Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Am J Pathol ; 192(9): 1321-1335, 2022 09.
Article in English | MEDLINE | ID: mdl-35750257

ABSTRACT

Toll-like receptor 3 (TLR3) is an endosomal receptor expressed in several immune and epithelial cells. Recent studies have highlighted its expression also in solid tumors, including prostate cancer (PCa), and have described its role primarily in the proinflammatory response and induction of apoptosis. It is up-regulated in some castration-resistant prostate cancers. However, the role of TLR3 in prostate cancer progression remains largely unknown. The current study experimentally demonstrated that exogenous TLR3 activation in PCa cell lines leads to a significant induction of secretion of the cytokines IL-6, IL-8, and interferon-ß, depending on the model and chemoresistance status. Transcriptomic analysis of TLR3-overexpressing cells revealed a functional program that is enriched for genes involved in the regulation of cell motility, migration, and tumor invasiveness. Increased motility, migration, and invasion in TLR3-overexpressing cell line were confirmed by several in vitro assays and using an orthotopic prostate xenograft model in vivo. Furthermore, TLR3-ligand induced apoptosis via cleavage of caspase-3/7 and poly (ADP-ribose) polymerase, predominantly in TLR3-overexpressing cells. These results indicate that TLR3 may be involved in prostate cancer progression and metastasis; however, it might also represent an Achilles heel of PCa, which can be exploited for targeted therapy.


Subject(s)
Prostatic Neoplasms , Toll-Like Receptor 3 , Animals , Apoptosis , Cell Line, Tumor , Humans , Male , Poly I-C/pharmacology , Prostate/pathology , Prostatic Neoplasms/pathology , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism
2.
Carcinogenesis ; 39(11): 1411-1418, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30010814

ABSTRACT

The cell surface glycoprotein Trop-2 is commonly overexpressed in carcinomas and represents an exceptional antigen for targeted therapy. Here, we provide evidence that surface Trop-2 expression is functionally connected with an epithelial phenotype in breast and prostate cell lines and in patient tumor samples. We further show that Trop-2 expression is suppressed epigenetically or through the action of epithelial-to-mesenchymal transition transcription factors and that deregulation of Trop-2 expression is linked with cancer progression and poor patient prognosis. Moreover, our data suggest that the cancer plasticity-driven intratumoral heterogeneity in Trop-2 expression may significantly contribute to response and resistance to therapies targeting Trop-2-expressing cells.


Subject(s)
Antigens, Neoplasm/metabolism , Breast Neoplasms/pathology , Carcinoma/pathology , Cell Adhesion Molecules/metabolism , Epithelial Cells/metabolism , Prostatic Neoplasms/pathology , Animals , Antigens, CD/biosynthesis , Antigens, Neoplasm/genetics , Breast Neoplasms/mortality , Cadherins/biosynthesis , Cell Adhesion Molecules/genetics , Cell Line, Tumor , DNA Methylation/genetics , Disease Progression , Epithelial-Mesenchymal Transition/physiology , Female , Humans , Male , Mice , Mice, Inbred BALB C , Prostatic Neoplasms/mortality , Xenograft Model Antitumor Assays
3.
Br J Cancer ; 118(6): 813-819, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29462126

ABSTRACT

Background:The intratumoural heterogeneity, often driven by epithelial-to-mesenchymal transition (EMT), significantly contributes to chemoresistance and disease progression in adenocarcinomas. Methods:We introduced a high-throughput screening platform to identify surface antigens that associate with epithelial­mesenchymal plasticity in well-defined pairs of epithelial cell lines and their mesenchymal counterparts. Using multicolour flow cytometry, we then analysed the expression of 10 most robustly changed antigens and identified a 10-molecule surface signature, in pan-cytokeratin-positive/EpCAM-positive and -negative fractions of dissociated breast tumours. Results:We found that surface CD9, CD29, CD49c, and integrin ß5 are lost in breast cancer cells that underwent EMT in vivo. The tetraspanin family member CD9 was concordantly downregulated both in vitro and in vivo and associated with epithelial phenotype and favourable prognosis. Conclusions:We propose that overall landscape of 10-molecule surface signature expression reflects the epithelial­mesenchymal plasticity in breast cancer.


