Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 162
Filter
1.
J Immunother Cancer ; 10(8)2022 08.
Article in English | MEDLINE | ID: mdl-35918123

ABSTRACT

BACKGROUND: One of the main difficulties of adoptive cell therapies with chimeric antigen receptor (CAR)-T cells in solid tumors is the identification of specific target antigens. The tumor microenvironment can present suitable antigens for CAR design, even though they are not expressed by the tumor cells. We have generated a CAR specific for the splice variant extra domain A (EDA) of fibronectin, which is highly expressed in the tumor stroma of many types of tumors but not in healthy tissues. METHODS: EDA expression was explored in RNA-seq data from different human tumor types and by immunohistochemistry in paraffin-embedded tumor biopsies. Murine and human anti-EDA CAR-T cells were prepared using recombinant retro/lentiviruses, respectively. The functionality of EDA CAR-T cells was measured in vitro in response to antigen stimulation. The antitumor activity of EDA CAR-T cells was measured in vivo in C57BL/6 mice challenged with PM299L-EDA hepatocarcinoma cell line, in 129Sv mice-bearing F9 teratocarcinoma and in NSG mice injected with the human hepatocarcinoma cell line PLC. RESULTS: EDA CAR-T cells recognized and killed EDA-expressing tumor cell lines in vitro and rejected EDA-expressing tumors in immunocompetent mice. Notably, EDA CAR-T cells showed an antitumor effect in mice injected with EDA-negative tumor cells lines when the tumor stroma or the basement membrane of tumor endothelial cells express EDA. Thus, EDA CAR-T administration delayed tumor growth in immunocompetent 129Sv mice challenged with teratocarcinoma cell line F9. EDA CAR-T treatment exerted an antiangiogenic effect and significantly reduced gene signatures associated with epithelial-mesenchymal transition, collagen synthesis, extracellular matrix organization as well as IL-6-STAT5 and KRAS pathways. Importantly, the human version of EDA CAR, that includes the human 41BB and CD3ζ endodomains, exerted strong antitumor activity in NSG mice challenged with the human hepatocarcinoma cell line PLC, which expresses EDA in the tumor stroma and the endothelial vasculature. EDA CAR-T cells exhibited a tropism for EDA-expressing tumor tissue and no toxicity was observed in tumor bearing or in healthy mice. CONCLUSIONS: These results suggest that targeting the tumor-specific fibronectin splice variant EDA with CAR-T cells is feasible and offers a therapeutic option that is applicable to different types of cancer.


Subject(s)
Receptors, Chimeric Antigen , Teratocarcinoma , Animals , Endothelial Cells , Fibronectins , Humans , Mice , Mice, Inbred C57BL , T-Lymphocytes , Teratocarcinoma/metabolism , Tumor Microenvironment , Xenograft Model Antitumor Assays
2.
Semin Cancer Biol ; 81: 193-205, 2022 06.
Article in English | MEDLINE | ID: mdl-33940178

ABSTRACT

The completion-of-tumor hypothesis involved in the dynamic interplay between the initiating oncogenic event and progression is essential to better recognize the foundational framework of tumors. Here we review and extend the gametogenesis-related hypothesis of tumors, because high embryonic/germ cell traits are common in tumors. The century-old gametogenesis-related hypothesis of tumors postulated that tumors arise from displaced/activated trophoblasts, displaced (lost) germ cells, and the reprogramming/reactivation of gametogenic program in somatic cells. Early primordial germ cells (PGCs), embryonic stem (ES) cells, embryonic germ cells (EGCs), and pre-implantation embryos at the stage from two-cell stage to blastocysts originating from fertilization or parthenogenesis have the potential to develop teratomas/teratocarcinomas. In addition, the teratomas/teratocarcinomas/germ cells occur in gonads and extra-gonads. Undoubtedly, the findings provide strong support for the hypothesis. However, it was thought that these tumor types were an exception rather than verification. In fact, there are extensive similarities between somatic tumor types and embryonic/germ cell development, such as antigens, migration, invasion, and immune escape. It was documented that embryonic/germ cell genes play crucial roles in tumor behaviors, e.g. tumor initiation and metastasis. Of note, embryonic/germ cell-like tumor cells at different developmental stages including PGC and oocyte to the early embryo-like stage were identified in diverse tumor types by our group. These embryonic/germ cell-like cancer cells resemble the natural embryonic/germ cells in morphology, gene expression, the capability of teratoma formation, and the ability to undergo the process of oocyte maturation and parthenogenesis. These embryonic/germ cell-like cancer cells are derived from somatic cells and contribute to tumor formation, metastasis, and drug resistance, establishing asexual meiotic embryonic life cycle. p53 inhibits the reactivation of embryonic/germ cell state in somatic cells and oocyte-like cell maturation. Based on earlier and our recent studies, we propose a novel model to complete the gametogenesis-related hypothesis of tumors, which can be applied to certain somatic tumors. That is, tumors tend to establish a somatic asexual meiotic embryonic cycle through the activation of somatic female gametogenesis and parthenogenesis in somatic tumor cells during the tumor progression, thus passing on corresponding embryonic/germ cell traits leading to the malignant behaviors and enhancing the cells' independence. This concept may be instrumental to better understand the nature and evolution of tumors. We rationalize that targeting the key events of somatic pregnancy is likely a better therapeutic strategy for cancer treatment than directly targeting cell mitotic proliferation, especially for those tumors with p53 inactivation.


Subject(s)
Teratocarcinoma , Teratoma , Female , Gametogenesis , Germ Cells/metabolism , Humans , Pregnancy , Teratocarcinoma/metabolism , Teratocarcinoma/pathology , Teratoma/metabolism , Teratoma/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
3.
Int J Mol Sci ; 22(22)2021 Nov 17.
Article in English | MEDLINE | ID: mdl-34830279

ABSTRACT

About 8% of our genome is composed of sequences from Human Endogenous Retroviruses (HERVs). The HERV-K (HML.2) family, here abbreviated HML.2, is able to produce virus particles that were detected in cell lines, malignant tumors and in autoimmune diseases. Parameters and properties of HML.2 released from teratocarcinoma cell lines GH and Tera-1 were investigated in detail. In most experiments, analyzed viruses were purified by density gradient centrifugation. HML.2 structural proteins, reverse transcriptase (RT) activity, viral RNA (vRNA) and particle morphology were analyzed. The HML.2 markers were predominantly detected in fractions with a buoyant density of 1.16 g/cm3. Deglycosylation of TM revealed truncated forms of transmembrane (TM) protein. Free virions and extracellular vesicles (presumably microvesicles-MVs) with HML.2 elements, including budding intermediates, were detected by electron microscopy. Viral elements and assembled virions captured and exported by MVs can boost specific immune responses and trigger immunomodulation in recipient cells. Sequencing of cDNA clones demonstrated exclusive presence of HERV-K108 env in HML.2 from Tera-1 cells. Not counting two recombinant variants, four known env sequences were found in HML.2 from GH cells. Obtained results shed light on parameters and morphology of HML.2. A possible mechanism of HML.2-induced diseases is discussed.


Subject(s)
Capsid/metabolism , Endogenous Retroviruses/genetics , Endogenous Retroviruses/metabolism , Extracellular Vesicles/virology , Gene Products, env/metabolism , Genes, env , RNA, Viral/genetics , Teratocarcinoma/metabolism , Teratocarcinoma/virology , Viral Envelope/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cell Membrane/virology , Centrifugation, Density Gradient/methods , Endogenous Retroviruses/isolation & purification , Gene Products, env/genetics , HEK293 Cells , Humans , Membrane Proteins/metabolism , RNA, Viral/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction/methods , Teratocarcinoma/pathology , Transfection , Virus Assembly/genetics
4.
Methods Mol Biol ; 2311: 25-38, 2021.
Article in English | MEDLINE | ID: mdl-34033075

ABSTRACT

The lack of a convenient, easily maintained, and inexpensive in vitro human neuronal model to study neurodegenerative diseases prompted us to develop a rapid, 1-h differentiated neuronal cell model based on human NT2 cells and C3 transferase. Here, we describe the rapid differentiation of human neuronal NT2 cells, and the differentiation, transduction, and transfection of human SK-N-MC cells and rat PC12 cells to obtain cells with the morphology of differentiated neurons that can express exogenous genes of interest at high level.


Subject(s)
Adrenal Gland Neoplasms/pathology , Neuroblastoma/metabolism , Neurogenesis , Neurons/pathology , Pheochromocytoma/pathology , Teratocarcinoma/pathology , ADP Ribose Transferases/pharmacology , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/metabolism , Animals , Botulinum Toxins/pharmacology , Cell Culture Techniques , Humans , Neuroblastoma/genetics , Neuroblastoma/pathology , Neurogenesis/drug effects , Neuronal Outgrowth , Neurons/drug effects , Neurons/metabolism , PC12 Cells , Phenotype , Pheochromocytoma/genetics , Pheochromocytoma/metabolism , Rats , Teratocarcinoma/genetics , Teratocarcinoma/metabolism , Transfection , Tretinoin/pharmacology
5.
Methods Mol Biol ; 2311: 39-49, 2021.
Article in English | MEDLINE | ID: mdl-34033076

ABSTRACT

This chapter describes the culture and propagation of murine embryonic stem cells, F9 and P19, and strategies for differentiation of these stem cells into neurons. Additional techniques are described for obtaining enriched populations of mature neurons from P19 cells and differentiation of F9 cells into serotonergic or catecholaminergic neurons. The protocols described herein can be used for dissection of the pathways such as gliogenesis and neurogenesis that are involved in differentiation of pluripotent stem cells such as F9 and P19 into glial cells or terminally differentiated neurons.


