Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Acta Neuropathol Commun ; 12(1): 72, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38711090

ABSTRACT

The RE1-silencing transcription factor (REST) acts either as a repressor or activator of transcription depending on the genomic and cellular context. REST is a key player in brain cell differentiation by inducing chromatin modifications, including DNA methylation, in a proximity of its binding sites. Its dysfunction may contribute to oncogenesis. Mutations in IDH1/2 significantly change the epigenome contributing to blockade of cell differentiation and glioma development. We aimed at defining how REST modulates gene activation and repression in the context of the IDH mutation-related phenotype in gliomas. We studied the effects of REST knockdown, genome wide occurrence of REST binding sites, and DNA methylation of REST motifs in IDH wild type and IDH mutant gliomas. We found that REST target genes, REST binding patterns, and TF motif occurrence proximal to REST binding sites differed in IDH wild-type and mutant gliomas. Among differentially expressed REST targets were genes involved in glial cell differentiation and extracellular matrix organization, some of which were differentially methylated at promoters or gene bodies. REST knockdown differently impacted invasion of the parental or IDH1 mutant glioma cells. The canonical REST-repressed gene targets showed significant correlation with the GBM NPC-like cellular state. Interestingly, results of REST or KAISO silencing suggested the interplay between these TFs in regulation of REST-activated and repressed targets. The identified gene regulatory networks and putative REST cooperativity with other TFs, such as KAISO, show distinct REST target regulatory networks in IDH-WT and IDH-MUT gliomas, without concomitant DNA methylation changes. We conclude that REST could be an important therapeutic target in gliomas.


Subject(s)
Brain Neoplasms , DNA Methylation , Gene Regulatory Networks , Glioma , Isocitrate Dehydrogenase , Mutation , Isocitrate Dehydrogenase/genetics , Glioma/genetics , Glioma/pathology , Glioma/metabolism , Humans , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Cell Line, Tumor , Repressor Proteins/genetics , Repressor Proteins/metabolism , Gene Expression Regulation, Neoplastic/genetics
2.
Cell Death Dis ; 15(3): 228, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38509074

ABSTRACT

Glioblastoma (GBM) is the most common and lethal brain tumor in adults. Due to its fast proliferation, diffusive growth and therapy resistance survival times are less than two years for patients with IDH-wildtype GBM. GBM is noted for the considerable cellular heterogeneity, high stemness indices and abundance of the glioma stem-like cells known to support tumor progression, therapeutic resistance and recurrence. Doublesex- and mab-3-related transcription factor a2 (DMRTA2) is involved in maintaining neural progenitor cells (NPC) in the cell cycle and its overexpression suppresses NPC differentiation. Despite the reports showing that primary GBM originates from transformed neural stem/progenitors cells, the role of DMRTA2 in gliomagenesis has not been elucidated so far. Here we show the upregulation of DMRTA2 expression in malignant gliomas. Immunohistochemical staining showed the protein concentrated in small cells with high proliferative potential and cells localized around blood vessels, where it colocalizes with pericyte-specific markers. Knock-down of DMRTA2 in human glioma cells impairs proliferation but not viability of the cells, and affects the formation of the tumor spheres, as evidenced by strong decrease in the number and size of spheres in in vitro cultures. Moreover, the knockdown of DMRTA2 in glioma spheres affects the stabilization of the glioma stem-like cell-dependent tube formation in an in vitro angiogenesis assay. We conclude that DMRTA2 is a new player in gliomagenesis and tumor neovascularization and due to its high expression in malignant gliomas could be a biomarker and potential target for new therapeutic strategies in glioblastoma.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Neural Stem Cells , Adult , Humans , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Glioblastoma/metabolism , Glioma/pathology , Neoplastic Stem Cells/metabolism , Neural Stem Cells/metabolism , Transcription Factors/metabolism
3.
RSC Adv ; 13(18): 11889-11902, 2023 Apr 17.
Article in English | MEDLINE | ID: mdl-37077262

ABSTRACT

This research aimed at designing and fabricating a smart thermosensitive injectable methylcellulose/agarose hydrogel system loaded with short electrospun bioactive PLLA/laminin fibers as a scaffold for tissue engineering applications or 3D cell culture models. Considering ECM-mimicking morphology and chemical composition, such a scaffold is capable of ensuring a hospitable environment for cell adhesion, proliferation, and differentiation. Its viscoelastic properties are beneficial from the practical perspective of minimally invasive materials that are introduced to the body via injection. Viscosity studies showed the shear-thinning character of MC/AGR hydrogels enabling the potential injection ability of highly viscous materials. Injectability tests showed that by tuning the injection rate, even a high amount of short fibers loaded inside of hydrogel could be efficiently injected into the tissue. Biological studies showed the non-toxic character of composite material with excellent viability, attachment, spreading, and proliferation of fibroblasts and glioma cells. These findings indicate that MC/AGR hydrogel loaded with short PLLA/laminin fibers is a promising biomaterial for both tissue engineering applications and 3D tumor culture models.

4.
Cancers (Basel) ; 15(5)2023 Mar 02.
Article in English | MEDLINE | ID: mdl-36900355

ABSTRACT

Glioblastomas (GBM) are the most common, primary brain tumors in adults. Despite advances in neurosurgery and radio- and chemotherapy, the median survival of GBM patients is 15 months. Recent large-scale genomic, transcriptomic and epigenetic analyses have shown the cellular and molecular heterogeneity of GBMs, which hampers the outcomes of standard therapies. We have established 13 GBM-derived cell cultures from fresh tumor specimens and characterized them molecularly using RNA-seq, immunoblotting and immunocytochemistry. Evaluation of proneural (OLIG2, IDH1R132H, TP53 and PDGFRα), classical (EGFR) and mesenchymal markers (CHI3L1/YKL40, CD44 and phospho-STAT3), and the expression of pluripotency (SOX2, OLIG2, NESTIN) and differentiation (GFAP, MAP2, ß-Tubulin III) markers revealed the striking intertumor heterogeneity of primary GBM cell cultures. Upregulated expression of VIMENTIN, N-CADHERIN and CD44 at the mRNA/protein levels suggested increased epithelial-to-mesenchymal transition (EMT) in most studied cell cultures. The effects of temozolomide (TMZ) or doxorubicin (DOX) were tested in three GBM-derived cell cultures with different methylation status of the MGMT promoter. Amongst TMZ- or DOX-treated cultures, the strongest accumulation of the apoptotic markers caspase 7 and PARP were found in WG4 cells with methylated MGMT, suggesting that its methylation status predicts vulnerability to both drugs. As many GBM-derived cells showed high EGFR levels, we tested the effects of AG1478, an EGFR inhibitor, on downstream signaling pathways. AG1478 caused decreased levels of phospho-STAT3, and thus inhibition of active STAT3 augmented antitumor effects of DOX and TMZ in cells with methylated and intermediate status of MGMT. Altogether, our findings show that GBM-derived cell cultures mimic the considerable tumor heterogeneity, and that identifying patient-specific signaling vulnerabilities can assist in overcoming therapy resistance, by providing personalized combinatorial treatment recommendations.

5.
Clin Epigenetics ; 15(1): 29, 2023 02 27.
Article in English | MEDLINE | ID: mdl-36850002

ABSTRACT

BACKGROUND: Glioblastoma (GBM, WHO grade IV) is an aggressive, primary brain tumor. Despite extensive tumor resection followed by radio- and chemotherapy, life expectancy of GBM patients did not improve over decades. Several studies reported transcription deregulation in GBMs, but regulatory mechanisms driving overexpression of GBM-specific genes remain largely unknown. Transcription in open chromatin regions is directed by transcription factors (TFs) that bind to specific motifs, recruit co-activators/repressors and the transcriptional machinery. Identification of GBM-related TFs-gene regulatory networks may reveal new and targetable mechanisms of gliomagenesis. RESULTS: We predicted TFs-regulated networks in GBMs in silico and intersected them with putative TF binding sites identified in the accessible chromatin in human glioma cells and GBM patient samples. The Cancer Genome Atlas and Glioma Atlas datasets (DNA methylation, H3K27 acetylation, transcriptomic profiles) were explored to elucidate TFs-gene regulatory networks and effects of the epigenetic background. In contrast to the majority of tumors, c-Jun expression was higher in GBMs than in normal brain and c-Jun binding sites were found in multiple genes overexpressed in GBMs, including VIM, FOSL2 or UPP1. Binding of c-Jun to the VIM gene promoter was stronger in GBM-derived cells than in cells derived from benign glioma as evidenced by gel shift and supershift assays. Regulatory regions of the majority of c-Jun targets have distinct DNA methylation patterns in GBMs as compared to benign gliomas, suggesting the contribution of DNA methylation to the c-Jun-dependent gene expression. CONCLUSIONS: GBM-specific TFs-gene networks identified in GBMs differ from regulatory pathways attributed to benign brain tumors and imply a decisive role of c-Jun in controlling genes that drive glioma growth and invasion as well as a modulatory role of DNA methylation.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Humans , Brain Neoplasms/genetics , Chromatin/genetics , DNA Methylation , Epigenesis, Genetic , Glioblastoma/genetics , Proto-Oncogene Proteins c-jun/metabolism
6.
Adv Exp Med Biol ; 1202: 223-241, 2020.
Article in English | MEDLINE | ID: mdl-32034716