Subject(s)
Antigens, Neoplasm/biosynthesis , Antigens, Surface/biosynthesis , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Antigens, Neoplasm/immunology , Antigens, Surface/immunology , Biomarkers, Tumor , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Plasticity/immunology , Cellular Reprogramming/physiology , Epithelial-Mesenchymal Transition/immunology , Female , Flow Cytometry , High-Throughput Screening Assays , Humans , Neoplasm Metastasis , Tetraspanin 29/biosynthesis , Tetraspanin 29/immunology , Transcription, Genetic
4.
Cytometry A ; 93(2): 239-248, 2018 02.
Article in English | MEDLINE | ID: mdl-29220555

ABSTRACT

Complex analysis of cellular responses after experimental treatment is important for screening, mechanistic understanding of treatment effects, and the identification of sensitive and resistant cell phenotypes. Modern multicolor flow cytometry has demonstrated its power for such analyses. Here, we introduce a multiparametric protocol for complex analysis of cytokinetics by the simultaneous detection of seven fluorescence parameters. This analysis includes the detection of two surface markers for immunophenotyping, analysis of proliferation based on the cell cycle and the measurement of incorporated nucleoside analogue 5-ethynyl-2'-deoxyuridine (EdU) in newly synthesized DNA, analysis of DNA damage using an anti-phospho-histone H2A.X (Ser139) antibody, and determination of cell death using a fixable viability probe and intracellular detection of caspase-3 activation. To demonstrate the applicability of this protocol for the analysis of heterogeneous and complex cell responses, we used different treatments and model cell lines. We demonstrated that this protocol has the potential to provide complex and simultaneous analysis of cytokinetics and analyze the heterogeneity of the response at the single-cell level. © 2017 International Society for Advancement of Cytometry.


Subject(s)
Apoptosis/physiology , Cell Proliferation/physiology , DNA Damage/physiology , Flow Cytometry/methods , Immunophenotyping/methods , Cell Line, Tumor , Humans
5.
J Assist Reprod Genet ; 35(8): 1407-1417, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29948426

ABSTRACT

PURPOSE: The purpose of the study was to determine whether the GDF-15 is present in follicular fluid; to evaluate if there is a relation between follicular and serum levels of GDF-15 and fertility status of study subjects; and to test whether granulosa cells, oocytes, or both produce GDF-15. METHODS: This study used follicular fluid (FF, serum, and oocytes obtained under informed consent from women undergoing oocyte retrieval for in vitro fertilization. It also used ovaries from deceased preterm newborns. Collection of FF and blood at the time of oocyte retrieval, ELISA and western blot were performed to determine levels and forms of GDF-15. Concentrations of GDF-15 in FF and serum, its expression in ovarian tissue, and secretion from granulosa cells were analyzed. RESULTS: GDF-15 concentration in FF ranged from 35 to 572 ng/ml, as determined by ELISA. Western blot analysis revealed the GDF-15 pro-dimer only in FF. Both normal healthy and cancerous granulosa cells secreted GDF-15 into culture media. Primary oocytes displayed cytoplasmic GDF-15 positivity in immunostained newborn ovaries, and its expression was also observed in fully grown human oocytes. CONCLUSIONS: To the best of our knowledge, this is the first documentation of cytokine GDF-15 presence in follicular fluid. Its concentration was not associated with donor/patient fertility status. Our data also show that GDF-15 is expressed and inducible in both normal healthy and cancerous granulosa cells, as well as in oocytes.


Subject(s)
Cell Differentiation/genetics , Follicular Fluid/metabolism , Granulosa Cells/metabolism , Growth Differentiation Factor 15/genetics , Adult , Female , Fertilization in Vitro , Gene Expression Regulation, Developmental , Growth Differentiation Factor 15/isolation & purification , Humans , Oocyte Retrieval , Oocytes/metabolism
6.
Mol Cancer ; 13: 113, 2014 May 20.
Article in English | MEDLINE | ID: mdl-24884804

