Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Stem Cells ; 39(5): 536-550, 2021 05.
Article in English | MEDLINE | ID: mdl-33470499

ABSTRACT

Glioblastoma (GBM) is the most common and deadliest tumor of the central nervous system. GBM has poor prognosis and glioma stem cells (GSCs) are implicated in tumor initiation and therapy resistance. Estrogen receptor ß (ERß) is expressed in GBM and exhibit tumor suppressive function. However, the role of ERß in GSCs and the therapeutic potential of ERß agonists on GSCs remain largely unknown. Here, we examined whether ERß modulates GSCs stemness and tested the utility of two ERß selective agonists (LY500307 and Liquiritigenin) to reduce the stemness of GSCs. The efficacy of ERß agonists was examined on GSCs isolated from established and patient derived GBMs. Our results suggested that knockout of ERß increased the proportion of CD133+ and SSEA+ positive GSCs and overexpression of ERß reduced the proportion of GSCs in GBM cells. Overexpression of ERß or treatment with ERß agonists significantly inhibited the GSCs cell viability, neurosphere formation, self-renewal ability, induced the apoptosis and reduced expression of stemness markers in GSCs. RNA sequencing analysis revealed that ERß agonist modulate pathways related to stemness, differentiation and apoptosis. Mechanistic studies showed that ERß overexpression or agonist treatment reduced glutamate receptor signaling pathway and induced apoptotic pathways. In orthotopic models, ERß overexpression or ERß agonists treatment significantly reduced the GSCs mediated tumor growth and improved the mice overall survival. Immunohistochemical studies demonstrated that ERß overexpression decreased SOX2 and GRM3 expression and increased expression of GFAP in tumors. These results suggest that ERß activation could be a promising therapeutic strategy to eradicate GSCs.


Subject(s)
Cell Differentiation/genetics , Cell Proliferation/drug effects , Estrogen Receptor beta/genetics , Glioma/genetics , Neoplastic Stem Cells/metabolism , AC133 Antigen/genetics , Animals , Apoptosis/drug effects , Benzopyrans/pharmacology , Cell Differentiation/drug effects , Cell Line, Tumor , Estrogen Receptor beta/agonists , Flavanones/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Glial Fibrillary Acidic Protein/genetics , Glioma/drug therapy , Glioma/pathology , Humans , Mice , Neoplastic Stem Cells/drug effects , Receptors, Glutamate/genetics , SOXB1 Transcription Factors/genetics , Signal Transduction/drug effects , Stage-Specific Embryonic Antigens/genetics , Xenograft Model Antitumor Assays
2.
Anticancer Res ; 40(10): 5567-5575, 2020 10.
Article in English | MEDLINE | ID: mdl-32988880

ABSTRACT

BACKGROUND/AIM: Stage-specific embryonic antigen-4 (SSEA-4) expression is associated with malignant aggressiveness and is useful as a marker for identifying cancer stem cells. Our aim was to assess the relationship between hormonal therapy and SSEA-4 expression in prostate cancer (PC). MATERIALS AND METHODS: SSEA-4 expression in paired specimens from PC patients who underwent neoadjuvant hormonal therapy (NHT) and radical prostatectomy (60 pre-NHT specimens and 60 post-NHT specimens) was evaluated using immunohistochemistry. Proliferation index (PI) and apoptotic index (AI) were also evaluated. RESULTS: Post-NHT tissues had significantly elevated SSEA-4 expression whereas anti-tumor effects of NHT were inversely correlated with SSEA-4 expression level. SSEA-4 expression in post-NHT tissues was significantly associated with biochemical recurrence-free survival. SSEA-4 expression in the post-NHT tissues was positively associated with PI and negatively done with AI. CONCLUSION: SSEA-4 is a potential therapeutic target for limiting the malignant potential in hormone-naïve PC when considering the use of NHT.


Subject(s)
Antineoplastic Agents, Hormonal/administration & dosage , Biomarkers, Tumor/genetics , Prostatic Neoplasms/drug therapy , Stage-Specific Embryonic Antigens/genetics , Aged , Humans , Male , Middle Aged , Neoadjuvant Therapy , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prostate/pathology , Prostatectomy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery
3.
Int J Mol Sci ; 21(15)2020 Jul 29.
Article in English | MEDLINE | ID: mdl-32751344

ABSTRACT

Human breast adenocarcinoma cells (MCF7) grow in three-dimensional culture as spheroids that represent the structural complexity of avascular tumors. Therefore, spheroids offer a powerful tool for studying cancer development, aggressiveness, and drug resistance. Notwithstanding the large amount of data regarding the formation of MCF7 spheroids, a detailed description of the morpho-functional changes during their aggregation and maturation is still lacking. In this study, in addition to the already established role of gap junctions, we show evidence of tunneling nanotube (TNT) formation, amyloid fibril production, and opening of large stable cellular bridges, thus reporting the sequential events leading to MCF7 spheroid formation. The variation in cell phenotypes, sustained by dynamic expression of multiple proteins, leads to complex networking among cells similar to the sequence of morphogenetic steps occurring in embryogenesis/organogenesis. On the basis of the observation that early events in spheroid formation are strictly linked to the redox homeostasis, which in turn regulate amyloidogenesis, we show that the administration of N-acetyl-l-cysteine (NAC), a reactive oxygen species (ROS) scavenger that reduces the capability of cells to produce amyloid fibrils, significantly affects their ability to aggregate. Moreover, cells aggregation events, which exploit the intrinsic adhesiveness of amyloid fibrils, significantly decrease following the administration during the early aggregation phase of neutral endopeptidase (NEP), an amyloid degrading enzyme.


