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










Publication year range
1.
Discov Med ; 35(178): 733-749, 2023 10.
Article in English | MEDLINE | ID: mdl-37811612

ABSTRACT

Pediatric brain tumors currently show the highest incidence among solid childhood malignancies and, together with leukemia, are the leading cause of death from cancer in childhood. Embryonal brain tumors are the most common and frequent type of childhood brain cancer and are usually characterized by an extremely aggressive course of the disease with the worst outcomes in most cases. There is an urgent need for specific refined molecular diagnostics, which would help to develop personalized treatment. In the present review paper, the latest molecular characteristics of various classified forms of embryonal brain tumors were analyzed in detail. Overexpression of the MYC and MYCN genes is characteristic of many embryonal brain tumors, leading to enhanced cell proliferation and disturbances in the cell cycle. The functioning of the SWI2/SNF2 chromatin remodeling complex are distorted in such malignancies as well. Noteworthy, LIN28 and MYC discussed here are involved in the induction of pluripotency. We have to mention that molecular mechanisms underlying the development of embryonal brain tumors of the central nervous system (CNS) are still not well understood. Thus, it is important to uncover such mechanisms with the aim to provide a better prognosis of the course of disease and to create personalized therapy.


Subject(s)
Brain Neoplasms , Neoplasms, Germ Cell and Embryonal , Child , Humans , Neoplasms, Germ Cell and Embryonal/diagnosis , Neoplasms, Germ Cell and Embryonal/genetics , Neoplasms, Germ Cell and Embryonal/therapy , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Prognosis , Brain , Pathology, Molecular
2.
Sci Rep ; 12(1): 22154, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36550267

ABSTRACT

In a previous study, we showed that serine/threonine-protein kinase 4 (STK4) is involved in the control on proliferation and migration of endometrial cancer (EC) cells in vitro. In the present paper, we studied STK4 expression in EC tissues from a large cohort of patients to determine whether STK4 can serve as a marker for the aggressiveness and prognosis of EC. Tissue samples from patients with EC were examined for tumor type, grade, and stage. The STK4 protein expression in EC cells was assessed by immunohistochemistry and related to clinicopathological data of patients, such as progression and patient survival rate. The STK4 mRNA levels and its relation to the survival rate were analyzed also in publicly available databases. The STK4 gene expression was low at both, the mRNA and protein levels in EC, especially in serous tumors. Comparison of STK4 expression with the patient survival rate shows that the higher expression is associated with worse prognosis in serous EC, while no such dependence was found in endometrioid EC. Hence, the determination of the SKT4 expression pattern could be used as a putative prognostic marker for serous EC.


Subject(s)
Carcinoma, Endometrioid , Cystadenocarcinoma, Serous , Endometrial Neoplasms , Female , Humans , Endometrial Neoplasms/pathology , Carcinoma, Endometrioid/pathology , Endometrium/metabolism , Prognosis , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Protein Serine-Threonine Kinases/genetics , Intracellular Signaling Peptides and Proteins
3.
Cancer Control ; 29: 10732748221094797, 2022.
Article in English | MEDLINE | ID: mdl-35533253

ABSTRACT

BACKGROUND: Several markers of survival among endometrial cancer (EC) patients have been proposed, namely, the oncoprotein stathmin, RAF kinase inhibitor (RKIP), Cyclin A, GATA-binding protein 3 (GATA3), and growth and differentiation factor-15 (GDF-15). Their elevated expression correlated significantly with a high stage, serous papillary/clear cell subtypes, and aneuploidy. In a previous study, we reported the elevated expression of the serine/threonine protein kinase N1 (PKN1) in cancerous cells. In the present paper, we studied PKN1 expression in EC tissues from a large cohort of patients, to determine whether PKN1 can serve as a marker for the aggressiveness and prognosis of EC, and/or as a marker of survival among EC patients. METHODS: Tissue samples from EC patients were examined retrospectively for tumor type, tumor size, FIGO stage and grade, depth of invasion in the myometrium, and presence of lymph node metastasis. The PKN1 protein expression in EC cells was assessed by immunohistochemistry. PKN1 mRNA levels were analyzed in publicly available databases, using bioinformatic tools. RESULTS: We found that expression of PKN1 at the mRNA and proteins levels tended to increase in high-grade EC samples (P = .0001 and P = .06, respectively). In addition, patients with metastatic disease had higher PKN1 mRNA levels (P = .02). Moreover, patients with high PKN1 expression could be characterized by poorer survival. CONCLUSIONS: We have shown a trend of the higher PKN1 expression levels in EC patients with poor prognosis. Therefore, PKN1 might be considered as a candidate prognostic marker for EC.


