English | MEDLINE | ID: mdl-28043144

ABSTRACT

The activation of AKT is one of the causes of resistance to epidermal growth factor receptor (EGFR)- tyrosine kinase inhibitors (TKIs). Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) combines with related receptors to trigger apoptosis or protect the cells against TRAIL apoptosis. This research focused on the association of EGFR and KRAS mutations with expression of AKT, p-AKT, DR5 and DcR1 in non-small cell lung cancer. 82 NSCLC patients were included in the study. xTAG liquichip techonolgy (xTAG-LCT) was applied to investigate the genetic mutation of EGFR and KRAS, Quantitative Real-time PCR was used to test the mRNA expression of AKT, DR5 and DcR1 and Western Blot was applied to test the protein expression of AKT, p-AKT, DR5, and DcR1. We found that of 82 patients, 31 cases had EGFR-activating mutations, more common in female, adenocarcinoma, and non-smoker patients; 9 cases had KRAS mutations, frequently found in patients with smoking history. The expression of AKT and p-AKT correlated with staging, tumor differentiation, and lymph node metastasis. The expression of DR5 in phase III and low differentiation tumor was significantly higher than that in phase I+II and high and median differentiation tumor; the expression of DcR1 in phase III and low differentiation tumor was significantly lower than that in phase I+II and high and median differentiation tumor. Compared with EGFR and KRAS wild type, in NSCLC tissue with EGFR and KRAS mutations, the expression of AKT and p-AKT was significantly higher. These results suggest that EGFR and KRAS mutation status was associated with the expression of AKT and p-AKT. AKT, p-AKT, DR5, and DcR1 all took part in the occurrence and development of NSCLC, and may become a reference index to evaluate the prognosis of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , ErbB Receptors/genetics , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Male , Mutation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, Tumor Necrosis Factor, Member 10c/genetics , Receptors, Tumor Necrosis Factor, Member 10c/metabolism
7.
Genes Chromosomes Cancer ; 55(2): 177-89, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26542757

ABSTRACT

Multiple chromosomal regions are affected by deletions in cervical cancer (CC) genomes, but their consequence and target gene involvement remains unknown. Our single nucleotide polymorphism (SNP) array identified 8p copy number losses localized to an 8.4 Mb minimal deleted region (MDR) in 36% of CC. The 8p MDR was associated with tumor size, treatment outcome, and with multiple HPV infections. Genetic, epigenetic, and expression analyses of candidate genes at MDR identified promoter hypermethylation and/or inactivation of decoy receptors TNFRSF10C and TNFRSF10D in the majority of CC patients. TNFRSF10C methylation was also detected in precancerous lesions suggesting that this change is an early event in cervical tumorigenesis. We further demonstrate here that CC cell lines exhibiting downregulated expression of TNFRSF10C and/or TNFRSF10D effectively respond to TRAIL-induced apoptosis and this affect was synergistic in combination with DNA damaging chemotherapeutic drugs. We show that the CC cell lines harboring epigenetic inactivation of TRAIL decoy receptors effectively activate downstream caspases suggesting a critical role of inactivation of these genes in efficient execution of extrinsic apoptotic pathway and therapy response. Therefore, these findings shed new light on the role of genetic/epigenetic defects in TRAIL decoy receptor genes in the pathogenesis of CC and provide an opportunity to explore strategies to test decoy receptor gene inactivation as a biomarker of response to Apo2L/TRAIL-combination therapy.


Subject(s)
Cisplatin/pharmacology , DNA Methylation , Receptors, Tumor Necrosis Factor, Member 10c/genetics , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Tumor Necrosis Factor Decoy Receptors/genetics , Uterine Cervical Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols , Base Sequence , Cell Line, Tumor , Cell Survival/drug effects , Chromosomes, Human, Pair 8/genetics , Cisplatin/therapeutic use , Epigenesis, Genetic , Female , GPI-Linked Proteins/genetics , HeLa Cells , Humans , Middle Aged , Polymorphism, Single Nucleotide , Sequence Deletion , Uterine Cervical Neoplasms/genetics
8.
Cell Physiol Biochem ; 36(3): 1151-62, 2015.
Article in English | MEDLINE | ID: mdl-26111475