Subject(s)
Acetylcysteine/pharmacology , Amyloid/chemistry , Free Radical Scavengers/pharmacology , Gap Junctions/ultrastructure , Homeostasis/drug effects , Spheroids, Cellular/ultrastructure , Amyloid/drug effects , Amyloid/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Aggregation/drug effects , Connexin 43/genetics , Connexin 43/metabolism , Gap Junctions/drug effects , Gap Junctions/metabolism , Gene Expression , Homeostasis/genetics , Humans , Interleukin-18/genetics , Interleukin-18/metabolism , MCF-7 Cells , Neprilysin/pharmacology , Oxidation-Reduction , Phenotype , Proteolysis , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Stage-Specific Embryonic Antigens/genetics , Stage-Specific Embryonic Antigens/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , gp100 Melanoma Antigen/genetics , gp100 Melanoma Antigen/metabolism
4.
Stem Cell Res Ther ; 10(1): 211, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31315669

ABSTRACT

BACKGROUND: Autosomal recessive osteopetrosis is a genetically and phenotypically heterogeneous disease, caused by defects in osteoclast formation and function. The only available treatment is allogeneic stem cell transplantation that has still high morbidity and mortality. The goal of the present study was to generate iPSCs from bone marrow-derived MSCs of osteopetrosis patients with three most common mutations by using two different integration-free gene transfer methods and compare their efficiencies. The secondary objective was to select the most appropriate integration-free production method for our institutional iPSC bank using this rare disease as a prototype. METHODS: Two different integration-free gene transfer methods (episomal and Sendai viral vectors) were tested and compared on the same set of patient samples exhibiting three different mutations associated with osteopetrosis. Generated iPSCs were characterized by standard assays, including immunophenotyping, immunocytochemistry, RT-PCR, embryoid body, and teratoma assays. Karyotype analyses were performed to evaluate genetic stability. RESULTS: iPSC lines exhibiting typical ESC-like colony morphology were shown to express pluripotency markers by immunofluorescence staining. Over 90% of the cells were found positive for SSEA-4 and OCT3/4 and negative/weak positive for CD29 by flow cytometry. Immunohistochemical staining of teratoma and spontaneously differentiated embryoid body sections confirmed their trilineage differentiation potential. All iPSC lines expressed pluripotency-related genes. Karyotype analyses were found normal. Direct sequencing of PCR-amplified DNA showed that disease-related mutations were retained in the patient-specific iPSCs. CONCLUSION: Generation of iPSC using SeV and episomal DNA vectors have several advantages over other methods like the ease of production, reliability, high efficiency, and safety, which is required for translational research. Furthermore, owing to the pluripotency and self-renewal capacity, patient-specific iPSCs seem to be ideal cell source for the modeling of a rare genetic bone disease like osteopetrosis to identify osteoclast defects, leading to clinical heterogeneity in osteopetrosis patients, especially among those with different mutations in the same gene.


Subject(s)
Gene Transfer Techniques , Mesenchymal Stem Cell Transplantation , Osteopetrosis/congenital , Pluripotent Stem Cells/transplantation , Cellular Reprogramming/genetics , Child , Child, Preschool , Chloride Channels/genetics , Female , Flow Cytometry , Humans , Induced Pluripotent Stem Cells/transplantation , Infant , Integrin beta1/genetics , Karyotype , Male , Mesenchymal Stem Cells/metabolism , Mutation/genetics , Octamer Transcription Factor-3/genetics , Osteoclasts/pathology , Osteoclasts/transplantation , Osteopetrosis/genetics , Osteopetrosis/pathology , Osteopetrosis/therapy , Pluripotent Stem Cells/metabolism , Sorting Nexins/genetics , Stage-Specific Embryonic Antigens/genetics , Transplantation, Homologous/methods , Vacuolar Proton-Translocating ATPases/genetics
5.
Int J Mol Sci ; 20(5)2019 Mar 04.
Article in English | MEDLINE | ID: mdl-30836626

ABSTRACT

A deeper understanding of the detailed mechanism of in vivo tissue healing is necessary for the development of novel regenerative therapies. Among several external factors, environmental pH is one of the crucial parameters that greatly affects enzyme activity and cellular biochemical reactions involving tissue repair and homeostasis. In this study, in order to analyze the microenvironmental conditions during bone healing, we first measured the pH in vivo at the bone healing site using a high-resolution fiber optic pH microsensor directly in femur defects and tooth extraction sockets. The pH was shown to decrease from physiological 7.4 to 6.8 during the initial two days of healing (inflammatory phase). In the same initial stages of the inflammatory phase of the bone healing process, mesenchymal stem cells (MSCs) are known to migrate to the healing site to contribute to tissue repair. Therefore, we investigated the effect of a short-term acidic (pH 6.8) pre-treatment on the stemness of bone marrow-derived MSCs (BMSCs). Interestingly, the results showed that pre-treatment of BMSCs with acidic pH enhances the expression of stem cell markers (OCT-4, NANOG, SSEA-4), as well as cell viability and proliferation. On the other hand, acidic pH decreased BMSC migration ability. These results indicate that acidic pH during the initial stages of bone healing is important to enhance the stem cell properties of BMSCs. These findings may enable the development of novel methods for optimization of stem cell function towards tissue engineering or regenerative medicine.


Subject(s)
Acids/pharmacology , Bone Regeneration/genetics , Osteogenesis/drug effects , Tissue Engineering/methods , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Regeneration/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cellular Microenvironment/drug effects , Humans , Hydrogen-Ion Concentration/drug effects , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Nanog Homeobox Protein/genetics , Octamer Transcription Factor-3/genetics , Regenerative Medicine , Stage-Specific Embryonic Antigens/genetics , Stem Cells/cytology , Stem Cells/drug effects , Wound Healing/genetics
6.
Proc Natl Acad Sci U S A ; 116(9): 3518-3523, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30808745

ABSTRACT

The globo-series glycosphingolipids (GSLs) SSEA3, SSEA4, and Globo-H specifically expressed on cancer cells are found to correlate with tumor progression and metastasis, but the functional roles of these GSLs and the key enzyme ß1,3-galactosyltransferase V (ß3GalT5) that converts Gb4 to SSEA3 remain largely unclear. Here we show that the expression of ß3GalT5 significantly correlates with tumor progression and poor survival in patients, and the globo-series GSLs in breast cancer cells form a complex in membrane lipid raft with caveolin-1 (CAV1) and focal adhesion kinase (FAK) which then interact with AKT and receptor-interacting protein kinase (RIP), respectively. Knockdown of ß3GalT5 disrupts the complex and induces apoptosis through dissociation of RIP from the complex to interact with the Fas death domain (FADD) and trigger the Fas-dependent pathway. This finding provides a link between SSEA3/SSEA4/Globo-H and the FAK/CAV1/AKT/RIP complex in tumor progression and apoptosis and suggests a direction for the treatment of breast cancer, as demonstrated by the combined use of antibodies against Globo-H and SSEA4.


Subject(s)
Breast Neoplasms/genetics , Galactosyltransferases/genetics , Glycosphingolipids/genetics , Membrane Microdomains/genetics , Antigens, Tumor-Associated, Carbohydrate/genetics , Antigens, Tumor-Associated, Carbohydrate/metabolism , Apoptosis/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caveolin 1/genetics , Caveolin 1/metabolism , Disease Progression , Fas-Associated Death Domain Protein/genetics , Female , Focal Adhesion Protein-Tyrosine Kinases/genetics , Gene Expression Regulation, Neoplastic/genetics , Glycosphingolipids/metabolism , Humans , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Membrane Microdomains/metabolism , Middle Aged , Proto-Oncogene Proteins c-akt/genetics , Saporins/genetics , Signal Transduction/genetics , Stage-Specific Embryonic Antigens/genetics , Stage-Specific Embryonic Antigens/metabolism
7.
Cell Death Dis ; 9(5): 502, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29717132

ABSTRACT

The adult olfactory mucosa, a highly regenerative tissue with unique life-long neurogenesis ability, is thought to harbor a naïve yet tightly controlled stem cell population. It will provide unique benefits in various stem cell-based therapies, such as stroke treatment. Here, we identified a subpopulation of adult pluripotent-like olfactory stem cells (APOSCs), which were modulated by an epigenetic repressor of CBX7. APOSCs form a floating sphere, express pluripotency markers Nanog, Oct-4, Sox-2, and SSEA-4 and show alkaline phosphatase activity. In addition, APOSCs display self-renewal and a pluripotent potential to differentiate into all three germ layers. Moreover, APOSCs coexpress pluripotency markers with CBX7. Within their natural niche, APOSCs from CBX7+/+ mice responded promptly to either spontaneous or injury-induced tissue regeneration. However, APOSCs from CBX7-/- mice manifested an impaired self-renewal and differentiation potential. Similarly, in vitro-cultivated CBX7-/- APOSCs underwent premature senescence, whereas CBX7+/+ APOSCs still actively divided, indicating that CBX7 is required for the self-renewal of APOSCs. Intracerebral implantation of APOSCs improved the stroke-mediated neurological dysfunction in rodents. These findings indicate that CBX7 plays a critical role in the regenerative properties of APOSCs and indicate the safety and feasibility of implantation of autologous APOSCs in stroke treatment.


Subject(s)
Epigenesis, Genetic , Olfactory Mucosa/metabolism , Pluripotent Stem Cells/metabolism , Polycomb Repressive Complex 1/genetics , Stroke/genetics , Animals , Cell Differentiation , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Olfactory Mucosa/cytology , Pluripotent Stem Cells/cytology , Polycomb Repressive Complex 1/metabolism , Rats , Rats, Sprague-Dawley , Regeneration/genetics , Signal Transduction , Stage-Specific Embryonic Antigens/genetics , Stage-Specific Embryonic Antigens/metabolism , Stem Cell Transplantation , Stroke/metabolism , Stroke/pathology , Stroke/therapy , Transplantation, Autologous
8.
Sci Rep ; 8(1): 5907, 2018 04 12.
Article in English | MEDLINE | ID: mdl-29651156

ABSTRACT

Nonhuman primate (NHP) models are more predictive than rodent models for developing induced pluripotent stem cell (iPSC)-based cell therapy, but robust and reproducible NHP iPSC-cardiomyocyte differentiation protocols are lacking for cardiomyopathies research. We developed a method to differentiate integration-free rhesus macaque iPSCs (RhiPSCs) into cardiomyocytes with >85% purity in 10 days, using fully chemically defined conditions. To enable visualization of intracellular calcium flux in beating cardiomyocytes, we used CRISPR/Cas9 to stably knock-in genetically encoded calcium indicators at the rhesus AAVS1 safe harbor locus. Rhesus cardiomyocytes derived by our stepwise differentiation method express signature cardiac markers and show normal electrochemical coupling. They are responsive to cardiorelevant drugs and can be successfully engrafted in a mouse myocardial infarction model. Our approach provides a powerful tool for generation of NHP iPSC-derived cardiomyocytes amenable to utilization in basic research and preclinical studies, including in vivo tissue regeneration models and drug screening.