Subject(s)
Biomarkers, Tumor , Endometrial Neoplasms , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Female , Humans , Prognosis , Protein Kinase C , RNA, Messenger , Retrospective Studies , Up-Regulation
4.
Life (Basel) ; 12(3)2022 Mar 05.
Article in English | MEDLINE | ID: mdl-35330127

ABSTRACT

Terahertz (THz) electromagnetic radiation is commonly used in astronomy, security screening, imaging, and biomedicine, among other applications. Such approach has raised the question of the influence of THz irradiation on biological objects, especially the human body. However, the results obtained to date are quite controversial. Therefore, we performed a comparative study on the viability of normal cells and cancer cells upon irradiation with a steady beam of THz rays. We used human peripheral blood mononuclear cells and cancer cell lines. Primary human mononuclear blood cells (monocytes, and B-, and T-cells) showed an increased death rate, determined by cell counting and fluorescence microscopy, upon 0.14 THz irradiation. The effect of THz radiation was different among malignant cells of B- and T-cell origin (Ramos and Jurkat cells) and epithelial cancer cells (MCF7 and LNCaP). This was demonstrated by cell counting and by the alamarBlue assay. In conclusion, THz radiation can result in the death of human primary and malignant cells. However, the mechanism of this phenomenon is largely unknown. Hence, more work should be done to shed some light on the mechanism of action of THz irradiation in living organisms to enhance technologic developments.

5.
Proc Natl Acad Sci U S A ; 117(27): 15673-15683, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32571933

ABSTRACT

Stemness encompasses the capability of a cell for self-renewal and differentiation. The stem cell maintains a balance between proliferation, quiescence, and regeneration via interactions with the microenvironment. Previously, we showed that ectopic expression of the mitochondrial ribosomal protein S18-2 (MRPS18-2) led to immortalization of primary fibroblasts, accompanied by induction of an embryonic stem cell (ESC) phenotype. Moreover, we demonstrated interaction between S18-2 and the retinoblastoma-associated protein (RB) and hypothesized that the simultaneous expression of RB and S18-2 is essential for maintaining cell stemness. Here, we experimentally investigated the role of S18-2 in cell stemness and differentiation. Concurrent expression of RB and S18-2 resulted in immortalization of Rb1-/- primary mouse embryonic fibroblasts and in aggressive tumor growth in severe combined immunodeficiency mice. These cells, which express both RB and S18-2 at high levels, exhibited the potential to differentiate into various lineages in vitro, including osteogenic, chondrogenic, and adipogenic lineages. Mechanistically, S18-2 formed a multimeric protein complex with prohibitin and the ring finger protein 2 (RNF2). This molecular complex increased the monoubiquitination of histone H2ALys119, a characteristic trait of ESCs, by enhanced E3-ligase activity of RNF2. Furthermore, we found enrichment of KLF4 at the S18-2 promoter region and that the S18-2 expression is positively correlated with KLF4 levels. Importantly, knockdown of S18-2 in zebrafish larvae led to embryonic lethality. Collectively, our findings suggest an important role for S18-2 in cell stemness and differentiation and potentially also in cancerogenesis.


Subject(s)
Mitochondria/genetics , Mouse Embryonic Stem Cells/metabolism , Retinoblastoma Binding Proteins/genetics , Ribosomal Proteins/genetics , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Cell Self Renewal/genetics , Fibroblasts/metabolism , Gene Expression Regulation, Developmental/genetics , Histones/genetics , Human Embryonic Stem Cells/metabolism , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Mice , Mitochondria/metabolism , Polycomb Repressive Complex 1/genetics , Ribosomal Proteins/chemistry , Tumor Microenvironment/genetics , Ubiquitin-Protein Ligases/genetics
6.
Cell Mol Immunol ; 17(3): 300-301, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31863080

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

7.
Clin Immunol ; 204: 14-22, 2019 07.
Article in English | MEDLINE | ID: mdl-30616923

ABSTRACT

SLAMF1/CD150 receptor is a founder of signaling lymphocyte activation molecule (SLAM) family of cell-surface receptors. It is widely expressed on cells within hematopoietic system. In hematologic malignancies CD150 cell surface expression is restricted to cutaneous T-cell lymphomas, few types of B-cell non-Hodgkin's lymphoma, near half of cases of chronic lymphocytic leukemia, Hodgkin's lymphoma, and multiple myeloma. Differential expression among various types of hematological malignancies allows considering CD150 as diagnostical and potential prognostic marker. Moreover, CD150 may be a target for antibody-based or measles virus oncolytic therapy. Due to CD150 signaling properties it is involved in regulation of malignant cell fate decision and tumor microenvironment in Hodgkin's lymphoma and chronic lymphocytic leukemia. This review summarizes evidence for the important role of CD150 in pathogenesis of hematologic malignancies.


Subject(s)
Hematologic Neoplasms , Signaling Lymphocytic Activation Molecule Family Member 1 , Animals , Humans
8.
Oncotarget ; 7(16): 22150-8, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-26959119

ABSTRACT

Endometrial cancer (EC) is one of the most frequent causes of cancer death among women in developed countries. Histopathological diagnosis and imaging techniques for EC are limited, thus new prognostic markers are needed to offer patients the best treatment and follow-up.In the present paper we showed that the level of mitochondrial ribosomal protein MRPS18-2 (S18-2) increased in EC compared with the normal endometrium and hyperplasia, based on a study of 42 patient biopsies. Importantly, high expression of free E2F1 in EC correlates well with high S18-2 expression. The EC cell line HEC-1-A, which overexpresses S18-2 constitutively, showed an increased proliferation capacity in vitro and in vivo (in SCID mice). Moreover, pan-keratin, beta-catenin and E-cadherin signals are diminished in these cells, compared to the parental HEC-1-A line, in contrast to vimentin signal that is increased. This may be associated with epithelial-mesenchymal cell transition (EMT).We conclude that high expression of S18-2 and free E2F1, and low pan-keratin, beta-catenin, and E-cadherin signals might be a good set of prognostic markers for EC.


Subject(s)
Adenocarcinoma/pathology , Biomarkers, Tumor/analysis , E2F1 Transcription Factor/biosynthesis , Endometrial Neoplasms/pathology , Ribosomal Proteins/biosynthesis , Adenocarcinoma/metabolism , Animals , Antigens, CD , Cadherins/biosynthesis , Endometrial Neoplasms/metabolism , Female , Heterografts , Humans , Keratins/biosynthesis , Mice , Mice, SCID , beta Catenin/biosynthesis
9.
Cell Mol Immunol ; 13(6): 828-838, 2016 11.
Article in English | MEDLINE | ID: mdl-26073466

ABSTRACT

Measles virus (MV) is highly contagious pathogen, which causes a profound immunosuppression, resulting in high infant mortality. This virus infects dendritic cells (DCs) following the binding of MV hemagglutinin (MV-H) to CD150 receptor and alters DC functions by a mechanism that is not completely understood. We have analyzed the effect of MV-H interaction with CD150-expressing DCs on the DC signaling pathways and consequent phenotypic and functional changes in the absence of infectious context. We demonstrated that contact between CD150 on human DCs and MV-H expressed on membrane of transfected CHO cells was sufficient to modulate the activity of two major regulatory pathways of DC differentiation and function: to stimulate Akt and inhibit p38 MAPK phosphorylation, without concomitant ERK1/2 activation. Furthermore, interaction with MV-H decreased the expression level of DC activation markers CD80, CD83, CD86, and HLA-DR and strongly downregulated IL-12 production but did not modulate IL-10 secretion. Moreover, contact with MV-H suppressed DC-mediated T-cell alloproliferation, demonstrating profound alteration of DC maturation and functions. Finally, engagement of CD150 by MV-H in mice transgenic for human CD150 decreased inflammatory responses, showing the immunosuppressive effect of CD150-MV-H interaction in vivo. Altogether, these results uncover novel mechanism of MV-induced immunosuppression, implicating modulation of cell signaling pathways following MV-H interaction with CD150-expressing DCs and reveal anti-inflammatory effects of CD150 stimulation.


