Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Breast Cancer ; 30(5): 758-771, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37199905

ABSTRACT

BACKGROUND: The therapy concepts that target several members of krüppel like factor (KLF) family have been achieved in breast cancer (BC). However, the role of KLF11 in BC remains unclear. This study explored the prognostic significance of KLF11 in BC patients and investigated its functional roles in this malignancy. METHODS: Immunohistochemistry (IHC) staining of KLF11 in 298 patients' samples was performed to determine the prognostic role of the KLF11. Then the protein level was correlated to clinicopathological characteristics and survival outcomes. Afterward, the function of KLF11 was explored in vitro with siRNA-mediated loss-of-function of cell viability, proliferation, and apoptosis. RESULTS: From the cohort study, we found that the expression of KLF11 was positively associated with highly proliferative BC of BC. Furthermore, prognostic analysis demonstrated that KLF11 was an independent negative factor for disease-free survival (DFS) and distant-metastasis-free survival (DMFS) of BC. The KLF11-related prognostic model for DFS and DMFS showed high accuracy in predicting the 3-,5- and 10 -year survival probability of BC patients. Additionally, the knockdown of KLF11 inhibited cell viability and proliferation, as well as induced cell apoptosis in MCF7 and MDA-MB-231 cells, while only inhibited cell viability and induced cell apoptosis in SK-BR-3 cells. CONCLUSIONS: Our study indicated that targeting KLF11 is an interesting therapeutic concept and further research could lead to a new therapeutic improvement in BC, especially in highly aggressive molecular subtypes.


Subject(s)
Breast Neoplasms , Repressor Proteins , Humans , Female , Repressor Proteins/metabolism , Breast Neoplasms/genetics , Prognosis , Cohort Studies , Apoptosis Regulatory Proteins , Apoptosis , Cell Proliferation
2.
Biomedicines ; 10(2)2022 Jan 25.
Article in English | MEDLINE | ID: mdl-35203468

ABSTRACT

Aging is the main cause of decline in oocyte quality, which can further trigger the failure of assisted reproductive technology (ART). Exploring age-related genes in oocytes is an important way to investigate the molecular mechanisms involved in oocyte aging. To provide novel insight into this field, we performed a pooled analysis of publicly available datasets, using the overlapping results of two statistical methods on two Gene Expression Omnibus (GEO) datasets. The methods utilized in the current study mainly include Spearman rank correlation, the Wilcoxon signed-rank test, t-tests, Venn diagrams, Gene Ontology (GO), Protein-Protein Interaction (PPI), Gene Set Enrichment Analysis (GSEA), Gene Set Variation Analysis (GSVA), and receiver operating characteristic (ROC) curve analysis. We identified hundreds of age-related genes across different gene expression datasets of in vitro maturation-metaphase II (IVM-MII) oocytes. Age-related genes in IVM-MII oocytes were involved in the biological processes of cellular metabolism, DNA replication, and histone modifications. Among these age-related genes, FAM111A expression presented a robust correlation with age, seen in the results of different statistical methods and different datasets. FAM111A is associated with the processes of chromosome segregation and cell cycle regulation. Thus, this enzyme is potentially an interesting novel marker for the aging of oocytes, and warrants further mechanistic study.

3.
Biomedicines ; 9(12)2021 Dec 14.
Article in English | MEDLINE | ID: mdl-34944731

ABSTRACT

In vitro maturation (IVM) of oocytes is a promising assisted reproductive technology (ART) deemed as a simple and safe procedure. It is mainly used in patients with impaired oocyte maturation and in fertility preservation for women facing the risk of losing fertility. However, to date, it is still not widely used in clinical practice because of its underperformance. The influencing factors, such as biphasic IVM system, culture medium, and the supplementation, have a marked effect on the outcomes of oocyte IVM. However, the role of different culture media, supplements, and follicular priming regimens in oocyte IVM have yet to be fully clarified and deserve further investigation.

4.
Int J Mol Sci ; 22(17)2021 Aug 24.
Article in English | MEDLINE | ID: mdl-34502044

ABSTRACT

Implantation consists of a complex process based on coordinated crosstalk between the endometrium and trophoblast. Furthermore, it is known that the microenvironment of this fetal-maternal interface plays an important role in the development of extravillous trophoblast cells. This is mainly due to the fact that tissues mediate embryonic signaling biologicals, among other molecules, prostaglandins. Prostaglandins influence tissue through several cell processes including differentiation, proliferation, and promotion of maternal immune tolerance. The aim of this study is to investigate the potential pathological mechanism of the prostaglandin E2 receptor 4 (EP4) in modulating extravillous trophoblast cells (EVTs) in unexplained recurrent marriage (uRM). Our results indicated that the expression of EP4 in EVTs was decreased in women experiencing uRM. Furthermore, silencing of EP4 showed an inhibition of the proliferation and induced apoptosis in vitro. In addition, our results demonstrated reductions in ß- human chorionic gonadotropin (hCG), progesterone, and interleukin (IL)-6, which is likely a result from the activation of the cyclic adenosine monophosphate (cAMP)- cAMP-dependent protein kinase A (PKA)-phosphorylating CREB (pCREB) pathway. Our data might provide insight into the mechanisms of EP4 linked to trophoblast function. These findings help build a more comprehensive understanding of the effects of EP4 on the trophoblast at the fetal-maternal interface in the first trimester of pregnancy.


Subject(s)
Abortion, Habitual/metabolism , Cyclic AMP/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Signal Transduction , Trophoblasts/metabolism , Abortion, Habitual/pathology , Adult , Apoptosis , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Gonadotropins/metabolism , Humans , Interleukin-6/metabolism , Middle Aged , Pregnancy , Progesterone/metabolism
5.
Int J Mol Sci ; 22(16)2021 Aug 09.
Article in English | MEDLINE | ID: mdl-34445278

ABSTRACT

Epigenetics play a vital role in early embryo development. Offspring conceived via assisted reproductive technologies (ARTs) have a three times higher risk of epigenetic diseases than naturally conceived children. However, investigations into ART-associated placental histone modifications or sex-stratified analyses of ART-associated histone modifications remain limited. In the current study, we carried out immunohistochemistry, chip-sequence analysis, and a series of in vitro experiments. Our results demonstrated that placentas from intra-cytoplasmic sperm injection (ICSI), but not in vitro fertilization (IVF), showed global tri-methylated-histone-H3-lysine-4 (H3K4me3) alteration compared to those from natural conception. However, for acetylated-histone-H3-lysine-9 (H3K9ac) and acetylated-histone-H3-lysine-27 (H3K27ac), no significant differences between groups could be found. Further, sex -stratified analysis found that, compared with the same-gender newborn cord blood mononuclear cell (CBMC) from natural conceptions, CBMC from ICSI-boys presented more genes with differentially enriched H3K4me3 (n = 198) than those from ICSI-girls (n = 79), IVF-girls (n = 5), and IVF-boys (n = 2). We also found that varying oxygen conditions, RNA polymerase II subunit A (Polr2A), and lysine demethylase 5A (KDM5A) regulated H3K4me3. These findings revealed a difference between IVF and ICSI and a difference between boys and girls in H3K4me3 modification, providing greater insight into ART-associated epigenetic alteration.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Epigenesis, Genetic , Histones/metabolism , Retinoblastoma-Binding Protein 2/metabolism , Sex Characteristics , Sperm Injections, Intracytoplasmic , Adult , Female , Humans , Infant, Newborn , Male , Methylation , Pregnancy
6.
Int J Mol Sci ; 22(4)2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33670444

ABSTRACT

Sialic acid-binding immunoglobulin-like lectins (Siglecs) are involved in various immune cell-mediated diseases. Their role in cancer is poorly investigated, and research focusses on Siglec-expression on immune cells interacting with tumor cells. This study evaluates the role of Siglec-8 in breast cancer (BC). Siglec-8 expression was analyzed immunohistochemically on 235 primary BC cases and was correlated with clinical and pathological parameters and outcome. Cell culture experiments were performed with various BC cell lines. Siglec-8 was expressed in 215 BC cases and expression was lowest in triple-negative BC. It correlated with estrogen receptor-status, grading and the prognostic factors galectin (Gal)-7 and tumor-associated mucin-1 (TA-MUC1). However, Gal-7 and TA-MUC1 were only prognosticators for clinical outcome in the cohort expressing high (Immunoreactivity score IRS > 3) Siglec-8 levels but not in the low-expressing cohort. Siglec-8 knockdown led to a significantly reduced Gal-7 expression in MCF7 cells. All BC cell lines expressed low Siglec-8-levels, that could be elevated in MCF7 by Peroxisome proliferator-activated receptor (PPARγ)-stimulation. This study demonstrates that Siglec-8 is expressed in BC cells and correlates with known clinical and prognostic parameters. It is probably associated with Gal-7 and TA-MUC1 and might be regulated via PPARγ. Further analyses focusing on functional associations will clarify Siglec-8's eligibility as a possible therapeutic target.


Subject(s)
Antigens, CD/biosynthesis , Antigens, Differentiation, B-Lymphocyte/biosynthesis , Breast Neoplasms/metabolism , Lectins/biosynthesis , Receptors, Estrogen/metabolism , Adult , Aged , Antigens, CD/genetics , Antigens, Differentiation, B-Lymphocyte/genetics , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Cell Line, Tumor , Galectins/biosynthesis , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry/methods , Lectins/genetics , MCF-7 Cells , Middle Aged , Mucin-1/biosynthesis , Neoplasm Grading , Prognosis
7.
Int J Mol Sci ; 22(1)2021 Jan 04.
Article in English | MEDLINE | ID: mdl-33406768

ABSTRACT

Peroxisome proliferator-activated receptors (PPARα, PPARß/δ, and PPARγ) belong to the transcription factor family, and they are highly expressed in all types of trophoblast during pregnancy. The present review discusses currently published papers that are related to the regulation of PPARs via lipid metabolism, glucose metabolism, and amino acid metabolism to affect trophoblast physiological conditions, including differentiation, maturation, secretion, fusion, proliferation, migration, and invasion. Recent pieces of evidence have proven that the dysfunctions of PPARs in trophoblast lead to several related pregnancy diseases such as recurrent miscarriage, preeclampsia, intrauterine growth restriction, and gestational diabetes mellitus. Moreover, the underlying mechanisms of PPARs in the control of these processes have been discussed as well. Finally, this review's purposes are to provide more knowledge about the role of PPARs in normal and disturbed pregnancy with trophoblast, so as to find PPAR ligands as a potential therapeutic target in the treatment and prevention of adverse pregnancy outcomes.


Subject(s)
Gene Expression Regulation , Peroxisome Proliferator-Activated Receptors/metabolism , Trophoblasts/physiology , Animals , Female , Humans , Pregnancy , Trophoblasts/cytology
8.
J Reprod Immunol ; 142: 103210, 2020 11.
Article in English | MEDLINE | ID: mdl-33011635

ABSTRACT

BACKGROUD: Prostaglandin E2 (PGE2), an inflammatory mediator, modulates cytokines, regulates immune responses in reproductive processes and stimulates inflammatory reactions via the prostaglandin E2 receptor 2 (EP2). However, the regulatory effects of EP2 signaling on trophoblasts and its role in unexplained recurrent miscarriage (uRM) remains unclear. PATIENTS AND METHODS: A total of 19 placentas from patients with a history of more than two consecutive pregnancy losses of unknown cause (uRM group) and placentas of 19 healthy patients following a legal termination of their pregnancy were used for PGE2 receptor (EP1, EP2 and EP4) expression analyses via immunohistochemistry. Double immunofluorescence was also used to identify EP2 expressing cells in the decidua. Finally, HTR-8/SVneo cells were used to clarify the role of EP2 in in vitro experiments. RESULTS: The expression of EP2 and EP4 was found to be reduced in the syncytiotrophoblast and decidua of uRM patients. A selective EP2 receptor antagonist (PF-04,418,948) reduced the proliferation and secretion of ß-hCG, inhibited interleukin -6 (IL-6) and interleukin-8 (IL-8) and up-regulated the production of the tumor necrosis factor-α (TNF-α) and plasminogen activator inhibitor type 1 (PAI-1) in HTR-8/SVneo cells in vitro. CONCLUSION: PGE2-EP2 signaling pathway may represent a novel therapy option for uRM. The involvement of EP2 in uRM acts perhaps via inflammatory cytokines and indicates that the PGE2-EP2 signaling pathway might represent an unexplored etiology for uRM.


Subject(s)
Abortion, Habitual/immunology , Cytokines/metabolism , Dinoprostone/metabolism , Receptors, Prostaglandin E, EP2 Subtype/genetics , Adult , Cell Line , Cell Proliferation/drug effects , Decidua/immunology , Decidua/metabolism , Down-Regulation/immunology , Female , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/immunology , Humans , Immunohistochemistry , Middle Aged , Pregnancy , Receptors, Prostaglandin E, EP2 Subtype/analysis , Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/analysis , Receptors, Prostaglandin E, EP4 Subtype/genetics , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Trophoblasts/drug effects , Trophoblasts/immunology , Trophoblasts/metabolism
9.
Onco Targets Ther ; 12: 6053-6068, 2019.
Article in English | MEDLINE | ID: mdl-31534346

ABSTRACT

Purpose: COX-2 overexpression and elevated levels of prostaglandin E2 (PGE2) play an important role in breast cancer carcinogenesis. Recently, expression of the PGE2 receptor EP3 has been shown to be a positive prognostic factor in breast cancer. This study analyzes the functional aspects of targeting EP3 in breast cancer cell lines. Material and methods: EP3 and EP1 expressions were determined in five breast cancer cell lines on the mRNA- and the protein-level. The selected cell lines were subsequently stimulated for 24-72 hrs with 10-1,000 nM of PGE2, the EP1/EP3 agonist sulprostone and the EP3 antagonist L798,106. Cell proliferation was determined via BrdU-assay, migration via scratch assay, EP3, Gi-protein and p-ERK1/2 expressions via Western blot and cAMP concentrations via ELISA. The Mann-Whitney-U-test was used to test for statistical significance. Results: The cell lines T-47D (EP3 expression 77.7%) and SK-BR-3 (EP3 expression 48.7%) were chosen. EP3 antagonism reduced its expression on SK-BR-3 significantly, while no effect was observed on T-47D. The proliferation and migration of SK-BR-3 cells were significantly reduced due to treatment with the EP1/3 agonist, the EP3 antagonist or a combination of both. Neither agonism nor antagonism influenced cell proliferation or migration in T-47D. In SK-BR-3, EP3 antagonism showed a significant decrease in Gi-protein levels, an increase in cAMP levels, and no significant change in p-ERK1/2 expression. Conclusion: Antagonism of the EP3 receptor results in a reduced proliferation and migration of SK-BR-3 breast cancer cells, potentially mediated via a Gi-protein-cAMP pathway. The results suggest that EP3 plays a role in tumorigenesis. This is in accordance with the cell culture data of other gynecological tumors, but it is conflicting in so far, as positive EP3 expression is clinically a positive prognostic marker in breast cancer. Therefore, other factors may be important in explaining this contradiction.

10.
Arch Gynecol Obstet ; 298(2): 457, 2018 08.
Article in English | MEDLINE | ID: mdl-29948166

ABSTRACT

In the original publication of the article, the name of first author was misspelled. The correct name has been copied below.

11.
Endocr Connect ; 7(5): 749-761, 2018 May.
Article in English | MEDLINE | ID: mdl-29700097

ABSTRACT

Although an inflammatory microenvironment is required for successful implantation, an inflammatory overreaction is one of the causes of unexplained recurrent pregnancy losses (uRPL). Prostaglandin E2 (PGE2) plays a pivotal role in regulating immune balance during early pregnancy, and it can stimulate inflammatory reactions via prostaglandin E2 receptor 3 (EP3). However, the role of PGE2 receptor signaling in the uRPL remains unknown. We aimed to investigate whether EP3 signaling is involved in the mechanism of uRPL. Via immunohistochemistry we could show that the expression of cyclooxygenase-2, EP3 and G protein alpha inhibitor 1 (Gi1) was enhanced in the decidua of the uRPL group in comparison to the control group in first-trimester placentas. In vitro, we demonstrated that sulprostone (an EP1/EP3 agonist) inhibited the secretion of beta-hCG and progesterone in JEG-3 cells and the secretion of beta-hCG in HTR-8/SVneo cells while it induced the expression of plasminogen activator inhibitor type 1 in JEG-3 cells. In addition, PGE2/sulprostone was able to stimulate the expression of Gi1, phosphorylated-extracellular signal-regulated kinases 1/2 (p-ERK1/2) and p53. L-798,106 (an EP3-specific antagonist) suppressed the expression of EP3 and p-ERK1/2 without affecting the secretion of beta-hCG. Elevated activation of EP3 signaling in first-trimester placentas plays an important role in regulating the inflammatory microenvironment, the hormone secretion of extravillous trophoblasts and the remodeling of extracellular matrix in the fetal-maternal interface. L-798,106 might be a 'potential therapeutic candidate' for the treatment of uRPL.

12.
Oncotarget ; 9(1): 982-994, 2018 Jan 02.
Article in English | MEDLINE | ID: mdl-29416671

ABSTRACT

BACKGROUND: Prostaglandin E2 (PGE2) receptor 3 (EP3) regulates tumor cell proliferation, migration, and invasion in numerous cancers. The role of EP3 as a prognostic biomarker in endometrial cancer remains unclear. The primary aim of this study was to analyze the prognostic significance of EP3 expression in endometrial cancer. METHODS: We analyzed the EP3 expression of 140 endometrial carcinoma patients by immunohistochemistry. RL95-2 endometrial cancer cell line was chosen from four endometrial cancer cell lines (RL95-2, Ishikawa, HEC-1-A, and HEC-1-B) according to EP3 expression level. Treated with PGE2 and EP3 antagonist, RL95-2 cells were investigated by MTT, BrdU, and wound healing assay for functional assessment of EP3. RESULTS: EP3 staining differed significantly according to WHO tumor grading in both whole cohort (p = 0.01) and the subgroup of endometrioid carcinoma (p = 0.01). Patients with high EP3 expression in their respective tumors had impaired progression-free survival as well as overall survival in both cohorts above. EP3 expression in the overall cohort was identified as an independent prognostic marker for progression-free survival (HR 1.014, 95%CI 1.003-1.024, p = 0.01) when adjusted for age, stage, grading, and recurrence. Treatment with EP3 antagonists induced upregulation of estrogen receptor ß and decreased activity of Ras and led to attenuated proliferation and migration of RL95-2 cells. CONCLUSIONS: EP3 seems to play a crucial role in endometrial cancer progression. In the context of limited systemic treatment options for endometrial cancer, this explorative analysis identifies EP3 as a potential target for diagnostic workup and therapy.

13.
Arch Gynecol Obstet ; 296(2): 269-276, 2017 08.
Article in English | MEDLINE | ID: mdl-28578502

ABSTRACT

PURPOSE: The nuclear hormone receptor estrogen receptor α (ERα) is pivotal for numerous processes in the cell. As a transcription factor, it regulates eukaryotic gene expression and affects cellular proliferation and differentiation in target tissues. Moreover, ERα is known for its influence on various gynecological diseases and carcinogenesis. Since its expression is often altered in diseased tissues and this alteration was found to be caused by hypermethylation of the ESR1 promotor region in cancer, including breast and colorectal cancer, the aim of this study is to elucidate if the expression of ERα is also regulated epigenetically in endometriosis and endometrial cancer. METHODS: Using real-time methylation-specific PCR (rt-MSP), we examined endometrial and endometriotic tissues as well as five endometrial cancer cell lines and compared the methylation status with the actual expression of ERα. RESULTS: The results of our study indicate that, though its expression is altered in endometrial and endometriotic tissue, ERα is not regulated by methylation of the promotor region in endometriosis. In contrast, three of the five endometrial cancer cell lines are methylated in the promotor region of ESR1. CONCLUSIONS: Thus, further investigation of the connection between ERα and endometrial cancer will be the next step.


Subject(s)
Endometrial Neoplasms/genetics , Endometriosis/genetics , Estrogen Receptor alpha/genetics , Promoter Regions, Genetic , Cell Line, Tumor , DNA Methylation , Endometriosis/metabolism , Endometrium/metabolism , Estrogen Receptor alpha/chemistry , Female , Gene Expression Regulation , Humans , Transcription, Genetic
14.
Mol Oncol ; 7(6): 1012-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23916134

ABSTRACT

Endoplasmic reticulum (ER) stress and autophagy are two basic cell survival mechanisms often occurring in concert. Extensive ER stress in cancer cells deliberately induced by chemotherapeutic drugs may lead to growth arrest and cell death. However, the link between ER stress and autophagy is not well understood. In this study, the treatment of cancer cells with ER stress-inducing drug nelfinavir resulted in the expression of endogenous mTOR inhibitor sestrin-2 (SESN2). Upregulation of SESN2 expression was associated with expression of ER stress markers ATF4, ATF3, and CHOP. SESN2 upregulation also occurred in cells treated with the proteasome inhibitor bortezomib. Ectopic expression of ATF4, but not of ATF3 or CHOP, caused transcriptional upregulation of SESN2 expression, indicating expressional regulation of SESN2 by ATF4. Transient overexpression of ectopic SESN2 resulted in mTOR inhibition and autophagy, confirming a link between ER stress, SESN2 upregulation, and mTOR inhibition. Accordingly, cancer cells treated with the ER stress-inducing agent nelfinavir showed reduced mTOR activity and associated increases in the expression levels of ATF4 and SESN2. These results show that ATF4-regulated SESN2 expression presents a new link between ER stress and mTOR inhibition and autophagy. mTOR inhibition by nelfinavir, which is currently in clinical trials for cancer patients, may also explain its observed ability to induce autophagy, growth arrest, and radiosensitization in cancer cells.


Subject(s)
Activating Transcription Factor 4/metabolism , Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , HIV Protease Inhibitors/pharmacology , Nelfinavir/pharmacology , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Nuclear Proteins/biosynthesis , Pyrazines/pharmacology , TOR Serine-Threonine Kinases/metabolism , Activating Transcription Factor 4/genetics , Autophagy/drug effects , Autophagy/genetics , Bortezomib , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , HeLa Cells , Humans , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Proteins/genetics , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
15.
Toxicol Appl Pharmacol ; 264(2): 300-4, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22935518

ABSTRACT

Inhibins and activins are gonadal peptide hormones of the transforming growth factor-ß super family with important functions in the reproductive system. By contrast, the recently identified inhibin ßE subunit, primarily expressed in liver cells, appears to exert functions unrelated to the reproductive system. Previously shown downregulation of inhibin ßE in hepatoma cells and anti-proliferative effects of ectopic inhibin ßE overexpression indicated growth-regulatory effects of inhibin ßE. We observed a selective re-expression of the inhibin ßE subunit in HepG2 hepatoblastoma cells, MCF7 breast cancer cells, and HeLa cervical cancer cells under endoplasmic reticulum stress conditions induced by tunicamycin, thapsigargin, and nelfinavir. Analysis of XPB1 splicing and ATF4 activation revealed that inhibin ßE re-expression was associated with induction of the endoplasmic reticulum stress reaction by these drugs. Transfection of an ATF4 expression plasmid specifically induced inhibin ßE expression in HeLa cells and indicates inhibin ßE as a hitherto unidentified target gene of ATF4, a key transcription factor of the endoplasmic reticulum stress response. Therefore, the inhibin ßE subunit defines not only a new player but also a possible new marker for drug-induced endoplasmic reticulum stress.


Subject(s)
Activating Transcription Factor 4/genetics , Antiviral Agents/pharmacology , Endoplasmic Reticulum Stress/physiology , Inhibin-beta Subunits/biosynthesis , Nelfinavir/pharmacology , Blotting, Western , Cell Line, Tumor , Endoplasmic Reticulum Stress/drug effects , Fluorescent Antibody Technique , Gene Targeting , HeLa Cells , Humans , Inhibin-beta Subunits/genetics , MCF-7 Cells , Plasmids/genetics , Polymerase Chain Reaction , RNA Splicing , Up-Regulation/drug effects
16.
Mol Cancer ; 9: 19, 2010 Jan 27.
Article in English | MEDLINE | ID: mdl-20105315

ABSTRACT

BACKGROUND: Nelfinavir is an HIV protease inhibitor that has been used for a long period of time to treat HIV-infected individuals. It has recently emerged that nelfinavir could represent a prospective new anti-cancer drug, prompting us to test the effect of nelfinavir on leukemia cells. METHODS: By combining in vitro and ex vivo studies, the effect of nelfinavir on leukemia cells and non-malignant, bone marrow-derived tissue cells was analyzed. RESULTS: At a concentration of 9 microg/ml, nelfinavir induced death of 90% of HL60, IM9, and Jurkat cells. At the same concentration and treatment conditions, less than 10% of aspirated human bone marrow cells showed nelfinavir-induced cell damage. Nelfinavir-induced death of leukemia cells was accompanied by activation of caspases 3, 7, and 8. Despite caspase activation, the upregulation of the anti-apoptotic bcl-2 family member protein mcl-1 that resulted from nelfinavir treatment stabilized the mitochondrial membrane potential, resulting in primarily mitochondria-independent cell death. Pharmacological downregulation of mcl-1 expression by treatment with sorafenib (2 microg/ml) significantly enhanced nelfinavir-induced apoptosis even at lower nelfinavir concentrations (5 microg/ml), but did not have additional detrimental effects on non-malignant bone marrow cells. CONCLUSIONS: The ability of nelfinavir to induce apoptosis in leukemia cells as a single agent in a mitochondria-independent manner might suggest it could be used as a second or third line of treatment for leukemia patients for whom standard mitochondria-directed treatment strategies have failed. Combination treatment with nelfinavir and sorafenib might further enhance the efficacy of nelfinavir even on chemo-resistant leukemia cells.


Subject(s)
Apoptosis/drug effects , Benzenesulfonates/pharmacology , Down-Regulation/drug effects , Leukemia/pathology , Mitochondrial Membranes/drug effects , Nelfinavir/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Pyridines/pharmacology , Bone Marrow Cells/drug effects , CDC2 Protein Kinase/metabolism , Caspase 8/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cyclin B/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Activation/drug effects , Gene Expression Regulation, Leukemic/drug effects , Humans , Leukemia/enzymology , Leukemia/genetics , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/enzymology , Myeloid Cell Leukemia Sequence 1 Protein , Niacinamide/analogs & derivatives , Phenylurea Compounds , Proto-Oncogene Proteins c-bcl-2/metabolism , Sorafenib , Up-Regulation/drug effects
17.
Invest New Drugs ; 27(6): 543-51, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19039521

ABSTRACT

Bortezomib, an approved drug for the treatment of certain haematological neoplasms, is currently being tested in clinical trials as a potential therapeutic agent against several types of solid cancer, including ovarian cancer. We have analyzed the effect of bortezomib on ovarian cancer cells and tissue explants either as a single agent or in combination with carboplatin, taxol, or TRAIL (tumor necrosis factor-related apoptosis-inducing ligand). Bortezomib alone efficiently induced apoptosis in ovarian cancer cells. Apoptosis was preceded by an upregulation of the endoplasmic reticulum stress sensor ATF3, and increased the expression of cytoplasmic heat shock proteins. Bortezomib enhanced the sensitivity of ovarian cancer cells and tissue explants to an apoptosis-inducing TRAIL receptor antibody by upregulating the TRAIL receptor DR5. In contrast to the synergistic effect observed for TRAIL, the efficacy of the taxol treatment was reduced by bortezomib, and bortezomib inhibited the G2/M phase accumulation of ovarian cancer cells treated with taxol. Bortezomib alone or in combination with taxol induced a cell cycle arrest within the S phase, and downregulation of cdk1, a cyclin-dependent kinase that is necessary for the entry into the M phase. Thus, bortezomib can be regarded as a promising agent for the treatment of ovarian cancer and could either be administered as a single agent or in combination with TRAIL. However, a combination treatment with taxanes may not be beneficial and may even be less effective.


Subject(s)
Apoptosis/drug effects , Boronic Acids/pharmacology , Boronic Acids/therapeutic use , Cell Cycle Checkpoints/drug effects , Endoplasmic Reticulum Stress/drug effects , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Pyrazines/pharmacology , Pyrazines/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bortezomib , CDC2 Protein Kinase/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , M Phase Cell Cycle Checkpoints/drug effects , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Protein Array Analysis , Stress, Physiological/drug effects , Time Factors , Up-Regulation/drug effects
18.
Cancer Biol Ther ; 8(3): 226-32, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19106637

ABSTRACT

Proteasome inhibitors and protease inhibitors are currently being discussed to be useful to sensitize drug-resistant cancer cells to chemotherapeutic agents or to act independently as single agents on drug-resistant cancer cells. We tested the effect of the clinically applied HIV protease inhibitor nelfinavir on ovarian cancer cells. Nelfinavir efficiently induced cell death in carboplatin-sensitive (SKOV3, OV-GH-5) and carboplatin-resistant (OVCAR3, OV-GH-1) ovarian cancer cell lines as well as in cancer biopsies and ascites samples from patients with recurrent ovarian cancer. Nelfinavir significantly changed the morphology of ovarian cancer cells, resulting in formation of large ER-derived vacuoles and induced upregulation of the hsp70 heat shock family member BiP (GRP78) which accumulated within swollen ER membranes. Upregulation of BiP and phosphorylation of eIF2alpha indicated induction of the unfolded protein response, which can cause cell cycle arrest and apoptosis. Correspondingly, we observed downregulation of cell cycle regulatory proteins after nelfinavir treatment, especially that of cyclin D3, and induction of apoptosis as confirmed by annexin binding. Because nelfinavir represents an already approved drug for use in humans with HIV infection, it could rapidly be tested in clinical studies as a potential treatment strategy against drug-resistant ovarian cancer.


Subject(s)
Apoptosis/drug effects , Cell Cycle/drug effects , Endoplasmic Reticulum/drug effects , HIV Protease Inhibitors/pharmacology , Nelfinavir/pharmacology , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/metabolism , Protein Folding , Cell Culture Techniques , Cell Line, Tumor , Cyclin D3/biosynthesis , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum Chaperone BiP , Eukaryotic Initiation Factor-2/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Heat-Shock Proteins/biosynthesis , Humans , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Phosphorylation , Vacuoles/pathology
19.
Biochem Biophys Res Commun ; 377(4): 1309-14, 2008 Dec 26.
Article in English | MEDLINE | ID: mdl-19000651

ABSTRACT

HIV protease inhibitors are currently being discussed to be useful as new and alternative anti-cancer agents, especially as second line treatments for chemo-resistant human cancer types. Among three clinically applied HIV protease inhibitors tested, we found a high efficacy of nelfinavir on ovarian cancer cells, accompanied by apoptosis (annexin binding) and necrosis (propidium iodide permeability). In vitro, at concentrations used to induce cell death in ovarian cancer cells, nelfinavir had no effect on the cellular viability of fibroblasts or peripheral blood mononuclear leukocytes. Nelfinavir sensitized ovarian cancer cells to treatment with an apoptosis-inducing TRAIL receptor antibody due to upregulation of the TRAIL receptor DR5 as shown by RT-PCR and FACScan analysis. We conclude that nelfinavir, an already approved drug, is a highly efficient agent against ovarian cancer cells and could sensitize ovarian cancer cells to TRAIL treatment, either therapeutically applied or endogenously produced by cells of the immune system.


Subject(s)
Antineoplastic Agents/pharmacology , HIV Protease Inhibitors/pharmacology , Nelfinavir/pharmacology , Ovarian Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Apoptosis , Cell Line, Tumor , Female , Humans , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Up-Regulation
20.
Cancer Res ; 66(3): 1630-9, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16452222

ABSTRACT

Lapatinib (GW572016) is a selective inhibitor of both epidermal growth factor receptor (EGFR) and HER-2 tyrosine kinases. Here, we explore the therapeutic potential of lapatinib by testing its effect on tumor cell growth in a panel of 31 characterized human breast cancer cell lines, including trastuzumab-conditioned HER-2-positive cell lines. We further characterize its activity in combination with trastuzumab and analyze whether EGFR and HER-2 expression or changes induced in the activation of EGFR, HER-2, Raf, AKT, or extracellular signal-regulated kinase (ERK) are markers of drug activity. We report that concentration-dependent antiproliferative effects of lapatinib were seen in all breast cancer cell lines tested but varied significantly between individual cell lines with up to 1,000-fold difference in the IC(50)s (range, 0.010-18.6 micromol/L). Response to lapatinib was significantly correlated with HER-2 expression and its ability to inhibit HER-2, Raf, AKT, and ERK phosphorylation. Long-term in vivo lapatinib studies were conducted with human breast cancer xenografts in athymic mice. Treatment over 77 days resulted in a sustained and significant reduction in xenograft volume compared with untreated controls. For the combination of lapatinib plus trastuzumab, synergistic drug interactions were observed in four different HER-2-overexpressing cell lines. Moreover, lapatinib retained significant in vitro activity against cell lines selected for long-term outgrowth (>9 months) in trastuzumab-containing (100 microg/mL) culture medium. These observations provide a clear biological rationale to test lapatinib as a single agent or in combination with trastuzumab in HER-2-overexpressing breast cancer and in patients with clinical resistance to trastuzumab.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Quinazolines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Drug Interactions , Drug Screening Assays, Antitumor , ErbB Receptors/biosynthesis , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Lapatinib , Mice , Mice, SCID , Oncogene Protein v-akt/metabolism , Phosphorylation , Quinazolines/administration & dosage , Receptor, ErbB-2/biosynthesis , Trastuzumab , Xenograft Model Antitumor Assays , raf Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...