Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Mol Med (Berl) ; 97(8): 1155-1167, 2019 08.
Article in English | MEDLINE | ID: mdl-31183506

ABSTRACT

Upon ligand binding, plasma membrane-located TNF-related apoptosis-inducing ligand (TRAIL)-receptors 1 and 2 induce apoptosis as well as cancer-promoting signaling in cancer cells. TRAIL-R3 and TRAIL-R4 are believed to negatively regulate TRAIL-mediated apoptosis. Intracellular localization of TRAIL-receptors, as observed in many tumor cells, has been associated with oncogenic features, which are distinct from membrane-associated TRAIL-R signaling. Here, analyzing a panel of 354 breast cancer specimens, we found that an unfavorable outcome correlating with cancer-promoting properties of TRAIL-R1, TRAIL-R2, and TRAIL-R4 was most significantly defined by their intracellular distribution and mutual co-expression. A nuclear or cytoplasmic heterogeneous expression pattern correlated with markedly decreased overall survival and discriminated high-risk breast cancer patients from low-risk patients with a homogeneous distribution of expression, i.e., nuclear and cytoplasmic expression. The homogeneous TRAIL-R expression was associated with favorable breast cancer surrogate markers corresponding with excellent survival prognoses at 5 years after diagnosis (hazard ratio, 0.043) and over the complete course of follow-up (hazard ratio, 0.098; both p < 0.001). No associations with specific intrinsic breast cancer subtypes were found. Our data suggest that the determination of intracellular co-expression patterns of TRAIL-R1, TRAIL-R2, and TRAIL-R4 provides an innovative and robust method for risk stratification in breast cancer patients beyond conventional prognostic markers. KEY MESSAGES: A total of 70% of breast cancer specimens show comparably high levels of intracellular TRAIL-Rs. Nuclear or cytoplasmic TRAIL-R co-expression occurs in the majority of tumors. A total of 25% of tumors show a heterogeneous expression of cytoplasmic or nuclear TRAIL-Rs. Patients with a heterogeneous TRAIL-R expression present with poor prognoses. Additive TRAIL-R-based risk stratification comprises different breast cancer subtypes.


Subject(s)
Biomarkers, Tumor/biosynthesis , Breast Neoplasms , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Tumor Necrosis Factor Decoy Receptors/biosynthesis , Aged , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Disease-Free Survival , Female , Humans , MicroRNAs/biosynthesis , Middle Aged , RNA, Neoplasm/biosynthesis , Retrospective Studies , Survival Rate
2.
J Hepatol ; 64(5): 1108-1117, 2016 May.
Article in English | MEDLINE | ID: mdl-26708145

ABSTRACT

BACKGROUND & AIMS: The liver exhibits a unique capacity for regeneration in response to injury. Lymphotoxin-ß receptor (LTßR), a core member of the tumor necrosis factor (TNF)/tumor necrosis factor receptor (TNFR) superfamily is known to play an important role in this process. However, the function of LTßR during pathophysiological alterations and its molecular mechanisms during liver regeneration are so far ill-characterized. METHODS: LTßR(-/-) mice were subjected to 70% hepatectomy and liver regeneration capacity, bile acid profiles, and transcriptome analysis were performed. RESULTS: LTßR(-/-) deficient mice suffered from increased and prolonged liver tissue damage after 70% hepatectomy, accompanied by deregulated bile acid homeostasis. Pronounced differences in the expression patterns of genes relevant for bile acid synthesis and recirculation were observed. LTßR and TNFRp55 share downstream signalling elements. Therefore, LTßR(-/-) mice were treated with etanercept to create mice functionally deficient in both signalling pathways. Strikingly, the combined blockade of TNFRp55 and LTßR signalling leads to complete failure of liver regeneration resulting in death within 24 to 48h after PHx. Transcriptome analysis revealed a marked disparity in gene expression programs in livers of LTßR(-/-) and etanercept-treated LTßR(-/-) vs. wild-type animals after PHx. Murinoglobulin 2 was identified as a significantly differentially regulated gene. CONCLUSIONS: LTßR is essential for efficient liver regeneration and cooperates with TNFRp55 in this process. Differences in survival kinetics strongly suggest distinct functions for these two cytokine receptors in liver regeneration. Failure of TNFR and LTßR signalling renders liver regeneration impossible.