Subject(s)
Dendritic Cells/immunology , Hemagglutinins/immunology , Immunity , Intracellular Space/metabolism , Measles virus/immunology , Signal Transduction , Signaling Lymphocytic Activation Molecule Family Member 1/metabolism , Animals , Cell Line , Humans , Inflammation/pathology , Mice, Inbred C57BL , Models, Biological , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Oncotarget ; 6(25): 21016-28, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26023799

ABSTRACT

We have shown earlier that overexpression of the human mitochondrial ribosomal protein MRPS18-2 (S18-2) led to immortalization of primary rat embryonic fibroblasts. The derived cells expressed the embryonic stem cell markers, and cellular pathways that control cell proliferation, oxidative phosphorylation, cellular respiration, and other redox reactions were activated in the immortalized cells.Here we report that, upon overexpression of S18-2 protein, primary rat skin fibroblasts underwent cell transformation. Cells passed more than 300 population doublings, and two out of three tested clones gave rise to tumors in experimental animals. Transformed cells showed anchorage-independent growth and loss of contact inhibition; they expressed epithelial markers, such as E-cadherin and ß-catenin. Transformed cells showed increased telomerase activity, disturbance of the cell cycle, and chromosomal instability. Taken together, our data suggest that S18-2 is a newly identified oncoprotein that may be involved in cancerogenesis.


Subject(s)
Cell Transformation, Neoplastic , Fibroblasts/metabolism , Ribosomal Proteins/metabolism , Skin/metabolism , Animals , Carcinogenesis , Cell Proliferation , Chromosomal Instability , Flow Cytometry , Green Fluorescent Proteins/metabolism , Humans , Karyotyping , Lipids/chemistry , Mice , Mice, SCID , Mitochondria/metabolism , Oxidation-Reduction , Oxygen/chemistry , Rats , Telomerase/metabolism
11.
PLoS One ; 10(2): e0118302, 2015.
Article in English | MEDLINE | ID: mdl-25710480

ABSTRACT

CD150 (IPO3/SLAM) belongs to the SLAM family of receptors and serves as a major entry receptor for measles virus. CD150 is expressed on normal and malignant cells of the immune system. However, little is known about its expression outside the hematopoietic system, especially tumors of the central nervous system (CNS). Although CD150 was not found in different regions of normal brain tissues, our immunohistochemical study revealed its expression in 77.6% of human CNS tumors, including glioblastoma, anaplastic astrocytoma, diffuse astrocytoma, ependymoma, and others. CD150 was detected in the cytoplasm, but not on the cell surface of glioma cell lines, and it was colocalized with the endoplasmic reticulum and Golgi complex markers. In addition to the full length mRNA of the mCD150 splice isoform, in glioma cells we found a highly expressed novel CD150 transcript (nCD150), containing an 83 bp insert. The insert is derived from a previously unrecognized exon designated Cyt-new, which is located 510 bp downstream of the transmembrane region exon, and is a specific feature of primate SLAMF1. Both mCD150 and nCD150 cDNA variants did not contain any mutations and had the leader sequence. The nCD150 transcript was also detected in normal and malignant B lymphocytes, primary T cells, dendritic cells and macrophages; however, in glioma cells nCD150 was found to be the predominant CD150 isoform. Similarly to mCD150, cell surface expression of nCD150 allows wild type measles virus entry to the cell. Our data indicate that CD150 expression in CNS tumors can be considered a new diagnostic marker and potential target for novel therapeutic approaches.


Subject(s)
Antigens, CD/genetics , Gene Expression Regulation , Receptors, Cell Surface/genetics , Amino Acid Sequence , Antigens, CD/chemistry , Antigens, CD/metabolism , B-Lymphocytes/metabolism , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Central Nervous System Neoplasms/metabolism , Central Nervous System Neoplasms/pathology , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , HEK293 Cells , Humans , Immunohistochemistry , Measles virus/physiology , Molecular Sequence Data , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Splicing , RNA, Messenger/metabolism , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1
12.
Immunol Cell Biol ; 88(5): 565-74, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20231852

ABSTRACT

The CD150 receptor is expressed on thymocytes, activated and memory T cells, B cells, platelets, natural killer T cells, and mature dendritic cells, and is also detected on tumor cells of Hodgkin's lymphoma (HL) and diffuse large B-cell lymphoma with an activated B cell phenotype. Here, we report that the level of CD150 expression is elevated during B cell differentiation toward plasma cells. In primary tonsillar B cells and HL cell lines, CD150 signaling regulates the phosphorylation of three types of mitogen-activated protein kinases (MAPKs): extracellular signal-regulated kinase 1/2 (ERK1/2), p38 MAPK, and Jun N-terminal kinase 1/2 (JNK1/2). CD150 induced ERK1/2 activation in primary tonsillar B cells and in two HL cell lines. CD150 mediated activation of JNK1/2 p54 and JNK2-gamma kinase isoforms in all CD150(+) B cell lines we tested. CD150 associated with the serine/threonine kinase hematopoetic progenitor kinase 1 (HPK1) regardless of CD150 tyrosine phosphorylation or binding of the SH2D1A adaptor protein to CD150, and HPK1 overexpression enhanced CD150-mediated JNK1/2 phosphorylation. CD150 ligation inhibited cell proliferation of all studied HL cell lines and induced apoptosis in L1236 HL cells that did not depend on JNK activity. As signaling through CD150 modulates MAPK activity in HL tumor cells, CD150 may contribute to regulation of tumor cell maintenance in low-rate proliferating HLs.


Subject(s)
Antigens, CD/immunology , B-Lymphocytes/immunology , Enzyme Activation/immunology , Hodgkin Disease/immunology , Mitogen-Activated Protein Kinases/immunology , Receptors, Cell Surface/immunology , Signal Transduction/immunology , Antigens, CD/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Blotting, Western , Cell Differentiation/immunology , Cell Line, Tumor , Cell Separation , Flow Cytometry , Hodgkin Disease/metabolism , Humans , Immunohistochemistry , Mitogen-Activated Protein Kinase 8/immunology , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/immunology , Mitogen-Activated Protein Kinase 9/metabolism , Mitogen-Activated Protein Kinases/metabolism , Receptors, Cell Surface/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1 , Transfection
13.
Exp Oncol ; 26(3): 210-6, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15494689

ABSTRACT

AIM: To understand the biochemical basis of cell sensitivity to cytotoxic effect of doxorubicine (DOX), we investigated signaling cascades mediated by c-Jun N-terminal protein kinases (JNK1/2), p38 mitogen-activated protein kinases (MAPK), extracellular signal-regulated protein kinase (ERK1/2) and protein kinase B/Akt in both DOX-sensitive BL41 and the DOX-resistant DG75 Burkitt's lymphoma (BL) cell lines. METHODS: To test the effect of DOX on different signaling cascades, BL41 and DG75 cells were treated with DOX for varying lengths of time. Cytotoxic effect of DOX was analyzed by Hoechst 33342 staining. Total amount of JNK1/2, ERK1/2, p38 MARK, Akt proteins, and also phosphorylated/activated forms of these enzymes were detected using Western blot analysis with specific antibodies. Immunophenotypic analysis of BL41 and DG75 cells was performed by indirect immunofluorescence staining. RESULTS: Our findings demonstrated that DOX treatment of the BL41 cells led to sustained activation of JNK1/2 and p38 MAPK. This activation/phosphorylation did not result from increased expression of either JNK1/2 or p38 MAPK since protein levels of JNK1/2 and p38 MAPK in DOX-treated and untreated cells were unaltered. Apoptotic signaling cascade induced by DOX in BL41 cell was accompanied by Akt dephosphorylation. The effect of DOX in drug-resistant cell line DG75 convoyed by dephosphorylation of JNK1/2, p38 MAPK and activation of Akt. Fate of BL cells did not depend from ERK activity. CONCLUSION: The outcome of cellular response to DOX in BL cell lines is determined by interference of at least three signaling pathways: JNK1/2, p38 MAPK and PKB/Akt. The balance between Akt/PKB and MAPK pathways is important in determining whether BL cells survive or undergo apoptosis in response to DOX treatment.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Burkitt Lymphoma/pathology , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Signal Transduction , Apoptosis/drug effects , Burkitt Lymphoma/metabolism , Cell Line, Tumor/chemistry , Cell Line, Tumor/drug effects , Humans , Immunophenotyping , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/analysis , Mitogen-Activated Protein Kinase 3/analysis , Mitogen-Activated Protein Kinase 8/analysis , Mitogen-Activated Protein Kinase 9/analysis , Neoplasm Proteins/analysis , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...