Subject(s)
Mouse Embryonic Stem Cells/pathology , Neural Stem Cells/pathology , Neurogenesis , Neurons/pathology , Teratocarcinoma/pathology , Animals , Cell Culture Techniques , Cell Line, Tumor , Cell Proliferation , Mice , Mouse Embryonic Stem Cells/drug effects , Mouse Embryonic Stem Cells/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Neurons/drug effects , Neurons/metabolism , Phenotype , Teratocarcinoma/metabolism , Tretinoin/pharmacology
6.
Mol Biol Rep ; 48(2): 1985-1994, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33619662

ABSTRACT

Reck (REversion-inducing Cysteine-rich protein with Kazal motifs) tumor suppressor gene encodes a multifunctional glycoprotein which inhibits the activity of several matrix metalloproteinases (MMPs), and has the ability to modulate the Notch and canonical Wnt pathways. Reck-deficient neuro-progenitor cells undergo precocious differentiation; however, modulation of Reck expression during progression of the neuronal differentiation process is yet to be characterized. In the present study, we demonstrate that Reck expression levels are increased during in vitro neuronal differentiation of PC12 pheochromocytoma cells and P19 murine teratocarcinoma cells and characterize mouse Reck promoter activity during this process. Increased Reck promoter activity was found upon induction of differentiation in PC12 cells, in accordance with its increased mRNA expression levels in mouse in vitro models. Interestingly, Reck overexpression, prior to the beginning of the differentiation protocol, led to diminished efficiency of the neuronal differentiation process. Taken together, our findings suggest that increased Reck expression at early stages of differentiation diminishes the number of neuron-like cells, which are positive for the beta-3 tubulin marker. Our data highlight the importance of Reck expression evaluation to optimize in vitro neuronal differentiation protocols.


Subject(s)
GPI-Linked Proteins/metabolism , Genes, Tumor Suppressor , Neurogenesis/genetics , Teratocarcinoma/metabolism , Animals , Binding Sites , Flow Cytometry , GPI-Linked Proteins/genetics , Gene Expression Regulation, Neoplastic/genetics , Mice , PC12 Cells , Promoter Regions, Genetic , Rats , Real-Time Polymerase Chain Reaction , Teratocarcinoma/genetics , Tubulin/metabolism , Up-Regulation
7.
Int J Mol Sci ; 22(4)2021 Feb 08.
Article in English | MEDLINE | ID: mdl-33567764

ABSTRACT

Cripto-1 is a member of the EGF-CFC/FRL1/Cryptic family and is involved in embryonic development and carcinogenesis. We designed a novel anti-Cripto-1 artificial antibody and assessed the recognition to the antigen and the potential to suppress the growth of cancer stem cells. First, single chain antibody clones were isolated by bio-panning with the affinity to recombinant Cripto-1 protein from our original phage-display library. Then, the variable regions of heavy chain VH and light chain VL in each clone were fused to constant regions of heavy chain CH and light chain CL regions respectively. These fused genes were expressed in ExpiCHO-S cells to produce artificial humanized antibodies against Cripto-1. After evaluation of the expression levels, one clone was selected and the anti-Cripto-1 antibody was produced and purified. The purified antibody showed affinity to recombinant Cripto-1 at 1.1 pmol and immunoreactivity to cancer tissues and cell lines. The antibody was available to detect the immunoreactivity in tissue microarrays of malignant tumors as well as in Cripto-1 overexpressing cells. Simultaneously, the antibody exhibited the potential to suppress the growth of human colon cancer derived GEO cells overexpressing Cripto-1 with IC50 at approximately 110 nM. The artificially humanized antibody is proposed to be a good candidate to target cancer cells overexpressing Cripto-1.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , GPI-Linked Proteins/immunology , Intercellular Signaling Peptides and Proteins/immunology , Neoplasm Proteins/immunology , Teratocarcinoma/drug therapy , Testicular Neoplasms/drug therapy , Amino Acid Sequence , Antibodies, Monoclonal, Humanized/immunology , Antineoplastic Agents/immunology , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation , Female , Humans , Male , Sequence Homology , Teratocarcinoma/immunology , Teratocarcinoma/metabolism , Teratocarcinoma/pathology , Testicular Neoplasms/immunology , Testicular Neoplasms/metabolism , Testicular Neoplasms/pathology , Tumor Cells, Cultured
8.
Int J Mol Med ; 47(1): 397-407, 2021 01.
Article in English | MEDLINE | ID: mdl-33416104

ABSTRACT

Immature ovarian teratocarcinoma (IOT) is a rare and malignant type of ovarian teratoma, and the molecular mechanisms underlying the pathogenesis and malignant phenotype of IOT remain uncharacterized. The present study examined a long non­coding RNA (lncRNA), long­chain intergenic non­coding RNA324 (LINC00324), which may serve a crucial role in pathogenesis of IOT. According to the results, LINC00324 was upregulated in IOT tissues and cells, as determined by reverse transcription­quantitative PCR, and its depletion impaired cell proliferation ability and improved cell apoptosis ability in IOT. Furthermore, LINC00324 acted as a miR­214­5p sponge to derepress cyclin dependent kinase 6 (CDK6), cyclin D1 (CCND1), murine double minute homolog 2 (MDM2), and mouse double minute 4 (MDM4) expression, thus increasing IOT cell proliferation and repressing apoptosis. Taken together, these results demonstrated that LINC00324 could serve as a competing endogenous RNA to facilitate IOT cell proliferation by regulation of miR­214­5p­CDK6/CCND1/MDM2/MDM4 network, which possibly provide a novel therapeutic target for IOT.