Subject(s)
DNA/genetics , Gene Expression Regulation , Liver Diseases/genetics , Liver Regeneration/genetics , Lymphotoxin beta Receptor/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Tumor Necrosis Factor Decoy Receptors/genetics , Animals , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Liver Diseases/metabolism , Liver Diseases/pathology , Lymphotoxin beta Receptor/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Receptors, Tumor Necrosis Factor, Type I/biosynthesis , Tumor Necrosis Factor Decoy Receptors/biosynthesis
3.
PLoS One ; 10(9): e0138248, 2015.
Article in English | MEDLINE | ID: mdl-26378933

ABSTRACT

Loss of the chromosomal region 8p21 negatively effects survival in patients with multiple myeloma (MM) that undergo autologous stem cell transplantation (ASCT). In this study, we aimed to identify the immunological and molecular consequences of del(8)(p21) with regards to treatment response and bortezomib resistance. In patients receiving bortezomib as a single first line agent without any high-dose therapy, we have observed that patients with del(8)(p21) responded poorly to bortezomib with 50% showing no response while patients without the deletion had a response rate of 90%. In vitro analysis revealed a higher resistance to bortezomib possibly due to an altered gene expression profile caused by del(8)(p21) including genes such as TRAIL-R4, CCDC25, RHOBTB2, PTK2B, SCARA3, MYC, BCL2 and TP53. Furthermore, while bortezomib sensitized MM cells without del(8)(p21) to TRAIL/APO2L mediated apoptosis, in cells with del(8)(p21) bortezomib failed to upregulate the pro-apoptotic death receptors TRAIL-R1 and TRAIL-R2 which are located on the 8p21 region. Also expressing higher levels of the decoy death receptor TRAIL-R4, these cells were largely resistant to TRAIL/APO2L mediated apoptosis. Corroborating the clinical outcome of the patients, our data provides a potential explanation regarding the poor response of MM patients with del(8)(p21) to bortezomib treatment. Furthermore, our clinical analysis suggests that including immunomodulatory agents such as Lenalidomide in the treatment regimen may help to overcome this negative effect, providing an alternative consideration in treatment planning of MM patients with del(8)(p21).


Subject(s)
Bortezomib/therapeutic use , Drug Resistance, Neoplasm/genetics , Multiple Myeloma/genetics , Tumor Necrosis Factor Decoy Receptors/biosynthesis , Antineoplastic Agents/therapeutic use , Apoptosis/genetics , Chromosome Deletion , Chromosomes, Human, Pair 8/genetics , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Multiple Myeloma/drug therapy , Multiple Myeloma/mortality , Prognosis , RNA, Messenger/genetics , Tumor Necrosis Factor Decoy Receptors/genetics
4.
Oncol Rep ; 33(4): 1947-55, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25672292

ABSTRACT

Non-steroidal anti-inflammatory drugs (NSAIDs) have been suggested as the potential new class of preventive or therapeutic antitumor agents. The aim of the present study was to evaluate the antitumor activity of the novel NSAID, CG100649. CG100649 is a novel NSAID dual inhibitor for COX-2 and carbonic anhydrase (CA)-I/-II. In the present study, we investigated the alternative mechanism by which CG100649 mediated suppression of the colon cancer growth and development. The anchorage­dependent and -independent clonogenic assay showed that CG100649 inhibited the clonogenicity of human colon cancer cells. The flow cytometric analysis showed that CG100649 induced the G2/M cell cycle arrest in colon cancer cells. Animal studies showed that CG100649 inhibited the tumor growth in colon cancer xenograft in nude mice. Furthermore, quantitative PCR and FACS analysis demonstrated that CG100649 upregulated the expression of TNF-related apoptosis-inducing ligand (TRAIL) receptors (DR4 and DR5) but decreased the expression of decoy receptors (DcR1 and DcR2) in colon cancer cells. The results showed that CG100649 treatment sensitized TRAIL­mediated growth suppression and apoptotic cell death. The combination treatment resulted in significant repression of the intestinal polyp formation in APCmin/+ mice. Our data clearly demonstrated that CG100649 contains preventive and therapeutic activity for colon cancer. The present study may be useful for identification of the potential benefit of the NSAID CG100649, for the achievement of a better treatment response in colon cancer.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Colonic Neoplasms/drug therapy , Furans/therapeutic use , Lung Neoplasms/drug therapy , Sulfonamides/therapeutic use , TNF-Related Apoptosis-Inducing Ligand/therapeutic use , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anticarcinogenic Agents/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Colonic Neoplasms/pathology , Drug Synergism , Female , Furans/administration & dosage , Furans/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , GPI-Linked Proteins/biosynthesis , GPI-Linked Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Intestinal Polyps/prevention & control , Intestine, Small/pathology , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, Tumor Necrosis Factor, Member 10c/biosynthesis , Receptors, Tumor Necrosis Factor, Member 10c/genetics , Specific Pathogen-Free Organisms , Sulfonamides/administration & dosage , Sulfonamides/pharmacology , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Tumor Necrosis Factor Decoy Receptors/biosynthesis , Tumor Necrosis Factor Decoy Receptors/genetics , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
5.
Biomed Res Int ; 2013: 293874, 2013.
Article in English | MEDLINE | ID: mdl-24324958

ABSTRACT

The TNF-related apoptosis-inducing ligand (TRAIL) is a TNF family member which has been under intense focus because of its remarkable ability to induce apoptosis in malignant human cells while leaving normal cells unscathed. However, many cancer cells remain resistant to TRAIL. In this study, we had investigated the synergistic effects of low dose fluorouracil (5-Fu) and TRAIL on TRAIL-resistant human gastric adenocarcinoma AGS cells and explored the potential mechanisms. Cell viability was analyzed by sulforhodamine B (SRB) assay and the synergistic effects were evaluated by Jin's formula and confirmed by both morphological changes under inverted microscope and flow cytometry. The expression of TRAIL-R1 (death receptor 4, DR4), TRAIL-R2 (DR5), TRAIL-R3 (decoy receptor, DcR1), TRAIL-R4 (DcR2), procaspase-3, procaspase-8, and procaspase-9 was detected by western blotting. Our results showed that there were significant synergistic effects of low dose 5-Fu and TRAIL on TRAIL-resistant AGS cells, and this effect was supposed to be mediated by decreasing DcR2 expression and increasing DR5 expression. The extrinsic and intrinsic apoptosis pathways were both activated. The data suggest that combined treatment of low dose 5-Fu and TRAIL can be an effective therapeutic approach for gastric adenocarcinoma.


Subject(s)
Adenocarcinoma/drug therapy , Drug Synergism , Fluorouracil/administration & dosage , Stomach Neoplasms/drug therapy , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , Adenocarcinoma/pathology , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , GPI-Linked Proteins/biosynthesis , Gene Expression Regulation, Neoplastic , Humans , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Receptors, Tumor Necrosis Factor, Member 10c , Rhodamines/pharmacology , Stomach Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Necrosis Factor Decoy Receptors/biosynthesis
6.
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
7.
J Biol Regul Homeost Agents ; 25(3): 375-86, 2011.
Article in English | MEDLINE | ID: mdl-22023762

ABSTRACT

The biological activity of TNF-related apoptosis inducing ligand (TRAIL) was analyzed in primary human erythroblasts derived from mononuclear cells of blood donors, kept in culture in the presence of 20 percent foetal calf serum, growth factors (EPO, SCF, IL-3) and glucocorticoids (10-6 M dexamethasone, 10-6 M oestradiol) or under growth factor and serum starvation. In the presence of growth factors and serum, primary erythroblasts showed a differential expression of TRAIL-Receptors (Rs) at various degrees of maturation and responded to TRAIL treatment with a mild cytotoxicity. On the other hand, in the absence of serum and growth factors, TRAIL treatment unexpectedly up-regulated TRAIL-R4 decoy receptor and promoted erythroblast survival. The concomitant activation of NF-kB/IkB survival pathway was detected with Western blotting and immunofluorescence procedures and confirmed by experiments performed with SN50, a pharmacological inhibitor of the NF-kB/IkB pathway. Our study indicates that TRAIL has a twofold activity on erythroid lineages: it induces a mild erythroid cell cytotoxicity in the presence of serum and growth factors, while it promotes erythroid cell survival through the activation of the NF-kB/IkB pathway under starvation conditions.


Subject(s)
Erythroblasts/metabolism , Erythropoietin , I-kappa B Kinase/metabolism , NF-kappa B/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Cell Survival/drug effects , Cell Survival/physiology , Erythroblasts/cytology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Jurkat Cells , NF-kappa B/antagonists & inhibitors , Peptides/pharmacology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Necrosis Factor Decoy Receptors/biosynthesis
8.
Leuk Res ; 35(6): 741-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21281967

ABSTRACT

Activation of the TNF-related apoptosis-inducing ligand (TRAIL) pathway can induce apoptosis in a broad range of human cancer cells. Four membrane-bound receptors have been identified. TRAIL-R1 and TRAIL-R2 contain a functional death domain; TRAIL-R3 and TRAIL-R4 lack a functional death domain and function as decoy receptors. Flow-cytometric analysis revealed that acute myeloid leukemic (AML) blasts expressed significantly more pro-apoptotic receptors compared to normal blasts. However, about 20% of AML patients highly expressed decoy receptor TRAIL-R3, which was strongly correlated to a shortened overall survival. TRAIL-R3 expression was also high on CD34+/CD38- cells, the compartment that harbors the leukemia initiating stem cell. Expression levels of pro-apoptotic TRAIL receptors were not correlated to the susceptibility for soluble TRAIL, which was generally low (mean level of cell death induction 14%). Cell death could be enhanced by down-modulation of TRAIL-R3, confirming its decoy function on AML blasts. Bypassing of TRAIL-R3 by treatment with antibodies directly targeting TRAIL-R2 resulted in higher rates of induced cell death (max. 80%). In conclusion, AML blasts do express pro-apoptotic TRAIL receptors. However, co-expression of decoy receptor TRAIL-R3 results in significant shortened overall survival. AML blasts could be targeted by anti-TRAIL-R2 antibodies, yielding a new therapeutic option for AML patients.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Neoplastic Stem Cells/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Tumor Necrosis Factor Decoy Receptors/biosynthesis , Adolescent , Adult , Aged , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Apoptosis/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Line , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Female , GPI-Linked Proteins/biosynthesis , HL-60 Cells , Humans , Kaplan-Meier Estimate , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Neoplastic Stem Cells/pathology , Prognosis , Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Member 10c , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Treatment Outcome , U937 Cells , Young Adult
9.
Cancer Res ; 71(2): 538-49, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21127198

ABSTRACT

Because tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) selectively kills tumor cells, it is being tested in cancer patients. Unfortunately, patients develop resistance to the cytokine, therefore, agents that can sensitize cells to TRAIL are urgently needed. In this study, we investigated whether dibenzylideneacetone (DBA) can sensitize cancer cells to TRAIL and potentiates TRAIL-induced apoptosis. As indicated by accumulation of the membrane phospholipid phosphatidylserine, DNA breaks, intracellular esterase activity, and activation of caspase-8, -9, and -3, we concluded that DBA potentiated TRAIL-induced apoptosis in colon cancer cells. DBA also converted TRAIL resistant-cells to TRAIL-sensitive. When examined for the mechanism, we found that DBA decreased the expression of antiapoptotic proteins and decoy receptor-2 and increased proapoptotic proteins. DBA also induced both death receptor (DR)-5 and DR4. Knockdown of DR5 and DR4 by small interfering RNA (SiRNA) reduced the sensitizing effect of DBA on TRAIL-induced apoptosis. In addition, DBA increased the expression of CHOP proteins. Knockdown of CHOP by siRNA decreased the induction of DBA-induced DR5 expression and apoptosis. Induction of receptors by DBA, however, was p53-independent, as deletion of p53 had no effect on receptor induction. We observed that DBA-induced induction of DR5 and DR4 was mediated through generation of reactive oxygen species (ROS), as N-acetylcysteine blocked the induction of death receptors and suppression of cell survival proteins by DBA. Overall, our results show that DBA potentiates TRAIL-induced apoptosis through downregulation of cell survival proteins and upregulation of death receptors via activation of ROS and CHOP mediated pathways.


Subject(s)
Pentanones/pharmacology , Reactive Oxygen Species/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Transcription Factor CHOP/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Down-Regulation/drug effects , Drug Synergism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HCT116 Cells , HT29 Cells , Humans , Mitogen-Activated Protein Kinases/metabolism , PPAR gamma/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Death Domain/biosynthesis , Receptors, Death Domain/genetics , Tumor Necrosis Factor Decoy Receptors/biosynthesis , Tumor Necrosis Factor Decoy Receptors/genetics , Up-Regulation/drug effects
10.
BMC Musculoskelet Disord ; 11: 192, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20799941

ABSTRACT

BACKGROUND: Rheumatoid Arthritis (RA) is a chronic autoimmune inflammatory disorder. Although the pathogenesis of disease is unclear, it is well known that T cells play a major role in both development and perpetuation of RA through activating macrophages and B cells. Since the lack of TNF-Related Apoptosis Inducing Ligand (TRAIL) expression resulted in defective thymocyte apoptosis leading to an autoimmune disease, we explored evidence for alterations in TRAIL/TRAIL receptor expression on peripheral T lymphocytes in the molecular mechanism of RA development. METHODS: The expression of TRAIL/TRAIL receptors on T cells in 20 RA patients and 12 control individuals were analyzed using flow cytometry. The correlation of TRAIL and its receptor expression profile was compared with clinical RA parameters (RA activity scored as per DAS28) using Spearman Rho Analysis. RESULTS: While no change was detected in the ratio of CD4+ to CD8+ T cells between controls and RA patient groups, upregulation of TRAIL and its receptors (both death and decoy) was detected on both CD4+ and CD8+ T cells in RA patients compared to control individuals. Death Receptor-4 (DR4) and the decoy receptors DcR1 and DcR2 on CD8+ T cells, but not on CD4+ T cells, were positively correlated with patients' DAS scores. CONCLUSIONS: Our data suggest that TRAIL/TRAIL receptor expression profiles on T cells might be important in revelation of RA pathogenesis.


Subject(s)
Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , CD8-Positive T-Lymphocytes/immunology , Receptors, Tumor Necrosis Factor/biosynthesis , Tumor Necrosis Factor Decoy Receptors/biosynthesis , Arthritis, Rheumatoid/pathology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Female , GPI-Linked Proteins/biosynthesis , Humans , Male , Predictive Value of Tests , Receptors, Tumor Necrosis Factor, Member 10c , Sensitivity and Specificity , Severity of Illness Index
11.
J Med Virol ; 82(8): 1392-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20572077

ABSTRACT

The hallmark of the dengue hemorrhagic fever/dengue shock syndrome is hematologic abnormality. The pathogenesis of dengue hemorrhagic fever/dengue shock syndrome remains unknown. Our work showed that the dengue virus serotype-2 induced apoptosis in human umbilical vein endothelial cells. Fas (CD95), Tumor Necrosis Factor receptors, and Tumor Necrosis Factor-related apoptosis-inducing ligand receptors are the most common death receptors, which can induce apoptosis. Compared with the untreated human umbilical vein endothelial cells, Fas expression was increased both in the mRNA level and on the surface of infected human umbilical vein endothelial cells. FasL was expressed at similar levels on human umbilical vein endothelial cells over a course of dengue virus serotype-2 infection, but the expression in mRNA level was increased in infected human umbilical vein endothelial cells. It is possible that there is soluble FasL secreted from human umbilical vein endothelial cells in the supernatant. Tumor Necrosis Factor-related apoptosis-inducing ligand receptor 1 and Tumor Necrosis Factor receptors 1-2 were constantly very low, whereas Tumor Necrosis Factor-related apoptosis-inducing ligand receptors 2-4 decreased after dengue virus serotype-2 infection. This result suggested that dengue virus serotype-2 may inhibit Tumor Necrosis Factor-related apoptosis-inducing ligand receptors-induced apoptosis. The apoptotic rates in human umbilical vein endothelial cells were decreased upon the addition of caspase family inhibitors. In addition, activated caspase 8 and caspase 3 were also observed by Western blot following dengue virus serotype-2 infection. Thus, it is shown that the Fas/FasL pathway may participate in dengue virus-induced apoptosis of vascular endothelial cells in vitro.


Subject(s)
Apoptosis , Dengue Virus/pathogenicity , Endothelial Cells/virology , Fas Ligand Protein/biosynthesis , fas Receptor/biosynthesis , Down-Regulation , Humans , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor, Type I/biosynthesis , Receptors, Tumor Necrosis Factor, Type II/biosynthesis , Tumor Necrosis Factor Decoy Receptors/biosynthesis , Up-Regulation
12.
J Allergy Clin Immunol ; 125(6): 1261-1268.e9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20471070

ABSTRACT

BACKGROUND: Previous work has identified CD11c(+)CD1c(-) dendritic cells (DCs) as the major "inflammatory" dermal DC population in patients with psoriasis vulgaris and CD1c(+) DCs as the "resident" cutaneous DC population. OBJECTIVE: We sought to further define molecular differences between these 2 myeloid dermal DC populations. METHODS: Inflammatory and resident DCs were single-cell sorted from lesional skin biopsy specimens of patients with psoriasis, and the transcriptome of CD11c(+)CD1c(-) versus CD1c(+) DCs was determined. Results were confirmed with RT-PCR, flow cytometry, immunohistochemistry, and double-labeled immunofluorescence. Human keratinocytes were cultured for functional studies. RESULTS: TNF-related apoptosis-inducing ligand (TRAIL), Toll-like receptors 1 and 2, S100A12/ENRAGE, CD32, and many other inflammatory products were differentially expressed in inflammatory DCs compared with resident DCs. Flow cytometry and immunofluorescence confirmed higher protein expression on CD1c(-) versus CD1c(+) DCs. TRAIL receptors, death receptor 4, and decoy receptor 2 were expressed in keratinocytes and dermal cells. In vitro culture of keratinocytes with TRAIL induced CCL20 chemokine. CONCLUSIONS: CD11c(+)CD1c(-) inflammatory DCs in psoriatic lesional skin express a wide range of inflammatory molecules compared with skin-resident CD1c(+) DCs. Some molecules made by inflammatory DCs, including TRAIL, could have direct effects on keratinocytes or other skin cell types to promote disease pathogenesis.


