Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Cell Tissue Res ; 393(2): 321-342, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37249709

ABSTRACT

Development of clinical-grade, cell preparations is central to cGMP (good manufacturing practice compliant) conditions. This study aimed to investigate the potential of two serum/xeno-free, cGMP (StemPro, StemMacs) culture media to maintain "stemness" of human minor salivary gland stem cell (mSG-SC) cultures compared to a complete culture medium (CCM). Overall, StemMacs resulted in higher proliferation rates after p.6 compared to the conventional serum-based medium, while StemPro showed substantial delays in cell proliferation after p.9. The mSG-SCs cultures exhibited two distinct cell populations at early passages a mesenchymal subpopulation and an epithelial-like subpopulation. Expression of several markers (CD146, STRO-1, SSEA-4, CD105, CD106, CD34, K 7/8, K14, K18) variably decreased with prolonged passaging (all three media). The percentage of SA-ß-gal positive cells was initially higher for StemMacs compared to StemPro/CCM and increased with prolonged passaging in all cases. The telomere fragment length decreased with prolonged passaging in all three media but more pronouncedly for the CCM. Expansion under serum-free conditions caused pronounced upregulation of ALP and BMP-2, with parallel complete elimination of the baseline expressions of LPL (all three media) and ACAN (serum-free media), therefore, showing a preferential shift of the mSG-SCs towards osteogenic phenotypes. Finally, several markers (Nanog, SOX-2, PDX-1, OTX2, GSC, HCG) decreased with prolonged culture, indicating successive loss of "stemness". Based on the findings, it seems that StemPro preserve stemness of the mSG-SCs after prolonged culture. Nevertheless, there is still a vacant role for the ideal development of clinical-grade culture conditions.


Subject(s)
Mesenchymal Stem Cells , Humans , Cell Differentiation , Salivary Glands, Minor , Stem Cells , Cell Culture Techniques/methods , Biomarkers/metabolism , Cell Proliferation , Culture Media/pharmacology , Cells, Cultured
2.
Cell Signal ; 108: 110693, 2023 08.
Article in English | MEDLINE | ID: mdl-37141926

ABSTRACT

Colorectal cancer (CRC) is a common malignant tumor of the human digestive tract. Inorganic pyrophosphatase 1 (PPA1) plays an imperative role in the advancement of malignant tumors, but its function in CRC is ill-defined. In this study, we inspected the functions of PPA1 in CRC. The abundance of PPA1 in CRC tissues was analyzed by utilizing publicly available data from the The Cancer Genome Atlas and Human Protein Atlas project. Cell counting kit-8 assay and 5-ethynyl-2'-deoxyuridine assay were used to evaluate the viability and proliferation of CRC cells. Bioinformatics analysis was used to forecast the PPA1 related genes and signal pathways in CRC. The protein expression was examined by western blot. The xenograft model was implemented to determine the influence of PPA1 in CRC in vivo. Proliferating cell nuclear antigen, CD133, and CD44 contents in xenograft tumors were evaluated by immunohistochemistry. In the present study, we found that the PPA1 content was heightened in CRC, and the diagnostic value of PPA1 in CRC was enormous. Overexpression of PPA1 enhanced cell proliferation and stemness properties in CRC cells, while downregulation of PPA1 had the opposite effects. PPA1 promoted the activation of the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway. Activation of the PI3K/Akt signaling reversed the effect of PPA1 silencing on cell proliferation and stemness properties in CRC cells. Silencing of PPA1 reduced xenograft tumor growth via modulating the PI3K/Akt signaling pathway in vivo. In conclusion, PPA1 promoted cell proliferation and stemness properties in CRC by activating the PI3K/Akt signaling pathway.


Subject(s)
Colorectal Neoplasms , Proto-Oncogene Proteins c-akt , Animals , Humans , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Inorganic Pyrophosphatase/genetics , Inorganic Pyrophosphatase/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
3.
Kaohsiung J Med Sci ; 39(3): 234-243, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36495291

ABSTRACT

We aimed to study the regulatory roles and mechanism of circular nuclear factor IX (circNFIX) in cancer growth and stemness properties of ovarian cancer (OC). CircNFIX and SH3RF3 levels in OC tissues and cells were tested by quantitative real-time PCR. RNase R treatment quantified circNFIX RNA stability. Molecular interaction among circNFIX, LIN28B, and SH3RF3 was predicted by bioinformatics software and validated through RNA immunoprecipitation (RIP) assay. The gain- or loss-experiments of circNFIX on capabilities of metastasis and stemness in vitro were assessed using Cell Counting Kit-8, Transwell, western blot, and sphere-formation assays. CircNFIX and SH3RF3 were markedly elevated in OC tissues and OC cells. Knocking down circNFIX repressed the proliferation, migration, invasion, and stemness properties of A2780 and SKOV3 cells. The RIP assay verified the direct binding relationship between LIN28B, circNFIX, and SH3RF3. Additionally, overexpression of circNFIX elevated the SH3RF3 expression, while this effect was reversed by LIN28B silence. Rescue experiments demonstrated that the overexpression of SH3RF3 reversed the knockdown of circNFIX on OC cells' proliferation, metastasis, and stemness properties. CircNFIX improved the mRNA stability and translation of SH3RF3 via recruiting LIN28B, thus promoting the proliferation, invasion, and stemness properties of OC cells in vitro.


Subject(s)
MicroRNAs , Ovarian Neoplasms , Humans , Female , Ovarian Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , MicroRNAs/genetics , Gene Expression Regulation, Neoplastic , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ubiquitin-Protein Ligases/genetics
4.
Cell Tissue Bank ; 24(3): 523-533, 2023 Sep.
Article in English | MEDLINE | ID: mdl-36526818

ABSTRACT

Mesenchymal stem cells (MSCs), derived from various tissues, are served as a promising source of cells in clinic and regenerative medicine. Umbilical cord-Wharton's jelly (WJ-MSCs)-derived MSCs exhibit advantages over those from adult tissues, such as no ethical concerns, shorter population doubling time, broad differentiation potential, readily available non-invasive source, prolonged maintenance of stemness properties. The aim of this study was to evaluate the effect of MRI (1.5 T, 10 min) on stemness gene expression patterns (OCT-4, SOX-2, NANOG) of WJ-MSCs. Additionally, we assessed cell viability, growth kinetics and apoptosis of WJ-MSCs after MRI treatment. The quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) data showed that transcript levels of SOX-2, NANOG in MRI-treated WJ-MSCs were increased 32- and 213-fold, respectively. MTT assay was performed at 24, 48, and 72 h post-treatment and the viability was not significantly different between the two groups. The doubling time of the MRI group was markedly higher than the control group. In addition, the colony formation ability of WJ-MSCs after MRI treatment significantly increased. Furthermore, no change in apoptosis was seen before or after MRI treatment. Our results suggest that the use of MRI can improve the quality of MSCs and enhance the efficacy of mesenchymal stem cell-based therapies.


Subject(s)
Mesenchymal Stem Cells , Wharton Jelly , Wharton Jelly/metabolism , Umbilical Cord , Cell Differentiation , Cells, Cultured , Cell Proliferation
5.
Ecotoxicol Environ Saf ; 213: 112031, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33578097

ABSTRACT

BACKGROUND: Nickel is a component of biomedical alloys that is released during corrosion or friction and causes cytotoxicity, mutation, differentiation or even carcinogenesis in tissues. However, the mechanisms underlying the potential hazards of Nickel-containing alloys implanted in the human body by surgery remain uncertain. OBJECTIVE: To study the effect of Ni(II) (NiCl2•6H2O) on cancer cells. METHODS: A549 and RKO cells were treated with various concentrations of Ni(II) to determine the effect of Ni(II) on cellular viability using a CCK8 assay. Flow cytometry was performed to analyze the effect of Ni(II) on apoptosis and the cell cycle. Sphere-forming assays were conducted to examine the stemness properties of A549 and RKO cells. Western blotting was to evaluate the expression levels of SOX2, IDH1, HIF-1ɑ and ß-catenin. The expression of isocitrate dehydrogenase (IDH1) in rectum adenocarcinoma (READ) was analyzed by Gene Expression Profiling Interactive Analysis (GEPIA). Kaplan-Meier analysis was used to calculate the correlation between survival and IDH1 expression. RESULTS: Long-term exposure (120 days) to 100 µM Ni(II) significantly repressed cell proliferation, decreased colony formation and arrested the cell cycle at the G0/G1 phase. In addition, the stem-like traits of A549 and RKO cells were significantly augmented. Ni(II) also significantly decreased the protein expression of IDH1 and the synthesis rate of NAPDH, which competitively inhibited α-ketoglutarate (α-KG) generation. The downregulation of IDH1 not only promoted ß-catenin accumulation in the cell nucleus in a HIF-1ɑ signaling-dependent manner but also induced the expression of the transcription factor SOX2 to maintain the stemness properties of cancer cells. Moreover, IDH1 expression negatively correlated with the clinicopathologic characteristics of READ. CONCLUSION: These findings demonstrate that chronic and continuous release of Ni(II) to the microenvironment suppresses IDH1 expression and augments the stemness properties of cancer cells via the activation HIF-1ɑ/ß-catenin/SOX2 pathway to enhance local tumor recurrence in patients with implanted Nickel-containing alloys at surgical sites.


Subject(s)
Isocitrate Dehydrogenase/metabolism , Nickel/toxicity , Apoptosis , Cell Differentiation , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival/drug effects , Humans , Mutation , Neoplasms , Signal Transduction , beta Catenin
6.
Biochim Biophys Acta Rev Cancer ; 1875(2): 188525, 2021 04.
Article in English | MEDLINE | ID: mdl-33600824

ABSTRACT

Gynecological cancers are characterized by a high mortality rate when chemoresistance develops. Autophagy collaborates with apoptosis and participates in homeostasis of chemoresistance. Recent findings supported that crosstalk of necrotic, apoptotic and autophagic factors, and chemotherapy-driven hypoxia, oxidative stress and ER stress play critical roles in chemoresistance in gynecological cancers. Meanwhile, current studies have shown that autophagy could be regulated by and cooperate with metabolic regulator, survival factors, stemness factors and specific post-translation modification in chemoresistant tumor cells. Meanwhile, non-coding RNA and autophagy crosstalk also contribute to the chemoresistance. Until now, analysis of individual autophagy factors towards the clinical significance and chemoresistance in gynecological cancer is still lacking. We suggest comprehensive integrated analysis of cellular homeostasis and tumor microenvironment to clarify the role of autophagy and the associated factors in cancer progression and chemoresistance. Panel screening of pan-autophagic factors will pioneer the development of risk models for predicting efficacy of chemotherapy and guidelines for systematic treatment and precision medicine.


Subject(s)
Autophagy , Drug Resistance, Neoplasm , Genital Neoplasms, Female/metabolism , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Oxidative Stress , Signal Transduction , Tumor Microenvironment
7.
J Cell Mol Med ; 24(22): 13020-13035, 2020 11.
Article in English | MEDLINE | ID: mdl-33026174

ABSTRACT

Although miR-148a-3p has been reported to function as a tumour suppressor in various cancers, the molecular mechanism of miR-148a-3p in regulating epithelial-to-mesenchymal transition (EMT) and stemness properties of pancreatic cancer (PC) cells remains to be elucidated. In the present study, we demonstrated that miR-148a-3p expression was remarkably down-regulated in PC tissues and cell lines. Moreover, low expression of miR-148a-3p was associated with poorer overall survival (OS) in patients with PC. In vitro, gain-of-function and loss-of-function experiments showed that miR-148a-3p suppressed EMT and stemness properties as well as the proliferation, migration and invasion of PC cells. A dual-luciferase reporter assay demonstrated that Wnt1 was a direct target of miR-148a-3p, and its expression was inversely associated with miR-148a-3p in PC tissues. Furthermore, miR-148a-3p suppressed the Wnt/ß-catenin pathway via down-regulation of Wnt1. The effects of ectopic miR-148a-3p were rescued by Wnt1 overexpression. These biological functions of miR-148a-3p in PC were also confirmed in a nude mouse xenograft model. Taken together, these findings suggest that miR-148a-3p suppresses PC cell proliferation, invasion, EMT and stemness properties via inhibiting Wnt1-mediated Wnt/ß-catenin pathway and could be a potential prognostic biomarker as well as a therapeutic target in PC.


Subject(s)
Epithelial-Mesenchymal Transition , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/metabolism , Wnt Signaling Pathway , Wnt1 Protein/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , beta Catenin/metabolism
8.
Int J Mol Sci ; 20(8)2019 Apr 16.
Article in English | MEDLINE | ID: mdl-31014017

ABSTRACT

Peri-implantitis-an infection caused by bacterial deposition of biofilm-is a common complication in dentistry which may lead to implant loss. Several decontamination procedures have been investigated to identify the optimal approach being capable to remove the bacterial biofilm without modifying the implant surface properties. Our study evaluated whether two different systems-Ni-Ti Brushes (Brush) and Air-Polishing with 40 µm bicarbonate powder (Bic40)-might alter the physical/chemical features of two different titanium surfaces-machined (MCH) and Ca++ nanostructured (NCA)-and whether these decontamination systems may affect the biological properties of human STRO-1+/c-Kit+ dental pulp stem cells (hDPSCs) as well as the bacterial ability to produce biofilm. Cell morphology, proliferation and stemness markers were analysed in hDPSCs grown on both surfaces, before and after the decontamination treatments. Our findings highlighted that Bic40 treatment either maintained the surface characteristics of both implants and allowed hDPSCs to proliferate and preserve their stemness properties. Moreover, Bic40 treatment proved effective in removing bacterial biofilm from both titanium surfaces and consistently limited the biofilm re-growth. In conclusion, our data suggest that Bic40 treatment may operatively clean smooth and rough surfaces without altering their properties and, consequently, offer favourable conditions for reparative cells to hold their biological properties.


Subject(s)
Antigens, Surface/metabolism , Detergents/chemistry , Proto-Oncogene Proteins c-kit/metabolism , Titanium/chemistry , Antigens, Surface/genetics , Biofilms/drug effects , Cell Proliferation , Dental Pulp/cytology , Dental Pulp/metabolism , Humans , Microscopy, Atomic Force , Proto-Oncogene Proteins c-kit/genetics , Pseudomonas aeruginosa/physiology , Stem Cells/cytology , Stem Cells/metabolism , Surface Properties , Titanium/pharmacology
9.
J Ovarian Res ; 11(1): 36, 2018 May 02.
Article in English | MEDLINE | ID: mdl-29716628

ABSTRACT

BACKGROUND: Ovarian cancer stem cells (OCSCs) contribute to the poor prognosis of ovarian cancer. Involvement of the androgen receptor (AR) in the malignant behaviors of other tumors has been reported. However, whether AR associates with Nanog (a stem cell marker) and participates in OCSC functions remain unclear. In this study, we investigated the interaction of Nanog with AR and examined whether this interaction induced stem-like properties in ovarian cancer cells. METHODS: AR and Nanog expression in ovarian tumors was evaluated. Using the CRISPR/Cas9 system, we constructed a Nanog green fluorescent protein (GFP) marker cell model to investigate the expression and co-localization of Nanog and AR. Then, we examined the effect of androgen on the Nanog promoter in ovarian cancer cell lines (A2780 and SKOV3). After androgen or anti-androgen treatment, cell proliferation, migration, sphere formation, colony formation and tumorigenesis were assessed in vitro and in vivo. RESULTS: Both AR and Nanog expression were obviously high in ovarian tumors. Our results showed that Nanog expression was correlated with AR expression. The androgen 5α-dihydrotestosterone (DHT) activated Nanog promoter transcription. Meanwhile, Nanog GFP-positive cells treated with DHT exhibited higher levels of proliferation, migration, sphere formation and colony formation. We also observed that the tumorigenesis of Nanog GFP-positive cells was significantly higher than that of the GFP-negative cells. Xenografts of Nanog GFP-positive cells showed significant differences when treated with androgen or anti-androgen drugs in vivo. CONCLUSIONS: The interaction of Nanog with the AR signaling axis might induce or contribute to OCSC regulation. In addition, androgen might promote stemness characteristics in ovarian cancer cells by activating the Nanog promoter. This finding merits further study because it may provide a new understanding of OCSC regulation from a hormone perspective and lead to the reevaluation of stem cell therapy for ovarian cancer.


Subject(s)
Carcinogenesis/genetics , Nanog Homeobox Protein/genetics , Ovarian Neoplasms/genetics , Receptors, Androgen/genetics , Androgens/genetics , Androgens/metabolism , Animals , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/pathology , Ovary/growth & development , Ovary/pathology , Promoter Regions, Genetic/genetics , Signal Transduction/genetics , Xenograft Model Antitumor Assays
10.
Stem Cell Res Ther ; 8(1): 247, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-29096714

ABSTRACT

BACKGROUND: Development of clinical-grade cell preparations is central to meeting the regulatory requirements for cellular therapies under good manufacturing practice-compliant (cGMP) conditions. Since addition of animal serum in culture media may compromise safe and efficient expansion of mesenchymal stem cells (MSCs) for clinical use, this study aimed to investigate the potential of two serum/xeno-free, cGMP culture systems to maintain long-term "stemness" of oral MSCs (dental pulp stem cells (DPSCs) and alveolar bone marrow MSCs (aBMMSCs)), compared to conventional serum-based expansion. METHODS: DPSC and aBMMSC cultures (n = 6/cell type) were established from pulp and alveolar osseous biopsies respectively. Three culture systems were used: StemPro_MSC/SFM_XenoFree (Life Technologies); StemMacs_MSC/XF (Miltenyi Biotek); and α-MEM (Life Technologies) with 15% fetal bovine serum. Growth (population doublings (PDs)), immunophenotypic (flow cytometric analysis of MSC markers) and senescence (ß-galactosidase (SA-ß-gal) activity; telomere length) characteristics were determined during prolonged expansion. Gene expression patterns of osteogenic (ALP, BMP-2), adipogenic (LPL, PPAR-γ) and chondrogenic (ACAN, SOX-9) markers and maintenance of multilineage differentiation potential were determined by real-time PCR. RESULTS: Similar isolation efficiency and stable growth dynamics up to passage 10 were observed for DPSCs under all expansion conditions. aBMMSCs showed lower cumulative PDs compared to DPSCs, and when StemMacs was used substantial delays in cell proliferation were noted after passages 6-7. Serum/xeno-free expansion produced cultures with homogeneous spindle-shaped phenotypes, while serum-based expansion preserved differential heterogeneous characteristics of each MSC population. Prolonged expansion of both MSC types but in particular the serum/xeno-free-expanded aBMMSCs was associated with downregulation of CD146, CD105, Stro-1, SSEA-1 and SSEA-4, but not CD90, CD73 and CD49f, in parallel with an increase of SA-gal-positive cells, cell size and granularity and a decrease in telomere length. Expansion under both serum-free systems resulted in "osteogenic pre-disposition", evidenced by upregulation of osteogenic markers and elimination of chondrogenic and adipogenic markers, while serum-based expansion produced only minor changes. DPSCs retained a diminishing (CCM, StemPro) or increasing (StemMacs) mineralization potential with passaging, while aBMMSCs lost this potential after passages 6-7 under all expansion conditions. CONCLUSIONS: These findings indicate there is still a vacant role for development of qualified protocols for clinical-grade expansion of oral MSCs; a key milestone achievement for translation of research from the bench to clinics.


Subject(s)
Bone Marrow Cells/drug effects , Culture Media, Serum-Free/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , Mesenchymal Stem Cells/drug effects , Osteogenesis/drug effects , Adipogenesis/drug effects , Adipogenesis/genetics , Aggrecans/genetics , Aggrecans/metabolism , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Alveolar Process/cytology , Alveolar Process/drug effects , Alveolar Process/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Biomarkers/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chondrogenesis/drug effects , Chondrogenesis/genetics , Culture Media, Serum-Free/chemistry , Dental Pulp/cytology , Dental Pulp/drug effects , Dental Pulp/metabolism , Drug Industry/legislation & jurisprudence , Gene Expression/drug effects , Humans , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteogenesis/genetics , PPAR gamma/genetics , PPAR gamma/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Telomere Homeostasis , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
11.
Oncotarget ; 8(33): 54388-54401, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28903350

ABSTRACT

Cancer stem cells (CSCs) are a subpopulation of cancer cells believed to be implicated in cancer initiation, progression, and recurrence. Here, we report that ectopic expression of zinc finger E-box binding homeobox 1 protein (ZEB1) results in the acquisition of CSC properties by breast cancer cells, leading to tumor initiation and progression in vitro and in vivo. The neurogenin 3 gene (Ngn3) is a bona fide target of ZEB1, and its repression is a key factor contributing to ZEB1-induced cancer cell stemness. ZEB1 suppressed Ngn3 transcription by forming a ZEB1/DNA methyltransferase (DNMT)3B/histone deacetylase 1 (HDAC1) complex on the Ngn3 promoter, leading to promoter hypermethylation and gene silencing. The rescue of Ngn3 expression attenuated ZEB1-induced cancer stemness and symmetric CSC division. Immunohistological analysis of human breast cancer specimens revealed a strong inverse relationship between ZEB1 and NGN3 protein expression. Thus, our findings suggest ZEB1-mediated silencing of Ngn3 is required for breast tumor initiation and maintenance. Targeted therapies against the ZEB1/Ngn3 axis may be highly valuable for the prevention and treatment of breast cancer.

SELECTION OF CITATIONS
SEARCH DETAIL