Subject(s)
Calcium/metabolism , Founder Effect , Induced Pluripotent Stem Cells/metabolism , Myocardial Infarction/therapy , Myocytes, Cardiac/metabolism , Animals , Biomarkers/metabolism , CRISPR-Cas Systems , Calcium/analysis , Cardiovascular Agents/pharmacology , Cell Differentiation , Cell Line , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Fluorescence , Gene Expression , Gene Knock-In Techniques , Genes, Reporter , Genetic Loci , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Macaca mulatta , Mice , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/transplantation , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Stage-Specific Embryonic Antigens/genetics , Stage-Specific Embryonic Antigens/metabolism , Transplantation, Heterologous
9.
Nat Commun ; 8(1): 2225, 2017 12 20.
Article in English | MEDLINE | ID: mdl-29263384

ABSTRACT

The power of human induced pluripotent stem cell (hiPSC)-based studies to resolve the smaller effects of common variants within the size of cohorts that can be realistically assembled remains uncertain. We identified and accounted for a variety of technical and biological sources of variation in a large case/control schizophrenia (SZ) hiPSC-derived cohort of neural progenitor cells and neurons. Reducing the stochastic effects of the differentiation process by correcting for cell type composition boosted the SZ signal and increased the concordance with post-mortem data sets. We predict a growing convergence between hiPSC and post-mortem studies as both approaches expand to larger cohort sizes. For studies of complex genetic disorders, to maximize the power of hiPSC cohorts currently feasible, in most cases and whenever possible, we recommend expanding the number of individuals even at the expense of the number of replicate hiPSC clones.


Subject(s)
Brain/metabolism , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/metabolism , Neurons/metabolism , RNA, Messenger/metabolism , Schizophrenia/genetics , Adolescent , Adult , Antigens, Surface/genetics , Autopsy , Case-Control Studies , Child , DNA Copy Number Variations , Female , Humans , Linear Models , Male , Nanog Homeobox Protein/genetics , Nestin/genetics , Octamer Transcription Factor-3/genetics , Proteoglycans/genetics , SOXB1 Transcription Factors/genetics , Sequence Analysis, RNA , Stage-Specific Embryonic Antigens/genetics , Synapsins/genetics , Transcriptome , Young Adult
10.
Stem Cell Res Ther ; 8(1): 239, 2017 Oct 27.
Article in English | MEDLINE | ID: mdl-29078802

ABSTRACT

BACKGROUND: Degenerative diseases are a major public health concern for the aging population and mesenchymal stem cells (MSCs) have great potential for treating many of these diseases. However, the quantity and quality of MSCs declines with aging, limiting the potential efficacy of autologous MSCs for treating the elderly population. METHODS: Human bone marrow (BM)-derived MSCs from young and elderly donors were obtained and characterized using standard cell surface marker criteria (CD73, CD90, CD105) as recommended by the International Society for Cellular Therapy (ISCT). The elderly MSC population was isolated into four subpopulations based on size and stage-specific embryonic antigen-4 (SSEA-4) expression using fluorescence-activated cell sorting (FACS), and subpopulations were compared to the unfractionated young and elderly MSCs using assays that evaluate MSC proliferation, quality, morphology, intracellular reactive oxygen species, ß-galactosidase expression, and adenosine triphosphate (ATP) content. RESULTS: The ISCT-recommended cell surface markers failed to detect any differences between young and elderly MSCs. Here, we report that elderly MSCs were larger in size and displayed substantially higher concentrations of intracellular reactive oxygen species and ß-galactosidase expression and lower amounts of ATP and SSEA-4 expression. Based on these findings, cell size and SSEA-4 expression were used to separate the elderly MSCs into four subpopulations by FACS. The original populations (young and elderly MSCs), as well as the four subpopulations, were then characterized before and after culture on tissue culture plastic and BM-derived extracellular matrix (BM-ECM). The small SSEA-4-positive subpopulation representing ~ 8% of the original elderly MSC population exhibited a "youthful" phenotype that was similar to that of young MSCs. The biological activity of this elderly subpopulation was inhibited by senescence-associated factors produced by the unfractionated parent population. After these "youthful" cells were isolated and expanded (three passages) on a "young microenvironment" (i.e., BM-ECM produced by BM cells from young donors), the number of cells increased ≈ 17,000-fold to 3 × 109 cells and retained their "youthful" phenotype. CONCLUSIONS: These results suggest that it is feasible to obtain large numbers of high-quality autologous MSCs from the elderly population and establish personal stem cell banks that will allow serial infusions of "rejuvenated" MSCs for treating age-related diseases.


