Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
J Exp Clin Cancer Res ; 43(1): 98, 2024 Apr 01.
Article En | MEDLINE | ID: mdl-38561856

BACKGROUND: The complement inhibitor CSMD1 acts as a tumor suppressor in various types of solid cancers. Despite its high level of expression in the brain, its function in gliomas, malignant brain tumors originating from glial cells, has not been investigated. METHODS: Three cohorts of glioma patients comprising 1500 patients were analyzed in our study along with their clinical data. H4, U-118 and U-87 cell lines were used to investigate the tumor suppressor function of CSMD1 in gliomas. PDGFB-induced brain tumor model was utilized for the validation of in vitro data. RESULTS: The downregulation of CSMD1 expression correlated with reduced overall and disease-free survival, elevated tumor grade, wild-type IDH genotype, and intact 1p/19q status. Moreover, enhanced activity was noted in the neuroinflammation pathway. Importantly, ectopic expression of CSMD1 in glioma cell lines led to decreased aggressiveness in vitro. Mechanically, CSMD1 obstructed the TNF-induced NF-kB and STAT3 signaling pathways, effectively suppressing the secretion of IL-6 and IL-8. There was also reduced survival in PDGFB-induced brain tumors in mice when Csmd1 was downregulated. CONCLUSIONS: Our study has identified CSMD1 as a tumor suppressor in gliomas and elucidated its role in TNF-induced neuroinflammation, contributing to a deeper understanding of glioma pathogenesis.


Brain Neoplasms , Glioma , Humans , Animals , Mice , Neuroinflammatory Diseases , Proto-Oncogene Proteins c-sis/genetics , Glioma/pathology , Brain Neoplasms/pathology , Disease-Free Survival , Isocitrate Dehydrogenase/genetics , Mutation , Membrane Proteins/genetics , Tumor Suppressor Proteins/genetics
3.
Mol Biol Rep ; 50(4): 3011-3022, 2023 Apr.
Article En | MEDLINE | ID: mdl-36652154

BACKGROUND: Eukaryotic elongation factor 2 kinase (eukaryotic elongation factor 2 kinase, eEF2K) is a calcium calmodulin dependent protein kinase that keeps the highest energy consuming cellular process of protein synthesis under check through negative regulation. eEF2K pauses global protein synthesis rates at the translational elongation step by phosphorylating its only kown substrate elongation factor 2 (eEF2), a unique translocase activity in ekaryotic cells enabling the polypeptide chain elongation. Therefore, eEF2K is thought to preserve cellular energy pools particularly upon acute development of cellular stress conditions such as nutrient deprivation, hypoxia, or infections. Recently, high expression of this enzyme has been associated with poor prognosis in an array of solid tumor types. Therefore, in a growing number of studies tremendous effort is being directed to the development of treatment methods aiming to suppress eEF2K as a novel therapeutic approach in the fight against cancer. METHODS: In our study, we aimed to investigate the changes in the tumorigenicity of chordoma cells in presence of gene silencing for eEF2K. Taking a transient gene silencing approach using siRNA particles, eEF2K gene expression was suppressed in chordoma cells. RESULTS: Silencing eEF2K expression was associated with a slight increase in cellular proliferation and a decrease in death rates. Furthermore, no alteration in the sensitivity of chordoma cells to chemotherapy was detected in response to the decrease in eEF2K expression which intriguingly promoted suppression of cell migratory and invasion related properties. CONCLUSION: Our findings indicate that the loss of eEF2K expression in chordoma cell lines results in the reduction of metastatic capacity.


Chordoma , Elongation Factor 2 Kinase , Humans , Elongation Factor 2 Kinase/genetics , Elongation Factor 2 Kinase/chemistry , Elongation Factor 2 Kinase/metabolism , Chordoma/genetics , Phosphorylation , Cell Line , Signal Transduction
4.
J Exp Clin Cancer Res ; 40(1): 258, 2021 Aug 17.
Article En | MEDLINE | ID: mdl-34404439

