Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 282
Filter
1.
Med Mol Morphol ; 57(1): 68-75, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37991604

ABSTRACT

Serpinb9 is an inhibitor of granzyme B and is potentially involved in the immune escape of tumor cells. In the present study, bioinformatics analysis using open databases suggested that SerpinB9 is overexpressed in testicular embryonal carcinoma. Immunohistological analysis was performed on 28 cases of testicular germ cell tumors to investigate the relationship between SerpinB9 expression in testicular germ cell tumors and the tumor immune environment. SerpinB9 was significantly upregulated in the non-seminoma group and inversely correlated with the number of tumor-infiltrating CD8-positive cells. In addition, yolk sac tumors were characterized by the loss of human leukocyte antigen-class I expression. These findings suggest that SerpinB9 contributes to the immune escape of testicular germ cell tumors. Targeting therapy for SerpinB9 might therefore be useful in immunotherapy for testicular germ cell tumors resistant to immune checkpoint inhibitors.


Subject(s)
Carcinoma, Embryonal , Neoplasms, Germ Cell and Embryonal , Testicular Neoplasms , Humans , Male , Biomarkers, Tumor/genetics , Biomarkers, Tumor/analysis , Carcinoma, Embryonal/metabolism , Carcinoma, Embryonal/pathology , Neoplasms, Germ Cell and Embryonal/genetics , Testicular Neoplasms/genetics , Testicular Neoplasms/metabolism
2.
Semin Diagn Pathol ; 40(1): 2-21, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35840444

ABSTRACT

Germ cell neoplasia in situ (GCNIS) is the precursor of both seminomatous and non-seminomatous germ cell tumors. It consists of distended tubules that may have either intratubular seminoma or intratubular embryonal carcinoma cells. Many invasive non-seminomatous tumors contain a mixture of tumor types, which are reviewed here. Morphology, aided by a panel of immunostains, can determine the presence and percent of embryonal carcinoma, yolk sac tumor, choriocarcinoma, or teratoma in such tumors. Use of immunostains, required for diagnosis in perhaps 25% of testicular neoplasms, is reviewed. Changes of classification in the AJCC (8th edition) in 2016 are discussed, including the partitioning of two tumor types: the central role of chromosome 12p amplification allows both teratoma and yolk sac tumor to be divided into prepubertal types (lacking amplification) and post-pubertal types. Occasionally, sex cord-stromal tumors, hematolymphoid tumors, or epididymal adenomatoid tumors enter the differential diagnosis of germ cell neoplasms.


Subject(s)
Carcinoma, Embryonal , Endodermal Sinus Tumor , Neoplasms, Germ Cell and Embryonal , Seminoma , Teratoma , Testicular Neoplasms , Male , Humans , Endodermal Sinus Tumor/pathology , Testicular Neoplasms/genetics , Testicular Neoplasms/diagnosis , Testicular Neoplasms/pathology , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/metabolism , Carcinoma, Embryonal/pathology , Teratoma/diagnosis , Teratoma/genetics , Teratoma/pathology , Seminoma/diagnosis , Seminoma/genetics , Seminoma/pathology
3.
Pathol Res Pract ; 237: 154044, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35930824

ABSTRACT

BACKGROUND: In recent years, several studies investigated the complex process called "reprogramming" of seminoma (S) cells. The accepted pathogenetic model is a complex network including SOX2, SOX17, OCT3/4 and PRAME, which modulates the epigenetic transcription of numerous downstream genes and drives a divergent gene expression profile resulting in the transition from pure S (P-S) to S component (S-C) of mixed germ cell tumors of the testis (M-GCTT), and finally to embryonal carcinoma (EC). Herein, we tested a large cohort of GCTT with SOX2 and PRAME to evaluate their expression in the evolutionary steps of GCTT and verify if the modulation in the expression of these two molecules could be relevant for the fate of GCTT. METHODS: We tested 43, 19 and 17 consecutive and retrospectively enrolled cases of GCTT, germ cell neoplasia in situ (GCNIS) and uninvolved background testes (UBT), respectively. SOX2 and PRAME expressions have been evaluated with H-score and compared by adopting the appropriate statistic tests (Student's t-test and Mann-Whitney U test). RESULTS: We found that SOX2 was more expressed by nonseminomatous-GCTT (NS-GCTT) (p < 0.001) and EC (p < 0.001) rather than S; by contrast, PRAME showed an opposite expression profile being expressed by S but not by NS-GCTT (p < 0.001) and EC (p < 0.001). S-C showed different expressions of SOX2 and PRAME compared to both P-S (p = 0.002 and <0.001, respectively) and EC (p < 0.001 and 0.042, respectively), with intermediate values between these latter two categories. GCNIS and UBT showed no expression of SOX2 (scattered positive Leydig cells) but high H-score levels of PRAME. CONCLUSIONS: SOX2 and PRAME are differentially expressed and specularly modulated during the "reprogramming" of S cells [P-S (high levels of PRAME, no expression/low levels of SOX2) → S-C (intermediate levels of PRAME, intermediate levels of SOX2) → EC (no expression/low levels of PRAME, high levels of SOX2)], therefore supporting a complex pathogenetic model where the interactions between these two molecules are crucial in determining the fate of GCTT.


Subject(s)
Carcinoma, Embryonal , Neoplasms, Germ Cell and Embryonal , Seminoma , Testicular Neoplasms , Male , Humans , Seminoma/genetics , Seminoma/metabolism , Seminoma/pathology , Testicular Neoplasms/pathology , Retrospective Studies , SOXB1 Transcription Factors/metabolism , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/metabolism , Carcinoma, Embryonal/pathology , Antigens, Neoplasm/metabolism
4.
Methods Mol Biol ; 2454: 625-641, 2022.
Article in English | MEDLINE | ID: mdl-32833131

ABSTRACT

Induced pluripotent stem cells (iPSCs) are useful for the development of therapies in regenerative medicine, analysis of pathogenesis, and exploration of candidate drugs. We developed an alternative usage of iPSCs of which the MHC haplotype is matched to transplantable recipients in a cynomolgus macaque model. We established two cancer cell lines, embryonal carcinoma, and glioblastoma cell lines from cynomolgus monkey iPSCs. Here, we describe a method to induce the cancer cell lines including a technique for culture of the monkey iPSCs on feeder cells and the induction of hematopoietic stem cells and neural progenitor cells from monkey iPSCs.


Subject(s)
Carcinoma, Embryonal , Glioblastoma , Induced Pluripotent Stem Cells , Animals , Carcinoma, Embryonal/metabolism , Cell Differentiation , Cell Line , Cells, Cultured , Glioblastoma/metabolism , Macaca fascicularis
5.
J Cancer Res Clin Oncol ; 148(3): 609-631, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34837545

ABSTRACT

INTRODUCTION: Melanoma-associated antigen A2 (MAGE-A2) is a member of the cancer-testis antigen family differentially overexpressed in a variety of malignancies and is associated with tumor development. However, clinical significance and prognostic value of MAGE-A2 in different histological subtypes of testicular germ cell tumors (TGCTs) have not been explored. MATERIALS AND METHODS: Here, we aimed to investigate the clinical significance and prognostic impact of MAGE-A2 expression in TGCTs compared to benign tumors as well as adjacent normal tissues and then between seminomas and non-seminomas groups using immunohistochemistry on tissue microarrays. RESULTS: The results indicated a statistically significant difference between overexpression of MAGE-A2 and histological subtypes of TGCTs. A statistically significant association was found between a high level of nuclear expression of MAGE-A2 protein and advanced pT stage (P = 0.022), vascular invasion (P = 0.037), as well as involvement of rete testis (P = 0.022) in embryonal carcinomas. Increased nuclear expression of MAGE-A2 was observed to be associated with more aggressive behaviors and tumor progression rather than cytoplasmic expression in these cases. Further, high level nuclear expression of MAGE-A2 had shorter disease-specific survival (DSS) or progression-free survival (PFS) compared to patients with moderate and low expression of MAGE-A2, however, without a statistically significant association. CONCLUSION: Our results confirm that increased nuclear expression of MAGE-A2 has a clinical significance in embryonal carcinomas and is associated with progression of disease. Moreover, MAGE-A2 may act as a potential predictive biomarker for the prognosis in embryonal carcinomas if follow-up period becomes longer. Further investigations for the biological function of MAGE-A2 are required in future studies.


Subject(s)
Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Embryonal/pathology , Neoplasms, Germ Cell and Embryonal/pathology , Seminoma/pathology , Testicular Neoplasms/pathology , Adolescent , Adult , Aged , Carcinoma, Embryonal/metabolism , Carcinoma, Embryonal/surgery , Case-Control Studies , Child , Child, Preschool , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Invasiveness , Neoplasms, Germ Cell and Embryonal/metabolism , Neoplasms, Germ Cell and Embryonal/surgery , Prognosis , Retrospective Studies , Seminoma/metabolism , Seminoma/surgery , Survival Rate , Testicular Neoplasms/metabolism , Testicular Neoplasms/surgery , Young Adult
6.
Front Immunol ; 13: 1096494, 2022.
Article in English | MEDLINE | ID: mdl-36713456

ABSTRACT

Background: Testicular germ cell tumor (TGCT) is the most common tumor in young men, but molecular signatures, especially the alternative splicing (AS) between its subtypes have not yet been explored. Methods: To investigate the differences between TGCT subtypes, we comprehensively analyzed the data of gene expression, alternative splicing (AS), and somatic mutation in TGCT patients from the TCGA database. The gene ontology (GO) enrichment analyses were used to explore the function of differentially expressed genes and spliced genes respectively, and Spearman correlation analysis was performed to explore the correlation between differential genes and AS events. In addition, the possible patterns in which AS regulates gene expression were elaborated by the ensemble database transcript atlas. And, we identified important transcription factors that regulate gene expression and AS and functionally validated them in TGCT cell lines. Results: We found significant differences between expression and AS in embryonal carcinoma and seminoma, while mixed cell tumors were in between. GO enrichment analyses revealed that both differentially expressed and spliced genes were enriched in transcriptional regulatory pathways, and obvious correlation between expression and AS events was determined. By analyzing the transcript map and the sites where splicing occurs, we have demonstrated that AS regulates gene expression in a variety of ways. We further identified two pivot AS-related molecules (SOX2 and HDAC9) involved in AS regulation, which were validated in embryonal carcinoma and seminoma cell lines. Differences in somatic mutations between subtypes are also of concern, with our results suggesting that mutations in some genes (B3GNT8, CAPN7, FAT4, GRK1, TACC2, and TRAM1L1) occur only in embryonal carcinoma, while mutations in KIT, KARS, and NRAS are observed only in seminoma. Conclusions: In conclusion, our analysis revealed the differences in gene expression, AS and somatic mutation among TGCT subtypes, providing a molecular basis for clinical diagnosis and precise therapy of TGCT patients.


Subject(s)
Carcinoma, Embryonal , Seminoma , Testicular Neoplasms , Male , Humans , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/metabolism , Carcinoma, Embryonal/pathology , Alternative Splicing , Seminoma/genetics , Seminoma/metabolism , Seminoma/pathology , DNA, Recombinant , Testicular Neoplasms/diagnosis , Mutation , Gene Expression
7.
Anticancer Res ; 41(9): 4211-4214, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34475040

ABSTRACT

BACKGROUND: Testicular cancer constitutes 1.0% of male cancer and typically carries a good prognosis. As far as we are aware, the role for hydrogen sulfide in testicular cancer and the level of hydrogen sulfide-synthesizing enzyme have never been addressed. Here we examined cystathionine gamma-lyase (CSE) expression in several germ-cell testicular tumors. MATERIALS AND METHODS: Tissue microarrays were employed to examine CSE expression in 32 benign testicular samples, 88 testicular seminomas, 34 embryonal carcinomas, 4 mature teratomas, and 16 yolk sac tumors, and CSE expression was compared to that seen in benign testicular tissue. RESULTS: Compared to benign testicular tissue, CSE expression was increased in all three types of testicular neoplasm but not in mature teratomas. Highest CSE expression was identified in embryonal carcinomas, which often show a relatively aggressive clinical course. CONCLUSION: For the first time, we show that CSE is increased in several common testicular germ-cell tumor types.


Subject(s)
Carcinoma, Embryonal/metabolism , Cystathionine gamma-Lyase/metabolism , Endodermal Sinus Tumor/metabolism , Testicular Neoplasms/metabolism , Up-Regulation , Case-Control Studies , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Male , Seminoma , Tissue Array Analysis
8.
Biochimie ; 186: 33-42, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33865902

ABSTRACT

The epithelium-specific ETS transcription factor-1 (ESE-1) plays multiple roles in pathogenesis and normal development of epithelial tissues. NANOG, a key mediator of stem cell self-renewal and pluripotency, is also expressed in various cancers and pluripotent cells. In this study, we investigated how ESE-1 influences NANOG expression and NANOG-induced proliferation in human germ cell-derived embryonic carcinoma NCCIT cells. Endogenous ESE-1 expression in NCCIT cells significantly increased during differentiation, whereas NANOG expression decreased. In addition, NANOG expression was downregulated by exogenous overexpression of ESE-1, and increased by shRNA-mediated knockdown of ESE-1. NANOG transcriptional activity was reduced by dose-dependent ESE-1 overexpression and a putative ESE-1 binding site (EBS) was mapped within conserved region 2. Site-directed mutagenesis of the putative EBS abrogated the repressive effect of ESE-1 on NANOG promoter activity. ESE-1 directly interacted with the putative EBS to regulate transcriptional activity of NANOG. Furthermore, NANOG-induced proliferation and colony formation of NCCIT cells were inhibited by ESE-1 overexpression and stimulated by ESE-1 shRNA-mediated knockdown. Altogether, our results suggest that ESE-1 exerts an anti-proliferative effect on NCCIT cells by acting as a novel transcriptional repressor of NANOG.


Subject(s)
Carcinoma, Embryonal/metabolism , Cell Proliferation , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Nanog Homeobox Protein/metabolism , Proto-Oncogene Proteins c-ets/metabolism , Transcription Factors/metabolism , Carcinoma, Embryonal/genetics , Cell Line, Tumor , DNA-Binding Proteins/genetics , Humans , Nanog Homeobox Protein/genetics , Proto-Oncogene Proteins c-ets/genetics , Transcription Factors/genetics
9.
Int J Mol Sci ; 21(22)2020 Nov 17.
Article in English | MEDLINE | ID: mdl-33212946

ABSTRACT

Overactivation of the c-MET/HGF system is a feature of many cancers. We previously reported that type II testicular germ cell tumor (TGCT) cells express the c-MET receptor, forming non-seminomatous lesions that are more positive compared with seminomatous ones. Notably, we also demonstrated that NT2D1 non-seminomatous cells (derived from an embryonal carcinoma lesion) increase their proliferation, migration, and invasion in response to HGF. Herein, we report that HGF immunoreactivity is more evident in the microenvironment of embryonal carcinoma biopsies with respect to seminomatous ones, indicating a tumor-dependent modulation of the testicular niche. PI3K/AKT is one of the signaling pathways triggered by HGF through the c-MET activation cascade. Herein, we demonstrated that phospho-AKT increases in NT2D1 cells after HGF stimulation. Moreover, we found that this pathway is involved in HGF-dependent NT2D1 cell proliferation, migration, and invasion, since the co-administration of the PI3K inhibitor LY294002 together with HGF abrogates these responses. Notably, the inhibition of endogenous PI3K affects collective cell migration but does not influence proliferation or chemotactic activity. Surprisingly, LY294002 administered without the co-administration of HGF increases cell invasion at levels comparable to the HGF-administered samples. This paradoxical result highlights the role of the testicular microenvironment in the modulation of cellular responses and stimulates the study of the testicular secretome in cancer lesions.


Subject(s)
Carcinoma, Embryonal/metabolism , Hepatocyte Growth Factor/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Testicular Neoplasms/metabolism , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/pathology , Cell Line, Tumor , Hepatocyte Growth Factor/genetics , Humans , Male , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Testicular Neoplasms/genetics
10.
Molecules ; 25(14)2020 Jul 08.
Article in English | MEDLINE | ID: mdl-32650569

ABSTRACT

The pharmacological effects of BST204-a fermented ginseng extract-on several types of cancers have been reported. However, the effects of ginseng products or single ginsenosides against cancer stem cells are still poorly understood. In this study, we identified the anti-tumorigenic and anti-invasive activities of BST204 through the suppression of the cancer stem cell marker, CD133. The treatment of embryonic carcinoma cells with BST204 induced the expression of the tumor suppressor protein, p53, which decreased the expression of cell cycle regulatory proteins and downregulated the expression of CD133 and several stemness transcription factors. These changes resulted in both the inhibition of tumor cell proliferation and tumorigenesis. The knockdown of CD133 suggests that it has a role in tumorigenesis, but not in cancer cell proliferation or cell cycle arrest. Treatment with BST204 resulted in the reduced expression of the mesenchymal marker, N-cadherin, and the increased expression of the epithelial marker, E-cadherin, leading to the suppression of tumor cell migration and invasion. The knockdown of CD133 also exhibited an anti-invasive effect, indicating the role of CD133 in tumor invasion. The single ginsenosides Rg3 and Rh2-major components of BST204-exhibited limited effects against cancer stem cells compared to BST204, suggesting possible synergism among several ginsenoside compounds.


Subject(s)
Carcinogenesis , Carcinoma, Embryonal , Cell Movement/drug effects , Neoplastic Stem Cells , Plant Extracts/pharmacology , AC133 Antigen/biosynthesis , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Embryonal/drug therapy , Carcinoma, Embryonal/metabolism , Carcinoma, Embryonal/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Suppressor Protein p53/biosynthesis
11.
Mod Pathol ; 33(11): 2354-2360, 2020 11.
Article in English | MEDLINE | ID: mdl-32561848

ABSTRACT

Hepatocyte Nuclear Factor 1 beta (HNF1ß) is a transcription factor which plays an important role during early organogenesis, especially of the pancreato-biliary and urogenital tract. Furthermore, HNF1ß is an established marker in the differential diagnosis of ovarian cancer and shows a distinct nuclear expression in the clear cell carcinoma subtype. Recently, it has been described in yolk sac tumor, which represents a common component in many non-seminomatous germ cell tumors. Due to its broad histologic diversity, the diagnosis may be challenging and additional tools are very helpful in the workup of germ cell tumors. Immunohistochemistry was used to study HNF1ß expression in a tissue microarray (TMA) of 601 testicular germ cell tumors including seminoma, embryonal carcinoma, yolk sac tumor, choriocarcinoma, teratoma, germ cell neoplasia in situ (GCNIS), and normal tissue. The expression pattern was compared to glypican 3 (GPC3) and α-fetoprotein (AFP), two markers currently in use for the detection of yolk sac tumor. HNF1ß showed a distinct nuclear staining in comparison to the cytoplasmic pattern of GPC3 and AFP. The sensitivity and specificity of HNF1ß were 85.4% and 96.5%, of GPC3 83.3% and 90.7%, of AFP 62.5% and 97.7%. We conclude that HNF1ß allows a reliable distinction of yolk sac tumor from other germ cell tumor components. Therefore, we propose HNF1ß as a novel and robust marker in the immunohistochemical workup of testicular germ cell tumors.


Subject(s)
Biomarkers, Tumor/metabolism , Endodermal Sinus Tumor/diagnosis , Hepatocyte Nuclear Factor 1-beta/metabolism , Testicular Neoplasms/diagnosis , Adult , Carcinoma, Embryonal/diagnosis , Carcinoma, Embryonal/metabolism , Carcinoma, Embryonal/pathology , Choriocarcinoma/diagnosis , Choriocarcinoma/metabolism , Choriocarcinoma/pathology , Diagnosis, Differential , Endodermal Sinus Tumor/metabolism , Endodermal Sinus Tumor/pathology , Humans , Immunohistochemistry , Male , Seminoma/diagnosis , Seminoma/metabolism , Seminoma/pathology , Sensitivity and Specificity , Teratoma/diagnosis , Teratoma/metabolism , Teratoma/pathology , Testicular Neoplasms/metabolism , Testicular Neoplasms/pathology , Testis/metabolism , Testis/pathology , Tissue Array Analysis
12.
J Cell Biochem ; 121(11): 4386-4396, 2020 11.
Article in English | MEDLINE | ID: mdl-31961017

ABSTRACT

The pluripotent mouse embryonal carcinoma cell line P19 is widely used as a model for research on all-trans-retinoid acid (RA)-induced neuronal differentiation; however, the signaling pathways involved in this process remain unclear. This study aimed to reveal the molecular mechanism underlying the RA-induced neuronal differentiation of P19 cells. Real-time quantitative polymerase chain reaction and Western blot analysis were used to determine the expression of neuronal-specific markers, whereas flow cytometry was used to analyze cell cycle and cell apoptosis. The expression profiles of messenger RNAs (mRNAs) in RA-induced neuronal differentiation of P19 cells were analyzed using high-throughput sequencing, and the functions of differentially expressed mRNAs (DEMs) were determined by bioinformatics analysis. RA induced an increase in both class III ß-tubulin (TUBB3) and neurofilament medium (NEFM) mRNA expression, indicating that RA successfully induces neuronal differentiation of P19 cells. Cell apoptosis was not affected; however, cell proliferation decreased. We found 4117 DEMs, which were enriched in the phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) signaling pathway, Wnt signaling pathway, and cell cycle. Particularly, a few DEMs could be identified in the PI3K/Akt signaling pathway networks, such as PI3K, Akt, glycogen synthase kinase-3ß (GSK3ß), cyclin-dependent kinase 4 (CDK4), P21, and Bax. RA significantly increased the protein expression of PI3K, Akt, phosphorylated Akt, GSK3ß, phosphorylated GSK3ß, CDK4, and P21, but it reduced Bax protein expression. The Akt inhibitor affected the increase of TUBB3 and NEFM mRNA expression in RA-induced P19 cells. The molecular mechanism underlying the RA-induced neuronal differentiation of P19 cells is potentially involved in the PI3K/Akt/GSK3ß signaling pathway. The decreased cell proliferation ability of neuronally differentiated P19 cells could be associated with the expression of cell cycle proteins.


Subject(s)
Carcinoma, Embryonal/pathology , Cell Differentiation , Glycogen Synthase Kinase 3 beta/metabolism , Neurons/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Tretinoin/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Embryonal/drug therapy , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/metabolism , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3 beta/genetics , Mice , Neurons/drug effects , Neurons/metabolism , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Tumor Cells, Cultured
13.
Cells ; 9(1)2020 01 10.
Article in English | MEDLINE | ID: mdl-31936807

ABSTRACT

B3GALT5 is involved in the synthesis of embryonic stem (ES) cell marker glycan, stage-specific embryonic antigen-3 (SSEA3). This gene has three native promoters and an integrated retroviral long terminal repeat (LTR) promoter. We found that B3GALT5-LTR is expressed at high levels in human ES cells. B3GALT5-LTR is also involved in the synthesis of the cancer-associated glycan, sialyl Lewis a. Sialyl Lewis a is expressed in ES cells and its expression decreases upon differentiation. Retinoic acid induced differentiation of ES cells, decreased the short form of NFYA (NFYAs), increased phosphorylation of STAT3, and decreased B3GALT5-LTR expression. NFYAs activated, and constitutively-active STAT3 (STAT3C) repressed B3GALT5-LTR promoter. The NFYAs and STAT3C effects were eliminated when their binding sites were deleted. Retinoic acid decreased the binding of NFYA to B3GALT5-LTR promoter and increased phospho-STAT3 binding. Lamin A repressed NFYAs and SSEA3 expression. SSEA3 repression mediated by a SIRT1 inhibitor was reversed by a STAT3 inhibitor. Repression of SSEA3 and sialyl Lewis a synthesis mediated by retinoic acid was partially reversed by lamin A short interfering RNA (siRNA) and a STAT3 inhibitor. In conclusion, B3GALT5-LTR is regulated by lamin A-NFYA and SIRT1-STAT3 signaling that regulates SSEA3 and sialyl Lewis a synthesis in ES cells, and sialyl Lewis a is also a ES cell marker.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate/metabolism , CA-19-9 Antigen/metabolism , CCAAT-Binding Factor/metabolism , Galactosyltransferases/genetics , Human Embryonic Stem Cells/metabolism , Lamin Type A/metabolism , STAT3 Transcription Factor/metabolism , Sirtuin 1/metabolism , Stage-Specific Embryonic Antigens/metabolism , Animals , Base Sequence , Carcinoma, Embryonal/metabolism , Cell Line , Humans , Mice , Polysaccharides/metabolism , Promoter Regions, Genetic/genetics , Signal Transduction , Terminal Repeat Sequences/genetics , Tretinoin/pharmacology
14.
Med Oncol ; 36(7): 61, 2019 May 29.
Article in English | MEDLINE | ID: mdl-31140031

ABSTRACT

Testicular germ cell tumors are the most frequent malignancies found in men between 15 and 44 years old. Although cellular apoptosis susceptibility (CAS) was demonstrated to be upregulated in breast cancer and colon cancer, the expression of CAS in the human testis and testicular germ cell tumors remained elusive. In the present study, CAS-positive signals were detected in the normal testicular tissues, cancer adjacent normal testicular tissues, seminoma, yolk sac tumor, and teratoma. Interestingly, the expression level of CAS in testicular germ cell tumors (TGCTs) (but not seminoma) was significantly lower than that of human testicular tissues and cancer adjacent normal testicular tissues, suggesting that decreased CAS contributed to the progression of TGCTs. Notably, the expression of CAS in seminoma was significantly higher than that of in the non-seminomas, consistent with the results from TCGA database. Furthermore, the localization of CAS is mainly restricted in the nucleus in the lesions of normal human testicular tissue and cancer adjacent normal testicular tissue. Although the expression of CAS was not significantly different between normal testicular tissue and seminoma, CAS was more enriched in cytoplasm in seminoma compared to the normal, cancer adjacent tissue and other types of TGCTs. The current results demonstrated reduced expression of CAS in the human testicular germ cell tumors and the CAS translocation from the nuclear to cytoplasm in seminoma, thereby supporting a possible role in normal testis function and in the development of seminoma.


Subject(s)
Cellular Apoptosis Susceptibility Protein/biosynthesis , Neoplasms, Germ Cell and Embryonal/metabolism , Testicular Neoplasms/metabolism , Testis/metabolism , Carcinoma, Embryonal/metabolism , Cytoplasm/metabolism , Endodermal Sinus Tumor/metabolism , Humans , Immunohistochemistry , Male , Seminoma/metabolism , Teratoma/metabolism , Tissue Array Analysis
15.
J Proteome Res ; 18(4): 1819-1826, 2019 04 05.
Article in English | MEDLINE | ID: mdl-30835130

ABSTRACT

Seminoma and embryonal carcinoma (EC), two typical types of testicular germ cell tumors (TGCTs), present significant differences in growth behavior, expression characteristics, differentiation potential, clinical features, therapy, and prognosis. The purpose of this study was to compare the distinctive or preference metabolic pathways between seminoma and EC. The Cancer Genome Atlas revealed that many genes encoding metabolic enzymes could distinguish between seminoma and EC. Using well-characterized cell line models for seminoma (Tcam-2 cells) and EC (NT2 cells), we characterized their metabolite profiles using ultraperformance liquid chromatography coupled to Q-TOF mass spectrometry (UPLC/Q-TOF MS). In general, the integrated results from transcriptome and metabolite profiling revealed that seminoma and EC exhibited distinctive characteristics in the metabolisms of amino acids, glucose, fatty acids, sphingolipids, nucleotides, and drugs. Notably, an attenuation of citric acid cycle/mitochondrial oxidative phosphorylation and sphingolipid biosynthesis as well as an increase in arachidonic acid metabolism and (very) long-chain fatty acid abundance occurred in seminoma as compared with EC. Our study suggests histologic subtype-dependent metabolic reprogramming in TGCTs and will lead to a better understanding of the metabolic signatures and biology of TGCT subtypes.


Subject(s)
Carcinoma, Embryonal/metabolism , Metabolome/genetics , Neoplasms, Germ Cell and Embryonal/metabolism , Seminoma/metabolism , Testicular Neoplasms/metabolism , Transcriptome/genetics , Carcinoma, Embryonal/genetics , Cell Line, Tumor , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Mass Spectrometry , Neoplasms, Germ Cell and Embryonal/genetics , Seminoma/genetics , Testicular Neoplasms/genetics
16.
J Biochem ; 165(6): 471-477, 2019 Jun 01.
Article in English | MEDLINE | ID: mdl-30576481

ABSTRACT

The protein phosphatase PPM1D (Wip1) was originally identified as a p53 target product. Activation of PPM1D through various mechanism promotes the tumorigenic potential of various cancers by suppressing p53 and other DNA damage response proteins. New functions of PPM1D have recently been revealed in physiological processes such as cell differentiation. However, the regulatory mechanisms of signalling pathway to maintain stemness and induce cell differentiation are still unclear. Here we report that PPM1D modulates retinoic acid (RA) signalling. PPM1D knockdown resulted in decreased alkaline phosphatase activity of the human teratocarcinoma cell line NT2/D1. Inhibition of PPM1D-induced cell differentiation and decreased gene expression of the stem cell marker Oct-4 (POU5F1). RA-induced cell differentiation was promoted by reducing PPM1D activity. RA treatment elicited activation of the MEK-ERK pathway and induced rapid and transient activation of the extracellular signal-regulated kinase 1/2 (ERK-1/2). PPM1D dephosphorylated a phosphopeptide with the TEY motif in ERK-1/2 in vitro. Moreover, phosphorylation of ERK-1/2 was facilitated by PPM1D inhibition. Our study shows that PPM1D plays an important role in maintaining the undifferentiation state and a new function in RA-induced ERK regulation and cell differentiation.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Embryonal/drug therapy , Cell Differentiation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Phosphatase 2C/antagonists & inhibitors , Tretinoin/pharmacology , Carcinoma, Embryonal/metabolism , Carcinoma, Embryonal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Protein Phosphatase 2C/metabolism , Structure-Activity Relationship
17.
Cell Death Dis ; 9(10): 990, 2018 09 24.
Article in English | MEDLINE | ID: mdl-30250195

ABSTRACT

Cancer cells with a less differentiated stem-like phenotype are more resistant to therapeutic manipulations than their differentiated counterparts, and are considered as one of the main causes of cancer persistence and relapse. As such, induction of differentiation in cancer stem-like cells (CSLCs) has emerged as an alternative strategy to enhance the efficacy of anticancer therapies. CSLCs are metabolically distinct from differentiated cells, and any aberration from the intrinsic metabolic state can induce differentiation of CSLCs. Therefore, metabolism-related molecular targets, with a capacity to promote differentiation within CSLCs, are of therapeutic importance. Here, we demonstrate that phosphoglycerate dehydrogenase (PHGDH), an essential enzyme catalyzing the synthesis of amino acid serine, is important for maintaining the poorly differentiated, stem-like state of CSLCs. Our data shows that PHGDH deficiency impairs the tumorsphere formation capacity in embryonal carcinoma stem-like cells (ECSLCs), breast cancer stem-like cells (BCSLCs) and patient-derived brain tumor-initiating cells (BTICs), which is accompanied by the reduced expression of characteristic stemness-promoting factors, such as Oct4, Nanog, Sox-2, and Bmi-1. Mechanistically, PHGDH deficiency in ECSLCs promotes differentiation to various lineages via degradation of Oct4 and by increasing the stability of differentiation marker ß3-tubulin. Furthermore, PHGDH inhibition promotes p-mTOR independent but Beclin-1-dependent autophagy, independent of apoptosis. When studied in combination, the inhibition of both PHGDH and p-mTOR in ECSLCs causes further augmentation of autophagy, and additionally promotes apoptosis, demonstrating the clinical applicability of PHGDH-based manipulations in cancer therapies. Recapitulating these in vitro findings in CSLC models, the intratumoral PHGDH expression in patient-derived tumors is positively correlated with the mRNA levels of stemness factors, especially Oct4, and cancer patients co-expressing high levels of PHGDH and Oct4 display significantly lower survival than those with low PHGDH/Oct4 co-expression. Altogether, this study identifies a clinically-relevant role for PHGDH in the regulation of stemness-differentiation axis within CSLCs.


Subject(s)
Autophagy , Brain Neoplasms/metabolism , Carcinoma, Embryonal/metabolism , Cell Differentiation , Embryonal Carcinoma Stem Cells/metabolism , Glioblastoma/metabolism , Phosphoglycerate Dehydrogenase/metabolism , TOR Serine-Threonine Kinases/metabolism , Testicular Neoplasms/metabolism , Beclin-1/metabolism , Brain Neoplasms/pathology , Carbohydrate Metabolism, Inborn Errors/metabolism , Carcinoma, Embryonal/pathology , Cell Line, Tumor , Glioblastoma/pathology , Humans , Male , Microcephaly/metabolism , Octamer Transcription Factor-3/metabolism , Phosphoglycerate Dehydrogenase/antagonists & inhibitors , Phosphoglycerate Dehydrogenase/deficiency , Phosphoglycerate Dehydrogenase/genetics , Proteolysis , Psychomotor Disorders/metabolism , Seizures/metabolism , Testicular Neoplasms/pathology , Transfection , Tubulin/metabolism , Ubiquitination
18.
Stem Cell Rev Rep ; 14(6): 823-836, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29980981

ABSTRACT

The paternally-imprinted genes insulin-like growth factor 2 (IGF2), H19, delta-like homologue 1 (DLK1), and maternally-expressed gene 3 (MEG3) are expressed from the tandem gene loci IGF2-H19 and DLK1-MEG3, which play crucial roles in initiating embryogenesis and development. The erasure of imprinting (EOI) at differentially methylated regions (DMRs) which regulate the expression of these genes maintains the developmental quiescence of primordial germ cells (PGCs) migrating through the embryo proper during embryogenesis and prevents them from forming teratomas. To address the potential involvement of the IGF2-H19 and DLK1-MEG3 loci in the pathogenesis of embryonal carcinoma (EC), we investigated their genomic imprinting at DMRs in the human PGC-derived EC cell line NTera-2 (NT2). We observed EOI at the IGF2-H19 locus and, somewhat to our surprise, a loss of imprinting (LOI) at the DLK1-MEG3 locus. As a result, NT2 cells express imprinted gene ratios from these loci such that there are i) low levels of the proliferation-promoting IGF2 relative to ii) high levels of the proliferation-inhibiting long noncoding RNA (lncRNA) H19 and iii) high levels of proliferation-promoting DLK1 relative to iv) low levels of the proliferation-inhibiting lncRNA MEG3. Consistent with this pattern of expression, the knockdown of DLK1 mRNA by shRNA resulted in decreased in vitro cell proliferation and in vivo tumor growth as well as decreased in vivo organ seeding by NT2 cells. Furthermore, treatment of NT2 cells with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine (5-azaD) inhibited their proliferation. This inhibition was accompanied by changes in expression of both tandem gene sets: a decrease in the expression of DLK1 and upregulation of the proliferation-inhibiting lncRNA MEG3, and at the same time upregulation of IGF2 and downregulation of the lncRNA H19. These results suggest that the DLK1-MEG3 locus, and not the IGF2-H19 locus, drives the tumorigenicity of NT2 cells. Based on these results, we identified DLK1 as a novel treatment target for EC that could be downregulated by 5-azaD.


Subject(s)
Carcinoma, Embryonal , Genetic Loci , Genomic Imprinting , Insulin-Like Growth Factor II , Intercellular Signaling Peptides and Proteins , Membrane Proteins , Neoplasm Proteins , RNA, Long Noncoding , RNA, Neoplasm , Calcium-Binding Proteins , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/metabolism , Carcinoma, Embryonal/pathology , Carcinoma, Embryonal/therapy , Cell Line, Tumor , Humans , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism
19.
Acta Biochim Biophys Sin (Shanghai) ; 50(8): 793-799, 2018 Aug 01.
Article in English | MEDLINE | ID: mdl-29945210

ABSTRACT

The ability to self-renew is one of the most important properties of embryonic stem (ES) cells. Pluripotin (SC1), a small molecule with high activity and low toxicity, promotes self-renewal in mouse ES cells. SC1 can noticeably change the morphology of retinoic acid (RA)-induced F9 embryonic carcinoma cells (F9 cells). However, in the long term, RA and SC1 together cause cell apoptosis. When being added after 18-24 h of RA-induced F9 cell differentiation, SC1 transitorily activated Nanog and Oct4. Both Nanog and Oct4 were downregulated when SC1 and RA were added simultaneously. On the other hand, Klf4 was continually activated when SC1 was added between 6 and 24 h. Phosphorylated Erk1/2 protein levels were reduced from 6 to 24 h, whereas unphosphorylated Erk1 protein levels remained unchanged. A higher concentration of SC1 promoted cell self-renewal by strengthening the inhibition of Erk1/2 protein phosphorylation in F9 cells. Furthermore, SC1 and RA affect global DNA methylation by influencing the expressions of methylation-associated proteins, including Dnmt3b, Dnmt3l, Tet1, Tet2, and Tet3. In conclusion, SC1 inhibits the differentiation of RA-induced F9 cells mainly by reducing the levels of phosphorylated Erk1/2 and enhancing the expression of Klf4, although it also reduces DNA methylation, which may have an additional effect on ES cell differentiation.


Subject(s)
Apoptosis/drug effects , Cell Differentiation/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Tretinoin/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/genetics , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/metabolism , Carcinoma, Embryonal/pathology , Cell Differentiation/genetics , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/drug effects , DNA Methylation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Kruppel-Like Factor 4 , Mice , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...