Subject(s)
Aging/physiology , Cell Separation/methods , Extracellular Matrix/chemistry , Mesenchymal Stem Cells/cytology , Adenosine Triphosphate/metabolism , Aging/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Biomarkers/metabolism , Bone Marrow Cells/classification , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Proliferation , Cell Size , Cellular Senescence , Gene Expression , Humans , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/classification , Mesenchymal Stem Cells/metabolism , Primary Cell Culture , Reactive Oxygen Species/metabolism , Stage-Specific Embryonic Antigens/genetics , Stage-Specific Embryonic Antigens/metabolism , Transplantation, Autologous , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
11.
Stem Cell Rev Rep ; 13(5): 603-610, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28624968

ABSTRACT

One of the important questions when studying established cancer cell lines is whether such cells contain a subpopulation of primitive cancer stem cells that maintains the expansion of the cell line. To address this issue, we performed studies on the established human embryonal carcinoma cell line NTera2 by evaluating the potential stemness of cells sorted according to their expression of the cell surface stem cell markers CD133 and SSEA4. By performing in vitro and in vivo assays, we observed different properties of cells expressing both, one, or neither of these antigens. While sorted SSEA4+ subpopulations exhibited the greatest propensity for migration toward normal serum and the highest seeding efficiency in the lungs of immunodeficient mice, CD133-SSEA4- cells displayed high seeding efficiency to the bone marrow after injection in vivo. It is worth noting that these properties did not depend on the size of the evaluated cells. To address the question of whether cancer stem cell phenotypes in cell lines are fixed or fluctuating, we sorted single cells according to their expression of CD133 and SSEA4 antigens and observed that cells which did not express these cancer stem cell markers gave rise to cells that express these markers after expansion in vitro. Therefore, our results support the idea that within established cancer cell lines, the phenotype of the cell subpopulation expressing cancer stem cell markers is not fixed but fluctuates during cell line expansion, and cells negative for these markers may acquire their expression.


Subject(s)
AC133 Antigen/genetics , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/metabolism , Phenotype , Pluripotent Stem Cells/metabolism , Stage-Specific Embryonic Antigens/genetics , AC133 Antigen/immunology , AC133 Antigen/metabolism , Animals , Biomarkers, Tumor/immunology , Biomarkers, Tumor/metabolism , Bone Marrow/immunology , Bone Marrow/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Embryo, Mammalian , Flow Cytometry , Gene Expression Profiling , Genomic Imprinting , Humans , Lung/immunology , Lung/pathology , Mice , Mice, SCID , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Pluripotent Stem Cells/immunology , Pluripotent Stem Cells/pathology , Stage-Specific Embryonic Antigens/immunology , Stage-Specific Embryonic Antigens/metabolism
12.
Sci Rep ; 7: 42138, 2017 02 06.
Article in English | MEDLINE | ID: mdl-28165055

ABSTRACT

Human pluripotent stem cells (hPSCs) are adhesion-dependent cells that require cultivation in colonies to maintain growth and pluripotency. Robust differentiation protocols necessitate single cell cultures that are achieved by use of ROCK (Rho kinase) inhibitors. ROCK inhibition enables maintenance of stem cell phenotype; its effects on metabolism are unknown. hPSCs were exposed to 10 µM ROCK inhibitor for varying exposure times. Pluripotency (TRA-1-81, SSEA3, OCT4, NANOG, SOX2) remained unaffected, until after prolonged exposure (96 hrs). Gas chromatography-mass spectrometry metabolomics analysis identified differences between ROCK-treated and untreated cells as early as 12 hrs. Exposure for 48 hours resulted in reduction in glycolysis, glutaminolysis, the citric acid (TCA) cycle as well as the amino acids pools, suggesting the adaptation of the cells to the new culture conditions, which was also reflected by the expression of the metabolic regulators, mTORC1 and tp53 and correlated with cellular proliferation status. While gene expression and protein levels did not reveal any changes in the physiology of the cells, metabolomics revealed the fluctuating state of the metabolism. The above highlight the usefulness of metabolomics in providing accurate and sensitive information on cellular physiological status, which could lead to the development of robust and optimal stem cell bioprocesses.


Subject(s)
Amides/pharmacology , Embryonic Stem Cells/drug effects , Enzyme Inhibitors/pharmacology , Induced Pluripotent Stem Cells/drug effects , Metabolome , Pyridines/pharmacology , rho-Associated Kinases/genetics , Antigens, Surface/genetics , Antigens, Surface/metabolism , Antigens, Tumor-Associated, Carbohydrate/genetics , Antigens, Tumor-Associated, Carbohydrate/metabolism , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Phenotype , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Stage-Specific Embryonic Antigens/genetics , Stage-Specific Embryonic Antigens/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
13.
Glycoconj J ; 34(6): 693-699, 2017 12.
Article in English | MEDLINE | ID: mdl-27318475

ABSTRACT

A variety of glycoconjugates, including glycosphingolipids (GSLs), expressed in mammalian tissues and cells were isolated and characterized in early biochemical studies. Later studies of virus-transformed fibroblasts demonstrated the association of GSL expression profiles with cell phenotypes. Changes of GSL expression profile were observed during mammalian embryogenesis. Cell surface molecules expressed on embryos in a stage-specific manner appeared to play key roles in regulation of cell-cell interaction and cell sorting during early development. Many mAbs showing stage-specific reactivity with mouse embryos were shown to recognize carbohydrate epitopes. Among various stage-specific embryonic antigens (SSEAs), SSEA-1 was found to react with neolacto-series GSL Lex, while SSEA-3 and SSEA-4 reacted with globo-series Gb5 and monosialyl-Gb5, respectively. GSL expression during mouse early development was shown to shift rapidly from globo-series to neolacto/lacto-series, and then to ganglio-series. We found that multivalent Lex caused decompaction of mouse embryos, indicating a functional role of Lex epitope in the compaction process. Autoaggregation of mouse embryonal carcinoma (EC) F9 cells provided a useful model of the compaction process. We showed that Lex-Lex interaction, a novel type of molecular interavction termed carbohydrate-carbohydrate interaction (CCI), was involved in cell aggregation. Similar shifting of GSL expression profiles from globo-series and neolacto/lacto-series to ganglio-series was observed during differentiation of human EC cells and embryonic stem (ES) cells, reflecting the essential role of cell surface glycoconjugates in early development.


Subject(s)
Embryonic Development , Glycosphingolipids/metabolism , Stage-Specific Embryonic Antigens/metabolism , Animals , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Glycosphingolipids/genetics , Humans , Stage-Specific Embryonic Antigens/genetics
14.
Sci Rep ; 6: 37004, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27845370

ABSTRACT

Oral submucous fibrosis (OSF), regarded as a precancerous condition, is characterized by juxta-epithelial inflammatory reaction followed by fibro-elastic change in the lamina properia and epithelial atrophy. The pathologic mechanisms of OSF still need to be further clarified. In the study, we investigated the functional expression of SSEA-4, which is a well-known stemness marker, in myofibroblast activity and the clinical significance in OSF tissues. The expression of SSEA-4 in OSF was evaluated by immunohistochemical staining. Functional analysis of SSEA-4 on myofibroblast activity of OSF was achieved by lentiviral silencing ST3GAL2. Immunohisitochemistry demonstrated that SSEA-4 expression was significantly higher expression in areca quid chewing-associated OSF tissues than those of normal oral mucosa tissues. From flow cytometry analysis, arecoline dose-dependently activated SSEA-4 expression in primary human normal buccal mucosal fibroblasts (BMFs). Sorted SSEA-4-positive cells from fibrotic BMFs (fBMFs) have higher colony-forming unit, collagen gel contraction, and α-smooth muscle actin (α-SMA) expression than SSEA-4-negative subset. Knockdown of ST3GAL2 in fBMFs suppressed SSEA-4 expression, collagen contraction, migration, invasiveness, and wound healing capability. Consistently, silencing ST3GAL2 was found to repress arecoline-induced myofibroblast activity in BMFs. The study highlights SSEA-4 as a critical marker for therapeutic intervention to mediate myofibroblast transdifferentiation in areca quid chewing-associated OSF.


Subject(s)
Biomarkers, Tumor , Gene Expression Regulation, Neoplastic , Mouth Neoplasms , Myofibroblasts , Precancerous Conditions , Stage-Specific Embryonic Antigens , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Female , Humans , Male , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Stage-Specific Embryonic Antigens/biosynthesis , Stage-Specific Embryonic Antigens/genetics
15.
Stem Cell Res Ther ; 7(1): 83, 2016 Jun 13.
Article in English | MEDLINE | ID: mdl-27296060

ABSTRACT

BACKGROUND: In regenerative medicine the maintenance of stem cell properties is of crucial importance. Ageing is considered a cause of reduced stemness capability. The limbus is a stem niche of easy access and harbors two stem cell populations: epithelial stem cells and fibroblast-like stem cells. Our aim was to investigate whether donor age and/or long-term culture have any influence on stem cell marker expression and the profiles in the fibroblast-like stem cell population. METHODS: Fibroblast-like stem cells were isolated and digested from 25 limbus samples of normal human corneo-scleral rings and long-term cultures were obtained. SSEA4 expression and sphere-forming capability were evaluated; cytofluorimetric assay was performed to detect the immunophenotypes HLA-DR, CD45, and CD34 and the principle stem cell markers ABCG2, OCT3/4, and NANOG. Molecular expression of the principal mesenchymal stem cell genes was investigated by real-time PCR. Two-dimensional gel electrophoresis and mass spectrometric sequencing were performed and a stable proteomic profile was identified. The proteins detected were explored by gene ontology and STRING analysis. The data were reported as means ± SD, compared by Student's unpaired t test and considering p < 0.05 as statistically significant. RESULTS: The isolated cells did not display any hematopoietic surface marker (CD34 and CD45) and HLA-DR and they maintained these features in long-term culture. The expression of the stemness genes and the multilineage differentiation under in-vitro culture conditions proved to be well maintained. Proteomic analysis revealed a fibroblast-like stem cell profile of 164 proteins with higher expression levels. Eighty of these showed stable expression levels and were involved in maintenance of "the stem gene profile"; 84 were differentially expressed and were involved in structural activity. CONCLUSIONS: The fibroblast-like limbal stem cells confirmed that they are a robust source of adult stem cells and that they have good plasticity, good proliferative capability, and long-term maintenance of stem cell properties, independently of donor age and long-term culture conditions. Our findings confirm that limbal fibroblast-like stem cells are highly promising for application in regenerative medicine and that in-vitro culture steps do not influence their stem cell properties. Moreover, the proteomic data enrich our knowledge of fibroblast-like stem cells.


Subject(s)
Epithelial Cells/cytology , Epithelium, Corneal/cytology , Fibroblasts/cytology , Limbus Corneae/cytology , Stem Cells/cytology , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Adult , Age Factors , Aged , Biomarkers/metabolism , Cell Differentiation , Cell Movement , Cell Proliferation , Epithelial Cells/metabolism , Epithelium, Corneal/metabolism , Female , Fibroblasts/metabolism , Gene Expression , HLA-DR Antigens/genetics , HLA-DR Antigens/metabolism , Humans , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Limbus Corneae/metabolism , Male , Middle Aged , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Primary Cell Culture , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Stage-Specific Embryonic Antigens/genetics , Stage-Specific Embryonic Antigens/metabolism , Stem Cells/metabolism
16.
Mol Biotechnol ; 58(6): 415-27, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27126695

ABSTRACT

To find out differences and similarities in phenotypic, proliferative, and trans-differentiation properties of stem cells isolated from pulp of deciduous (SHEDs) and permanent (DPSCs) teeth with human bone marrow stem cells (BMSCs), we examined the expression of mesenchymal and embryonic stem cell markers in relation to the proliferation and osteogenic differentiation potentials of these cells. In this way, after isolating SHEDs, DPSCs, and BMSCs, cell proliferation was evaluated and population doubling time was calculated accordingly. Expression patterns of mesenchymal, hematopoietic, and embryonic stem cell markers were assessed followed by examining differentiation potential toward osseous tissue through alizarin red staining and qRT-PCR. Based on the results, the proliferation rates of SHEDs and DPSCs were significantly higher than that of BMSCs (P < 0.0001). High expression of mesenchymal stem cell markers and weak expression of hematopoietic markers were observed in all the three groups. The mean expression of OCT-4 was significantly higher in SHEDs and DPSCs (P = 0.028), while the expression of SSEA-4 was lower (P = 0.006) compared to BMSCs. Osteogenic differentiation potential of SHEDs was greater than DPSCs; however, it was lower than that of BMSCs. Conclusively, the distinctive immunophenotyping, proliferation rate, and differentiation pattern of SHEDs and DPSCs discriminate these cells from BMSCs. Furthermore, dissimilarity in differentiation potential is evidence implying that SHEDs might be more primitive stem cell population compared to DPSCs.


Subject(s)
Dental Pulp/cytology , Mesenchymal Stem Cells/cytology , Osteogenesis , Stem Cells/cytology , Stem Cells/immunology , Tooth, Deciduous/cytology , Adult , Biomarkers/analysis , Cell Differentiation , Cell Proliferation , Cells, Cultured , Child , Dentition, Permanent , Gene Expression Regulation , Humans , Immunophenotyping , Octamer Transcription Factor-3/genetics , Stage-Specific Embryonic Antigens/genetics , Young Adult
17.
J Cell Mol Med ; 20(4): 721-30, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26849468

ABSTRACT

The long-term propagation of basal prostate progenitor cells ex vivo has been very difficult in the past. The development of novel methods to expand prostate progenitor cells in vitro allows determining their cell surface phenotype in greater detail. Mouse (Lin(-)Sca-1(+) CD49f(+) Trop2(high)-phenotype) and human (Lin(-) CD49f(+) TROP2(high)) basal prostate progenitor cells were expanded in vitro. Human and mouse cells were screened using 242 anti-human or 176 antimouse monoclonal antibodies recognizing the cell surface protein profile. Quantitative expression was evaluated at the single-cell level using flow cytometry. Differentially expressed cell surface proteins were evaluated in conjunction with the known CD49f(+)/TROP2(high) phenotype of basal prostate progenitor cells and characterized by in vivo sandwich-transplantation experiments using nude mice. The phenotype of basal prostate progenitor cells was determined as CD9(+)/CD24(+)/CD29(+)/CD44(+)/CD47(+)/CD49f(+)/CD104(+)/CD147(+)/CD326(+)/Trop2(high) of mouse as well as human origin. Our analysis revealed several proteins, such as CD13, Syndecan-1 and stage-specific embryonal antigens (SSEAs), as being differentially expressed on murine and human CD49f(+) TROP2(+) basal prostate progenitor cells. Transplantation experiments suggest that CD49f(+) TROP2(high) SSEA-4(high) human prostate basal progenitor cells to be more potent to regenerate prostate tubules in vivo as compared with CD49f(+) TROP2(high) or CD49f(+) TROP2(high) SSEA-4(low) cells. Determination of the cell surface protein profile of functionally defined murine and human basal prostate progenitor cells reveals differentially expressed proteins that may change the potency and regenerative function of epithelial progenitor cells within the prostate. SSEA-4 is a candidate cell surface marker that putatively enables a more accurate identification of the basal PESC lineage.


Subject(s)
Epithelial Cells/transplantation , Regeneration/genetics , Stage-Specific Embryonic Antigens/genetics , Stem Cell Transplantation , Stem Cells/cytology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Proliferation , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Male , Mice , Mice, Nude , Prostate/surgery , Signal Transduction , Stage-Specific Embryonic Antigens/metabolism , Stem Cells/metabolism , Syndecan-1/genetics , Syndecan-1/metabolism
18.
J Korean Med Sci ; 31(2): 171-7, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26839468

ABSTRACT

We investigated the potential of human dental pulp stem cells (hDPSCs) to differentiate into dopaminergic neurons in vitro as an autologous stem cell source for Parkinson's disease treatment. The hDPSCs were expanded in knockout-embryonic stem cell (KO-ES) medium containing leukemia inhibitory factor (LIF) on gelatin-coated plates for 3-4 days. Then, the medium was replaced with KO-ES medium without LIF to allow the formation of the neurosphere for 4 days. The neurosphere was transferred into ITS medium, containing ITS (human insulin-transferrin-sodium) and fibronectin, to select for Nestin-positive cells for 6-8 days. The cells were then cultured in N-2 medium containing basic fibroblast growth factor (FGF), FGF-8b, sonic hedgehog-N, and ascorbic acid on poly-l-ornithine/fibronectin-coated plates to expand the Nestin-positive cells for up to 2 weeks. Finally, the cells were transferred into N-2/ascorbic acid medium to allow for their differentiation into dopaminergic neurons for 10-15 days. The differentiation stages were confirmed by morphological, immunocytochemical, flow cytometric, real-time PCR, and ELISA analyses. The expressions of mesenchymal stem cell markers were observed at the early stages. The expressions of early neuronal markers were maintained throughout the differentiation stages. The mature neural markers showed increased expression from stage 3 onwards. The percentage of cells positive for tyrosine hydroxylase was 14.49%, and the amount was 0.526 ± 0.033 ng/mL at the last stage. hDPSCs can differentiate into dopaminergic neural cells under experimental cell differentiation conditions, showing potential as an autologous cell source for the treatment of Parkinson's disease.