Subject(s)
Cell Proliferation , MicroRNAs/metabolism , Ovarian Neoplasms/metabolism , RNA, Long Noncoding/metabolism , Teratocarcinoma/metabolism , Adolescent , Adult , Cell Line, Tumor , Child , Female , Humans , MicroRNAs/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , RNA, Long Noncoding/genetics , Teratocarcinoma/genetics , Teratocarcinoma/pathology
9.
Cells ; 8(12)2019 11 23.
Article in English | MEDLINE | ID: mdl-31771212

ABSTRACT

The transforming growth factor-ß (TGFß) family factors induce pleiotropic effects and are involved in the regulation of most normal and pathological cellular processes. The activity of different branches of the TGFß family signaling pathways and their interplay with other signaling pathways govern the fine regulation of the self-renewal, differentiation onset and specialization of pluripotent stem cells in various cell derivatives. TGFß family signaling pathways play a pivotal role in balancing basic cellular processes in pluripotent stem cells and their derivatives, although disturbances in their genome integrity induce the rearrangements of signaling pathways and lead to functional impairments and malignant transformation into cancer stem cells. Therefore, the identification of critical nodes and targets in the regulatory cascades of TGFß family factors and other signaling pathways, and analysis of the rearrangements of the signal regulatory network during stem cell state transitions and interconversions, are key issues for understanding the fundamental mechanisms of both stem cell biology and cancer initiation and progression, as well as for clinical applications. This review summarizes recent advances in our understanding of TGFß family functions in naїve and primed pluripotent stem cells and discusses how these pathways are involved in perturbations in the signaling network of malignant teratocarcinoma stem cells with impaired differentiation potential.


Subject(s)
Cell Differentiation , Cell Self Renewal , Neoplastic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Signal Transduction , Teratocarcinoma/metabolism , Testicular Neoplasms/metabolism , Transforming Growth Factor beta/metabolism , Animals , Humans , Male , Neoplastic Stem Cells/pathology , Teratocarcinoma/pathology , Testicular Neoplasms/pathology
10.
Cell Reprogram ; 21(4): 171-180, 2019 08.
Article in English | MEDLINE | ID: mdl-31298562

ABSTRACT

Transcription factors NANOG, OCT4, SOX2, and NESTIN are expressed in both human embryonic stem cells (hESCs) and cancer stem cells and they play a crucial role in maintaining characteristics of stemness such as self-renewal and pluripotency. This article evaluates the expression of variants of the main stem cell-specific transcription factors NANOG and OCT4 critically and accurately with specific primers designed for identifying the most important variants that maintain stemness. We have examined four variants of NANOG along with a processed pseudogene and seven variants of OCT4 in human teratocarcinoma cell lines (NTERA2D1, SuSa, GCT-27, and 833KE), hESCs, and ovarian cancer cells by reverse transcriptase-polymerase chain reaction. In addition, we have examined their expression in NTERA2D1 cells on differentiation with all-trans-retinoic-acid. We show that NANOG1 is expressed in all teratocarcinoma cells and can be distinguished from NANOGP8, which is an expressed pseudogene. NANOG2 was not expressed in any of the cell lines, including ESCs. OCT4A was expressed in all cells, whereas the variant OCT4B-variant 3 was expressed only in NTERA2D1 cells. On differentiation of NTERA2D1 with retinoic acid, only NANOGP8 and OCT4A were expressed. In ovarian cancer cells, only 3/6 expressed NANOG1 and OCT4A. All malignant cells from patients with ovarian cancer (N = 6) expressed NANOG1 and OCT4A. These results demonstrate the necessity to precisely evaluate the expression of stem cell transcription factors when defining stemness.


Subject(s)
Alternative Splicing , Human Embryonic Stem Cells/metabolism , Nanog Homeobox Protein/metabolism , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/metabolism , SOXB1 Transcription Factors/metabolism , Teratocarcinoma/metabolism , Cell Differentiation , Cells, Cultured , Female , Human Embryonic Stem Cells/cytology , Humans , Nanog Homeobox Protein/genetics , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Protein Isoforms , SOXB1 Transcription Factors/genetics , Teratocarcinoma/genetics , Teratocarcinoma/pathology
11.
Bioorg Med Chem ; 26(8): 1523-1537, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29439916

ABSTRACT

LSD1/KDM1 is a histone demethylase that preferentially removes methyl groups from the mono- and di-methylated lysine 4 in histone H3 (H3K4), key marks for active chromatin for transcriptional activation. LSD1 is essential for pluripotent embryonic stem cells and embryonic teratocarcinoma/carcinoma cells and its expression is often elevated in various cancers. We developed a new LSD1 inhibitor, CBB3001, which potently inhibited LSD1 activity both in vitro and in vivo. CBB3001 also selectively inhibited the growth of human ovarian teratocarcinoma PA-1 and mouse embryonic carcinoma F9 cells, caused the downregulation of pluripotent stem cell proteins SOX2 and OCT4. However, CBB3001 does not have significant inhibition on the growth of human colorectal carcinoma HCT116 cells or mouse fibroblast NIH3T3 cells that do not express these stem cell proteins. Our studies strongly indicate that CBB3001 is a specific LSD1 inhibitor that selectively inhibits teratocarcinoma and embryonic carcinoma cells that express SOX2 and OCT4.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Embryonal/drug therapy , Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Teratocarcinoma/drug therapy , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Carcinoma, Embryonal/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , HCT116 Cells , Histone Demethylases/metabolism , Humans , Mice , Molecular Structure , NIH 3T3 Cells , Structure-Activity Relationship , Teratocarcinoma/metabolism
12.
J Biol Chem ; 293(10): 3663-3674, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29358331

ABSTRACT

The pluripotency-controlling stem-cell protein SRY-box 2 (SOX2) plays a pivotal role in maintaining the self-renewal and pluripotency of embryonic stem cells and also of teratocarcinoma or embryonic carcinoma cells. SOX2 is monomethylated at lysine 119 (Lys-119) in mouse embryonic stem cells by the SET7 methyltransferase, and this methylation triggers ubiquitin-dependent SOX2 proteolysis. However, the molecular regulators and mechanisms controlling SET7-induced SOX2 proteolysis are unknown. Here, we report that in human ovarian teratocarcinoma PA-1 cells, methylation-dependent SOX2 proteolysis is dynamically regulated by the LSD1 lysine demethylase and a methyl-binding protein, PHD finger protein 20-like 1 (PHF20L1). We found that LSD1 not only removes the methyl group from monomethylated Lys-117 (equivalent to Lys-119 in mouse SOX2), but it also demethylates monomethylated Lys-42 in SOX2, a reaction that SET7 also regulated and that also triggered SOX2 proteolysis. Our studies further revealed that PHF20L1 binds both monomethylated Lys-42 and Lys-117 in SOX2 and thereby prevents SOX2 proteolysis. Down-regulation of either LSD1 or PHF20L1 promoted SOX2 proteolysis, which was prevented by SET7 inactivation in both PA-1 and mouse embryonic stem cells. Our studies also disclosed that LSD1 and PHF20L1 normally regulate the growth of pluripotent mouse embryonic stem cells and PA-1 cells by preventing methylation-dependent SOX2 proteolysis. In conclusion, our findings reveal an important mechanism by which the stability of the pluripotency-controlling stem-cell protein SOX2 is dynamically regulated by the activities of SET7, LSD1, and PHF20L1 in pluripotent stem cells.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Histone Demethylases/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Neoplasm Proteins/metabolism , Ovarian Neoplasms/metabolism , Protein Processing, Post-Translational , SOXB1 Transcription Factors/metabolism , Amino Acid Substitution , Animals , Cell Line, Tumor , Cells, Cultured , Chromosomal Proteins, Non-Histone/antagonists & inhibitors , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , HEK293 Cells , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/chemistry , Histone Demethylases/genetics , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/genetics , Humans , Methylation , Mice, Inbred C57BL , Mutation , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Protein Stability , Proteolysis , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , SOXB1 Transcription Factors/chemistry , SOXB1 Transcription Factors/genetics , Teratocarcinoma/enzymology , Teratocarcinoma/metabolism , Teratocarcinoma/pathology
13.
Oncol Rep ; 39(2): 851-859, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29207191

ABSTRACT

The pituitary sex hormones (SexHs): follicle­stimulating hormone (FSH), luteinizing hormone (LH), and prolactin (PRL) regulate several functions crucial for reproduction, including oogenesis, spermatogenesis, and lactation. An important source of prolactin-like hormones, known as lactogens, is the placenta, and lactogens bind to the PRL receptor (PRLR) with high affinity and thereby mimic the actions of PRL. Recently, it has been demonstrated that pituitary SexHs were involved in metastatic lung cancer, certain sarcomas, and leukemia. In the present study we aimed to investigate whether FSH, LH, and PRL were able to stimulate stem cells involved in early development. To address this issue we employed a murine embryonic stem cell line (ES-D3) as well as two teratocarcinoma cell lines, P19 (murine) and NTera2 (human). We determined that all these cells expressed SexH receptors at the mRNA and protein levels and that stimulation of these receptors induced phosphorylation of p42/44 MAPK, p38 MAPK, and AKT. Moreover, ES-D3, P19, and NTera2 cells responded with increased migration and adhesion to physiological concentrations of pituitary SexHs. In view of these findings we proposed that maternal-derived pituitary SexHs regulate the biology of stem cells involved in early development.


Subject(s)
Embryonic Stem Cells/cytology , Gonadotropins, Pituitary/pharmacology , Receptors, Gonadotropin/metabolism , Teratocarcinoma/metabolism , Testicular Neoplasms/metabolism , Animals , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Embryonic Stem Cells/drug effects , Follicle Stimulating Hormone/pharmacology , Humans , Luteinizing Hormone/pharmacology , Male , Mice , Prolactin/pharmacology , Receptors, Gonadotropin/genetics , Signal Transduction/drug effects , Teratocarcinoma/genetics , Testicular Neoplasms/genetics
14.
Biomed Res Int ; 2017: 2413197, 2017.
Article in English | MEDLINE | ID: mdl-28680880

ABSTRACT

Gedunin is one of the major compounds found in the neem tree (Azadirachta indica). In the present study, antiproliferative potential of gedunin was evaluated in human embryonal carcinoma cells (NTERA-2, a cancer stem cell model) and peripheral blood mononuclear cells (PBMCs), using Sulforhodamine (SRB) and WST-1 assays, respectively. The effects of gedunin on expression of heat shock protein 90 (HSP90), its cochaperone Cdc37, and HSP client proteins (AKT, ErbB2, and HSF1) were evaluated by real-time PCR. Effects of gedunin on apoptosis were evaluated by (a) apoptosis associated morphological changes, (b) caspase 3/7 expression, (c) DNA fragmentation, (d) TUNEL assay, and (e) real-time PCR of apoptosis related genes (Bax, p53, and survivin). Gedunin showed a promising antiproliferative effect in NTERA-2 cells with IC50 values of 14.59, 8.49, and 6.55 µg/mL at 24, 48, and 72 h after incubations, respectively, while exerting a minimal effect on PBMCs. Expression of HSP90, its client proteins, and survivin was inhibited and Bax and p53 were upregulated by gedunin. Apoptosis related morphological changes, DNA fragmentation, and increased caspase 3/7 activities confirmed the proapoptotic effects of gedunin. Collectively, results indicate that gedunin may be a good drug lead for treatment of chemo and radiotherapy resistant cancer stem cells.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Embryonal Carcinoma Stem Cells/metabolism , Limonins/pharmacology , Teratocarcinoma/drug therapy , Teratocarcinoma/metabolism , DNA Fragmentation/drug effects , DNA, Neoplasm/metabolism , Drug Screening Assays, Antitumor , Embryonal Carcinoma Stem Cells/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Limonins/chemistry , Neoplasm Proteins/biosynthesis , Teratocarcinoma/pathology
15.
Biochem Cell Biol ; 95(2): 251-262, 2017 04.
Article in English | MEDLINE | ID: mdl-28177772

ABSTRACT

Mouse F9 cells differentiate into primitive endoderm (PrE) following the activation of the canonical WNT-ß-catenin pathway. The upregulation of Wnt6 and activation of ß-catenin-TCF-LEF-dependent transcription is known to accompany differentiation, but the Frizzled (FZD) receptor responsible for transducing the WNT6 signal is not known. Eight of the 10 Fzd genes were found to be expressed in F9 cells, with Fzd7 being the most highly expressed, and chosen for further analysis. To alter steady-state Fzd7 levels and test the effect this has on differentiation, siRNA and overexpression approaches were used to knock-down and ectopically express the Fzd7 message, respectively. siRNA knock-down of Fzd7 resulted in reduced DAB2 levels, and the overexpression activated a TCF-LEF reporter, but neither approach affected differentiation. Our focus turned to how canonical WNT6 signaling was attenuated to allow PrE cells to form parietal endoderm (PE). Dkk1, encoding a WNT antagonist, was examined and results showed that its expression increased in F9 cells treated with retinoic acid (RA) or overexpressing Wnt6. F9 cells overexpressing human DKK1 or treated with DKK1-conditioned medium and then treated with RA failed to differentiate, indicating that a negative feedback loop involving WNT6 and DKK1 attenuates canonical WNT-ß-catenin signaling, thereby allowing PE cells to differentiate.


Subject(s)
Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/genetics , Proto-Oncogene Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Teratocarcinoma/genetics , Wnt Proteins/genetics , beta Catenin/genetics , Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Apoptosis Regulatory Proteins , Cell Differentiation/drug effects , Cell Line, Tumor , Culture Media, Conditioned/pharmacology , Endoderm/metabolism , Endoderm/pathology , Feedback, Physiological , Frizzled Receptors , Genes, Reporter , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Mice , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Teratocarcinoma/metabolism , Teratocarcinoma/pathology , Tretinoin/pharmacology , Wnt Proteins/metabolism , beta Catenin/metabolism
16.
Cell Mol Neurobiol ; 37(3): 437-443, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27116544

ABSTRACT

Cathepsin E (CatE), an aspartic protease, has a limited distribution in certain cell types such as gastric cells. CatE is not detectable in the normal brain, whereas it is increasingly expressed in damaged neurons and activated microglia of the pathological brain. Neurons expressing high levels of CatE showed apparent morphological changes, including a marked shrinkage of the cytoplasmic region and beading of neurites, suggesting neuronal damage. The intracellular level of CatE in neurons is strictly regulated at both transcriptional and translational levels. Although the up-regulation of CatE may cause pathological changes in neurons, little information is available about the precise outcome of the increased expression of CatE in neurons. In this study, we have attempted to clarify the outcome of up-regulated CatE gene expression in neurons using the P19 cell neuronal differentiation after the overexpression of CatE. We unexpectedly found that the overexpression of CatE interfered with neuronal differentiation of P19 cells through an impairment of cell aggregate formation. Pepstatin A, an aspartic protease inhibitor, restored the impaired cell aggregation of P19/CatE cells. The small number of P19 cells differentiated into neurons had abnormal morphology characterized by their fusiform cell bodies with short processes. Furthermore, CatE proteolytically cleaved the extracellular domain of N-cadherin. These observations suggest that the overexpression of CatE interferes with neuronal differentiation of P19 cells through an impairment of cell aggregate formation, possibly through proteolytic degradation of N-cadherin.


Subject(s)
Cadherins/metabolism , Cathepsin E/metabolism , Cell Differentiation , Neurons/pathology , Proteolysis , Teratocarcinoma/pathology , Animals , Cell Aggregation/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , DNA, Complementary/genetics , Immunoblotting , Mice , Neurons/metabolism , Protease Inhibitors/pharmacology , Proteolysis/drug effects , Rats , Teratocarcinoma/metabolism , Transfection
17.
Proc Natl Acad Sci U S A ; 113(35): 9822-7, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27535933

ABSTRACT

TP53 (which encodes the p53 protein) is the most frequently mutated gene among all human cancers, whereas tumors that retain the wild-type TP53 gene often use alternative mechanisms to repress the p53 tumor-suppressive function. Testicular teratocarcinoma cells rarely contain mutations in TP53, yet the transcriptional activity of wild-type p53 is compromised, despite its high expression level. Here we report that in the teratocarcinoma cell line NTera2, p53 is subject to lysine methylation at its carboxyl terminus, which has been shown to repress p53's transcriptional activity. We show that reduction of the cognate methyltransferases reactivates p53 and promotes differentiation of the NTera2 cells. Furthermore, reconstitution of methylation-deficient p53 mutants into p53-depleted NTera2 cells results in elevated expression of p53 downstream targets and precocious loss of pluripotent gene expression compared with re-expression of wild-type p53. Our results provide evidence that lysine methylation of endogenous wild-type p53 represses its activity in cancer cells and suggest new therapeutic possibilities of targeting testicular teratocarcinoma.


Subject(s)
Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/genetics , Lysine/metabolism , Protein Processing, Post-Translational , Tumor Suppressor Protein p53/genetics , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/metabolism , Humans , Male , Methylation , Protein Domains , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Teratocarcinoma/genetics , Teratocarcinoma/metabolism , Teratocarcinoma/pathology , Testicular Neoplasms/genetics , Testicular Neoplasms/metabolism , Testicular Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism
18.
Stem Cells Transl Med ; 5(5): 694-702, 2016 May.
Article in English | MEDLINE | ID: mdl-26987352

ABSTRACT

UNLABELLED: Human induced pluripotent stem cells (iPSCs) and derived progeny provide invaluable regenerative platforms, yet their clinical translation has been compromised by their biosafety concern. Here, we assessed the safety of transplanting patient-derived iPSC-generated pancreatic endoderm/progenitor cells. Transplantation of progenitors from iPSCs reprogrammed by lentiviral vectors (LV-iPSCs) led to the formation of invasive teratocarcinoma-like tumors in more than 90% of immunodeficient mice. Moreover, removal of primary tumors from LV-iPSC progeny-transplanted hosts generated secondary and metastatic tumors. Combined transgene-free (TGF) reprogramming and elimination of residual pluripotent cells by enzymatic dissociation ensured tumor-free transplantation, ultimately enabling regeneration of type 1 diabetes-specific human islet structures in vivo. The incidence of tumor formation in TGF-iPSCs was titratable, depending on the oncogenic load, with reintegration of the cMYC expressing vector abolishing tumor-free transplantation. Thus, transgene-free cMYC-independent reprogramming and elimination of residual pluripotent cells are mandatory steps in achieving transplantation of iPSC progeny for customized and safe islet regeneration in vivo. SIGNIFICANCE: Pluripotent stem cell therapy for diabetes relies on the safety as well as the quality of derived insulin-producing cells. Data from this study highlight prominent tumorigenic risks of induced pluripotent stem cell (iPSC) products, especially when reprogrammed with integrating vectors. Two major underlying mechanisms in iPSC tumorigenicity are residual pluripotent cells and cMYC overload by vector integration. This study also demonstrated that combined transgene-free reprogramming and enzymatic dissociation allows teratoma-free transplantation of iPSC progeny in the mouse model in testing the tumorigenicity of iPSC products. Further safety assessment and improvement in iPSC specification into a mature ß cell phenotype would lead to safe islet replacement therapy for diabetes.