BACKGROUND: Human CUB and Sushi multiple domains 1 (CSMD1) is a large membrane-bound tumor suppressor in breast cancer. The current study aimed to elucidate the molecular mechanism underlying the effect of CSMD1 in highly invasive triple negative breast cancer (TNBC). METHODS: We examined the antitumor action of CSMD1 in three TNBC cell lines overexpressing CSMD1, MDA-MB-231, BT-20 and MDA-MB-486, in vitro using scanning electron microscopy, proteome array, qRT-PCR, immunoblotting, proximity ligation assay, ELISA, co-immunoprecipitation, immunofluorescence, tumorsphere formation assays and flow cytometric analysis. The mRNA expression pattern and clinical relevance of CSMD1 were evaluated in 3520 breast cancers from a modern population-based cohort. RESULTS: CSMD1-expressing cells had distinct morphology, with reduced deposition of extracellular matrix components. We found altered expression of several cancer-related molecules, as well as diminished expression of signaling receptors including Epidermal Growth Factor Receptor (EGFR), in CSMD1-expressing cells compared to control cells. A direct interaction of CSMD1 and EGFR was identified, with the EGF-EGFR induced signaling cascade impeded in the presence of CSMD1. Accordingly, we detected increased  ubiquitination levels of EGFR upon activation in CSMD1-expressing cells, as well as increased degradation kinetics and chemosensitivity. Accordingly, CSMD1 expression rendered tumorspheres pretreated with gefitinib more sensitive to chemotherapy. In addition, higher mRNA levels of CSMD1 tend to be associated with better outcome of triple negative breast cancer patients treated with chemotherapy. CONCLUSIONS: Our results indicate that CSMD1 cross-talks with the EGFR endosomal trafficking cascade in a way that renders highly invasive breast cancer cells sensitive to chemotherapy. Our study unravels one possible underlying molecular mechanism of CSMD1 tumor suppressor function and may provide novel avenues for design of better treatment.


Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Membrane Proteins/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Cell Line, Tumor , Cells, Cultured , Drug Resistance, Neoplasm/genetics , Endosomes/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression , Humans , Membrane Proteins/genetics , Prognosis , Protein Binding , Protein Transport , Proteome , Proteomics/methods , Signal Transduction/drug effects , Tumor Suppressor Proteins/genetics
5.
Dev Biol ; 478: 212-221, 2021 10.
Article En | MEDLINE | ID: mdl-34245726

To date, the role of miRNAs on pluripotency and differentiation of ESCs into specific lineages has been studied extensively. However, the specific role of miRNAs during lateral and paraxial mesoderm cell fate decision is still unclear. To address this, we firstly determined miRNA profile of mouse ESCs differentiating towards lateral and paraxial lineages which were detected using Flk1 and PDGFαR antibodies, and of myogenic and hematopoietic differentiation potential of purified paraxial and lateral mesodermal cells within these populations. miRNAs associated with lateral and paraxial mesoderm, and their targets were identified using bioinformatics tools. The targets of the corresponding miRNAs were validated after transfection into mouse ESCs. The roles of the selected miRNAs in lateral, and paraxial mesoderm formation were assessed along with hematopoietic and myogenic differentiation capacity. Among the miRNAs, mmu-miR-126a-3p, mmu-miR-335-5p and mmu-miR-672-5p, upregulated in lateral mesoderm cells, and mmu-miR-10b-5p, mmu-miR-196a-5p and mmu-miR-615-3p, upregulated in paraxial mesoderm cells. While transient co-transfection of mmu-miR-126a-3p, mmu-miR-335-5p and mmu-miR-672-5p increased the number of lateral mesodermal cells, co-transfection of mmu-miR-10b-5p, mmu-miR-196a-5p and mmu-miR-615-3p increased the number of paraxial mesodermal cells. Moreover, differentiation potential of the lateral mesodermal cells into hematopoietic cell lineage increased upon co-transfection of mmu-miR-126a-3p, mmu-miR-335-5p and mmu-miR-672-5p and differentiation potential of the paraxial mesodermal cells into skeletal muscle lineage were increased upon co-transfection of mmu-miR-10b-5p, mmu-miR-196a-5p and mmu-miR-615-3p. In conclusion, we determined the miRNA profile of lateral and paraxial mesodermal cells and co-transfection of miRNAs increased differentiation potential of both lateral and paraxial mesodermal cells transiently.


Cell Differentiation , Embryonic Stem Cells/physiology , Mesoderm/cytology , MicroRNAs/genetics , Animals , Computational Biology , Embryoid Bodies/cytology , Embryonic Stem Cells/metabolism , Hematopoiesis , Mesoderm/embryology , Mesoderm/metabolism , Mice , MicroRNAs/metabolism , Muscle Development , Transfection , Up-Regulation
6.
J Cell Physiol ; 236(12): 8122-8136, 2021 12.
Article En | MEDLINE | ID: mdl-34101829

Hematopoietic stem cells (HSCs) are particularly characterized by their quiescence and self-renewal. Cell cycle regulators tightly control quiescence and self-renewal capacity. Studies suggest that modulation of ubiquitination and neddylation could contribute to HSC function via cyclin-dependent kinase inhibitors (CDKIs). S-phase kinase-associated protein 2 (SKP2) is responsible for ubiquitin-mediated proteolysis of CDKIs. Here, we modulated overall neddylation and SKP2-associated ubiquitination in HSCs by using SKP2-C25, an SKP2 inhibitor, and MLN4924 (Pevonedistat) as an inhibitor of the NEDD8 system. Treatments of SKP2-C25 and MLN4924 increased both murine and human stem and progenitor cell (HSPC) compartments. This is associated with the improved quiescence of murine HSC by upregulation of p27 and p57 CDKIs. A colony-forming unit assay showed an enhanced in vitro self-renewal potential post inhibition of ubiquitination and neddylation. In addition, MLN4924 triggered the mobilization of bone marrow HSPCs to peripheral blood. Intriguingly, MLN4924 treatment could decrease the proliferation of murine bone marrow mesenchymal stem cells or endothelial cells. These findings shed light on the contribution of SKP2, and associated ubiquitination and neddylation in HSC maintenance, self-renewal, and expansion.


Cell Proliferation/physiology , Endothelial Cells/cytology , Hematopoietic Stem Cells/metabolism , Ubiquitination/physiology , Animals , Cell Cycle/physiology , Humans , Mice , NEDD8 Protein/metabolism , S-Phase Kinase-Associated Proteins/metabolism
7.
Appl Physiol Nutr Metab ; 46(8): 866-876, 2021 Aug.
Article En | MEDLINE | ID: mdl-33493087

Calorie restriction (CR) is suggested to prevent the development of mammary tumors (MTs); however, the mechanism remains to be clarified. We aimed to determine the microRNA (miRNA) profile in mice applied to 2 different CR protocols; chronic (CCR) and intermittent (ICR) and follow the MT development. In addition, the roles of miRNAs involved in adiponectin and/or leptin signaling pathways were investigated. Mice were divided into 3 groups: ad-libitum (AL), CCR, or ICR, which comprised 3 weeks of AL feeding followed by 1 week of 60% CR in a cyclic manner. Blood and tissue collection were performed at weeks 10, 17/18, 49/50 and 81/82. Long-term CCR provided better protection compared with ICR for MT development with a delay in the MT occurrence. Adiponectin expression in mammary fat pad were significantly higher in CCR group compared with AL. Using GeneChip Array, 250 of 3195 miRNAs were differentially expressed among the dietary groups. Thirteen of 250 miRNAs were related to adiponectin and/or leptin signaling genes. Results were verified by reverse transcription polymerase chain reaction. Specifically, miR-326-3p, miR-500-3p and miR-129-5p, which are related to adiponectin and/or leptin signaling, may play important roles in the preventive effects of CR in MT development and in ageing. Thus, these miRNAs might be putative biomarkers to target for diagnostic and treatment purposes. Novelty: Type of CR and micro RNA interaction is related to ageing. miR-326-3p, miR-500-3p and miR-129-5p expression levels were differentially expressed in MT development and in ageing. The genes associated with adiponectin and/or leptin signaling pathways are regulated by certain miRNAs in the protective effects of CR.


Adiponectin/metabolism , Breast Neoplasms/metabolism , Caloric Restriction/methods , Leptin/metabolism , MicroRNAs/metabolism , Signal Transduction , Animals , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL
8.
Sci Rep ; 10(1): 7994, 2020 05 14.
Article En | MEDLINE | ID: mdl-32409701

Meis1, which belongs to TALE-type class of homeobox gene family, appeared as one of the key regulators of hematopoietic stem cell (HSC) self-renewal and a potential therapeutical target. However, small molecule inhibitors of MEIS1 remained unknown. This led us to develop inhibitors of MEIS1 that could modulate HSC activity. To this end, we have established a library of relevant homeobox family inhibitors and developed a high-throughput in silico screening strategy against homeodomain of MEIS proteins using the AutoDock Vina and PaDEL-ADV platform. We have screened over a million druggable small molecules in silico and selected putative MEIS inhibitors (MEISi) with no predicted cytotoxicity or cardiotoxicity. This was followed by in vitro validation of putative MEIS inhibitors using MEIS dependent luciferase reporter assays and analysis in the ex vivo HSC assays. We have shown that small molecules named MEISi-1 and MEISi-2 significantly inhibit MEIS-luciferase reporters in vitro and induce murine (LSKCD34l°w cells) and human (CD34+, CD133+, and ALDHhi cells) HSC self-renewal ex vivo. In addition, inhibition of MEIS proteins results in downregulation of Meis1 and MEIS1 target gene expression including Hif-1α, Hif-2α and HSC quiescence modulators. MEIS inhibitors are effective in vivo as evident by induced HSC content in the murine bone marrow and downregulation of expression of MEIS target genes. These studies warrant identification of first-in-class MEIS inhibitors as potential pharmaceuticals to be utilized in modulation of HSC activity and bone marrow transplantation studies.


Drug Development , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Myeloid Ecotropic Viral Integration Site 1 Protein/antagonists & inhibitors , Amino Acid Sequence , Animals , Biomarkers , Bone Marrow Cells , Cell Proliferation , Drug Evaluation, Preclinical , Flow Cytometry , Genes, Reporter , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Knockout , Models, Molecular , Myeloid Ecotropic Viral Integration Site 1 Protein/chemistry , Protein Conformation , Small Molecule Libraries , Structure-Activity Relationship
9.
Cell Oncol (Dordr) ; 42(5): 663-677, 2019 Oct.
Article En | MEDLINE | ID: mdl-31175552

PURPOSE: Chordomas are highly therapy-resistant primary bone tumors that exhibit high relapse rates and may induce local destruction. Here, we evaluated the effects of tumor necrosis factor-alpha (TNF-α) on chordoma progression and clinical outcome. METHODS: Chordoma cells were treated with TNF-α after which its short- and long-term effects were evaluated. Functional assays, qRT-PCR and microarray-based expression analyses were carried out to assess the effect of TNF-α on chemo-resistance, epithelial to mesenchymal transition (EMT), migration, invasion and cancer stem cell-like properties. Finally, relationships between TNF-α expression and clinicopathological features were assessed in a chordoma patient cohort. RESULTS: We found that TNF-α treatment increased the migration and invasion of chordoma cells. Also, NF-κB activation was observed along with increased EMT marker expression. In addition, enhanced tumor sphere formation and soft agar colony formation were observed, concomitantly with increased chemo-resistance and CD338 marker expression. The TNF-α and TNFR1 expression levels were found to be significantly correlated with LIF, PD-L1 and Ki67 expression levels, tumor volume and a short survival time in patients. In addition, a high neutrophil to lymphocyte ratio was found to be associated with recurrence and a decreased overall survival. CONCLUSIONS: From our data we conclude that TNF-α may serve as a prognostic marker for chordoma progression and that tumor-promoting inflammation may be a major factor in chordoma tumor progression.


Bone Neoplasms/pathology , Chordoma/pathology , Epithelial-Mesenchymal Transition/drug effects , Tumor Necrosis Factor-alpha/physiology , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Adult , Aged , B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Bone Neoplasms/drug therapy , Bone Neoplasms/mortality , Cell Line, Tumor , Cell Movement/drug effects , Child , Chordoma/drug therapy , Chordoma/mortality , Chordoma/secondary , Disease Progression , Drug Resistance, Neoplasm , Female , Humans , Inflammation , Leukemia Inhibitory Factor/metabolism , Lymphocytes/metabolism , Male , Middle Aged , Neoplasm Proteins/metabolism , Neutrophils/metabolism , Prognosis , Receptors, Tumor Necrosis Factor, Type I/metabolism , Retrospective Studies , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Young Adult
10.
Exp Cell Res ; 380(1): 9-19, 2019 07 01.
Article En | MEDLINE | ID: mdl-30951707

Chordoma is a rare, slow-growing tumor thought to arise from remnants of embryonic notochord associated with an aggressive outcome. Cancer stem-like cells (CSCs) are related to tumorigenesis, recurrence, and resistance in cancers. Therefore, chordoma CSCs are possible targets for chordoma treatment. In this study, dysregulated miRNAs were determined in chordoma CSCs and identified their role in chordoma. Dysregulated miRNAs were determined via miRNA microarray and validated through qPCR. miRNAs were transiently transfected to the chordoma cell lines and their roles in proliferation, apoptosis, migration and invasion capacities and stem-like properties were identified. Finally, a relationship between clinicopathological features and dysregulated miRNAs has been evaluated among 21 chordoma patients. CD133+CD15+ cells exhibited CSC phenotype with increased CSC- and Epithelial-Mesenchymal Transition (EMT)-related gene expression, invasion, migration, tumorosphere- and colony-forming abilities. In addition, WNT5A, TGF-α, BTG2 and MYCBP genes involved in CSC-related pathways, were targets of miR-140-3p, miR-148a-3p, miR-210-5p and miR-574-5p, respectively. Transfection of CSC-related miRNAs also increased migration and invasion along with stem cell phenotype. Finally, we determined that miR-140-3p and miR-148a-3p expressions correlated with Ki67 while miR-140-3p and TGF-α expressions were correlated with p53. Moreover, MYCBP expression was positively correlated with tumor volume, and metastasis was associated with the expression of miR-210-5p and TGF-α in our patient cohort. Through these findings, we conclude that chordoma CSCs have distinctive miRNA profile, which can regulate stem-like properties of chordoma CSCs.


Chordoma/genetics , MicroRNAs/genetics , Aged , Carcinogenesis/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Chordoma/pathology , DNA-Binding Proteins/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Ki-67 Antigen/genetics , Male , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Transcription Factors/genetics , Transforming Growth Factor alpha/genetics , Tumor Suppressor Protein p53/genetics
11.
Gene ; 701: 161-168, 2019 Jun 15.
Article En | MEDLINE | ID: mdl-30849537

Glial tumors are malignant brain tumors that arise from glial cells of brain or spine and have genetic aberrations in their genome. 1p/19q co-deletion is associated with increased Overall Survival (OS) time with enhanced response to chemo- and radio-therapy in oligodendrogliomas. However, prognostic significance of 1p/19q co-polysomy is still unclear. We evaluated 1p/19q status of 221 patients with glial tumor by Fluorescent in situ Hybridization (FISH). Records of the patients were collected retrospectively. Our results demonstrated that 1p/19q co-polysomy was associated with decreased OS time, high P53 expression and frequently located in temporal lobe, whereas 1p/19q co-deletion was associated with increased overall survival time, low P53 expression and frontal lobe location. Furthermore, classification of patients based on both 1p/19q status and P53 expression revealed that patients with 1p/19q co-polysomy and high P53 expression had the worst prognosis. Lastly, our bioinformatic survival analysis revealed that high expression of SRM, ICMT, and FTL located in 1p36.13-p36.31 and 19q13.2-q13.33 region were related with decreased OS time in patients with Low Grade Glioma (LGG). The study demonstrated that 1p/19q co-polysomy is a poor prognostic marker for glial tumor.


Biomarkers, Tumor , Brain Neoplasms , Chromosome Aberrations , Chromosomes, Human, Pair 1 , Gene Expression Regulation, Neoplastic , Neoplasm Proteins , Oligodendroglioma , Adult , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 1/metabolism , Disease-Free Survival , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Oligodendroglioma/genetics , Oligodendroglioma/metabolism , Oligodendroglioma/mortality , Oligodendroglioma/pathology , Retrospective Studies , Survival Rate
12.
Curr Cancer Drug Targets ; 19(6): 479-494, 2019.
Article En | MEDLINE | ID: mdl-30182856

BACKGROUND: c-Myc plays a major role in the maintenance of glycolytic metabolism and hematopoietic stem cell (HSC) quiescence. OBJECTIVE: Targeting modulators of HSC quiescence and metabolism could lead to HSC cell cycle entry with concomitant expansion. METHODS AND RESULTS: Here we show that c-Myc inhibitor 10074-G5 treatment leads to 2-fold increase in murine LSKCD34low HSC compartment post 7 days. In addition, c-Myc inhibition increases CD34+ and CD133+ human HSC number. c-Myc inhibition leads to downregulation of glycolytic and cyclindependent kinase inhibitor (CDKI) gene expression ex vivo and in vivo. In addition, c-Myc inhibition upregulates major HDR modulator Rad51 expression in hematopoietic cells. Besides, c-Myc inhibition does not alter proliferation kinetics of endothelial cells, fibroblasts or adipose-derived mesenchymal stem cells, however, it limits bone marrow derived mesenchymal stem cell proliferation. We further demonstrate that a cocktail of c-Myc inhibitor 10074-G5 along with tauroursodeoxycholic acid (TUDCA) and i-NOS inhibitor L-NIL provides a robust HSC maintenance and expansion ex vivo as evident by induction of all stem cell antigens analyzed. Intriguingly, the cocktail of c-Myc inhibitor 10074-G5, TUDCA and L-NIL improves HDR related gene expression. CONCLUSION: These findings provide tools to improve ex vivo HSC maintenance and expansion, autologous HSC transplantation and gene editing through modulation of HSC glycolytic and HDR pathways.


Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Oxadiazoles/pharmacology , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Rad51 Recombinase/metabolism , Animals , Antiviral Agents/pharmacology , Apoptosis/drug effects , Cell Culture Techniques , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Hematopoietic Stem Cells/drug effects , Humans , Lysine/analogs & derivatives , Lysine/pharmacology , Mice , Mice, Inbred BALB C , Mice, SCID , Nitric Oxide Synthase/antagonists & inhibitors , Rad51 Recombinase/biosynthesis , Rad51 Recombinase/genetics , Small Molecule Libraries/pharmacology , Taurochenodeoxycholic Acid/pharmacology
13.
Exp Mol Pathol ; 105(3): 395-403, 2018 12.
Article En | MEDLINE | ID: mdl-30414978

Amniotic fluid (AF) is the liquid layer that provides mechanical support and allows movement of the fetus during embryogenesis. Mesenchymal stem cells (MSCs), which have differentiation capacity, are also found in AF-derived cells at a low ratio. Smooth muscle cells (SMCs) play an important role in organ function and are frequently used in tissue engineering. We examined the differentiation of AF-derived MSCs (AMSCs) into SMCs. AMSCs were sorted from cultured amniotic cells and differentiated into SMCs using differentiation agents, including platelet-derived growth factor BB (PDGF-BB) and tumor growth factor ß (TGF-ß). Characterization of differentiated SMCs was confirmed morphologically, molecularly (via quantitative polymerase chain reaction [qPCR] and immunocytochemistry [ICC]), and functionally (using a contractile assay and fluo-4 calcium signaling assay). Poly(lactide-co-glycolide) (PLGA) scaffolds were fabricated, and the attachment capacity of AMSCs was assessed via scanning electron microscopy. AMSCs were successfully differentiated into SMCs. Our results indicate that AMSCs change their morphology and exhibit increased expression of ACTA2 and MYH11, which was confirmed via qPCR and ICC. Furthermore, functional experiments revealed that differentiated SMCs had both contraction ability and increased Ca2 concentration in the cytoplasm. Finally, PLGA scaffolds were prepared and AMSCs were successfully planted onto the scaffolds. The AMSCs fully differentiated into functional SMCs, and the PLGA polymer is a suitable scaffold material for AMSCs. With further clinical trials, AF-derived MSC-based SMC engineering may become a highly efficient treatment option.


Amniotic Fluid/cytology , Cell Differentiation/physiology , Mesenchymal Stem Cells/cytology , Muscle, Smooth/cytology , Animals , Female , Polylactic Acid-Polyglycolic Acid Copolymer , Pregnancy , Rabbits , Tissue Engineering/methods , Tissue Scaffolds/chemistry
14.
Brain Res ; 1700: 199-210, 2018 12 01.
Article En | MEDLINE | ID: mdl-30176243

Although glioblastomas are common, there remains a need to elucidate the underlying mechanisms behind their initiation and progression and identify molecular pathways for improving treatment. In this study, sixteen fresh-frozen glioblastoma samples and seven samples of healthy brain tissues were analyzed with miRNA and whole transcriptome microarray chips. Candidate miRNAs and mRNAs were selected to validate expression in fifty patient samples in total with the criteria of abundance, relevance and prediction scores. miRNA and target mRNA relationships were assessed by inhibiting selected miRNAs in glioblastoma cells. Functional tests have been conducted in order to see the effects of miRNAs on invasion, migration and apoptosis of GBM cells. Analyses were carried out to determine correlations between selected molecules and clinicopathological features. 1332 genes and 319 miRNAs were found to be dysregulated by the microarrays. The results were combined and analyzed with Transcriptome Analysis Console 3 software and the DAVID online database. Primary differential pathways included Ras, HIF-1, MAPK signaling and cell adhesion. OncomiR candidates 21-5p, 92b-3p, 182-5p and 339-5p for glioblastoma negatively correlated with notable mRNA targets both in tissues and in in vitro experiments. miR-21-5p and miR-339-5p significantly affected migration, invasion and apoptosis of GBM cells in vitro. Significant correlations with overall survival, tumor volume, recurrence and age at diagnosis were discovered. In this article we present valuable integrated microarray analysis of glioblastoma samples regarding miRNA and gene-expression levels. Notable biomarkers and miRNA-mRNA interactions have been identified, some of which correlated with clinicopathological features in our cohort.


Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , MicroRNAs/metabolism , RNA, Messenger/metabolism , Apoptosis/physiology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/mortality , Cell Line, Tumor , Cohort Studies , Computer Simulation , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Glioblastoma/mortality , Humans , Microarray Analysis , Prognosis , Survival Analysis , Transcriptome
15.
Mol Cell Biochem ; 437(1-2): 133-142, 2018 Jan.
Article En | MEDLINE | ID: mdl-28620820

Cell proliferation and migration are crucial in many physiological processes including development, cancer, tissue repair, and wound healing. Cell migration is regulated by several signaling molecules. Identification of genes related to cell migration is required to understand molecular mechanism of non-healing chronic wounds which is a major concern in clinics. In the current study, the role of cytoglobin (CYGB) gene in fibroblast cell migration and proliferation was described. L929 mouse fibroblast cells were transduced with lentiviral particles for CYGB and GFP, and analyzed for cell proliferation and migration ability. Fibroblast cells overexpressing CYGB displayed decreased cell proliferation, colony formation capacity, and cell migration. Phosphorylation levels of mTOR and two downstream effectors S6 and 4E-BP1 which take part in PI3K/AKT/mTOR signaling declined in CYGB-overexpressing cells. Microarray analysis indicated that CYGB overexpression leads to downregulation of cell proliferation, migration, and tumor growth associated genes in L929 cell line. This study demonstrated the role of CYGB in fibroblast cell motility and proliferation. CYGB could be a promising candidate for further studies as a potential target for diseases related to cell migration such as cancer and chronic wound treatment.


Cell Movement , Cell Proliferation , Fibroblasts/metabolism , Globins/metabolism , Signal Transduction , Animals , Cell Line , Cytoglobin , Fibroblasts/cytology , Globins/genetics , Mice , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
16.
Oncol Res ; 25(7): 1177-1188, 2017 Aug 07.
Article En | MEDLINE | ID: mdl-28247842

Chordomas are rare tumors of the spine and skull base that are locally destructive and resistant to chemotherapy and radiation therapy, with a poor prognosis and limited therapeutic options. Chordoma patients have a long life expectancy with high mortality from the disease. Cancer stem cells, which are known to exist in chordomas, have extensive proliferative and self-renewal potential and are responsible for maintaining tumor heterogeneity along with chemotherapy and radiotherapy resistance. Leukemia inhibitory factor (LIF) has multiple functions in stem cell biology, the immune response, and cancer, and is potentially a key molecule that allows cancer stem cells to self-renew. The purpose of this study was to determine whether LIF increases the aggressive traits of chordoma cells and leads to a poor prognosis in patients. Chordoma cell lines were treated with LIF, and functional tests were done. Twenty skull base chordoma samples were checked for levels of LIF and a correlation with clinicopathological features. The whole transcriptome microarray was used to observe changes in gene expression. We observed increased migration, invasion, tumorosphere formation, colony formation, epithelial-mesenchymal transition, and chemoresistance accompanied by a dramatic elevation in inflammatory gene networks and pathways in chordomas. The expression of LIF was associated with tumor size and a poorer overall survival. Microarray and quantitative real-time polymerase chain reaction assessments suggest that LIF can facilitate tumor-promoting inflammation. Results indicate that LIF plays a role in maintaining cancer stem cells in chordomas.


Chordoma/genetics , Chordoma/pathology , Leukemia Inhibitory Factor/genetics , Adolescent , Adult , Aged , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Chordoma/metabolism , Chordoma/mortality , Combined Modality Therapy , Cytokines/genetics , Cytokines/metabolism , Disease Progression , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Leukemia Inhibitory Factor/metabolism , Leukemia Inhibitory Factor/pharmacology , Leukemia Inhibitory Factor Receptor alpha Subunit/genetics , Leukemia Inhibitory Factor Receptor alpha Subunit/metabolism , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Young Adult
17.
BMC Cancer ; 17(1): 207, 2017 03 21.
Article En | MEDLINE | ID: mdl-28327132

BACKGROUND: Although meningioma is a common disease, there is a lack of understanding of the underlying molecular mechanisms behind its initiation and progression. We used combined miRNA-mRNA transcriptome analysis to discover dysregulated genes and networks in meningiomas. METHODS: Fourteen fresh-frozen meningioma samples and one human meningeal cell line were analyzed by using miRNA and whole transcriptome microarray chips. Data was filtered and analyzed. Candidate miRNAs and mRNAs were selected for validation in fifty-eight patient samples. miRNA and target mRNA relationships were assessed by inhibiting miRNA in meningioma cells. Apoptosis and viability assays were also used as functional tests. RESULTS: With the whole transcriptome microarray, 3753 genes were found to be dysregulated, and 891 miRNAs were found to be dysregulated as a result of miRNA microarray. Results were combined and analyzed with bioinformatics tools. Top differential pathways included those of inflammation, cancer, and cellular growth and survival. The oncosupressor PTX3 was constitutively low in meningioma samples. Moreover, PTX3 negatively correlated with miR-29c in our samples. Inhibiting miR-29c upregulated the PTX3 level, induced apoptosis of meningioma cells, and decreased cell viability. CABIN1, miR-29c, TMOD1, PTX3, RPL22, SPARCL1 and RELA were correlated with clinicopathological features in patient samples. CONCLUSIONS: Our results present the first integrated mRNA-miRNA analysis in meningiomas. miR-29c-3p and PTX3 are inversely correlated in tissues and meningioma cells, hinting that PTX3 can be regulated by miR-29c-3p. Furthermore, we determined potential clinicopathological markers.


C-Reactive Protein/genetics , Gene Expression Profiling/methods , Meningeal Neoplasms/genetics , Meningioma/genetics , MicroRNAs/genetics , RNA, Messenger/genetics , Serum Amyloid P-Component/genetics , Apoptosis/genetics , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Survival/genetics , Computational Biology/methods , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans
18.
Metabolism ; 69: 130-142, 2017 04.
Article En | MEDLINE | ID: mdl-28285642

Obesity is a worldwide medical problem resulting in serious morbidity and mortality involving differentiation of pre-adipocytes into mature adipocytes (adipogenesis). Boron treatment has been reported to be associated with weight reduction in experimental animals; however, its effects on pre-adipocyte differentiation and anti-adipogenic molecular mechanisms are unknown. In this study, we demonstrate the inhibitory activities of boric acid (BA) and sodium pentaborate pentahydrate (NaB) on adipogenesis using common cellular models. Boron treatment repressed the expression of adipogenesis-related genes and proteins, including CCAAT-enhancer-binding protein α and peroxisome proliferator-activated receptor γ, by regulating critical growth factors and the ß-catenin, AKT, and extracellular signal-regulated kinase signaling pathways. In addition, although boron treatment did not induce apoptosis in pre-adipocytes, it depressed mitotic clonal expansion by regulation of cell cycle genes. Overall, these data offer promising insights into the prevention/treatment of obesity and associated diseases.


Adipogenesis/drug effects , Anti-Obesity Agents/therapeutic use , Boron Compounds/pharmacology , Obesity/drug therapy , Stem Cells/drug effects , Wnt Signaling Pathway/drug effects , 3T3-L1 Cells , Adipocytes/drug effects , Adipogenesis/genetics , Adiponectin/biosynthesis , Adiponectin/genetics , Animals , Borates/pharmacology , Boric Acids/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cytokines/biosynthesis , Cytokines/genetics , Gene Expression Regulation/drug effects , Genes, cdc/drug effects , Leptin/biosynthesis , Leptin/genetics , Mice , Mitosis/drug effects
19.
Gene ; 585(1): 76-83, 2016 Jul 01.
Article En | MEDLINE | ID: mdl-27016303

Little is known about the molecular biology of chordomas, which are rare, chemoresistant tumors with no well-established treatment. miRNAs regulate gene networks and pathways. We aimed to evaluate the effects of dysregulated miRNA in chordomas would help reveal the underlying mechanisms of chordoma initiation and progression. In this study, miR-31, anti-miR-140-3p, anti-miR148a, and miR-222 were transiently transfected to chordoma cell lines and an MTS assay, apoptosis assay, and cell-cycle analysis were conducted to evaluate the effects. The mRNA level of predicted and confirmed targets of each miRNA, as well as the EMT and MET markers of U-CH1 and MUG-Chor1, were assessed with real-time polymerase chain reaction. Transient transfection of miRNA mimics was achieved, as each mimic increased or decreased the level of its corresponding miRNA. miR-31 decreased cell viability in MUG-Chor1 and U-CH2 after 72h, which is consistent with previous findings for U-CH1. Both miR-31 and anti-miR-148a induced apoptosis in all three cell lines. Although each miRNA had a similar pattern, miR-31 had the most effective S-phase arrest in all three cell lines. RDX, MET, DNMT1, DNMT3B, TRPS1, BIRC5, and KIT were found to be targeted by the selected miRNAs. The level of miR-222 in chordoma cell lines U-CH1 and MUG-Chor1 correlated positively with EMT markers and negatively with MET markers. This study uncovered the potential of miR-31, miR-140-3p, miR-148a, and miR-222-3p to be key molecules in the cell viability, cell cycle, and apoptosis in chordomas, as well as initiation, differentiation, and progression.


Apoptosis/genetics , Chordoma/genetics , MicroRNAs/genetics , S Phase Cell Cycle Checkpoints/genetics , Bone Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Survival , Gene Expression Regulation, Neoplastic/genetics , Humans , MicroRNAs/pharmacology , Oligonucleotide Array Sequence Analysis , Oligonucleotides/genetics , Oligonucleotides/pharmacology , Real-Time Polymerase Chain Reaction , S Phase Cell Cycle Checkpoints/drug effects
...