Subject(s)
Cell Differentiation , Dental Pulp/cytology , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Brain/pathology , Cell Differentiation/drug effects , Cells, Cultured , Culture Media/chemistry , Culture Media/pharmacology , Dopaminergic Neurons/pathology , Enzyme-Linked Immunosorbent Assay , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Humans , Mice , Mice, Inbred ICR , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism , Real-Time Polymerase Chain Reaction , Stage-Specific Embryonic Antigens/genetics , Stage-Specific Embryonic Antigens/metabolism , Stem Cells/pathology , Tubulin/genetics , Tubulin/metabolism , Tyrosine 3-Monooxygenase/analysis , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
19.
Cell Cycle ; 15(2): 234-49, 2016.
Article in English | MEDLINE | ID: mdl-26654216

ABSTRACT

Amniotic fluid stem cells (AFSC) represent an attractive potential cell source for fetal and pediatric cell-based therapies. However, upgrading them to pluripotency confers refractoriness toward senescence, higher proliferation rate and unlimited differentiation potential. AFSC were observed to rapidly and efficiently reacquire pluripotency which together with their easy recovery makes them an attractive cell source for reprogramming. The reprogramming process as well as the resulting iPSC epigenome could potentially benefit from the unspecialized nature of AFSC. iPSC derived from AFSC also have potential in disease modeling, such as Down syndrome or ß-thalassemia. Previous experiments involving AFSC reprogramming have largely relied on integrative vector transgene delivery and undefined serum-containing, feeder-dependent culture. Here, we describe non-integrative oriP/EBNA-1 episomal plasmid-based reprogramming of AFSC into iPSC and culture in fully chemically defined xeno-free conditions represented by vitronectin coating and E8 medium, a system that we found uniquely suited for this purpose. The derived AF-iPSC lines uniformly expressed a set of pluripotency markers Oct3/4, Nanog, Sox2, SSEA-1, SSEA-4, TRA-1-60, TRA-1-81 in a pattern typical for human primed PSC. Additionally, the cells formed teratomas, and were deemed pluripotent by PluriTest, a global expression microarray-based in-silico pluripotency assay. However, we found that the PluriTest scores were borderline, indicating a unique pluripotent signature in the defined condition. In the light of potential future clinical translation of iPSC technology, non-integrating reprogramming and chemically defined culture are more acceptable.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , Plasmids/chemistry , Transfection/methods , Amniotic Fluid/cytology , Amniotic Fluid/drug effects , Antigens, Surface/genetics , Antigens, Surface/metabolism , Biomarkers/metabolism , Cell Culture Techniques , Cell Differentiation , Cell Proliferation , Culture Media/pharmacology , Gene Expression , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Lewis X Antigen/genetics , Lewis X Antigen/metabolism , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Plasmids/metabolism , Protein Array Analysis , Proteoglycans/genetics , Proteoglycans/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Stage-Specific Embryonic Antigens/genetics , Stage-Specific Embryonic Antigens/metabolism
20.
Article in English | WPRIM (Western Pacific) | ID: wpr-133743

ABSTRACT

We investigated the potential of human dental pulp stem cells (hDPSCs) to differentiate into dopaminergic neurons in vitro as an autologous stem cell source for Parkinson's disease treatment. The hDPSCs were expanded in knockout-embryonic stem cell (KO-ES) medium containing leukemia inhibitory factor (LIF) on gelatin-coated plates for 3-4 days. Then, the medium was replaced with KO-ES medium without LIF to allow the formation of the neurosphere for 4 days. The neurosphere was transferred into ITS medium, containing ITS (human insulin-transferrin-sodium) and fibronectin, to select for Nestin-positive cells for 6-8 days. The cells were then cultured in N-2 medium containing basic fibroblast growth factor (FGF), FGF-8b, sonic hedgehog-N, and ascorbic acid on poly-l-ornithine/fibronectin-coated plates to expand the Nestin-positive cells for up to 2 weeks. Finally, the cells were transferred into N-2/ascorbic acid medium to allow for their differentiation into dopaminergic neurons for 10-15 days. The differentiation stages were confirmed by morphological, immunocytochemical, flow cytometric, real-time PCR, and ELISA analyses. The expressions of mesenchymal stem cell markers were observed at the early stages. The expressions of early neuronal markers were maintained throughout the differentiation stages. The mature neural markers showed increased expression from stage 3 onwards. The percentage of cells positive for tyrosine hydroxylase was 14.49%, and the amount was 0.526 ± 0.033 ng/mL at the last stage. hDPSCs can differentiate into dopaminergic neural cells under experimental cell differentiation conditions, showing potential as an autologous cell source for the treatment of Parkinson's disease.


Subject(s)
Animals , Humans , Mice , Brain/pathology , Cell Differentiation/drug effects , Cells, Cultured , Culture Media/chemistry , Dental Pulp/cytology , Dopaminergic Neurons/cytology , Enzyme-Linked Immunosorbent Assay , Glial Fibrillary Acidic Protein/genetics , Mice, Inbred ICR , Myelin Basic Protein/genetics , Real-Time Polymerase Chain Reaction , Stage-Specific Embryonic Antigens/genetics , Stem Cells/cytology , Tubulin/genetics , Tyrosine 3-Monooxygenase/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...