Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Cells ; 10(11)2021 10 22.
Article in English | MEDLINE | ID: mdl-34831070

ABSTRACT

Embryonic cancer stem cells (CSCs) can differentiate into any cancer type. Targeting CSC using natural compounds is a good approach as it suppresses cancer recurrence with fewer adverse effects, and methylsulfonylmethane (MSM) is a sulfur-containing compound with well-known anticancer activities. This study determined the mechanistic aspects of the anticancer activity of MSM. We used Western blotting and real-time qPCR for molecular signaling studies and conducted flow cytometry for analyzing the processes in cells. Our results suggested an inhibition in the expression of CSC markers and Wnt/ß-catenin signaling. MSM induced TRAIL-mediated extrinsic apoptosis in NCCIT and NTERA-2 cells rather than an intrinsic pathway. Inhibition of iron metabolism-dependent reactive oxygen species (ROS) generation takes part in TRAIL-mediated apoptosis induction by MSM. Suppressing iron metabolism by MSM also regulated p38/p53/ERK signaling and microRNA expressions, such as upregulating miR-130a and downregulating miR-221 and miR-222, which resulted in TRAIL induction and thereby extrinsic pathway of apoptosis. Hence, MSM could be a good candidate for neoadjuvant therapy by targeting CSCs by inhibiting iron metabolism.


Subject(s)
Apoptosis , Dimethyl Sulfoxide/pharmacology , Embryonal Carcinoma Stem Cells/pathology , Iron/metabolism , Sulfones/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Cell Cycle Checkpoints/drug effects , DNA Damage , Embryonal Carcinoma Stem Cells/drug effects , Embryonal Carcinoma Stem Cells/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Models, Biological , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , Wnt Signaling Pathway/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Sci Rep ; 11(1): 20075, 2021 10 08.
Article in English | MEDLINE | ID: mdl-34625606

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder that leads to dementia and behavioral changes. Extracellular deposition of amyloid plaques (Aß) and intracellular deposition of neurofibrillary tangles in neurons are the major pathogenicities of AD. However, drugs targeting these therapeutic targets are not effective. Therefore, novel targets for the treatment of AD urgently need to be identified. Expression of the mesoderm-specific transcript (Mest) is regulated by genomic imprinting, where only the paternal allele is active for transcription. We identified hypermethylation on the Mest promoter, which led to a reduction in Mest mRNA levels and activation of Wnt signaling in brain tissues of AD patients. Mest knockout (KO) using the CRIPSR/Cas9 system in mouse embryonic stem cells and P19 embryonic carcinoma cells leads to neuronal differentiation arrest. Depletion of Mest in primary hippocampal neurons via lentivirus expressing shMest or inducible KO system causes neurodegeneration. Notably, depletion of Mest in primary cortical neurons of rats leads to tau phosphorylation at the S199 and T231 sites. Overall, our data suggest that hypermethylation of the Mest promoter may cause or facilitate the progression of AD.


Subject(s)
Alzheimer Disease/pathology , DNA Methylation , Embryonic Stem Cells/pathology , Neurons/pathology , Promoter Regions, Genetic , Proteins/genetics , Wnt Signaling Pathway , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Embryonal Carcinoma Stem Cells/metabolism , Embryonal Carcinoma Stem Cells/pathology , Embryonic Stem Cells/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Humans , Mice , Neurons/metabolism , Phosphorylation , Proteins/metabolism , tau Proteins/genetics , tau Proteins/metabolism
3.
Biochem Biophys Res Commun ; 570: 169-174, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34284143

ABSTRACT

Glycosaminoglycan polysaccharides are components of animal extracellular matrices and regulate cell functions based on their various sulfation and epimerization pattern structures. The present study aimed to find glycosaminoglycan structures to promote neural differentiation. We investigated the effect of exogenous glycosaminoglycans with well-defined structures on the all-trans-retinoic acid-induced neural differentiation of P19 embryonal carcinoma cells, which is an ideal model culture system for studying neural differentiation. We found that chondroitin sulfate E and heparin, but not any other glycosaminoglycans, upregulated the expressions of neural specific markers but not a grail specific marker. Chondroitin sulfate E was suggested to function during spheroid formation, however, equimolar concentration of its oligosaccharide did not show promotive effect on the neural differentiation. Another finding was that hyaluronan oligosaccharide mixture markedly downregulated the expressions of a myelin specific marker. These findings suggested that the specific sulfation pattern and/or chain length of exogenous added glycosaminoglycan is important to regulate neural differentiation and myelination.


Subject(s)
Cell Differentiation/drug effects , Embryonal Carcinoma Stem Cells/pathology , Glycosaminoglycans/chemistry , Glycosaminoglycans/pharmacology , Neurons/pathology , Tretinoin/pharmacology , Animals , Biomarkers/metabolism , Cattle , Mice , Neurons/drug effects , Oligosaccharides/metabolism , Swine
4.
Int J Mol Sci ; 22(5)2021 Feb 24.
Article in English | MEDLINE | ID: mdl-33668324

ABSTRACT

FOXC1, a transcription factor involved in cell differentiation and embryogenesis, is demonstrated to be a negative regulator of Nanog in this study. FOXC1 is up-regulated in retinoic acid-induced differentiation of F9 Embryonal Carcinoma (EC) cells; furthermore, FOXC1 specifically inhibits the core pluripotency factor Nanog by binding to the proximal promoter. Overexpression of FOXC1 in F9 or knockdown in 3T3 results in the down-regulation or up-regulation of Nanog mRNA and proteins, respectively. In order to explain the mechanism by which FOXC1 inhibits Nanog expression, we identified the co-repressor HDAC2 from the FOXC1 interactome. FOXC1 recruits HDAC2 to Nanog promoter to decrease H3K27ac enrichment, resulting in transcription inhibition of Nanog. To the best of our knowledge, this is the first report that FOXC1 is involved in the epigenetic regulation of gene expression.


Subject(s)
Embryonal Carcinoma Stem Cells/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase 2/metabolism , Nanog Homeobox Protein/genetics , Promoter Regions, Genetic , Tretinoin/pharmacology , Animals , Antineoplastic Agents/pharmacology , Embryonal Carcinoma Stem Cells/drug effects , Embryonal Carcinoma Stem Cells/pathology , Epigenesis, Genetic , Forkhead Transcription Factors/genetics , HEK293 Cells , Histone Deacetylase 2/genetics , Humans , Mice , NIH 3T3 Cells , Nanog Homeobox Protein/metabolism
5.
RNA Biol ; 17(11): 1613-1624, 2020 11.
Article in English | MEDLINE | ID: mdl-32372724

ABSTRACT

PIWI homologs constitute a subclass of the Argonaute family. Traditionally, they have been shown to associate with a specific class of small RNAs, piRNAs, to suppress transposable elements and protect genomic integrity in germ cells. Recent studies imply that PIWI proteins may also exert important biological functions in somatic contexts, including the brain. However, their exact role in neural development remains unknown. Hence we investigated whether PIWI proteins are involved in neuronal differentiation. By using an established cell model for studying neurogenesis, NTera2/D1 (NT2) cells, we found that a particular PIWI homolog, PIWIL4 was increasingly upregulated throughout the course of all-trans retinoic acid (RA)-mediated neuronal differentiation. During this process, PIWIL4 knockdown led to partial recovery of embryonic stem cell markers, while suppressing RA-induced expression of neuronal markers. Consistently, PIWIL4 overexpression further elevated their expression levels. Furthermore, co-immunoprecipitation revealed an RA-induced interaction between PIWIL4 and the H3K27me3 demethylase UTX. Chromatin immunoprecipitation showed that this interaction could be essential for the removal of H3K27me3 from the promoters of RA-inducible genes. By a similar mechanism, PIWIL4 knockdown also suppressed the expression of PTN and NLGN3, two important neuronal factors secreted to regulate glioma activity. We further noted that the conditioned medium collected from PIWIL4-silenced NT2 cells significantly reduced the proliferation of glioma cells. Thus, our data suggest a novel somatic role of PIWIL4 in modulating the expression of neuronal genes that can be further characterized to promote neuronal differentiation and to modulate the activity of glioma cells.


Subject(s)
Cell Differentiation/genetics , Embryonal Carcinoma Stem Cells/metabolism , Embryonal Carcinoma Stem Cells/pathology , Neurons/metabolism , RNA-Binding Proteins/genetics , Cell Differentiation/drug effects , Cell Line , Cell Proliferation , Cell Survival/drug effects , Cell Survival/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental/drug effects , Gene Knockdown Techniques , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Histone Demethylases/metabolism , Histones/metabolism , Humans , Neurons/cytology , Protein Binding , RNA-Binding Proteins/metabolism , Transcriptome
6.
Sci Rep ; 9(1): 11928, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31417131

ABSTRACT

Human embryonal carcinoma (EC) cells comprise the pluripotent stem cells of malignant non-seminomatous germ cell tumors (GCTs) and represent the malignant counterpart of embryonic stem cells (ESCs). WNT/ß-catenin signaling has been implicated in regulating adult and embryonic stem cells although its role in EC cells is less investigated. Here, we studied WNT signaling in a panel of representative pluripotent and nullipotent human EC cell lines. We found that EC cell lines show distinct levels of intrinsic WNT signaling and respond differently to ectopic WNT activation. Short-term activation of WNT signaling induced a differentiation-response in the pluripotent EC cells (NT2 and NCCIT) whereas the nullipotent EC cells (TERA1 and 2102Ep) were refractory and maintained high levels of OCT4 and SSEA4 expression. Long-term activation of WNT signaling in NCCIT and, to a lesser extent, TERA1 cells led to (re)gain of OCT4 expression and a switch from SSEA4 to SSEA1 surface antigens ultimately resulting in OCT4+/SSEA4-/SSEA1+ profile. Cisplatin treatment indicated that the OCT4+/SSEA4-/SSEA1+ NCCIT cells became more resistant to chemotherapy treatment. Our findings are of particular interest for the GCT and ES cell biology and shed light on the role of WNT signaling in human EC cells.


Subject(s)
Cell Culture Techniques , Embryonal Carcinoma Stem Cells/metabolism , Embryonal Carcinoma Stem Cells/pathology , Wnt Signaling Pathway , Cell Differentiation/drug effects , Cell Line, Tumor , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Humans , Stage-Specific Embryonic Antigens/metabolism , Time Factors , Wnt Signaling Pathway/drug effects
7.
J Vis Exp ; (146)2019 04 27.
Article in English | MEDLINE | ID: mdl-31081818

ABSTRACT

The P19 cell line derived from a mouse embryo-derived teratocarcinoma has the ability to differentiate into the three germ layers. In the presence of retinoic acid (RA), the suspension cultured P19 cell line is induced to differentiate into neurons. This phenomenon is extensively investigated as a neurogenesis model in vitro. Therefore, the P19 cell line is very useful for molecular and cellular studies associated with neurogenesis. However, protocols for neuronal differentiation of P19 cell line described in the literature are very complex. The method developed in this study are simple and will play a part in elucidating the molecular mechanisms in neurodevelopmental abnormalities and neurodegenerative diseases.


Subject(s)
Embryonal Carcinoma Stem Cells/pathology , Neurogenesis , Animals , Cell Differentiation/drug effects , Embryonal Carcinoma Stem Cells/metabolism , Image Processing, Computer-Assisted , Mice , Neurogenesis/drug effects , Tretinoin/pharmacology
8.
Biochem Biophys Res Commun ; 512(3): 629-634, 2019 05 07.
Article in English | MEDLINE | ID: mdl-30914194

ABSTRACT

During brain development, the expression of promyelocytic leukemia zinc finger (Plzf) in neural stem cells is precisely controlled to maintain the balance between neural stem cell self-renewal and differentiation. However, the mechanism underlying transcriptional regulation of Plzf in neural stem cell is still unclear. Herein, using P19 embryonal carcinoma cells as a model, we observed that Plzf expression was induced in the P19-derived embryonic bodies, which enrich neural stem-like cell populations, as demonstrated by the expression of neural stem cell markers, Nestin and Sox2. We then characterized the Plzf promoter and identified two E2f1 binding sites (-755/-751 and -53/-49, the transcription start site was designated as +1) are important for the activation of Plzf promoter. Finally, we found that the induction of Plzf in the neural stem-like cells derived from pluripotent P19 cells is decrease by E2f1 knockdown. Taken together, we conclude that E2f1 is an important transcription factor that regulates Plzf transcription and may involve in maintaining the self-renewal ability of neural stem cells.


Subject(s)
E2F1 Transcription Factor/metabolism , Embryonal Carcinoma Stem Cells/pathology , Gene Expression Regulation, Neoplastic , Neural Stem Cells/pathology , Promyelocytic Leukemia Zinc Finger Protein/genetics , Animals , Cell Line, Tumor , Embryonal Carcinoma Stem Cells/metabolism , Mice , Neural Stem Cells/metabolism , Neurogenesis , Promoter Regions, Genetic , Zinc Fingers
9.
J Biol Chem ; 294(15): 5914-5922, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30808711

ABSTRACT

Embryonic stem cells can self-renew and differentiate, holding great promise for regenerative medicine. They also employ multiple mechanisms to preserve the integrity of their genomes. Nucleotide excision repair, a versatile repair mechanism, removes bulky DNA adducts from the genome. However, the dynamics of the capacity of nucleotide excision repair during stem cell differentiation remain unclear. Here, using immunoslot blot assay, we measured repair rates of UV-induced DNA damage during differentiation of human embryonic carcinoma (NTERA-2) cells into neurons and muscle cells. Our results revealed that the capacity of nucleotide excision repair increases as cell differentiation progresses. We also found that inhibition of the apoptotic signaling pathway has no effect on nucleotide excision repair capacity. Furthermore, RNA-Seq-based transcriptomic analysis indicated that expression levels of four core repair factors, xeroderma pigmentosum (XP) complementation group A (XPA), XPC, XPG, and XPF-ERCC1, are progressively up-regulated during differentiation, but not those of replication protein A (RPA) and transcription factor IIH (TFIIH). Together, our findings reveal that increase of nucleotide excision repair capacity accompanies cell differentiation, supported by the up-regulated transcription of genes encoding DNA repair enzymes during differentiation of two distinct cell lineages.


Subject(s)
Cell Differentiation , DNA Repair , Embryonal Carcinoma Stem Cells/metabolism , Muscle Cells/metabolism , Neoplasm Proteins/metabolism , Neurons/metabolism , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryonal Carcinoma Stem Cells/pathology , Endonucleases/genetics , Endonucleases/metabolism , Humans , Muscle Cells/pathology , Neoplasm Proteins/genetics , Neurons/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Replication Protein A/genetics , Replication Protein A/metabolism , Transcription Factor TFIIH/genetics , Transcription Factor TFIIH/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Xeroderma Pigmentosum Group A Protein/genetics , Xeroderma Pigmentosum Group A Protein/metabolism
10.
Biochem Biophys Res Commun ; 509(2): 603-610, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30606481

ABSTRACT

MicroRNA-135a-5p has been reported to play a potential role in the generation of new neurons. However, the underlying targets of miR-135a-5p in regulating neuronal differentiation have been poorly understood. Our study recently has uncovered that Sox6 and CD44 genes were significantly downregulated during neuronal differentiation of P19 cells, a multipotent cell type. We then found that Sox6 directly bound to the promoter of CD44. Importantly, we identified Sox6 as a direct target of miR-135a-5p. Additionally, we demonstrated that miR-135a-5p is crucial for the neuronal differentiation of P19 cells. More significantly, we found that Sox6 overexpression could overturn miR-135a-5p-mediated neuronal differentiation and dendrite development. In conclusion, these findings indicated that miR-135a-5p/Sox6/CD44 axis provides an important molecular target mechanism for neurodifferentiation.


Subject(s)
Embryonal Carcinoma Stem Cells/pathology , Hyaluronan Receptors/genetics , MicroRNAs/genetics , Neurogenesis , SOXD Transcription Factors/genetics , Animals , Cell Line, Tumor , Embryonal Carcinoma Stem Cells/cytology , Embryonal Carcinoma Stem Cells/metabolism , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Hyaluronan Receptors/metabolism , Mice , SOXD Transcription Factors/metabolism , Signal Transduction
11.
Chemosphere ; 216: 372-378, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30384306

ABSTRACT

Ambient fine particulate matter (PM2.5) has been found to be associated with congenital heart defects, but the molecular mechanisms remain to be elucidated. Our previous study revealed that extractable organic matter (EOM) from PM2.5 exerted cardiac developmental toxicity in zebrafish embryos. The aim of the current study is to explore the effects of EOM on cardiac differentiation of P19 mouse embryonic carcinoma stem cells. We found that EOM at 10 µg/ml (a non-cytotoxic dose level) significantly reduced the proportion of cardiac muscle troponin (cTnT) positive cells and the percentage of spontaneously beating embryoid bodies, indicating a severe inhibition of cardiac differentiation. Immunofluorescence and qPCR data demonstrated that EOM increased the expression levels of the aryl hydrocarbon receptor (AhR) and its target gene Cyp1A1 and diminished the expression level of ß-catenin. Furthermore, EOM treatment significantly upregulated cell proliferation rate and elevated the percentage of γH2A.X positive cells without affecting apoptosis. It is worth noting that the EOM-induced changes in gene expression, cellular proliferation and DNA double strain breaks were attenuated by the AhR antagonist CH223191. In conclusion, our data indicate that AhR mediates the inhibitory effects of EOM (from PM2.5) on the cardiac differentiation of P19 cells.


Subject(s)
Cardiotoxicity/drug therapy , Embryonal Carcinoma Stem Cells/metabolism , MicroRNAs/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Cell Differentiation , Cell Proliferation , Embryonal Carcinoma Stem Cells/pathology , Gene Expression , Mice
12.
Proc Natl Acad Sci U S A ; 115(41): 10434-10439, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30249655

ABSTRACT

HERV-K HML-2 (HK2) has been proliferating in the germ line of humans at least as recently as 250,000 years ago, with some integrations that remain polymorphic in the modern human population. One of the solitary HK2 LTR polymorphic integrations lies between exons 17 and 18 of RASGRF2, a gene that affects dopaminergic activity and is thus related to addiction. Here we show that this antisense HK2 integration (namely RASGRF2-int) is found more frequently in persons who inject drugs compared with the general population. In a Greek HIV-1-positive population (n = 202), we found RASGRF2-int 2.5 times (14 versus 6%) more frequently in patients infected through i.v. drug use compared with other transmission route controls (P = 0.03). Independently, in a United Kingdom-based hepatitis C virus-positive population (n = 184), we found RASGRF2-int 3.6 times (34 versus 9.5%) more frequently in patients infected during chronic drug abuse compared with controls (P < 0.001). We then tested whether RASGRF2-int could be mechanistically responsible for this association by modulating transcription of RASGRF2 We show that the CRISPR/Cas9-mediated insertion of HK2 in HEK293 cells in the exact RASGRF2 intronic position found in the population resulted in significant transcriptional and phenotypic changes. We also explored mechanistic features of other intronic HK2 integrations and show that HK2 LTRs can be responsible for generation of cis-natural antisense transcripts, which could interfere with the transcription of nearby genes. Our findings suggest that RASGRF2-int is a strong candidate for dopaminergic manipulation, and emphasize the importance of accurate mapping of neglected HERV polymorphisms in human genomic studies.


Subject(s)
Embryonal Carcinoma Stem Cells/metabolism , Endogenous Retroviruses/genetics , Substance Abuse, Intravenous/genetics , Transcription, Genetic , Virus Integration/genetics , ras Guanine Nucleotide Exchange Factors/genetics , Case-Control Studies , Child , Cohort Studies , Embryonal Carcinoma Stem Cells/pathology , Female , Genome, Human , Humans , Male , Tumor Cells, Cultured
13.
J Tissue Eng Regen Med ; 12(9): 1909-1924, 2018 09.
Article in English | MEDLINE | ID: mdl-29905008

ABSTRACT

Tissue engineering, as a novel transplantation therapy, aims to create biomaterial scaffolds resembling the extracellular matrix in order to regenerate the damaged tissues. Adding bioactive factors to the scaffold would improve cell-tissue interactions. In this study, the effect of chitosan polyvinyl alcohol nanofibres containing carbon nanotube scaffold with or without active bioglass (BG+ /BG- ), in combination with neonatal rat brain extract on cell viability, proliferation, and neural differentiation of P19 embryonic carcinoma stem cells was investigated. To induce differentiation, the cells were cultured in α-MEM supplemented with neonatal rat brain extract on the scaffolds. The expression of undifferentiated stem cell markers as well as neuroepithelial and neural-specific markers was evaluated and confirmed by real-time Reverse transcription polymerase chain reaction (RT-PCR) and immunofluorescence procedures. Finally, the three-dimensional (3D) cultured cells were implanted into the damaged neural tubes of chick embryos, and their fates were followed in ovo. Based on the histological and immunofluorescence observations, the transplanted cells were able to survive, migrate, and penetrate into the host embryonic tissues. Gene network analysis suggested the possible involvement of neurotransmitters as a downstream target of synaptophysin and tyrosine hydroxylase. Overall, the results of this study indicated that combining the effects of 3D cell culture and natural brain tissue extract can accelerate the differentiation of P19 embryonic carcinoma cells into neuronal phenotype cells.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation/drug effects , Embryonal Carcinoma Stem Cells/pathology , Neurons/pathology , Tissue Extracts/pharmacology , Animals , Cell Proliferation/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , Chick Embryo , Chickens , Female , Gene Expression Regulation/drug effects , Male , Neurites/drug effects , Neurites/metabolism , Neurons/drug effects , Rats, Wistar
14.
Biosci Rep ; 38(4)2018 08 31.
Article in English | MEDLINE | ID: mdl-29769412

ABSTRACT

Intermediate-sized non-coding RNAs (imsncRNAs) have been shown to play important regulatory roles in the development of several eukaryotic organisms. In the present research, we selected imsncRNA 761 (imsnc761) as a research target. Expression analyses in a previous study showed that imsnc761 was down-regulated in maturation-arrested testis tissues as compared with the level in normal controls. In the present study, we found that imsnc761 could interact with DEAD-box helicase 6 (DDX6) to induce NTERA-2 (NT2 (testicular embryonal carcinoma cell)) cell apoptosis and proliferation inhibition via the p53 pathway. This interaction between imsnc761 and DDX6 also inhibited mitochondrial function and specific gene transcription and translation. To facilitate further research, we used label-free quantitation method to analyze the associated differences in Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathways and biological processes. This confirmed the changes in several specific pathways, which matched our molecular experimental results.


Subject(s)
Apoptosis , DEAD-box RNA Helicases/metabolism , Embryonal Carcinoma Stem Cells/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Untranslated/metabolism , Testicular Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Cell Proliferation , Embryonal Carcinoma Stem Cells/pathology , HEK293 Cells , Humans , Male , Testicular Neoplasms/pathology , Testis/metabolism , Testis/pathology
15.
Int J Nanomedicine ; 12: 7529-7549, 2017.
Article in English | MEDLINE | ID: mdl-29066898

ABSTRACT

BACKGROUND: Silver nanoparticles (AgNPs) exhibit strong antibacterial and anticancer activity owing to their large surface-to-volume ratios and crystallographic surface structure. Owing to their various applications, understanding the mechanisms of action, biological interactions, potential toxicity, and beneficial effects of AgNPs is important. Here, we investigated the toxicity and differentiation-inducing effects of AgNPs in teratocarcinoma stem cells. MATERIALS AND METHODS: AgNPs were synthesized and characterized using various analytical techniques such as UV-visible spectroscopy, X-ray diffraction, energy-dispersive X-ray spectroscopy, and transmission electron microscopy. The cellular responses of AgNPs were analyzed by a series of cellular and biochemical assays. Gene and protein expressions were analyzed by reverse transcription-quantitative polymerase chain reaction and western blotting, respectively. RESULTS: The AgNPs showed typical crystalline structures and spherical shapes (average size =20 nm). High concentration of AgNPs induced cytotoxicity in a dose-dependent manner by increasing lactate dehydrogenase leakage and reactive oxygen species. Furthermore, AgNPs caused mitochondrial dysfunction, DNA fragmentation, increased expression of apoptotic genes, and decreased expression of antiapoptotic genes. Lower concentrations of AgNPs induced neuronal differentiation by increasing the expression of differentiation markers and decreasing the expression of stem cell markers. Cisplatin reduced the viability of F9 cells that underwent AgNPs-induced differentiation. CONCLUSION: The results showed that AgNPs caused differentially regulated cytotoxicity and induced neuronal differentiation of F9 cells in a concentration-dependent manner. Therefore, AgNPs can be used for differentiation therapy, along with chemotherapeutic agents, for improving cancer treatment by targeting specific chemotherapy-resistant cells within a tumor. Furthermore, understanding the molecular mechanisms of apoptosis and differentiation in stem cells could also help in developing new strategies for cancer stem cell (CSC) therapies. The findings of this study could significantly contribute to the nanomedicine because this study is the first of its kind, and our results will lead to new strategies for cancer and CSC therapies.


Subject(s)
Apoptosis , Cell Differentiation , Embryonal Carcinoma Stem Cells/pathology , Metal Nanoparticles/chemistry , Models, Biological , Silver/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Down-Regulation/drug effects , Down-Regulation/genetics , Embryonal Carcinoma Stem Cells/drug effects , Extracellular Matrix/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/pharmacology , Silver/chemistry , Up-Regulation/drug effects , Up-Regulation/genetics , X-Ray Diffraction
16.
Cell Death Differ ; 24(11): 1975-1986, 2017 11.
Article in English | MEDLINE | ID: mdl-28885616

ABSTRACT

We have previously reported that myeloid differentiation primary response gene 88 (MyD88) is downregulated during all-trans retinoic acid (RA)-induced differentiation of pluripotent NTera2 human embryonal carcinoma cells (hECCs), whereas its maintained expression is associated with RA differentiation resistance in nullipotent 2102Ep hECCs. MyD88 is the main adapter for toll-like receptor (TLR) signalling, where it determines the secretion of chemokines and cytokines in response to pathogens. In this study, we report that loss of MyD88 is essential for RA-facilitated differentiation of hECCs. Functional analysis using a specific MyD88 peptide inhibitor (PepInh) demonstrated that high MyD88 expression in the self-renewal state inhibits the expression of a specific set of HOX genes. In NTera2 cells, MyD88 is downregulated during RA-induced differentiation, a mechanism that could be broadly replicated by MyD88 PepInh treatment of 2102Ep cells. Notably, MyD88 inhibition transitioned 2102Ep cells into a stable, self-renewing state that appears to be primed for differentiation upon addition of RA. At a molecular level, MyD88 inhibition combined with RA treatment upregulated HOX, RA signalling and TLR signalling genes. These events permit differentiation through a standard downregulation of Oct4-Sox2-Nanog mechanism. In line with its role in regulating secretion of specific proteins, conditioned media experiments demonstrated that differentiated (MyD88 low) NTera2 cell media was sufficient to differentiate NTera2 cells. Protein array analysis indicated that this was owing to secretion of factors known to regulate angiogenesis, neurogenesis and all three branches of TGF-ß Superfamily signalling. Collectively, these data offer new insights into RA controlled differentiation of pluripotent cells, with notable parallels to the ground state model of embryonic stem cell self-renewal. These data may provide insights to facilitate improved differentiation protocols for regenerative medicine and differentiation-therapies in cancer treatment.


Subject(s)
Cell Differentiation/drug effects , Embryonal Carcinoma Stem Cells/pathology , Myeloid Differentiation Factor 88/metabolism , Pluripotent Stem Cells/pathology , Tretinoin/pharmacology , Cell Differentiation/genetics , Cell Self Renewal/drug effects , Cell Self Renewal/genetics , Embryonal Carcinoma Stem Cells/drug effects , Embryonal Carcinoma Stem Cells/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mesoderm/pathology , Models, Biological , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
17.
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
18.
J Biol Chem ; 292(10): 3970-3976, 2017 03 10.
Article in English | MEDLINE | ID: mdl-28179426

ABSTRACT

Lin28a, originally discovered in the nematode Caenorhabditis elegans and highly conserved across species, is a well characterized regulator of let-7 microRNA (miRNA) and is implicated in cell proliferation and pluripotency control. However, little is known about how Lin28a function is modulated at the post-translational level and thereby responds to major signaling pathways. Here we show that Lin28a is directly phosphorylated by ERK1/2 kinases at Ser-200. By editing lin28a gene with the CRISPR/Cas9-based method, we generated P19 mouse embryonic carcinoma stem cells expressing Lin28a-S200A (phospho-deficient) and Lin28a-S200D (phospho-mimetic) mutants, respectively, to study the functional impact of Ser-200 phosphorylation. Lin28a-S200D-expressing cells, but not Lin28a-S200A-expressing or control P19 embryonic carcinoma cells, displayed impaired inhibition of let-7 miRNA and resulted in decreased cyclin D1, whereas Lin28a-S200A knock-in cells expressed less let-7 miRNA, proliferated faster, and exhibited differentiation defect upon retinoic acid induction. Therefore our results support that ERK kinase-mediated Lin28a phosphorylation may be an important mechanism for pluripotent cells to facilitate the escape from the self-renewal cycle and start the differentiation process.


Subject(s)
Cell Differentiation , Cell Proliferation , Embryonal Carcinoma Stem Cells/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , RNA-Binding Proteins/metabolism , Animals , Embryonal Carcinoma Stem Cells/metabolism , HeLa Cells , Humans , Mice , Phosphorylation , RNA-Binding Proteins/genetics , Signal Transduction
19.
Anticancer Agents Med Chem ; 17(9): 1184-1198, 2017.
Article in English | MEDLINE | ID: mdl-27306567

ABSTRACT

Retinoic acid (RA), especially all-trans retinoic acid is the most potent natural metabolite of vitamin A. RA is involved in a variety of biological functions including embryogenesis, cell differentiation and apoptosis. RA acts through its nuclear receptors to induce transcription of specific target genes. Mouse P19 embryonic carcinoma (EC) stem cells (ES) are one of the most studied in vitro systems for RA-induced differentiation. P19 ES cells can differentiate to endodermal-like, mesodermal-like, and neuronal-like phenotypes in response to specific morphogens including RA and dimethyl sulfoxide (DMSO). At low concentrations, RA directs P19 ES cells to differentiate into cells displaying an endodermal phenotype, whereas at higher concentrations it induces differentiation to neuroectoderm. In the past, many RA---regulated genes have been discovered in EC and ES cells and efforts are ongoing to elucidate the exact mechanisms of RA-induced ES cell differentiation and apoptosis. In the RA-triggered differentiation process of the P19 ES cells, several proteins belonging to different families participate, some being obligatory while others, dispensable. Revealing the mechanisms behind RA-induced effects on ES cells has a bearing on understanding how cells proliferate, differentiate and undergo apoptosis that can provide greater insight into cancer biology and therapy. In addition to summarizing the reports on gene/protein targets of RA in stem cells, the signaling pathways driven by some of the specific class of proteins in the presence or absence of RA in P19 ES cell differentiation, especially to an endodermal phenotype, are the focus of this review.


Subject(s)
Embryonal Carcinoma Stem Cells/metabolism , Signal Transduction , Tretinoin/metabolism , Animals , Cell Differentiation/drug effects , Embryonal Carcinoma Stem Cells/drug effects , Embryonal Carcinoma Stem Cells/pathology , Mice , Tretinoin/pharmacology
20.
Mol Cell Probes ; 32: 46-54, 2017 04.
Article in English | MEDLINE | ID: mdl-28013042

ABSTRACT

Murine P19 embryonal carcinoma (EC) cells are convenient to differentiate into all germ layer derivatives. One of the advantages of P19 cells is that the exogenous DNA can be easily inserted into them. Here, at the first part of this study, we generated stable GFP-expressing P19 cells (P19-GFP+). FACS and western-blot analysis confirmed stable expression of GFP in the cells. We previously demonstrated the efficient induction of neuronal differentiation from mouse ES and EC cells by application of a neuroprotective drug, selegiline In the second part of this study selegiline was used to induce differentiation of P19-GFP+ into stable GFP-expressing neuron-like cells. Cresyl violet staining confirmed neuronal morphology of the differentiated cells. Furthermore, real-time PCR and immunoflourescence approved the expression of neuron specific markers. P19-GFP+ cells were able to survive, migrate and integrated into host tissues when transplanted to developing chick embryo CNS. The obtained live GFP-expressing cells can be used as an abundant source of developmentally pluripotent material for transplantation studies, investigating the cellular and molecular aspects of early differentiation.


Subject(s)
Cell Culture Techniques/methods , Embryonal Carcinoma Stem Cells/pathology , Green Fluorescent Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Animals , Cell Differentiation/drug effects , Chickens , Embryonal Carcinoma Stem Cells/drug effects , Embryonal Carcinoma Stem Cells/transplantation , Fluorescence , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/drug effects , Mice , Neurons/drug effects , Selegiline/pharmacology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...