ABSTRACT

BACKGROUND: Tumor heterogeneity and the plasticity of cancer cells present challenges for effective clinical diagnosis and therapy. Such challenges are epitomized by neuroendocrine transdifferentiation (NED) and the emergence of neuroendocrine-like cancer cells in prostate tumors. This phenomenon frequently arises from androgen-depleted prostate adenocarcinoma and is associated with the development of castration-resistant prostate cancer and poor prognosis. RESULTS: In this study, we showed that NED was evoked in both androgen receptor (AR)-positive and AR-negative prostate epithelial cell lines by growing the cells to a high density. Androgen depletion and high-density cultivation were both associated with cell cycle arrest and deregulated expression of several cell cycle regulators, such as p27Kip1, members of the cyclin D protein family, and Cdk2. Dual inhibition of Cdk1 and Cdk2 using pharmacological inhibitor or RNAi led to modulation of the cell cycle and promotion of NED. We further demonstrated that the cyclic adenosine 3', 5'-monophosphate (cAMP)-mediated pathway is activated in the high-density conditions. Importantly, inhibition of cAMP signaling using a specific inhibitor of adenylate cyclase, MDL-12330A, abolished the promotion of NED by high cell density. CONCLUSIONS: Taken together, our results imply a new relationship between cell cycle attenuation and promotion of NED and suggest high cell density as a trigger for cAMP signaling that can mediate reversible NED in prostate cancer cells.


Subject(s)
Cell Transdifferentiation , Neuroendocrine Cells/pathology , Prostatic Neoplasms/pathology , Androgens/pharmacology , CDC2 Protein Kinase , Cell Count , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Transdifferentiation/drug effects , Cyclic AMP/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinases/metabolism , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Epithelial Cells/pathology , Humans , Immunohistochemistry , Male , Neuroendocrine Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Receptors, Androgen/metabolism , Signal Transduction/drug effects
7.
Cytometry A ; 83(5): 472-82, 2013 May.
Article in English | MEDLINE | ID: mdl-23450810

ABSTRACT

The clonogenic assay is a well-established in vitro method for testing the survival and proliferative capability of cells. It can be used to determine the cytotoxic effects of various treatments including chemotherapeutics and ionizing radiation. However, this approach can also characterize cells with different phenotypes and biological properties, such as stem cells or cancer stem cells. In this study, we implemented a faster and more precise method for assessing the cloning efficiency of cancer stem-like cells that were characterized and separated using a high-speed cell sorter. Cell plating onto a microplate using an automatic cell deposition unit was performed in a single-cell or dilution rank mode by the fluorescence-activated cell sorting method. We tested the new automatic cell-cloning assay (ACCA) on selected cancer cell lines and compared it with the manual approach. The obtained results were also compared with the results of the limiting dilution assay for different cell lines. We applied the ACCA to analyze the cloning capacity of different subpopulations of prostate and colon cancer cells based on the expression of the characteristic markers of stem (CD44 and CD133) and cancer stem cells (TROP-2, CD49f, and CD44). Our results revealed that the novel ACCA is a straightforward approach for determining the clonogenic capacity of cancer stem-like cells identified in both cell lines and patient samples.


Subject(s)
Cell Proliferation , Colonic Neoplasms/pathology , Flow Cytometry/methods , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/pathology , Tumor Stem Cell Assay/methods , AC133 Antigen , Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Survival , Colonic Neoplasms/metabolism , Glycoproteins/metabolism , Humans , Hyaluronan Receptors/metabolism , In Vitro Techniques , Integrin alpha6/metabolism , Male , Neoplastic Stem Cells/metabolism , Peptides/metabolism , Prostatic Neoplasms/metabolism
8.
Article in English | MEDLINE | ID: mdl-23320042

ABSTRACT

Since the ability of cancer cells to evade apoptosis often limits the efficacy of radiotherapy and chemotherapy, autophagy is emerging as an alternative target to promote cell death. Therefore, we wondered whether Rottlerin, a natural polyphenolic compound with antiproliferative effects in several cell types, can induce cell death in MCF-7 breast cancer cells. The MCF-7 cell line is a good model of chemo/radio resistance, being both apoptosis and autophagy resistant, due to deletion of caspase 3 gene, high expression of the antiapoptotic protein Bcl-2, and low expression of the autophagic Beclin-1 protein. The contribution of autophagy and apoptosis to the cytotoxic effects of Rottlerin was examined by light, fluorescence, and electron microscopic examination and by western blotting analysis of apoptotic and autophagic markers. By comparing caspases-3-deficient (MCF-7(3def)) and caspases-3-transfected MCF-7 cells (MCF-7(3trans)), we found that Rottlerin induced a noncanonical, Bcl-2-, Beclin 1-, Akt-, and ERK-independent autophagic death in the former- and the caspases-mediated apoptosis in the latter, in not starved conditions and in the absence of any other treatment. These findings suggest that Rottlerin could be cytotoxic for different cancer cell types, both apoptosis competent and apoptosis resistant.

9.
Prostate ; 71(12): 1332-43, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21321977

ABSTRACT

BACKGROUND: Epithelial-mesenchymal transition (EMT) underlying cancer cell invasion and metastasis has been thoroughly studied in prostate cancer. Although EMT markers have been clinically observed in benign prostate hyperplasia, molecular events underlying the onset and progression of EMT in benign prostate cells have not been described. METHODS: EMT in BPH-1 cells was induced by TGF-ß1 treatment and the kinetics of expression of EMT markers, regulators, and selected miRNAs was assessed by western blotting and quantitative RT-PCR. RESULTS: EMT in BPH-1 cells was accompanied by rapid up-regulation of SNAI2/Slug and ZEB1 transcription factors, while changes in expression levels of ZEB2 and miR-200 family members were observed after extended time intervals. Invasive phenotype with EMT hallmarks, characterizing tumorigenic clones derived from BPH-1 cells, was associated with increased mRNA levels of SNAI2, ZEB1, and ZEB2, but was not associated with significant changes in basal levels of miR-200 family members. RNA interference revealed that SNAI2/Slug is crucial for TGF-ß1-induced vimentin up-regulation and migration of BPH-1 cells. CONCLUSIONS: This study suggests that in BPH-1 cells the transcription factor SNAI2/Slug is important for EMT initiation, while the ZEB family of transcription factors in cooperation with the miR-200 family may oppose the reversal of the EMT phenotype.


Subject(s)
Epithelial-Mesenchymal Transition , Prostatic Hyperplasia/physiopathology , Transcription Factors/biosynthesis , Transforming Growth Factor beta1/pharmacology , Biomarkers/metabolism , Cell Line , Cell Movement , Epithelial-Mesenchymal Transition/genetics , Homeodomain Proteins/genetics , Humans , Kinetics , Male , MicroRNAs/metabolism , Neoplasm Invasiveness/genetics , Phenotype , RNA, Messenger/metabolism , Repressor Proteins/genetics , Snail Family Transcription Factors , Transcription Factors/genetics , Up-Regulation/drug effects , Vimentin/metabolism , Zinc Finger E-box Binding Homeobox 2 , Zinc Finger E-box-Binding Homeobox 1
10.
Noncoding RNA ; 7(4)2021 Dec 02.
Article in English | MEDLINE | ID: mdl-34940756

ABSTRACT

Neuroendocrine prostate cancer (NEPC) represents a variant of prostate cancer that occurs in response to treatment resistance or, to a much lesser extent, de novo. Unravelling the molecular mechanisms behind transdifferentiation of cancer cells to neuroendocrine-like cancer cells is essential for development of new treatment opportunities. This review focuses on summarizing the role of small molecules, predominantly microRNAs, in this phenomenon. A published literature search was performed to identify microRNAs, which are reported and experimentally validated to modulate neuroendocrine markers and/or regulators and to affect the complex neuroendocrine phenotype. Next, available patients' expression datasets were surveyed to identify deregulated microRNAs, and their effect on NEPC and prostate cancer progression is summarized. Finally, possibilities of miRNA detection and quantification in body fluids of prostate cancer patients and their possible use as liquid biopsy in prostate cancer monitoring are discussed. All the addressed clinical and experimental contexts point to an association of NEPC with upregulation of miR-375 and downregulation of miR-34a and miR-19b-3p. Together, this review provides an overview of different roles of non-coding RNAs in the emergence of neuroendocrine prostate cancer.

11.
Hum Reprod ; 25(12): 2962-71, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20884666

ABSTRACT

BACKGROUND: Transforming growth factor-ß cytokines have various biological effects in female reproductive tissue, including modulation of inflammatory response and induction of immune tolerance to seminal antigens in the reproductive tract. However, no studies have analyzed the presence of growth/differentiation factor-15 (GDF-15/macrophage inhibitory cytokine-1) in seminal fluid or demonstrated the quantity and form of GDF-15, its possible role or the relationship between its concentration and semen quality. METHODS: The form and the concentration of GDF-15 were determined in 53 seminal plasma samples of both fertile and infertile men by ELISA and western blot. The sperm cells of three volunteers were treated with recombinant GDF-15, and cell viability and apoptosis were assessed by flow cytometry. The effect of GDF-15 on vaginal epithelial cells and peripheral blood mononuclear cells (PBMCs) was analyzed by quantitative RT-PCR. RESULTS: The GDF-15 concentration in seminal plasma ranged from 0.2 to 6.6 µg/ml as determined by ELISA. Western blot analysis revealed that GDF-15 is present in the active form. In vitro cultivation of sperm cells with GDF-15 did not affect their viability or rates of apoptosis; however, it did inhibit proliferation of PBMCs and induce expression of FOXP3 in CD4+CD25+ cells. CONCLUSIONS: To the best of our knowledge, this is the first demonstration that GDF-15 is an abundant cytokine in seminal plasma, although its concentration is not associated with semen quality or the fertility/infertility status of the donors. Moreover, our data show that GDF-15 displays immunosuppressive characteristics.