Subject(s)
Biomarkers/metabolism , Langerhans Cells/metabolism , Psoriasis/diagnosis , Psoriasis/immunology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Differentiation , Cell Separation , Cells, Cultured , Chemokine CCL20/biosynthesis , Chemokine CCL20/genetics , Flow Cytometry , Gene Expression Profiling , Humans , Immunohistochemistry , Inflammation , Keratinocytes/immunology , Keratinocytes/metabolism , Keratinocytes/pathology , Langerhans Cells/immunology , Langerhans Cells/pathology , Microarray Analysis , Psoriasis/pathology , S100 Proteins/biosynthesis , S100 Proteins/genetics , Tumor Necrosis Factor Decoy Receptors/biosynthesis , Tumor Necrosis Factor Decoy Receptors/genetics
13.
Virchows Arch ; 456(1): 39-44, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19936781

ABSTRACT

The tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a member of the TNF family, which mediates apoptosis by the extrinsic pathway. Up-regulation of decoy receptors, DcR1 and DcR2, may result in diminished binding of TRAIL to their functional receptors. DcR1 expression was assessed in normal endometrial tissue (NE) and endometrial carcinoma (EC) samples by immunohistochemistry (IHC) and quantitative real-time polymerase chain reaction (PCR). IHC was performed in two tissue microarrays; one composed of 80 samples of NE and a second one constructed from paraffin-embedded blocks of 62 EC. For quantitative real-time RT-PCR analysis, RNA was obtained from 19 NE and 28 EC samples using Trizol. mRNA expression of DcR1 was assessed with Taqman-based assays in an Abi-Prism 700 SDS. Results were correlated with stage, histological type, and grade. By IHC, cytoplasmic expression of DcR1 was frequently seen in NE (79.6%) and varied according to the menstrual cycle. Positive DcR1 immunostaining was also detected in EC (98.1% of the cases) without any specific statistical association with histological type, grade, and stage. By quantitative real-time PCR, all NE had similar levels of DcR1expression (0.8-1.7 RQ), which were considered the basal levels of DcR1 expression in NE. Increased DcR1 expression (> or =5-fold higher than the basal levels) was detected in 13 of 28 EC (46.4%). High DcR1 expression levels were found in ECs of different stages: IA, four of 12 (33%); IB, two of four (50%); IC, four of six (66%); and IIA and IIB three of six (50%). Results suggest that DcR1 expression occurs in a subset of EC and may contribute to resistance to TRAIL-induced apoptosis.


Subject(s)
Endometrial Neoplasms/metabolism , Endometrium/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Apoptosis/physiology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Female , GPI-Linked Proteins , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Microarray Analysis , Receptors, Tumor Necrosis Factor, Member 10c , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor Decoy Receptors/biosynthesis , Tumor Necrosis Factor Decoy Receptors/genetics
14.
Gene Ther ; 16(7): 862-73, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19440225

ABSTRACT

Intraocular inflammation has been recognized as a major factor leading to blindness. Because tumor necrosis factor-alpha (TNF-alpha) enhances intraocular cytotoxic events, systemic anti-TNF therapies have been introduced in the treatment of severe intraocular inflammation, but frequent re-injections are needed and are associated with severe side effects. We have devised a local intraocular nonviral gene therapy to deliver effective and sustained anti-TNF therapy in inflamed eyes. In this study, we show that transfection of the ciliary muscle by plasmids encoding for three different variants of the p55 TNF-alpha soluble receptor, using electrotransfer, resulted in sustained intraocular secretion of the encoded proteins, without any detection in the serum. In the eye, even the shorter monomeric variant resulted in efficient neutralization of TNF-alpha in a rat experimental model of endotoxin-induced uveitis, as long as 3 months after transfection. A subsequent downregulation of interleukin (IL)-6 and iNOS and upregulation of IL-10 expression was observed together with a decreased rolling of inflammatory cells in anterior segment vessels and reduced infiltration within the ocular tissues. Our results indicate that using a nonviral gene therapy strategy, the local self-production of monomeric TNF-alpha soluble receptors induces a local immunomodulation enabling the control of intraocular inflammation.


Subject(s)
Ciliary Body/metabolism , Genetic Therapy/methods , Muscle, Smooth/metabolism , Receptors, Tumor Necrosis Factor, Type I/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Tumor Necrosis Factor Decoy Receptors/biosynthesis , Uveitis/therapy , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Electroporation/methods , Endotoxins/adverse effects , Eye/metabolism , Female , Gene Transfer Techniques , Genes, Reporter , Humans , Immunomodulation , Interleukin-10/metabolism , Interleukin-6/metabolism , Lac Operon/genetics , Leukocyte Rolling , Microscopy, Confocal , Nitric Oxide Synthase Type II/metabolism , Plasmids , Rats , Rats, Inbred Lew , Receptors, Tumor Necrosis Factor, Type I/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Transfection/methods , Tumor Necrosis Factor Decoy Receptors/metabolism , Tumor Necrosis Factor-alpha/adverse effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
15.
Urol Oncol ; 26(2): 158-65, 2008.
Article in English | MEDLINE | ID: mdl-18312935

ABSTRACT

OBJECTIVE: Tumor necrosis factor-related apoptosis inducing ligand (TRAIL) has recently been investigated because of its ability to selectively kill cancer cells. Despite recent publications mainly focusing on TRAIL resistance in cancer cells, little is known about how TRAIL contributes to the carcinogenesis process. Because the expression patterns of TRAIL and its receptors in patients with prostate carcinoma have recently been reported, this study investigated the significance of TRAIL and TRAIL receptor expression in connection to serum prostate-specific antigen (PSA) and Gleason scoring. MATERIALS AND METHODS: A total of 98 patients were included in the study. Gleason scores, PSA, TRAIL, and TRAIL receptor expressions were used for the comparison purposes. The Spearman rho correlation test was administered to reveal the correlations among the variants. The Kruskal Wallis-Mann Whitney U or Friedman-Wilcoxon signed ranks test determined the statistical significance between the pairs. Multinomial and/or multiple binary logistic regression analyses were deployed to test whether TRAIL markers were independent variables to predict the prognosis of prostate cancer. Kaplan-Meier and log-rank tests were used to determine the survival rates. RESULTS: High-serum PSA levels were correlated with higher levels of TRAIL and TRAIL receptor expressions. Patients with high Gleason scores had higher levels of TRAIL-R4 decoy receptor expression but lower levels of TRAIL death ligand expression. CONCLUSIONS: TRAIL-R4 decoy receptor expression is strongly correlated with PSA recurrence, which is suggestive of poor prognosis. High levels of TRAIL-R4 expression but low levels of TRAIL death ligand expression are connected to decreased survival.


Subject(s)
Prostate-Specific Antigen/blood , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/mortality , Tumor Necrosis Factor Decoy Receptors/biosynthesis , Humans , Male , Recurrence , Survival Rate
16.
Cancer Gene Ther ; 14(12): 976-84, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17853923

ABSTRACT

High levels of decoy receptor 2 (DcR2; TRAIL-R4) expression are correlated with TRAIL resistance in prostate cancer cells. In addition, upregulation of TRAIL death receptor (DR4 and DR5) expression, either by ionizing radiation or chemotherapy, can sensitize cancer cells to TRAIL. Considering more than half of human cancers are TRAIL resistant, modulation of surface TRAIL receptor expression appears to be an attractive treatment modality to counteract TRAIL resistance. In this study, three siRNA duplexes targeting DcR2 receptor were tested. Ad5hTRAIL infections were performed to overexpress human full-length TRAIL to induce cell death, and the in vitro tumorigenic potential of prostate cancer cells was assessed using colony-forming assays on soft agar. The DU145 and LNCaP prostate cancer cell lines, which express high levels of DcR2, were resistant to Ad5hTRAIL-induced death. Downregulation of surface DcR2 expression by siRNA sensitized these prostate cancer cell lines to Ad5hTRAIL. In addition, DcR2 siRNA-mediated knockdown of DcR2, followed by Ad5hTRAIL infection, dramatically reduced the in vitro tumorigenic potential of prostate cancer cells. Collectively, our results suggest the potential for combining receptor-specific siRNA with TRAIL in the treatment of certain cancers.


Subject(s)
Adenoviridae , Genetic Therapy , Neoplasm Proteins/antagonists & inhibitors , Prostatic Neoplasms/metabolism , RNA, Small Interfering/biosynthesis , Transduction, Genetic , Tumor Necrosis Factor Decoy Receptors/antagonists & inhibitors , Animals , COS Cells , Cell Death/genetics , Cell Line, Tumor , Chlorocebus aethiops , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , RNA, Small Interfering/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factor Decoy Receptors/biosynthesis , Tumor Necrosis Factor Decoy Receptors/genetics , Up-Regulation/genetics
17.
Clin Cancer Res ; 13(11): 3191-7, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17545522

ABSTRACT

PURPOSE: Epigenetic aberrations have been shown to play an important role in the pathogenesis of most cancers. To investigate the clinical significance of epigenetic changes in neuroblastoma, we evaluated the relationship between clinicopathologic variables and the pattern of gene methylation in neuroblastoma cell lines and tumors. EXPERIMENTAL DESIGN: Methylation-specific PCR was used to evaluate the gene methylation status of 19 genes in 14 neuroblastoma cell lines and 8 genes in 70 primary neuroblastoma tumors. Associations between gene methylation, established prognostic factors, and outcome were evaluated. Log-rank tests were used to identify the number of methylated genes that was most predictive of overall survival. RESULTS: Epigenetic changes were detected in the neuroblastoma cell lines and primary tumors, although the pattern of methylation varied. Eight of the 19 genes analyzed were methylated in >70% of the cell lines. Epigenetic changes of four genes were detected in only small numbers of cell lines. None of the cell lines had methylation of the other seven genes analyzed. In primary neuroblastoma tumors, high-risk disease and poor outcome were associated with methylation of DCR2, CASP8, and HIN-1 individually. Although methylation of the other five individual genes was not predictive of poor outcome, a trend toward decreased survival was seen in patients with a methylation phenotype, defined as > or =4 methylated genes (P = 0.055). CONCLUSION: Our study indicates that clinically aggressive neuroblastoma tumors have aberrant methylation of multiple genes and provides a rationale for exploring treatment strategies that include demethylating agents.


Subject(s)
Caspase 8/biosynthesis , Cytokines/biosynthesis , Gene Expression Regulation, Neoplastic , Methylation , Neuroblastoma/metabolism , Tumor Necrosis Factor Decoy Receptors/biosynthesis , Tumor Suppressor Proteins/biosynthesis , Caspase 8/genetics , Cell Line, Tumor , Cohort Studies , Cytokines/genetics , Epigenesis, Genetic , Female , Humans , Infant , Infant, Newborn , Male , Treatment Outcome , Tumor Necrosis Factor Decoy Receptors/genetics , Tumor Suppressor Proteins/genetics
18.
Hum Gene Ther ; 18(1): 39-50, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17187448

ABSTRACT

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) selectively induces apoptosis in cancer cells but not in normal cells. However, studies have indicated that more than half of human tumors exhibit TRAIL resistance. Although the mechanism of TRAIL resistance is not understood, it represents a barrier to any TRAIL-mediated gene therapy approach. In addition, no correlation between TRAIL receptor (TRAIL-R) expression profile and TRAIL resistance has been demonstrated in cancer cells. In this study, three different lung cancer cell lines and three different primary cell cultures established from patients with lung cancer (two patients with squamous cell lung carcinoma and one with adenocarcinoma) were screened for sensitivity to adenoviral delivery of TRAIL. Whereas TRAIL-resistant primary lung cell cultures and the A549 lung cancer cell line exhibited high levels of surface decoy receptor-2 (DcR2/TRAIL-R4) expression, TRAIL-sensitive lung cancer cell lines (HBE and H411) failed to express it. A DcR2 short interfering RNA (siRNA) approach involving three different siRNA constructs in combination downregulated DcR2/TRAIL-R4 expression and sensitized lung cancer cells to TRAIL-induced apoptosis. Immunohistochemical staining of samples from 10 patients with lung carcinoma suggested that high-level DcR2/TRAIL-R4 expression is a common phenotype observed in patients with non-small cell lung carcinoma.


Subject(s)
Adenocarcinoma/therapy , Adenoviridae , Apoptosis , Genetic Therapy , Lung Neoplasms/therapy , Neoplasms, Squamous Cell/therapy , RNA, Small Interfering , Tumor Necrosis Factor Decoy Receptors/biosynthesis , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Cell Line, Tumor , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Ligands , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Neoplasms, Squamous Cell/genetics , Neoplasms, Squamous Cell/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Necrosis Factor Decoy Receptors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...