ABSTRACT

Cannabinoids are a group of structurally heterogeneous but pharmacologically related compounds, including plant-derived cannabinoids, synthetic substances and endogenous cannabinoids, such as anandamide and 2-arachidonoylglycerol. Cannabinoids elicit a wide range of central and peripheral effects mostly mediated through cannabinoid receptors. There are two types of specific Gi/o-protein-coupled receptors cloned so far, called CB1 and CB2, although an existence of additional cannabinoid-binding receptors has been suggested. CB1 and CB2 differ in their predicted amino acid sequence, tissue distribution, physiological role and signaling mechanisms. Significant alterations of a balance in the cannabinoid system between the levels of endogenous ligands and their receptors occur during malignant transformation in various types of cancer, including gliomas. Cannabinoids exert anti-proliferative action in tumor cells. Induction of cell death by cannabinoid treatment relies on the generation of a pro-apoptotic sphingolipid ceramide and disruption of signaling pathways crucial for regulation of cellular proliferation, differentiation or apoptosis. Increased ceramide levels lead also to ER-stress and autophagy in drug-treated glioblastoma cells. Beyond blocking of tumor cells proliferation cannabinoids inhibit invasiveness, angiogenesis and the stem cell-like properties of glioma cells, showing profound activity in the complex tumor microenvironment. Advances in translational research on cannabinoid signaling led to clinical investigations on the use of cannabinoids in treatments of glioblastomas.


Subject(s)
Cannabinoids/metabolism , Glioma/metabolism , Glioma/pathology , Signal Transduction , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Receptors, Cannabinoid/metabolism , Tumor Microenvironment
7.
Adv Exp Med Biol ; 1202: 259-279, 2020.
Article in English | MEDLINE | ID: mdl-32034718

ABSTRACT

Signal transduction pathways directly communicate and transform chromatin to change the epigenetic landscape and regulate gene expression. Chromatin acts as a dynamic platform of signal integration and storage. Histone modifications and alteration of chromatin structure play the main role in chromatin-based gene expression regulation. Alterations in genes coding for histone modifying enzymes and chromatin modifiers result in malfunction of proteins that regulate chromatin modification and remodeling. Such dysregulations culminate in profound changes in chromatin structure and distorted patterns of gene expression. Gliomagenesis is a multistep process, involving both genetic and epigenetic alterations. Recent applications of next generation sequencing have revealed that many chromatin regulation-related genes, including ATRX, ARID1A, SMARCA4, SMARCA2, SMARCC2, BAF155 and hSNF5 are mutated in gliomas. In this review we summarize newly identified mechanisms affecting expression or functions of selected histone modifying enzymes and chromatin modifiers in gliomas. We focus on selected examples of pathogenic mechanisms involving ATRX, histone methyltransferase G9a, histone acetylases/deacetylases and chromatin remodeling complexes SMARCA2/4. We discuss the impact of selected epigenetics alterations on glioma pathobiology, signaling and therapeutic responses. We assess the attempts of targeting defective pathways with new inhibitors.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Chromatin/metabolism , Glioma/drug therapy , Glioma/pathology , Histones/metabolism , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Chromatin/genetics , Chromatin Assembly and Disassembly/drug effects , Epigenesis, Genetic/drug effects , Glioma/enzymology , Glioma/genetics , Histone Code/drug effects , Histones/chemistry , Humans
8.
Front Immunol ; 9: 1329, 2018.
Article in English | MEDLINE | ID: mdl-29963047

ABSTRACT

Immune cells accumulating in the microenvironment of malignant tumors are tumor educated and contribute to its growth, progression, and evasion of antitumor immune responses. Glioblastoma (GBM), the common and most malignant primary brain tumor in adults, shows considerable accumulation of resident microglia and peripheral macrophages, and their polarization into tumor-supporting cells. There are controversies regarding a functional phenotype of glioma-associated microglia/macrophages (GAMs) due to a lack of consistent markers. Previous categorization of GAM polarization toward the M2 phenotype has been found inaccurate because of oversimplification of highly complex and heterogeneous responses. In this study, we characterized functional responses and gene expression in mouse and human microglial cultures exposed to fresh conditioned media [glioma-conditioned medium (GCM)] from human U87 and LN18 glioma cells. Functional analyses revealed mutual communication reflected by strong stimulation of glioma invasion by microglial cells and increased microglial phagocytosis after GCM treatment. To define transcriptomic markers of GCM-activated microglia, we performed selected and global gene expression analyses of stimulated microglial cells. We found activated pathways associated with immune evasion and TGF signaling. We performed computational comparison of the expression patterns of GAMs from human GBMs and rodent experimental gliomas to select genes consistently changed in different datasets. The analyses of marker genes in GAMs from different experimental models and clinical samples revealed only a small set of common genes, which reflects variegated responses in clinical and experimental settings. Tgm2 and Gpnmb were the only two genes common in the analyzed data sets. We discuss potential sources of the observed differences and stress a great need for definitive elucidation of a functional state of GAMs.

9.
Oncotarget ; 8(10): 16340-16355, 2017 Mar 07.
Article in English | MEDLINE | ID: mdl-28030801

ABSTRACT

Osteopontin (SPP1, a secreted phosphoprotein 1) is primarily involved in immune responses, tissue remodelling and biomineralization. However, it is also overexpressed in many cancers and regulates tumour progression by increasing migration, invasion and cancer stem cell self-renewal. Mechanisms of SPP1 overexpression in gliomas are poorly understood. We demonstrate overexpression of two out of five SPP1 isoforms in glioblastoma (GBM) and differential isoform expression in glioma cell lines. Up-regulated SPP1 expression is associated with binding of the GLI1 transcription factor to the promoter and OCT4 (octamer-binding transcription factor 4) to the first SPP1 intron of the SPP1 gene in human glioma cells but not in non-transformed astrocytes. GLI1 knockdown reduced SPP1 mRNA and protein levels in glioma cells. GLI1 and OCT4 are known regulators of stem cell pluripotency. GBMs contain rare cells that express stem cell markers and display ability to self-renew. We reveal that SPP1 is overexpressed in glioma initiating cells defined by high rhodamine 123 efflux, sphere forming capacity and stemness marker expression. Forced differentiation of human glioma spheres reduced SPP1 expression. Knockdown of SPP1, GLI1 or CD44 with siRNAs diminished sphere formation. C6 glioma cells stably depleted of Spp1 displayed reduced sphere forming capacity and downregulated stemness marker expression. Overexpression of the wild type Spp1, but not Spp1 lacking a Cd44 binding domain, rescued cell ability to form spheres. Our findings show re-activation of the embryonic-type transcriptional regulation of SPP1 in malignant gliomas and point to the importance of SPP1-CD44 interactions in self-renewal and pluripotency glioma initiating cells.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Neoplastic Stem Cells/metabolism , Osteopontin/genetics , Transcriptional Activation , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Differentiation/genetics , Cell Line, Tumor , Cell Self Renewal , Glioblastoma/metabolism , Glioblastoma/pathology , HeLa Cells , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Microscopy, Fluorescence , Neoplastic Stem Cells/pathology , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Osteopontin/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Rats , Reverse Transcriptase Polymerase Chain Reaction , Spheroids, Cellular/metabolism , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism
10.
Mech Ageing Dev ; 133(6): 444-55, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22613224

ABSTRACT

Curcumin, a natural polyphenol derived from the rhizome of Curcuma longa, is a potent anticancer agent, which restricts tumor cell growth both in vitro and in vivo. Thus far curcumin was shown to induce death of cancer cells. This study reports the induction of cellular senescence of human colon cancer cells HCT116 upon curcumin treatment. The SA-ß-galactosidase activation was observed both in p53+/+ and p53-/- cells, however the latter ones were less sensitive to the prosenescent activity of curcumin. Upregulation of p53 and p21 proteins was observed in p53+/+ HCT116, while p53-independent induction of p21 was noticed in p53-/- HCT116. Moreover, the senescence of HCT116 cells was accompanied by autophagy, that was confirmed by electron microscopy observations of autophagosomes in the curcumin-treated cells as well as LC3-II expression, punctue staining of LC3 and increased content of acidic vacuoles. Inhibition of autophagy, due to the diminished expression of ATG5 by RNAi decreased the number of senescent cells induced by curcumin, but did not lead to increased cell death. Altogether, we demonstrated a new antitumor activity of curcumin leading to cancer cell senescence and revealed the presence of a functional link between senescence and autophagy in curcumin-treated cells.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Cellular Senescence/drug effects , Colorectal Neoplasms/pathology , Curcumin/pharmacology , Autophagy/genetics , Autophagy-Related Protein 5 , Cell Proliferation/drug effects , Cellular Senescence/genetics , Gene Silencing , HCT116 Cells , Humans , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/genetics , Tumor Suppressor Protein p53/metabolism , Vacuoles/drug effects , beta-Galactosidase/metabolism
11.
Autophagy ; 5(6): 880-1, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19535901

ABSTRACT

Beclin 1 binds to Bcl-2 through its BH3 domain and this interaction inhibits starvation-induced autophagy. However, we have found that when Beclin 1 binds thus to Bcl-2, it fails to inhibit Bcl-2-mediated protection against four different inducers of apoptosis. In this punctum, we discuss possible reasons why Beclin 1 fails to behave like other BH3-only proteins and induce apoptosis.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Autophagy , Beclin-1 , HeLa Cells , Humans , Mice , Models, Biological
12.
Methods Mol Biol ; 445: 175-93, 2008.
Article in English | MEDLINE | ID: mdl-18425451

ABSTRACT

Macroautophagy (herein referred to as autophagy) contributes to the control of life and death throughout the animal and plant kingdoms. Bilateral links have been found between apoptosis and autophagy where inducers of apoptosis also induce autophagy and vice versa. In some cases, autophagy delays the onset of apoptosis and thus prolongs life although it may also promote apoptosis and other forms of cell death. It is thus of great biological and medical interest to understand the molecular connections between these two pathways, and try to utilize-or block-them selectively to aid induction of cell death (e.g., cancer cells) or inhibit death (e.g., in degenerative disorders). This chapter describes methods for studying apoptotic induction of autophagy and its effects on cell function. We also discuss potential pitfalls. Although cell lines are used as model systems, the substances and methods described here can be applied to primary cells and tissues.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Caspases/metabolism , Cell Line, Tumor , Collagen Type XI/metabolism , Flow Cytometry , HeLa Cells , Humans , Microtubule-Associated Proteins/metabolism
13.
Autophagy ; 3(6): 586-90, 2007.
Article in English | MEDLINE | ID: mdl-17786021

ABSTRACT

Expression of GFP-LC3 is now in widespread use to visualize autophagy in cultured cells. Recently, Kuma et al. (Autophagy 2007; 3:323-8) highlighted some complications using GFP-LC3, demonstrating that punctate dots containing GFP-LC3 do not always represent autophagic structures. We report here that GFP-LC3 can also rapidly aggregate into autophagosome look-alike structures when cells are permeabilized with saponin before cell fixation. Treatment with saponin reduced diffuse cytosolic and nuclear GFP-LC3 but caused an increase in the number and intensity of fluorescent puncta per cell regardless of whether the cells were induced to undergo autophagy. Saponin also induced GFP-LC3 puncta in Atg5(-/-) MEF transfected with GFP-LC3, where no LC3-II is produced, demonstrating that the puncta are autophagosome-independent. The increase in GFP-LC3 puncta was not matched by an increase in endogenous LC3-II or GFP-LC3-II detected by immunoblotting when protein samples were normalized to cell number. A qualitatively similar effect was observed when cells were treated with other detergents commonly used for membrane permeabilization, such as CHAPS, Triton X-100 or digitonin. We also noted that tubulin could not be used to normalize for protein loading on blots after applying saponin as it was selectively extracted from untreated cells but not from cells treated with vinblastine. When using mild detergents to remove background fluorescence, we recommend using a membrane-associated protein such as ATP synthase beta for normalization. Thus, detergents used prior to fixation may precipitate GFP-LC3 aggregation into structures that appear autophagosomal and so should be used with caution.


Subject(s)
Artifacts , Autophagy/physiology , Detergents/pharmacology , Green Fluorescent Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Saponins/pharmacology , Animals , Autophagy/drug effects , Autophagy-Related Protein 5 , COS Cells , Cell Line , Chlorocebus aethiops , Cholic Acids/pharmacology , Digitonin/pharmacology , Dose-Response Relationship, Drug , Embryo, Mammalian/cytology , Fibroblasts/metabolism , Humans , Kidney/cytology , Mice , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Octoxynol/pharmacology , Transfection , Tubulin Modulators/pharmacology , Vinblastine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...