Subject(s)
Diabetes Mellitus, Type 1/surgery , Diabetes Mellitus, Type 2/surgery , Induced Pluripotent Stem Cells/transplantation , Islets of Langerhans Transplantation/methods , Islets of Langerhans/surgery , Keratinocytes/transplantation , Regeneration , Teratocarcinoma/prevention & control , Adult , Aged , Animals , Cell Differentiation , Cells, Cultured , Cellular Reprogramming Techniques , Diabetes Mellitus, Type 1/diagnosis , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Gene Expression Regulation, Neoplastic , Genetic Vectors , Heterografts , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Insulin/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Islets of Langerhans Transplantation/adverse effects , Keratinocytes/metabolism , Keratinocytes/pathology , Lentivirus/genetics , Male , Mice, SCID , Phenotype , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Teratocarcinoma/genetics , Teratocarcinoma/metabolism , Teratocarcinoma/pathology , Transfection
19.
Nucleic Acids Res ; 44(10): 4665-83, 2016 06 02.
Article in English | MEDLINE | ID: mdl-26883630

ABSTRACT

Cell differentiation is a central process in development and in cancer growth and dissemination. OCT4 (POU5F1) and NANOG are essential for cell stemness and pluripotency; yet, the mechanisms that regulate their expression remain largely unknown. Repetitive elements account for almost half of the Human Genome; still, their role in gene regulation is poorly understood. Here, we show that the dioxin receptor (AHR) leads to differentiation of human carcinoma cells through the transcriptional upregulation of Alu retrotransposons, whose RNA transcripts can repress pluripotency genes. Despite the genome-wide presence of Alu elements, we provide evidences that those located at the NANOG and OCT4 promoters bind AHR, are transcribed by RNA polymerase-III and repress NANOG and OCT4 in differentiated cells. OCT4 and NANOG repression likely involves processing of Alu-derived transcripts through the miRNA machinery involving the Microprocessor and RISC. Consistently, stable AHR knockdown led to basal undifferentiation, impaired Alus transcription and blockade of OCT4 and NANOG repression. We suggest that transcripts produced from AHR-regulated Alu retrotransposons may control the expression of stemness genes OCT4 and NANOG during differentiation of carcinoma cells. The control of discrete Alu elements by specific transcription factors may have a dynamic role in genome regulation under physiological and diseased conditions.


Subject(s)
Alu Elements , Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Differentiation/genetics , Gene Expression Regulation, Neoplastic , Receptors, Aryl Hydrocarbon/physiology , Teratocarcinoma/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Nucleus/metabolism , Humans , Mice , MicroRNAs/metabolism , Nanog Homeobox Protein/genetics , Octamer Transcription Factor-3/genetics , Promoter Regions, Genetic , RNA Polymerase III/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Teratocarcinoma/enzymology , Teratocarcinoma/metabolism , Teratocarcinoma/pathology , Teratoma/genetics , Teratoma/metabolism , Transcription, Genetic , Tretinoin/pharmacology
20.
An Acad Bras Cienc ; 87(1): 389-404, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25761220

ABSTRACT

The altered expression of the SOX2 transcription factor is associated with oncogenic or tumor suppressor functions in human cancers. This factor regulates the migration and invasion of different cancer cells. In this study we investigated the effect of constitutive SOX2 overexpression on the migration and adhesion capacity of embryonal teratocarcinoma NT2/D1 cells derived from a metastasis of a human testicular germ cell tumor. We detected that increased SOX2 expression changed the speed, mode and path of cell migration, but not the adhesion ability of NT2/D1 cells. Additionally, we demonstrated that SOX2 overexpression increased the expression of the tumor suppressor protein p53 and the HDM2 oncogene. Our results contribute to the better understanding of the effect of SOX2 on the behavior of tumor cells originating from a human testicular germ cell tumor. Considering that NT2/D1 cells resemble cancer stem cells in many features, our results could contribute to the elucidation of the role of SOX2 in cancer stem cells behavior and the process of metastasis.


Subject(s)
Cell Movement/physiology , SOXB1 Transcription Factors/metabolism , Teratocarcinoma/metabolism , Cell Adhesion/physiology , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Male , Teratocarcinoma/pathology , Tissue Array Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...