Subject(s)
Growth Differentiation Factor 15/physiology , Semen/metabolism , Adult , Apoptosis/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Epithelial Cells/metabolism , Female , Forkhead Transcription Factors/biosynthesis , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , Leukocytes, Mononuclear/drug effects , Male , Middle Aged , Semen Analysis , Spermatozoa
12.
Stem Cell Res ; 46: 101844, 2020 07.
Article in English | MEDLINE | ID: mdl-32590255

ABSTRACT

Deciphering the properties of adult stem cells is crucial for understanding of their role in healthy tissue and in cancer progression as well. Both stem cells and cancer stem cells have shown association with epithelial-to-mesenchymal transition (EMT) in various tissue types. Aiming to investigate the epithelial and mesenchymal phenotypic traits in adult mouse prostate, we sorted subpopulations of basal prostate stem cells (mPSCs) and assessed the expression levels of EMT regulators and markers with custom-designed gene expression array. The population of mPSCs defined by a Lin-/Sca-1+CD49fhi/Trop-2+ (LSC Trop-2+) surface phenotype was enriched in mesenchymal markers, especially EMT master regulator Slug, encoded by the Snai2 gene. To further dissect the role of Slug in mPSCs, we used transgenic Snai2tm1.1Wbg reporter mouse strain. Using this model, we confirmed the presence of mesenchymal traits and increase of organoid forming capacity in Slug+ population of mPSCs. The Slug+-derived organoids comprised all prostate epithelial cell types - basal, luminal, and neuroendocrine. Collectively, these data uncover the important role of Slug expression in the physiology of mouse prostate stem cells.


Subject(s)
Epithelial-Mesenchymal Transition , Prostate , Animals , Cell Line, Tumor , Cell Movement , Epithelial Cells , Male , Mice , Snail Family Transcription Factors/genetics
13.
Sci Rep ; 9(1): 5695, 2019 04 05.
Article in English | MEDLINE | ID: mdl-30952903

ABSTRACT

Skp2 is a crucial component of SCFSkp2 E3 ubiquitin ligase and is often overexpressed in various types of cancer, including prostate cancer (PCa). The epithelial-to-mesenchymal transition (EMT) is involved in PCa progression. The acquisition of a mesenchymal phenotype that results in a cancer stem cell (CSC) phenotype in PCa was described. Therefore, we aimed to investigate the expression and localization of Skp2 in clinical samples from patients with PCa, the association of Skp2 with EMT status, and the role of Skp2 in prostate CSC. We found that nuclear expression of Skp2 was increased in patients with PCa compared to those with benign hyperplasia, and correlated with high Gleason score in PCa patients. Increased Skp2 expression was observed in PCa cell lines with mesenchymal and CSC-like phenotype compared to their epithelial counterparts. Conversely, the CSC-like phenotype was diminished in cells in which SKP2 expression was silenced. Furthermore, we observed that Skp2 downregulation led to the decrease in subpopulation of CD44+CD24- cancer stem-like cells. Finally, we showed that high expression levels of both CD24 and CD44 were associated with favorable recurrence-free survival for PCa patients. This study uncovered the Skp2-mediated CSC-like phenotype with oncogenic functions in PCa.


Subject(s)
Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/physiology , Prostatic Neoplasms/genetics , S-Phase Kinase-Associated Proteins/genetics , Animals , CD24 Antigen/genetics , Cell Line, Tumor , Humans , Hyaluronan Receptors/genetics , Male , Mice , Mice, Nude , Neoplasm Grading , Neoplastic Stem Cells/metabolism , PC-3 Cells , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/physiopathology , Xenograft Model Antitumor Assays
14.
Stem Cell Res ; 35: 101405, 2019 03.
Article in English | MEDLINE | ID: mdl-30776675

ABSTRACT

Human induced pluripotent stem cell line was generated from commercially available primary human prostate fibroblasts HPrF derived from a fetus, aged 18-24 weeks of gestation. The fibroblast cell line was reprogrammed with Yamanaka factors (OCT4, SOX2, c-MYC, KLF4) using CytoTune™-iPS 2.0 Sendai Reprogramming Kit. Pluripotency of the derived transgene-free iPS cell line was confirmed both in vitro by detecting the expression of factors of pluripotency on a single-cell level, and in vivo using teratoma formation assay. This iPS cell line will be a useful tool for studying both normal prostate development and prostate cancer disease.


Subject(s)
Cellular Reprogramming Techniques , Fetus , Fibroblasts , Induced Pluripotent Stem Cells , Prostate , Cellular Reprogramming , Fetus/cytology , Fetus/embryology , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Male , Prostate/cytology , Prostate/embryology
15.
Cell Death Dis ; 9(8): 783, 2018 Jul 16.
Article in English | MEDLINE | ID: mdl-30013025

ABSTRACT

The PDF and HTML versions of the article have been updated to include the Creative Commons Attribution 4.0 International License information.

16.
Stem Cell Res ; 33: 255-259, 2018 12.
Article in English | MEDLINE | ID: mdl-30481741

ABSTRACT

A human induced pluripotent stem cell line was generated from cancer-associated fibroblasts of a 68-years old patient with diagnosed prostate adenocarcinoma (PCa). The fibroblast cell line was reprogrammed with Epi5™ Episomal iPSC Reprogramming Kit. Pluripotency of the derived transgene-free iPS cell line was confirmed both in vitro by detecting expression of factors of pluripotency on a single-cell level, and also in vivo using teratoma formation assay. This new iPS cell line may be used for differentiation into different prostate-specific cell types in differentiation studies.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/metabolism , Prostatic Neoplasms/genetics , Aged , Humans , Male
17.
Cell Death Dis ; 8(10): e3100, 2017 10 12.
Article in English | MEDLINE | ID: mdl-29022903

ABSTRACT

MicroRNA miR-34a is recognized as a master regulator of tumor suppression. The strategy of miR-34a replacement has been investigated in clinical trials as the first attempt of miRNA application in cancer treatment. However, emerging outcomes promote the re-evaluation of existing knowledge and urge the need for better understanding the complex biological role of miR-34a. The targets of miR-34a encompass numerous regulators of cancer cell proliferation, survival and resistance to therapy. MiR-34a expression is transcriptionally controlled by p53, a crucial tumor suppressor pathway, often disrupted in cancer. Moreover, miR-34a abundance is fine-tuned by context-dependent feedback loops. The function and effects of exogenously delivered or re-expressed miR-34a on the background of defective p53 therefore remain prominent issues in miR-34a based therapy. In this work, we review p53-independent mechanisms regulating the expression of miR-34a. Aside from molecules directly interacting with MIR34A promoter, processes affecting epigenetic regulation and miRNA maturation are discussed. Multiple mechanisms operate in the context of cancer-associated phenomena, such as aberrant oncogene signaling, EMT or inflammation. Since p53-dependent tumor-suppressive mechanisms are disturbed in a substantial proportion of malignancies, we summarize the effects of miR-34a modulation in cell and animal models in the clinically relevant context of disrupted or insufficient p53 function.


Subject(s)
Genes, Tumor Suppressor , MicroRNAs/genetics , Neoplasms/genetics , Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Animals , Epigenesis, Genetic/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Promoter Regions, Genetic/genetics
18.
Article in English | MEDLINE | ID: mdl-27605398