ABSTRACT

BACKGROUND/AIMS: Although Vitisin A, derived from wine grapes, is known to have cytotoxic, anti-adipogenic, anti-inflammatory and antioxidant effects, the underlying antitumor mechanism has not been investigated in prostate cancer cells to date. In the present study, the apoptotic mechanism of Vitisin A plus TNF-related apoptosis-inducing ligand (TRAIL) in prostate cancer cells was elucidated. METHODS: The cytotoxicity of Vitisin A and/or TRAIL against PC-3, DU145 and LNCaP prostate cancer cells was measured by MTT colorimetric assay. Annexin V-FITC Apoptosis Detection kit was used to detect apoptotic cells by flow cytometry. Intracellular levels of ROS were measured by flow cytometry using 2070-diacetyl dichlorofluorescein (DCFDA). RESULTS: Combined treatment with Vitisin A and TRAIL enhanced cytotoxicity and also increased sub-G1 population in PC-3 cells better than DU145 or LNCap prostate cancer cells. Similarly, Annexin V and PI staining revealed that combination increased early and late apoptosis in PC-3 cells compared to untreated control. Consistently, combination attenuated the expression of pro-caspases 7/8, DcR1, Bcl-XL or Bcl-2 and activated caspase 3, FADD, DR5 and DR4 in PC-3 cells. Also, combination increased DR5 promoter activity compared to untreated control. Furthermore, combination increased the production of reactive oxygen species (ROS) and DR5 cell surface expression. The ROS inhibitor NAC and silencing of DR5 by siRNA transfection inhibited the ability of combination to induce PARP cleavage and generate ROS. CONCLUSION: These findings provide evidence that Vitisin A can be used in conjunction with TRAIL as a potent TRAIL sensitizer for synergistic apoptosis induction via upregulation of DR5 and production of ROS in prostate cancer cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Benzofurans/pharmacology , Gene Expression Regulation, Neoplastic , Phenols/pharmacology , Prostate/drug effects , Reactive Oxygen Species/agonists , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Apoptosis/drug effects , Caspase 7/genetics , Caspase 7/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Drug Combinations , Drug Synergism , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Male , Prostate/metabolism , Prostate/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, Tumor Necrosis Factor, Member 10c/genetics , Receptors, Tumor Necrosis Factor, Member 10c/metabolism , Signal Transduction , bcl-X Protein/genetics , bcl-X Protein/metabolism
9.
J Obstet Gynaecol ; 34(4): 346-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24649804

ABSTRACT

We aimed to evaluate the membrane expression of DcR1 and DcR2 in the normal endometrium (NE), endometrial atypical hyperplasia (EAH) and endometrioid endometrial cancer (EEC). The study comprised 101 patients: 20 NE, 14 EAH and 67 EEC. Membrane expression of DcR1 and DcR2 was examined and presented as total score (TS). The membrane expression of both DcR1 and DcR2 was more common in EEC than in NE (p < 0.001; p < 0.001). A strong correlation was found between type of endometrial tissue (NE/EAH/EEC) and the TS of DcR1 (p = 0.001) and DcR2 (p < 0.001). In EEC, the TS of DcR1 and DcR2 was not related to grading and survival. The TS of DcR1 negatively correlated with staging (p = 0.018), but DcR2 did not. The membrane expression of decoy receptors for TRAIL DcR1 and DcR2 is greater in NE than EEC. In EEC patients, membrane expression of DcR1 and DcR2 are not independent predictors of survival.


Subject(s)
Carcinoma, Endometrioid/metabolism , Endometrial Hyperplasia/metabolism , Endometrial Neoplasms/metabolism , Endometrium/metabolism , Tumor Necrosis Factor Decoy Receptors/metabolism , Case-Control Studies , Female , GPI-Linked Proteins/metabolism , Humans , Receptors, Tumor Necrosis Factor, Member 10c
10.
Mol Biol (Mosk) ; 48(6): 963-9, 2014.
Article in Russian | MEDLINE | ID: mdl-25845236

ABSTRACT

Accumulating evidence suggests that proinflammatory cytokines play an important role in white matter injury in preterm infants, a condition in which oligodendrocyte (OL) progenitor cells are preferentially injured. We investigated the role of tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) and its death (TRAIL-R1, TRAIL-R2) and decoy (TRAIL-R3, TRAIL-R4) receptors in periventricular white matter injury (PWMI). We hypothesized that the maturation-dependent vulnerability of OLs to TRAIL is due to differential TRAIL receptor expression. We previously investigated TRAIL/TRAIL receptor expression levels in rat OLs in vivo in the context of PWMI. We found that during different developmental stages, human OLs differentially express TRAIL receptors; there is a progressive loss of sensitivity to TRAIL as OLs proceed through the maturation process. Our results show that both TRAIL-R1 and -R2 are highly expressed on human OL progenitors and pre-OLs, while TRAIL-R3 and TRAIL-R4 are mainly expressed on immature and mature human OLs. Our results suggest that TRAIL-R1 and TRAIL-R2 might mediate the death signal in human OL precursor cells and pre-OLs.


Subject(s)
Gene Expression Regulation, Developmental , Oligodendroglia/drug effects , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Apoptosis/genetics , Cell Differentiation/drug effects , Cell Proliferation/drug effects , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Humans , Oligodendroglia/cytology , Oligodendroglia/metabolism , Primary Cell Culture , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, Tumor Necrosis Factor, Member 10c , Signal Transduction , Spheroids, Cellular/cytology , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Necrosis Factor Decoy Receptors/genetics , Tumor Necrosis Factor Decoy Receptors/metabolism
11.
Biochem Biophys Res Commun ; 441(4): 743-50, 2013 Nov 29.
Article in English | MEDLINE | ID: mdl-24211571

ABSTRACT

Aberrant promoter methylation and resultant silencing of TRAIL decoy receptors were reported in a variety of cancers, but to date little is known about the relevance of this epigenetic modification in melanoma. In this study, we examined the methylation and the expression status of TRAIL receptor genes in cutaneous and uveal melanoma cell lines and specimens and their interaction with DNA methyltransferases (DNMTs) DNMT1, DNMT3a, and DNMT3b. DR4 and DR5 methylation was not frequent in cutaneous melanoma but on the contrary it was very frequent in uveal melanoma. No correlation between methylation status of DR4 and DR5 and gene expression was found. DcR1 and DcR2 were hypermethylated with very high frequency in both cutaneous and uveal melanoma. The concordance between methylation and loss of gene expression ranged from 91% to 97%. Here we showed that DNMT1 was crucial for DcR2 hypermethylation and that DNMT1 and DNMT3a coregulate the methylation status of DcR1. Our work also revealed the critical relevance of DcR1 and DcR2 expression in cell growth and apoptosis either in cutaneous or uveal melanoma. In conclusion, the results presented here claim for a relevant impact of aberrant methylation of decoy receptors in melanoma and allow to understand how the silencing of DcR1 and DcR2 is related to melanomagenesis.


Subject(s)
DNA-Cytosine Methylases/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Melanoma/genetics , Skin Neoplasms/genetics , Tumor Necrosis Factor Decoy Receptors/genetics , Uveal Neoplasms/genetics , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Line, Tumor , DNA Methylation , DNA-Cytosine Methylases/genetics , GPI-Linked Proteins/genetics , Humans , Melanoma/enzymology , Receptors, Tumor Necrosis Factor, Member 10c , Skin Neoplasms/enzymology , Uveal Neoplasms/enzymology
12.
Arch Gynecol Obstet ; 288(4): 889-99, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23584885

ABSTRACT

PURPOSE: To evaluate the membrane expression of DR4, DR5, DcR1 and DcR2 in the normal endometrium (NE), atypical endometrial hyperplasia (AEH) and endometrioid adenocarcinoma (EAC). METHODS: The study comprised 197 patients: 20 NE, 18 AEH and 159 EAC. Tissue microarrays were constructed. Membrane expression of DR4, DR5, DcR1 and DcR2 was examined and presented as total score (TS). RESULTS: In EAC, the membrane expression of DR4, DR5 and DcR2 was less common compared to NE (p < 0.001; p < 0.001; p = 0.018) and AEH (p < 0.001; p < 0.001; p = 0.004). In EAC the membrane expression of DcR1 did not differ when compared to NE (p = 0.055) and AEH (p = 0.173). A strong correlation was found between the type of endometrial tissue (NE/AEH/EAC) and the TS of DR4 (p < 0.001), DR5 (p < 0.001), DcR1 (p = 0.033) and DcR2 (p < 0.001). In EAC, the TS of DR4, DR5, DcR1 and DcR2 was not related to grading and staging. In EAC, the membrane expression of DR5, but not DR4, DcR1 and DcR2, was related to better disease-free survival (DFS). The overall survival (OS) was not related to membrane TRAIL receptors expression. CONCLUSIONS: The membrane expression of the receptors for TRAIL DR4, DR5, DcR1 and DcR2 is greater in NE than EAC. The level of membrane staining of the receptors in EAC is not dependent on grading and staging. In EAC patients, membrane expression of DR4, DR5, DcR1 and DcR2 are not independent predictors of survival.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Endometrioid/metabolism , Endometrial Hyperplasia/metabolism , Endometrial Neoplasms/metabolism , Endometrium/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Necrosis Factor Decoy Receptors/metabolism , Biomarkers/metabolism , Carcinoma, Endometrioid/mortality , Cell Membrane/metabolism , Endometrial Neoplasms/mortality , Female , GPI-Linked Proteins/metabolism , Humans , Immunohistochemistry , Receptors, Tumor Necrosis Factor, Member 10c , Survival Analysis , Tissue Array Analysis
13.
Nat Genet ; 36(11): 1174-80, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15475954

ABSTRACT

RNA interference is a conserved mechanism by which double-stranded RNA is processed into short interfering RNAs (siRNAs) that can trigger both post-transcriptional and transcriptional gene silencing. In fission yeast, the RNA-induced initiation of transcriptional gene silencing (RITS) complex contains Dicer-generated siRNAs and is required for heterochromatic silencing. Here we show that RITS components, including Argonaute protein, bind to all known heterochromatic loci. At the mating-type region, RITS is recruited to the centromere-homologous repeat cenH in a Dicer-dependent manner, whereas the spreading of RITS across the entire 20-kb silenced domain, as well as its subsequent maintenance, requires heterochromatin machinery including Swi6 and occurs even in the absence of Dicer. Furthermore, our analyses suggest that RNA interference machinery operates in cis as a stable component of heterochromatic domains with RITS tethered to silenced loci by methylation of histone H3 at Lys9. This tethering promotes the processing of transcripts and generation of additional siRNAs for heterochromatin maintenance.


Subject(s)
Gene Silencing , RNA Interference , Schizosaccharomyces/genetics , Animals , Chromosomal Proteins, Non-Histone/genetics , Chromosomes, Fungal , DNA Methylation , GPI-Linked Proteins , Heterochromatin , Models, Genetic , RNA, Small Interfering , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor, Member 10c , Schizosaccharomyces pombe Proteins/genetics , Tumor Necrosis Factor Decoy Receptors
14.
Gynecol Oncol ; 126(2): 268-73, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22555108

ABSTRACT

OBJECTIVE: To assess the expression of TRAIL-R3 and the methylation of a CpG island within the TRAIL-R3 promoter both in cystadenoma tumors and primary and metastatic epithelial ovarian carcinoma (EOC). METHODS: RNA was obtained from women with normal ovarian (NO) tissues (n=18), ovarian serous cystadenoma tumors (n=11) and EOC (n=16) using Trizol. Quantitative PCR (qRT-PCR) was performed to quantify the relative levels of TRAIL-R3. The methylation frequency of the CpG island in the TRAIL-R3 promoter was assessed using the methylation-specific PCR (MSP) assay after DNA bisulfite conversion. The differences between the groups were evaluated using the chi-square, Student's t, ANOVA, Mann-Whitney U, Wilcoxon or Kruskal-Wallis tests as indicated. The survival rates were calculated using the Kaplan-Meier method. RESULTS: Cystadenoma and metastatic EOC tumors expressed significantly more TRAIL-R3 mRNA than primary EOC tumors. Methylation of the TRAIL-R3 promoter was absent in NO tissues, while hemimethylation of the TRAIL-R3 promoter was frequently found in the neoplasia samples with 45.4% of the cystadenoma tumors, 8.3% of the primary EOC samples and 11.1% of the metastatic EOC samples showing at least partial methylation (p=0.018). Neither the expression of TRAIL-R3 nor alterations in the methylation profile were associated to cumulative progression-free survival or the overall survival in EOC patients. CONCLUSIONS: Primary EOC is associated to a lower TRAIL-R3 expression, which leads to a better understanding of the complex disease and highlighting potential therapeutic targets. Promoter DNA methylation was not related to this finding, suggesting the presence of other mechanisms to transcriptional control.


Subject(s)
Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Tumor Necrosis Factor Decoy Receptors/biosynthesis , Tumor Necrosis Factor Decoy Receptors/genetics , Apoptosis/physiology , Cell Line, Tumor , Cystadenocarcinoma, Serous/pathology , DNA Methylation , Disease-Free Survival , Epigenomics , Female , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Ovarian Neoplasms/pathology , Prospective Studies , Receptors, Tumor Necrosis Factor, Member 10c
15.
Cancer Lett ; 525: 67-75, 2022 01 28.
Article in English | MEDLINE | ID: mdl-34728311

ABSTRACT

Genotoxic agents are widely used anti-cancer therapies because of their ability to interfere with highly proliferative cells. An important outcome of these interventions is the induction of a state of permanent arrest also known as cellular senescence. However, senescent cancer cells are characterized by genomic instability and are at risk of escaping the growth arrest to eventually facilitate cancer relapse. The tumor necrosis factor related apoptosis inducing ligand (TRAIL) signals extrinsic apoptosis via Death Receptors (DR) 4 and 5, while Decoy Receptors (DcR) 1 and 2, and Osteoprotegerin (OPG) are homologous to death receptors but incapable of transducing an apoptotic signal. The use of recombinant TRAIL as an anti-cancer strategy in combination with chemotherapy is currently in development, and a major question remains whether senescent cancer cells respond to TRAIL. Here, we show variable sensitivity of cancer cells to TRAIL after senescence induction, and upregulation of both pro-apoptotic and anti-apoptotic receptors in therapy-induced senescent cancer cells. A DR5-selective TRAIL variant (DHER), unable to bind to DcR1 or OPG, was more effective in inducing apoptosis of senescent cancer cells compared to wild-type TRAIL. Importantly, no apoptosis induction was observed in non-cancerous cells, even at the highest concentrations tested. Our results suggest that targeting DR5 can serve as a novel therapeutic strategy for the elimination of therapy-induced senescent cancer cells.


Subject(s)
Breast Neoplasms/drug therapy , Osteoprotegerin/genetics , Ovarian Neoplasms/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , Apoptosis/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cellular Senescence/drug effects , Cellular Senescence/genetics , Doxorubicin/pharmacology , Female , GPI-Linked Proteins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Genomic Instability/drug effects , Humans , MCF-7 Cells , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Receptors, Tumor Necrosis Factor, Member 10c/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor Decoy Receptors/genetics
16.
Dev Neurosci ; 33(6): 519-30, 2011.
Article in English | MEDLINE | ID: mdl-22286051

ABSTRACT

The immature brain is susceptible to inflammatory injury induced by hypoxia-ischemia (HI) or infection, which causes serious neurodevelopmental disabilities in the survivors of preterm births. Recently, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and its receptors (death receptor DR4/5 and decoy receptor DcR1/2) were reported to mediate various neuroinflammatory responses. However, little information is available regarding the role of TRAIL and its receptors in the immature brain after HI. The purpose of this study was to evaluate the expression of TRAIL and its receptors in the immature brain after HI and relate this expression to neurological function. We performed right common carotid artery ligation followed by hypoxia (6% O(2), 37°C) for 2.5 h to induce HI in postnatal day 3 rats. The distribution of TRAIL and its receptors, caspase-3 and CD68-labeled microglia/macrophages was evaluated 24 h after HI by immunostaining. The protein and mRNA expression of TRAIL and DR5 was measured by Western blot and real-time PCR, respectively. Delayed neuronal loss was evaluated by NeuN and Nissl staining 7 days after HI. Furthermore, neurological deficits were evaluated by a righting reflex test, time of eye opening and T-maze test. The expression of TRAIL, DR5 and DcR1/2 receptors and caspase-3 was more pronounced in the ipsilateral hemisphere compared with the contralateral part and the control group 24 h after HI. DR5/active caspase-3 double-positive cells were observed at 24 h after HI in the ipsilateral hemisphere but not in the contralateral hemisphere. The TRAIL and CD68 double-labeled cells were more pronounced in the ipsilateral cortical regions compared with the corresponding regions of the contralateral part. HI also resulted in a significant increase in TRAIL and DR5 protein and mRNA expression at 24 h, which corresponded to neuronal cell loss 7 days after HI. Furthermore, the HI group displayed impaired neurobehavioral development compared with the control group (p < 0.05). Altogether our results show that the TNF-α superfamily ligand TRAIL is induced on CD68+ microglia/macrophages after perinatal HI and that one of its receptors, DR5, is induced on neocortical neurons and glial cells. That many DR5+ cells were also caspase-3+ strongly supports the conclusion that these signaling molecules are involved in the delayed loss of neurons in the neocortex and in the neurobehavioral deficits that are often seen after perinatal HI.


Subject(s)
Hypoxia-Ischemia, Brain/metabolism , Microglia/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Animals, Newborn , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Blotting, Western , Hypoxia-Ischemia, Brain/pathology , Immunohistochemistry , Neurons/metabolism , Neurons/pathology , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptors, Tumor Necrosis Factor, Member 10c/metabolism , Reverse Transcriptase Polymerase Chain Reaction
17.
Curr Opin Cell Biol ; 11(2): 255-60, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10209153

ABSTRACT

The death receptors Fas and tumor necrosis factor receptor 1 (TNFR1) trigger apoptosis upon engagement by their cognate death ligands. Recently, researchers have discovered several novel homologues of Fas and TNFR1: DR 3, 4, 5, and 6 function as death receptors that signal apoptosis, whereas DcR 1, 2, and 3 act as decoys that compete with specific death receptors for ligand binding. Further, mouse gene knockout studies have enabled researchers to delineate some of the signaling pathways that connect death receptors to the cell's apoptotic machinery.


Subject(s)
Antigens, CD/physiology , Apoptosis/physiology , Arabidopsis Proteins , DNA-Binding Proteins/physiology , Fatty Acid Desaturases/physiology , Membrane Glycoproteins/physiology , Receptors, Cell Surface/physiology , Receptors, Tumor Necrosis Factor/physiology , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/physiology , fas Receptor/physiology , Animals , Apoptosis Regulatory Proteins , Caspase 8 , Caspase 9 , Caspases/genetics , Caspases/physiology , Cytochrome c Group/physiology , Death Domain Receptor Signaling Adaptor Proteins , Expressed Sequence Tags , Fas Ligand Protein , GPI-Linked Proteins , Mice , Mice, Knockout , Mice, Transgenic , Models, Biological , Protein Kinases/physiology , Receptors, Tumor Necrosis Factor, Member 10c , Receptors, Tumor Necrosis Factor, Member 6b , Receptors, Tumor Necrosis Factor, Type I , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor Decoy Receptors
18.
Hepatobiliary Pancreat Dis Int ; 10(1): 95-100, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21269942

ABSTRACT

BACKGROUND: A growing body of evidence suggests that many tumors are initiated by both epigenetic abnormalities and gene mutations, which promote tumor progression. Epigenetic abnormalities include changes in DNA methylation and in the modification of histones. This study aimed to assess the status of methylation in the CpG island (CGI) of the tumor necrosis factor receptor superfamily member 10c (TNFRSF10C) with combined bisulfite restriction analysis (COBRA) and to evaluate its role in the progression of pancreatic cancer (PC). METHODS: The methylation status of four PC cell lines was assessed using COBRA and/or bisulfite genomic sequencing (BGS). Changes in methylation and TNFRSF10C expression in PC cell lines before and after treatment with 5-aza-2'-deoxycytidine (5-aza-dC) and/or trichostatin A (TSA) were assessed by BGS and real-time RT-PCR. Apoptosis in the four cell lines was tested by flow cytometry (FCM) and TUNEL assay. RESULTS: The methylation status of the TNFRSF10C promoter was assessed in PC cells (BxPC-3: 68.84+/-8.71%; CFPAC-1: 0; PANC-1: 96.77+/-4.57%; SW1990: 54.97+/-7.33%) with the COBRA assay, which was confirmed by the results of BGS. After treatment with 5-aza-dC and/or TSA, apoptosis was induced in PC cells to different degrees, and the levels of TNFRSF10C transcriptional expression in the PC cell lines (except CFPAC-1) increased markedly after 5-aza-dC treatment. CONCLUSIONS: A high frequency of CGI methylation in the TNFRSF10C promoter results in inactivation of the gene and enhancement of tumor growth in most PC cell lines (except CFPAC-1). Inactivation of TNFRSF10C by CGI hypermethylation can play an important role in PC progression and be potentially useful as a diagnostic marker and a new therapeutic approach for PC.


Subject(s)
DNA Methylation , Pancreatic Neoplasms/genetics , Tumor Necrosis Factor Decoy Receptors/genetics , Tumor Necrosis Factor Decoy Receptors/metabolism , Apoptosis/drug effects , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , DNA Methylation/drug effects , Decitabine , Disease Progression , Epigenesis, Genetic , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression/drug effects , Humans , Hydroxamic Acids/pharmacology , Promoter Regions, Genetic , Receptors, Tumor Necrosis Factor, Member 10c , Restriction Mapping , Sequence Analysis, DNA , Tumor Cells, Cultured
19.
J Mol Endocrinol ; 66(4): 325-338, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33875613

ABSTRACT

Strategies to increase functional pancreatic beta cell mass is of great interest in diabetes-related research. TNF-related apoptosis-inducing ligand (TRAIL) is well known to promote proliferation and survival in various cell types, including vascular smooth muscle and endothelial cells. Correlation between the protective nature of TRAIL on these cells and its proliferative effect is noteworthy. TRAIL's seemingly protective/therapeutic effect in diabetes prompted us to question whether it may act as an inducer of proliferation in pancreatic beta cells. We used rat primary islet cells and MIN6 mouse beta cell line to investigate TRAIL-induced proliferation. Cell viability and/or death was analyzed by MTT, WST-1, and Annexin-V/PI assays, while proliferation rates and pathways were assessed via immunocytochemical and Western blot analyses. Receptor neutralization antibodies identified the mediator receptors. Recombinant soluble TRAIL (sTRAIL) treatment led to 1.6-fold increased proliferation in insulin-positive cells in dispersed rat islets compared to the untreated group, while adenovirus-mediated overexpression of TRAIL increased the number of proliferating beta cells up to more than six-fold. sTRAIL or adenoviral vector-mediated TRAIL overexpression induced proliferation in MIN6 cells also. TRAIL's proliferative effect was mediated via AKT activation, which was suppressed upon specific inhibition. Neutralization of each TRAIL receptor reversed the proliferative effect to some degree, with the highest level of inhibition in death receptor 5 (DR5) blockage in MIN6 cells and in decoy receptor 1 (DcR1) blockage in primary rat beta cells. Thus, TRAIL induces proliferation in rodent pancreatic beta cells through activation of the AKT pathway.


Subject(s)
Diabetes Mellitus/genetics , Insulin-Secreting Cells/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, Tumor Necrosis Factor, Member 10c/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , Animals , Apoptosis/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Diabetes Mellitus/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Expression Regulation/genetics , Humans , Insulin-Secreting Cells/pathology , Mice , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Proto-Oncogene Proteins c-akt/genetics , Rats
20.
J Exp Med ; 186(7): 1165-70, 1997 Oct 06.
Article in English | MEDLINE | ID: mdl-9314565

ABSTRACT

TRAIL-R3, a new member of the TRAIL receptor family, has been cloned and characterized. TRAIL-R3 encodes a 299 amino acid protein with 58 and 54% overall identity to TRAIL-R1 and -R2, respectively. Transient expression and quantitative binding studies show TRAIL-R3 to be a plasma membrane-bound protein capable of high affinity interaction with the TRAIL ligand. The TRAIL-R3 gene maps to human chromosome 8p22-21, clustered with the genes encoding two other TRAIL receptors. In contrast to TRAIL-R1 and -R2, this receptor shows restricted expression, with transcripts detectable only in peripheral blood lymphocytes and spleen. The structure of TRAIL-R3 is unique when compared to the other TRAIL receptors in that it lacks a cytoplasmic domain and appears to be glycosyl-phosphatidylinositol-linked. Moreover, unlike TRAIL-R1 and -R2, in a transient overexpression system TRAIL-R3 does not induce apoptosis.


Subject(s)
Membrane Glycoproteins/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Receptors, Tumor Necrosis Factor , Tumor Necrosis Factor-alpha/metabolism , Amino Acid Sequence , Apoptosis/physiology , Apoptosis Regulatory Proteins , Base Sequence , Blotting, Northern , Chromosome Mapping , Chromosomes, Human, Pair 8/genetics , Cloning, Molecular , GPI-Linked Proteins , Gene Expression Regulation , Glycosylphosphatidylinositols , Humans , Immunoglobulin Fc Fragments/genetics , Jurkat Cells , Lymphocytes/chemistry , Lymphocytes/metabolism , Membrane Glycoproteins/pharmacology , Molecular Sequence Data , Protein Binding , Receptors, Cell Surface/genetics , Receptors, Tumor Necrosis Factor, Member 10c , Recombinant Fusion Proteins/pharmacology , Sequence Analysis, DNA , Sequence Homology, Amino Acid , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor Decoy Receptors , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL