Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 193
Filter
1.
Sci Rep ; 14(1): 11935, 2024 05 24.
Article in English | MEDLINE | ID: mdl-38789477

ABSTRACT

Carbohydrate markers of immature cells during prenatal human development can be aberrantly expressed in cancers and deserve evaluation as immune targets. A candidate target in Ewing sarcoma is the globo-series ganglioside stage-specific embryonic antigen-4 (SSEA-4). We detected SSEA-4 expression on the cell surface of all of 14 EwS cell lines and in 21 of 31 (68%) primary EwS tumor biopsies. Among paired subpopulations of tumor cells with low versus high SSEA-4 expression, SSEA-4high expression was significantly and consistently associated with functional characteristics of tumor aggressiveness, including higher cell proliferation, colony formation, chemoresistance and propensity to migrate. SSEA-4low versus SSEA-4high expression was not related to expression levels of the EWSR1-FLI1 fusion transcript or markers of epithelial/mesenchymal plasticity. SSEA-4low cells selected from bulk populations regained higher SSEA-4 expression in vitro and during in vivo tumor growth in a murine xenograft model. T cells engineered to express SSEA-4-specific chimeric antigen receptors (CARs) specifically interacted with SSEA-4 positive EwS cells and exerted effective antigen-specific tumor cell lysis in vitro. In conclusion, with its stable expression and functional significance in EwS, SSEA-4 is an attractive therapeutic immune target in this cancer that deserves further evaluation for clinical translation.


Subject(s)
Sarcoma, Ewing , Stage-Specific Embryonic Antigens , Humans , Sarcoma, Ewing/pathology , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/genetics , Stage-Specific Embryonic Antigens/metabolism , Animals , Mice , Cell Line, Tumor , Cell Proliferation , Female , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Xenograft Model Antitumor Assays
2.
Neurosci Res ; 203: 42-50, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38154662

ABSTRACT

Numerous evidences showed that human umbilical cord blood (UCB) mononuclear cells were a promising approach for the therapy of ischemic stroke(IS). The effect of stage-specific embryonic antigen 3 (SSEA3)positive subpopulation in UCB was not investigated in IS. In this study, we isolated SSEA3 positive cells from healthy UCB mononuclear cells, which comprised about 7.01% of the total UCB-mononuclear cells. Flow cytometry analysis revealed that SSEA3(+)UCB cells were almost positive for CD44 and CD45, and negative for CD73, CD90 and CD105. The expression of Oct3/4 in SSEA3(+)UCB cells were higher than that in UCB. SSEA3(+)UCB cells sorted by magnetic cell sorting were intravenously injected into the middle cerebral arterial occlusion(MCAO) rat model. Neurological score showed that SSEA3(+)UCB transplantation group exhibited significant improvements in the functional outcome of MCAO rats than UCB transplantation group. Nissl staining and microtubule association protein-2(MAP2) immunofluorescence staining showed that the SSEA3(+)UCB transplantation group decreased neuronal loss. SSEA3(+)UCB transplantation group reduced neuronal apoptosis, inhibited caspase3 expression, and decreased tumor necrosis factor α(TNF-α). These results indicate that SSEA3 positive cells are a novel subpopulation of UCB cells, which exhibit great potential for the treatment of ischemic stroke.


Subject(s)
Disease Models, Animal , Fetal Blood , Ischemic Stroke , Animals , Humans , Ischemic Stroke/therapy , Ischemic Stroke/metabolism , Fetal Blood/cytology , Leukocytes, Mononuclear/transplantation , Leukocytes, Mononuclear/metabolism , Male , Rats, Sprague-Dawley , Rats , Infarction, Middle Cerebral Artery/therapy , Cord Blood Stem Cell Transplantation/methods , Stage-Specific Embryonic Antigens/metabolism , Brain Ischemia/therapy , Apoptosis/physiology
3.
Cell Prolif ; 56(1): e13345, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36225120

ABSTRACT

OBJECTIVES: Multilineage differentiating Stress Enduring (MUSE) cells are endogenous, stress-resistant stem cells, expressing pluripotency master genes and able to differentiate in cells of the three embryonic sheets. Stage-Specific Embryonic Antigen 3 (SSEA-3), a glycosphingolipid (GSL), is the marker for identifying MUSE cells and is used to isolate this population from mesenchymal stromal cells. GSLs modulate signal transduction by interacting with plasma membrane components. The growth factor FGF2, important for MUSE cells biology, may interact with GSLs. Specific cell surface markers represent an invaluable tool for stem cell isolation. Nonetheless their role, if any, in stem cell biology is poorly investigated. Functions of stem cells, however, depend on niche external cues, which reach cells through surface markers. We addressed the role of SSEA-3 in MUSE cell behaviour, trying to define whether SSEA-3 is just a marker or if it plays a functional role in this cell population by determining if it has any relationship with FGF2 activity. RESULTS: We evidenced how the SSEA-3 and FGF2 cooperation affected the self-renewal and clonogenic capacity of MUSE cells. The block of SSEA-3 significantly reduced the multilineage potential of MUSE cells with production of nullipotent clones. CONCLUSIONS: We contributed to dissecting the mechanisms underlying MUSE cell properties for establishing successful stem-cell-based therapies and the promotion of MUSE cells as a tool for the in vitro disease model.


Subject(s)
Alprostadil , Fibroblast Growth Factor 2 , Cell Differentiation , Stage-Specific Embryonic Antigens/metabolism
4.
ACS Chem Biol ; 16(8): 1526-1537, 2021 08 20.
Article in English | MEDLINE | ID: mdl-34369155

ABSTRACT

The globo-series glycosphingolipids (SSEA3, SSEA4, and Globo H) were shown to express in many cancers selectively, and a combination of anti-SSEA4 and anti-Globo H antibodies was able to suppress tumor growth in mice inoculated with breast cancer cell lines. To further understand the effect, we focused on the combined effect of the two antibodies in target binding and antibody-dependent cellular cytotoxicity (ADCC) in vitro. Here, we report that the binding of anti-Globo H antibody (VK9) to MDA-MB231 breast cancer cells was influenced by anti-SSEA4 antibody (MC813-70), and a combination of both antibodies induced a similar effect as did anti-SSEA4 antibodies alone in a reporter-based ADCC assay, indicating that SSEA4 is a major target in breast cancer due to its higher expression than Globo H. Furthermore, we showed that a homogeneous anti-SSEA4 antibody (chMC813-70-SCT) designed to maximize the ADCC activity can be used to isolate a subpopulation of natural killer (NK) cells that exhibit an ∼23% increase in killing the target cells as compared to the unseparated NK cells. These findings can be used to predict a therapy outcome based on the expression levels of antigens and evaluate therapeutic antibody development.


Subject(s)
Antibodies/immunology , Antigens, Tumor-Associated, Carbohydrate/immunology , Breast Neoplasms/metabolism , Stage-Specific Embryonic Antigens/immunology , Animals , Antigens, Tumor-Associated, Carbohydrate/metabolism , Cell Line, Tumor , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Receptors, IgG/metabolism , Stage-Specific Embryonic Antigens/metabolism
5.
Anticancer Res ; 41(7): 3327-3335, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34230128

ABSTRACT

BACKGROUND/AIM: Stage-specific embryonic antigen (SSEA)-4 plays important roles in the malignant aggressiveness of various cancers. The aim of this study was to investigate the pathological characteristics of SSEA-4 in castration-resistant prostate cancer (CRPC). MATERIALS AND METHODS: SSEA-4 expression and its pathological roles were evaluated in five prostate cancer (PC) cell lines and 27 CRPC tissues. The relationship between SSEA-4 and the androgen receptor (AR) was also examined. RESULTS: SSEA-4 expression was detected in AR-negative cells (PC3, DU145, and AICaP1) but was not detected in AR-positive cells (LNCaP and AICaP2). SSEA-4 expression in human CRPC tissues was significantly higher than that in locally advanced or metastatic hormone sensitive PC (HSPC) tissues. A negative correlation was also detected between SSEA-4 and AR in CRPC tissues but not in HSPC tissues. CONCLUSION: SSEA-4 was over-expressed in CRPC and the changes were mediated by complex mechanisms that related to the AR and hormonal therapy.


Subject(s)
Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/metabolism , Stage-Specific Embryonic Antigens/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/physiology , Humans , Male , PC-3 Cells
6.
Stem Cells Dev ; 30(5): 227-233, 2021 03.
Article in English | MEDLINE | ID: mdl-33397195

ABSTRACT

Systemic lupus erythematosus (SLE) is a chronic inflammatory autoimmune disease characterized by the production of multiple autoimmune antibodies and potentially involves any organ or tissue with a broad range of clinical manifestations. Conventional therapy still utilizes glucocorticoids and immunosuppressants. However, some patients show inadequate responses to glucocorticoids and immunosuppression, which may induce secondary immune dysfunction and severe infection as well as lead to an increased tumor risk. The lack of in vitro models has hampered progress in understanding and treating SLE. Patient-derived induced pluripotent stem cells (iPSCs) may provide a unique opportunity for modeling in vitro diseases as well as a platform for drug screening in individual patients. We isolated peripheral blood mononuclear cells from blood to explore the establishment of an in vitro model platform for SLE and directly purified CD34+ cells and seeded them for expansion. CD34+ cells were forced to express seven pluripotency factors, OCT4, SOX2, NANOG, LIN28, c-MYC, KLF4, and SV40LT, through transduction in lentiviral vectors. The morphological characteristics of induced pluripotent stem-like cells, such as prominent nucleoli and a high nucleus-to-cytoplasm ratio, were observed. The pluripotency of established SLE patient-derived iPSCs was confirmed by the expression of embryonic stem cell (ESC) markers and the ability of cells to differentiate into multiple cell lines. SLE patient-derived iPSCs exhibited human ESC properties, including morphology; growth characteristics; expression of pluripotency, genes, and surface markers; and teratoma formation. In conclusion, we generated SLE patient-derived iPSCs and validated their pluripotency. This study is a first but critical step that can provide a model platform for research aimed at understanding the SLE mechanism, which may lead to the discovery of new targets or compounds for the treatment of this disease.


Subject(s)
Antigens, CD34/metabolism , Gene Expression Regulation , Induced Pluripotent Stem Cells/metabolism , Leukocytes, Mononuclear/metabolism , Lupus Erythematosus, Systemic/blood , Adult , Antigens, Surface/metabolism , Cells, Cultured , Female , Flow Cytometry , Humans , Induced Pluripotent Stem Cells/cytology , Karyotyping , Leukocytes, Mononuclear/cytology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Nanog Homeobox Protein/genetics , Octamer Transcription Factor-3/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stage-Specific Embryonic Antigens/metabolism
7.
Methods Mol Biol ; 2239: 235-249, 2021.
Article in English | MEDLINE | ID: mdl-33226623

ABSTRACT

The pluripotency of human induced pluripotent stem cells (HiPSCs) cannot be tested strictly in a similar way as we can do for the mouse ones because of ethical restrictions. One common and initial approach to prove the pluripotency of an established human iPSC line is to demonstrate expression of a set of established surface and intracellular pluripotency markers. This chapter provides procedures of immunocytochemistry of the established HiPSC lines for a set of the signature intracellular pluripotency proteins, OCT4, SOX2, NANOG, and LIN28. We also describe cell phenotyping by flow cytometry for the five established human pluripotency surface markers, SSEA3, SSEA4, TRA-1-60, TRA-1-81, and TRA2-49 (ALP). Numbers of ALP+ and TRA-1-60+ colonies are the most widely used parameters for evaluation of human iPSC reprogramming efficiency. Therefore, this chapter also provides detailed steps for substrate colorimetric reaction of the ALP activity, as well as the TRA-1-60 staining, of the iPSC colonies in the reprogramming population.


Subject(s)
Cellular Reprogramming , Immunophenotyping/methods , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Transcription Factors/metabolism , Alkaline Phosphatase/metabolism , Antigens, Surface/metabolism , Antigens, Tumor-Associated, Carbohydrate/metabolism , Biomarkers/metabolism , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Flow Cytometry , Humans , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/metabolism , Proteoglycans/metabolism , RNA-Binding Proteins/metabolism , SOXB1 Transcription Factors/metabolism , Stage-Specific Embryonic Antigens/metabolism
8.
Proc Natl Acad Sci U S A ; 117(44): 27435-27444, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33087559

ABSTRACT

Conversion of human pluripotent stem cells from primed to naïve state is accompanied by altered transcriptome and methylome, but glycosphingolipid (GSL) profiles in naïve human embryonic stem cells (hESCs) have not been systematically characterized. Here we showed a switch from globo-(SSEA-3, SSEA-4, and Globo H) and lacto-series (fucosyl-Lc4Cer) to neolacto-series GSLs (SSEA-1 and H type 2 antigen), along with marked down-regulation of ß-1,3-galactosyltransferase (B3GALT5) upon conversion to naïve state. CRISPR/Cas9-generated B3GALT5-knockout (KO) hESCs displayed an altered GSL profile, increased cloning efficiency and intracellular Ca2+, reminiscent of the naïve state, while retaining differentiation ability. The altered GSLs could be rescued through overexpression of B3GALT5. B3GALT5-KO cells cultured with 2iLAF exhibited naïve-like transcriptome, global DNA hypomethylation, and X-chromosome reactivation. In addition, B3GALT5-KO rendered hESCs more resistant to calcium chelator in blocking entry into naïve state. Thus, loss of B3GALT5 induces a distinctive state of hESCs displaying unique GSL profiling with expression of neolacto-glycans, increased Ca2+, and conducive for transition to naïve pluripotency.


Subject(s)
Cell Differentiation , Galactosyltransferases/metabolism , Glycosphingolipids/metabolism , Pluripotent Stem Cells/metabolism , Stage-Specific Embryonic Antigens/metabolism , CRISPR-Cas Systems/genetics , Cell Line , Embryonic Stem Cells , Galactosyltransferases/genetics , Gene Knockdown Techniques , Humans
9.
J Investig Dermatol Symp Proc ; 20(1): S16-S21, 2020 11.
Article in English | MEDLINE | ID: mdl-33099378

ABSTRACT

Alopecia areata (AA), which is defined as an autoimmune hair loss disease, has a serious impact on the quality of life for patients with AA worldwide. In this study, to our knowledge, a previously unreported method of AA induction in C3H mice has been established and validated. Using this method, we showed that dermal injection of 1-3 million of a mixture of skin cells freshly isolated from AA-affected skin induces AA in more than 80% of healthy mice. Contrary to the previous protocol, the induction of AA by this approach does not need any surgical AA skin grafting, cell manipulation, or high number of activated T cells. We also showed that dermal injection of adherent myeloid cells (mainly CD11b+) in healthy mice is as potent as a mixture of none adherent CD3+ T cells and CD19+ B cells in the induction of AA. Interestingly, most of the mice (7 out of 8) that received non-adherent cells developed AA universalis, whereas most of the mice (5 out of 7) that received adherent cells developed patchy AA. Finally, we found a high number of stage-specific embryonic antigen-expressing cells whose expression in monocytes in an inflammatory disease causes the release of inflammatory cytokines, TNF-α and IL-1ß, from these cells in AA-affected skin.


Subject(s)
Alopecia Areata/metabolism , Alopecia Areata/pathology , Myeloid Cells/metabolism , Myeloid Cells/transplantation , Animals , Antigens, Tumor-Associated, Carbohydrate/metabolism , CD11b Antigen/metabolism , Cell Adhesion , Cells, Cultured , Disease Models, Animal , Female , Lewis X Antigen/metabolism , Mice , Mice, Inbred C3H , Stage-Specific Embryonic Antigens/metabolism
10.
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
11.
Andrology ; 8(6): 1923-1934, 2020 11.
Article in English | MEDLINE | ID: mdl-32691968

ABSTRACT

BACKGROUND: Spermatogonial stem cells (SSCs), as tissue-specific stem cells, are capable of both self-renewal and differentiation and supporting the continual and robust spermatogenesis for male fertility. As a rare sub-fraction of undifferentiated spermatogonia, SSCs share most molecular markers with the progenitor spermatogonia. Thus, the heterogeneity of the progenitor cells often obscures the characteristics of stem cells. Distinguishing SSCs from the progenitors is of paramount importance to understand the regulatory mechanisms governing their actions. OBJECTIVES: The present study was designed to reveal that SSEA4 can be a marker for putative porcine SSCs that distinguished it from the progenitors and to build a sorting program for efficient enrichment of porcine SSCs. METHODS: To explore expression of SSEA4 within the undifferentiated spermatogonial population, we performed co-immunofluorescent staining for SSEA4 and common molecular markers (VASA, DBA, PLZF, c-KIT, and SOX9) in the 7-, 90-, and 150-day-old porcine testicular tissues. SSEA4-positive cells were isolated from the 90-day-old porcine testes by flow cytometry. Immunofluorescent, RNA-sequencing, and transplantation analysis were used to reveal that SSEA4-positive fraction holds the stem cell capacity. RESULTS: We found that SSEA4 was expressed in a rare sub-fraction of porcine undifferentiated spermatogonia, and RNA-sequencing analysis revealed that the genes for stem cell maintenance and SSC-specific markers (ID4 and PAX7) were up-regulated in the SSEA4-sorted fraction, compared with undifferentiated spermatogonia. In addition, germ cell transplantation assay demonstrated that SSEA4-positive spermatogonia colonized in the recipient testicular tubules. Sorting of the undifferentiated spermatogonia with anti-SSEA4 antibody resulted in a 2.5-fold enrichment of SSCs compared with the germ cell gate group, and 21-fold enrichment of SSCs compared with the SSEA4-negative spermatogonia group. CONCLUSIONS: Our findings revealed that SSEA4 is a new surface marker for porcine undifferentiated spermatogonia. This finding helps to elucidate the characteristics of porcine SSCs and facilitates the culture and manipulation of SSCs.


Subject(s)
Adult Germline Stem Cells/metabolism , Adult Germline Stem Cells/transplantation , Spermatogenesis/physiology , Spermatogonia/metabolism , Stage-Specific Embryonic Antigens/metabolism , Adult Germline Stem Cells/cytology , Animals , Biomarkers/metabolism , Cell Differentiation/physiology , Heterografts , Male , Mice , Mice, Inbred C57BL , Spermatogonia/cytology , Spermatozoa/growth & development , Swine , Testis/metabolism , Transplantation, Heterologous
12.
Genes Cells ; 25(7): 510-517, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32277725

ABSTRACT

Retinoblastoma is an infant cancer that results from loss of RB1 expression in both alleles. The RB1 gene was the first reported cancer suppressor gene; however, the mechanism by which RB1 loss causes cancer in the retina has not yet been clarified. Human-induced pluripotent stem cells (iPSCs) provide an ideal tool for mechanistic research regarding retinoblastoma. However, because RB1 is a tumor suppressor, loss of both alleles of RB1 in human iPS cells may affect the phenotype of the cells. To examine this possibility, we established human iPSCs with deletions in both alleles of RB1 by CRISPR/Cas9 technique to characterize the associated phenotype. We first examined the expression of RB1 transcripts by RT-qPCR, and RB1 transcripts were expressed in immature hiPSCs and then the expression levels of RB1 transcripts consistently increased during retinal organoid differentiation in human iPSCs. Expression levels of immature markers including SSEA4, OCT3/4 and NANOG were indistinguishable between control iPSCs and RB1 knockout iPSCs. Proliferative activity was also unaffected by homozygous RB1 deletion. Taken together, we showed that homozygous deletion of RB1 did not affect the maturation and proliferation statuses of human iPSCs.


Subject(s)
Cell Differentiation/genetics , Cell Proliferation/genetics , Induced Pluripotent Stem Cells/metabolism , Organoids/metabolism , Retina/metabolism , Retinoblastoma Binding Proteins/genetics , Ubiquitin-Protein Ligases/genetics , CRISPR-Cas Systems , Gene Deletion , Humans , Induced Pluripotent Stem Cells/cytology , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/metabolism , Phenotype , Retina/growth & development , Retinal Neoplasms/genetics , Retinal Neoplasms/metabolism , Retinoblastoma/genetics , Retinoblastoma/metabolism , Retinoblastoma Binding Proteins/metabolism , Stage-Specific Embryonic Antigens/metabolism , Ubiquitin-Protein Ligases/metabolism
13.
Cell Physiol Biochem ; 54(2): 271-286, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-32233339

ABSTRACT

BACKGROUND/AIMS: Pericytes (PCs) are multipotent vascular precursors that play a critical physiological role in the development and maintenance of blood vessel integrity. In this study, we aim to characterize PCs isolated from human abdominal adipose tissue and develop an integration-free induced pluripotent stem cells (iPSCs) using episomal vectors. METHODS: The ultrastructure of adipose tissue-derived PCs was determined using scanning and transmission electron microscopy. The expression of mesenchymal stem cells (MSCs) and pericyte markers were examined using flow cytometry and PCR analysis. PCs were induced to adipogenic, osteogenic and myogenic lineages, and their angiogenic potential was determined using tube formation assay. We further established pericyte reprogramming protocol using episomal vectors. RESULTS: Our data showed that human adipose tissue-derived PCs uniformly expressed MSCs, CD105 and CD73, and PCs markers, desmin, and alpha smooth muscle actin (α-SMA), while lacked the expression of HLA-DR and the hematopoietic markers CD34, CD11b and CD45. Ultrastructure analysis showed typical internal structure for the PCs with a characteristic prominent eccentric nuclei and cytoplasmic invaginations forming a caveolar system. Functional analysis showed efficient differentiation into adipocytes, osteocytes, and myocyte-like cells. Adipose tissue-derived PCs showed angiogenic potential using tube-forming assay. To determine further application of these cells for personalized therapy, we reprogrammed PCs into induced pluripotent stem cells (iPSCs) using episomal vectors. Reprogrammed cells gradually lost their fusiform shape, acquired the epithelial cell morphology and formed colonies. Furthermore, reprogrammed cells successfully expressed the pluripotency markers OCT4, Nanog, SSEA-4, and ß-catenin, an early reprogramming marker. CONCLUSION: The accessibility and abundance of human fat supports the application of adipose derived PCs as a novel and promising source of cell therapy and regenerative medicine.


Subject(s)
Adipose Tissue/cytology , Cellular Reprogramming Techniques/methods , Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , Pericytes/cytology , 5'-Nucleotidase/metabolism , Actins/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Adipose Tissue/ultrastructure , Cell Lineage , Cells, Cultured , Cellular Reprogramming/genetics , Cellular Reprogramming/physiology , Desmin/metabolism , Endoglin/metabolism , Flow Cytometry , GPI-Linked Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/ultrastructure , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Muscle Cells/cytology , Muscle Cells/metabolism , Muscle Development/genetics , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/metabolism , Osteocytes/cytology , Osteocytes/metabolism , Osteogenesis/genetics , Pericytes/metabolism , Pericytes/ultrastructure , Stage-Specific Embryonic Antigens/metabolism , beta Catenin/metabolism
14.
Cell Tissue Res ; 381(1): 71-81, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32043210

ABSTRACT

Periodontal ligament (PDL) stem cell properties are critical in the periodontal tissue regeneration for periodontitis. Previously, we have demonstrated that cigarette smoking attenuates PDL-derived stem cell (PDLSC) regenerative properties. Here, we report the findings on the regenerative properties of human PDLSCs with different donor ages and the underlying mechanisms. Human PDLSCs from 18 independent donors were divided into different age groups (≤ 20, 20-40, and > 40 years old). The proliferation of PDLSCs with donor age of ≤ 20 years old was significantly higher than that of the 20-40- and > 40-years-old groups, whereas the migration of PDLSCs with donor age of ≤ 20 and 20-40 years old was significantly higher than that of the > 40-years-old group. Moreover, the mesodermal lineage differentiation capabilities of PDLSCs were also higher in the donor age group of ≤ 20 years old than the donor age of > 40 years old. In addition, shorter telomere length and lower expression of SSEA4 were found in PDLSCs with donor age of > 40 years old, compared with those with donor age of ≤ 20-years-old group. Besides, PDLSCs with donor age of 20-40 and > 40 years old had higher IL6 and CXCL8 gene expressions. In summary, results from this study revealed the attenuated proliferation, migration, and mesodermal lineage differentiation properties in human PDLSCs with older donor ages. Donor age of PDLSCs should be considered as the selection criteria for the periodontal tissue regeneration treatment.


Subject(s)
Age Factors , Chronic Periodontitis/therapy , Periodontal Ligament/cytology , Stage-Specific Embryonic Antigens/metabolism , Stem Cells/cytology , Telomere/ultrastructure , Adult , Cell Proliferation , Cells, Cultured , Female , Guided Tissue Regeneration, Periodontal , Humans , Interleukin-6/metabolism , Interleukin-8/metabolism , Male , Osteogenesis , Young Adult
15.
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
16.
J Orthop Res ; 38(2): 258-268, 2020 02.
Article in English | MEDLINE | ID: mdl-31429977

ABSTRACT

Liver kinase B1 (LKB1), a serine/threonine protein, is a key regulator in stem cell function and energy metabolism. Herein, we describe the role of LKB1 in modulating the differentiation of synovium-derived stem cells (SDSCs) toward chondrogenic, adipogenic, and osteogenic lineages. Human fetal SDSCs were transduced with CRISPR associated protein 9 (Cas9)-single-guide RNA vectors to knockout or lentiviral vectors to overexpress the LKB1 gene. Analyses including ICE (Inference of CRISPR Edits) data from Sanger sequencing and quantitative polymerase chain reaction (qPCR) as well as Western blot demonstrated successful knockout (KO) or overexpression (OE) of LKB1 in human fetal SDSCs without any detectable side effects in morphology, proliferation rate, and cell cycle. LKB1 KO increased CD146 expression; interestingly, LKB1 OE increased SSEA4 level. The qPCR data showed that LKB1 KO upregulated the levels of SOX2 and NANOG while LKB1 OE lowered the expression of POU5F1 and KLF4. Furthermore, LKB1 KO enhanced, and LKB1 OE inhibited, chondrogenic and adipogenic differentiation potential. However, perhaps due to the inherent inability to achieve osteogenesis, LKB1 did not obviously affect osteogenic differentiation. These data demonstrate that LKB1 plays a significant role in determining human SDSCs' adipogenic and chondrogenic differentiation, which might provide an approach for fine-tuning the direction of stem cell differentiation in tissue engineering and regeneration. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:258-268, 2020.


Subject(s)
Cell Differentiation , Fetal Stem Cells/physiology , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Base Sequence , CD146 Antigen/metabolism , Cells, Cultured , Genetic Vectors , Humans , Kruppel-Like Factor 4 , Lentivirus , Protein Serine-Threonine Kinases/genetics , Stage-Specific Embryonic Antigens/metabolism , Synovial Membrane/cytology
17.
J Biol Chem ; 295(4): 1009-1020, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31831622

ABSTRACT

Cancer remains a leading cause of morbidity and mortality worldwide, requiring ongoing development of targeted therapeutics such as monoclonal antibodies. Carbohydrates on embryonic cells are often highly expressed in cancer and are therefore attractive targets for antibodies. Stage-specific embryonic antigen-4 (SSEA-4) is one such glycolipid target expressed in many cancers, including breast and ovarian carcinomas. Here, we defined the structural basis for recognition of SSEA-4 by a novel monospecific chimeric antibody (ch28/11). Five X-ray structures of ch28/11 Fab complexes with the SSEA-4 glycan headgroup, determined at 1.5-2.7 Å resolutions, displayed highly similar three-dimensional structures indicating a stable binding mode. The structures also revealed that by adopting a horseshoe-shaped conformation in a deep groove, the glycan headgroup likely sits flat against the membrane to allow the antibody to interact with SSEA-4 on cancer cells. Moreover, we found that the terminal sialic acid of SSEA-4 plays a dominant role in dictating the exquisite specificity of the ch28/11 antibody. This observation was further supported by molecular dynamics simulations of the ch28/11-glycan complex, which show that SSEA-4 is stabilized by its terminal sialic acid, unlike SSEA-3, which lacks this sialic acid modification. These high-resolution views of how a glycolipid interacts with an antibody may help to advance a new class of cancer-targeting immunotherapy.


Subject(s)
Antibodies, Neoplasm/immunology , N-Acetylneuraminic Acid/metabolism , Neoplasms/immunology , Stage-Specific Embryonic Antigens/metabolism , Antibodies, Neoplasm/chemistry , Antibody Specificity/immunology , Carbohydrate Conformation , Humans , Immunoglobulin Fab Fragments/metabolism , Ligands , Molecular Dynamics Simulation , Polysaccharides/chemistry , Polysaccharides/metabolism , Stage-Specific Embryonic Antigens/chemistry
18.
Int J Mol Sci ; 20(24)2019 Dec 07.
Article in English | MEDLINE | ID: mdl-31817926

ABSTRACT

Glycosphingolipids (GSLs), such as the globo-series GSLs stage-specific embryonic antigen 3 (SSEA-3), SSEA-4, and Globo-H, are specifically expressed on pluripotent stem cells and cancer cells, and are known to be associated with various biological processes such as cell recognition, cell adhesion, and signal transduction. However, the behavior and biological roles of these GSLs are still unclear. In our previous study, we observed the interactions between the lipid raft and GSLs in real-time using single-molecule imaging, where we successfully synthesized various fluorescent analogs of GSLs (e.g., GM1 and GM3). Here, we have developed fluorescent analogs of SSEA-3, SSEA-4, and Globo-H using chemical synthesis. The biophysical properties of these analogs as raft markers were examined by partitioning giant plasma membrane vesicles from RBL-2H3 cells into detergent-resistant membrane fractions and liquid-ordered/liquid-disordered phases. The results indicated that the analogs were equivalent to native-type GSLs. The analogs could be used to observe the behavior of globo-series GSLs for detailing the structure and biological roles of lipid rafts and GSL-enriched nanodomains during cell differentiation and cell malignancy.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate/metabolism , Cell Membrane/metabolism , Glycosphingolipids/metabolism , Leukemia, Basophilic, Acute/metabolism , Membrane Microdomains/metabolism , Stage-Specific Embryonic Antigens/metabolism , Animals , Cell Communication , Cell Differentiation , Leukemia, Basophilic, Acute/pathology , Molecular Structure , Rats , Signal Transduction , Tumor Cells, Cultured
19.
Cells ; 8(12)2019 11 21.
Article in English | MEDLINE | ID: mdl-31766521

ABSTRACT

: Stem cells isolated from the apical papilla of wisdom teeth (SCAPs) are an attractive model for tissue repair due to their availability, high proliferation rate and potential to differentiate in vitro towards mesodermal and neurogenic lineages. Adult stem cells, such as SCAPs, develop in stem cell niches in which the oxygen concentration [O2] is low (3-8% compared with 21% of ambient air). In this work, we evaluate the impact of low [O2] on the physiology of SCAPs isolated and processed in parallel at 21% or 3% O2 without any hyperoxic shock in ambient air during the experiment performed at 3% O2. We demonstrate that SCAPs display a higher proliferation capacity at 3% O2 than in ambient air with elevated expression levels of two cell surface antigens: the alpha-6 integrin subunit (CD49f) and the embryonic stem cell marker (SSEA4). We show that the mesodermal differentiation potential of SCAPs is conserved at early passage in both [O2], but is partly lost at late passage and low [O2], conditions in which SCAPs proliferate efficiently without any sign of apoptosis. Unexpectedly, we show that autophagic flux is active in SCAPs irrespective of [O2] and that this process remains high in cells even after prolonged exposure to 3% O2.


Subject(s)
Cell Culture Techniques/methods , Dental Papilla/metabolism , Stem Cells/cytology , Autophagy/physiology , Cell Differentiation/physiology , Cell Hypoxia/physiology , Cell Proliferation/physiology , Cells, Cultured , Humans , Integrin alpha6/metabolism , Membrane Proteins/metabolism , Molar, Third/cytology , Osteogenesis/physiology , Oxygen/metabolism , Proto-Oncogene Proteins/metabolism , Stage-Specific Embryonic Antigens/metabolism , Stem Cell Niche/physiology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...