ABSTRACT

BACKGROUND: Small leucine rich proteoglycans (SLRPs), major non-collagen components of the extracellular matrix (ECM), have multiple biological roles with diverse effects. Asporin, a member of the SLRPs class I, competes with other molecules in binding to collagen and affects its mineralization. Its role in cancer is only now being elucidated. METHODS: The PubMed online database was used to search relevant reviews and original articles. Furthermore, altered asporin expression was analysed in publicly available genome-wide expression data at the Gene Expression Omnibus database. RESULTS: Polymorphisms in the N-terminal polyaspartate domain, which binds calcium, are associated with osteoarthritis and prostate cancer. Asporin also promotes the progression of scirrhous gastric cancer where it is required for coordinated invasion by cancer associated fibroblasts and cancer cells. Besides the enhanced expression of asporin observed in multiple cancer types, such as breast, prostate, gastric, pancreas and colon cancer, tumour suppressive effects of asporin were described in triple-negative breast cancer. We also discuss a number of factors modulating asporin expression in different cell types relevant for alterations toing the tumour microenvironment. CONCLUSION: The apparent contradicting tumour promoting and suppressive effects of asporin require further investigation. Deciphering the role of asporin and other SLRPs in tumour-stroma interactions is needed for a better understanding of cancer progression and potentially also for novel tumour microenvironment based therapies.


Subject(s)
Extracellular Matrix Proteins/physiology , Neoplasms/etiology , Tumor Microenvironment/physiology , Adipose Tissue/physiology , Disease Progression , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Female , Humans , Male , MicroRNAs/physiology , Neoplasms/genetics , Polymorphism, Genetic/genetics , Tumor Microenvironment/genetics
19.
Oncotarget ; 6(34): 36156-71, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26416355

ABSTRACT

Plasticity of cancer cells, manifested by transitions between epithelial and mesenchymal phenotypes, represents a challenging issue in the treatment of neoplasias. Both epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET) are implicated in the processes of metastasis formation and acquisition of stem cell-like properties. Mouse double minute (MDM) 2 and MDMX are important players in cancer progression, as they act as regulators of p53, but their function in EMT and metastasis may be contradictory. Here, we show that the EMT phenotype in multiple cellular models and in clinical prostate and breast cancer samples is associated with a decrease in MDM2 and increase in MDMX expression. Modulation of EMT-accompanying changes in MDM2 expression in benign and transformed prostate epithelial cells influences their migration capacity and sensitivity to docetaxel. Analysis of putative mechanisms of MDM2 expression control demonstrates that in the context of defective p53 function, MDM2 expression is regulated by EMT-inducing transcription factors Slug and Twist. These results provide an alternative context-specific role of MDM2 in EMT, cell migration, metastasis, and therapy resistance.


Subject(s)
Breast Neoplasms/metabolism , Epithelial-Mesenchymal Transition/physiology , Nuclear Proteins/biosynthesis , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-mdm2/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Proteins , Cell Line, Tumor , Female , Heterografts , Humans , Male , Mice , Mice, Nude , Phenotype , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Transfection
20.
Cell Cycle ; 13(18): 2931-43, 2014.
Article in English | MEDLINE | ID: mdl-25486480

ABSTRACT

The product of the ecotropic virus integration site 1 (EVI1) gene, whose overexpression is associated with a poor prognosis in myeloid leukemias and some epithelial tumors, regulates gene transcription both through direct DNA binding and through modulation of the activity of other sequence specific transcription factors. Previous results from our laboratory have shown that EVI1 influenced transcription regulation in response to the myeloid differentiation inducing agent, all-trans retinoic acid (ATRA), in a dual manner: it enhanced ATRA induced transcription of the RARß gene, but repressed the ATRA induction of the EVI1 gene itself. In the present study, we asked whether EVI1 would modulate the ATRA regulation of a larger number of genes, as well as biological responses to this agent, in human myeloid cells. U937 and HL-60 cells ectopically expressing EVI1 through retroviral transduction were subjected to microarray based gene expression analysis, and to assays measuring cellular proliferation, differentiation, and apoptosis. These experiments showed that EVI1 modulated the ATRA response of several dozens of genes, and in fact reinforced it in the vast majority of cases. A particularly strong synergy between EVI1 and ATRA was observed for GDF15, which codes for a member of the TGF-ß superfamily of cytokines. In line with the gene expression results, EVI1 enhanced cell cycle arrest, differentiation, and apoptosis in response to ATRA, and knockdown of GDF15 counteracted some of these effects. The potential clinical implications of these findings are discussed.


Subject(s)
DNA-Binding Proteins/metabolism , Myeloid Cells/metabolism , Oncogenes , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Tretinoin/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Differentiation/drug effects , DNA-Binding Proteins/genetics , Down-Regulation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Knockdown Techniques , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/metabolism , HL-60 Cells , Humans , MDS1 and EVI1 Complex Locus Protein , Myeloid Cells/drug effects , Proto-Oncogenes/genetics , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL