Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Int J Mol Sci ; 24(12)2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37373296

ABSTRACT

Phosphorylation of the serine 139 of the histone variant H2AX (γH2AX) is a DNA damage marker that regulates DNA damage response and various diseases. However, whether γH2AX is involved in neuropathic pain is still unclear. We found the expression of γH2AX and H2AX decreased in mice dorsal root ganglion (DRG) after spared nerve injury (SNI). Ataxia telangiectasia mutated (ATM), which promotes γH2AX, was also down-regulated in DRG after peripheral nerve injury. ATM inhibitor KU55933 decreased the level of γH2AX in ND7/23 cells. The intrathecal injection of KU55933 down-regulated DRG γH2AX expression and significantly induced mechanical allodynia and thermal hyperalgesia in a dose-dependent manner. The inhibition of ATM by siRNA could also decrease the pain threshold. The inhibition of dephosphorylation of γH2AX by protein phosphatase 2A (PP2A) siRNA partially suppressed the down-regulation of γH2AX after SNI and relieved pain behavior. Further exploration of the mechanism revealed that inhibiting ATM by KU55933 up-regulated extracellular-signal regulated kinase (ERK) phosphorylation and down-regulated potassium ion channel genes, such as potassium voltage-gated channel subfamily Q member 2 (Kcnq2) and potassium voltage-gated channel subfamily D member 2 (Kcnd2) in vivo, and KU559333 enhanced sensory neuron excitability in vitro. These preliminary findings imply that the down-regulation of γH2AX may contribute to neuropathic pain.


Subject(s)
Neuralgia , Peripheral Nerve Injuries , Animals , Mice , Ganglia, Spinal/metabolism , Hyperalgesia/genetics , Hyperalgesia/metabolism , Neuralgia/etiology , Neuralgia/metabolism , Peripheral Nerve Injuries/metabolism , Potassium/metabolism , RNA, Small Interfering/metabolism , Sensory Receptor Cells/metabolism , Shal Potassium Channels/metabolism
2.
Front Immunol ; 13: 885685, 2022.
Article in English | MEDLINE | ID: mdl-35911694

ABSTRACT

Histone lysine crotonylation (KCR), a novel epigenetic modification, is important in regulating a broad spectrum of biological processes and various diseases. However, whether KCR is involved in neuropathic pain remains to be elucidated. We found KCR occurs in macrophages, sensory neurons, and satellite glial cells of trigeminal ganglia (TG), neurons, astrocytes, and microglia of the medulla oblongata. KCR in TG was detected mainly in small and medium sensory neurons, to a lesser extent in large neurons. Peripheral nerve injury elevated KCR levels in macrophages in the trigeminal and dorsal root ganglia and microglia in the medulla oblongata but reduced KCR levels in sensory neurons. Inhibition of histone crotonyltransferases (p300) by intra-TG or intrathecal administration of C646 significantly alleviated partial infraorbital nerve transection (pIONT)- or spinal nerve ligation (SNL)-induced mechanical allodynia and thermal hyperalgesia. Intra-TG or intrathecal administration of Crotonyl coenzyme A trilithium salt to upregulate KCR dose-dependently induced mechanical allodynia and thermal hyperalgesia in mice. Mechanismly, inhibition of p300 alleviated pIONT-induced macrophage activation and reduced the expression of pain-related inflammatory cytokines Tnfα, Il1ß and chemokines Ccl2 and Cxcl10. Correspondingly, exogenous crotonyl-CoA induced macrophage activation and the expression of Tnfα, Il1ß, Il6, Ccl2 and Ccl7 in TG, which C646 can repress. These findings suggest that histone crotonylation might be functionally involved in neuropathic pain and neuroinflammation regulation.


Subject(s)
Hyperalgesia , Neuralgia , Animals , Histones/metabolism , Hyperalgesia/etiology , Hyperalgesia/metabolism , Lysine , Mice , Neuralgia/etiology , Neuralgia/metabolism , Sensory Receptor Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism
3.
Asian J Androl ; 24(1): 90-96, 2022.
Article in English | MEDLINE | ID: mdl-34213488

ABSTRACT

Peroxisome proliferator-activated receptors γ (PPARγ) is a master regulator that controls energy metabolism and cell fate. PPARγ2, a PPARγ isoform, is highly expressed in the normal prostate but expressed at lower levels in prostate cancer tissues. In the present study, PC3 and LNCaP cells were used to examine the benefits of restoring PPARγ2 activity. PPARγ2 was overexpressed in PC3 and LNCaP cells, and cell proliferation and migration were detected. Hematoxylin and eosin (H&E) staining was used to detect pathological changes. The genes regulated by PPARγ2 overexpression were detected by microarray analysis. The restoration of PPARγ2 in PC3 and LNCaP cells inhibited cell proliferation and migration. PC3-PPARγ2 tissue recombinants showed necrosis in cancerous regions and leukocyte infiltration in the surrounding stroma by H&E staining. We found higher mixed lineage kinase domain-like (MLKL) and lower microtubule-associated protein 1 light chain 3 (LC3) expression in cancer tissues compared to controls by immunohistochemistry (IHC) staining. Microarray analysis showed that PPARγ2 gain of function in PC3 cells resulted in the reprogramming of lipid- and energy metabolism-associated signaling pathways. These data indicate that PPARγ2 exerts a crucial tumor-suppressive effect by triggering necrosis and an inflammatory reaction in human prostate cancer.


Subject(s)
PPAR gamma , Prostatic Neoplasms , Animals , Cell Proliferation , Humans , Male , Mice , PC-3 Cells , PPAR gamma/genetics , Prostatic Neoplasms/genetics , Signal Transduction , Xenograft Model Antitumor Assays
4.
Toxicology ; 463: 152991, 2021 11.
Article in English | MEDLINE | ID: mdl-34673133

ABSTRACT

4-Nitrophenol (PNP) has been extensively used in manufacturing for several decades. Its toxic effects on the male reproductive system have been reported, but the underlying mechanisms remain unclear. In this study, we utilized two testicular somatic cell lines (TM3 and TM4 cells) to explore the possible toxic effects of PNP on the male reproductive system. The activity of the cells after exposure to different doses of PNP (0.01, 0.1, 1, 10 and 100 µM) was evaluated. PNP treatment at 10 µM significantly inhibited cell viability, and 10 µM PNP was thus selected for subsequent experiments. Although PNP (10 µM) inhibited cell proliferation, promoted cell apoptosis, and changed the cell cycle distribution and ultrastructure in both types of cells, these effects were more significant in the TM4 cells. In addition, an Agilent mouse mRNA array was used to identify the gene expression differences between the control and PNP (10 µM) exposed TM3 and TM4 cells. The microarray analysis identified 67 and 1372 differentially expressed genes mainly concentrated in endothelial cell morphogenesis and anatomical structure development in TM3 cells and associated with cardiovascular system development and circulatory system development in TM4 cells. Moreover, a pathway analysis revealed that PNP not only predominately affected meiotic recombination and meiosis in TM3 cells, but also influenced axon guidance and developmental biology in TM4 cells. These results suggest that TM3 and TM4 cells exhibit different responses to PNP, which might mediate different toxic mechanisms.


Subject(s)
Leydig Cells/drug effects , Nitrophenols/toxicity , Sertoli Cells/drug effects , Animals , Apoptosis/drug effects , Axon Guidance/drug effects , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Leydig Cells/metabolism , Male , Meiosis/drug effects , Mice , Nitrophenols/administration & dosage , Reproduction/drug effects , Sertoli Cells/metabolism , Testis/cytology , Testis/drug effects
5.
Cell Death Dis ; 12(6): 528, 2021 05 22.
Article in English | MEDLINE | ID: mdl-34023860

ABSTRACT

Prostate cancer (PCA) is one of the most common male genitourinary tumors. However, the molecular mechanisms involved in the occurrence and progression of PCA have not been fully clarified. The present study aimed to investigate the biological function and molecular mechanism of the nuclear receptor peroxisome proliferator-activated receptor gamma 2 (PPARG2) in PCA. Our results revealed that PPARG2 was downregulated in PCA, and overexpression of PPARG2 inhibited cell migration, colony formation, invasion and induced cell cycle arrest of PCA cells in vitro. In addition, PPARG2 overexpression modulated the activation of the Akt signaling pathway, as well as inhibited tumor growth in vivo. Moreover, mechanistic analysis revealed that PPARG2 overexpression induced increased expression level of miR-200b-3p, which targeted 3' UTR of the downstream targets DNMT3A/3B, and facilitated interaction with demethylated AKAP12 gene promoter and suppressed cell proliferation in PCA. Our findings provided the first evidence for a novel PPARG2-AKAP12 axis mediated epigenetic regulatory network. The study identified a molecular mechanism involving an epigenetic modification that could be possibly targeted as an antitumoral strategy against prostate cancer.


Subject(s)
A Kinase Anchor Proteins/genetics , Cell Cycle Proteins/genetics , PPAR gamma/physiology , Prostatic Neoplasms/pathology , A Kinase Anchor Proteins/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , DNA Methylation , Down-Regulation/genetics , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , PC-3 Cells , PPAR gamma/genetics , Promoter Regions, Genetic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Up-Regulation/genetics
6.
J Pain Res ; 13: 3195-3206, 2020.
Article in English | MEDLINE | ID: mdl-33293856

ABSTRACT

BACKGROUND: Currently, medical treatment of inflammatory pain is limited by a lack of safe and effective therapies. Triptonide (TPN), a major component of Tripterygium wilfordii Hook.f. with low toxicity, has been shown to have good anti-inflammatory and neuroprotective effects. The present study aims to investigate the effects of TPN on chronic inflammatory pain. MATERIALS AND METHODS: Inflammatory pain was induced by intraplantar injection of complete Freund's adjuvant (CFA). TPN's three different doses were intravenously administered to compare the analgesic efficacy: 0.1 mg/kg, 0.5 mg/kg, and 2.0 mg/kg. The foot swelling was quantitated by measuring paw volume. Mechanical allodynia and thermal hyperalgesia were assessed with von Frey filament testing and Hargreaves' test, respectively. Western blots, qRT-PCR and immunofluorescence tests were used to analyze the expression of pAKT, tumor necrosis factor-α (TNF-α), interleukin 1 beta (IL-1ß), and interleukin 6 (IL-6). Two AKT inhibitors, AKT inhibitor Ⅳ and MK-2206, were used to examine AKT's effects on pain behavior and cytokines expression. RESULTS: Intravenous treatment with TPN attenuated CFA-induced paw edema, mechanical allodynia, and thermal hyperalgesia. Western blotting and immunofluorescence results showed that CFA induced AKT activation in the dorsal root ganglion (DRG) neurons. However, these effects were suppressed by treatment with TPN. Furthermore, TPN treatment inhibited CFA-induced increase of pro-inflammatory cytokines, including TNF-α, IL-1ß, and IL-6. Consistent with the in vivo data, TPN inhibited LPS-induced Akt phosphorylation and inflammatory mediator production in ND7/23 cells. Finally, intrathecal treatment with AKT inhibitor Ⅳ or MK-2206, attenuated CFA-induced mechanical allodynia and thermal hyperalgesia, and simultaneously decreased the mRNA expression of TNF-α, IL-1ß, and IL-6 in DRG. CONCLUSION: These data indicate that TPN attenuates CFA-induced pain potentially via inhibiting AKT-mediated pro-inflammatory cytokines production in DRG. TPN may be used for the treatment of chronic inflammatory pain.

7.
Mol Med Rep ; 21(2): 815-821, 2020 02.
Article in English | MEDLINE | ID: mdl-31974620

ABSTRACT

Circular RNAs (circRNAs) regulate several physiological and pathological processes, but their role in visceral lipid deposition has not been explored. In the present study, human preadipocytes from visceral fat tissue (HPA­v) were induced to form adipocytes, and the circRNA expression profiles in HPA­v and adipocytes were detected using circRNA microarrays. The microarray data revealed that 2,215 and 1,865 circRNAs were significantly up­ and downregulated, respectively, in adipocytes compared with HPA­v. Moreover, the parental genes of differentially expressed circRNAs were associated with fatty acid metabolism based on Kyoto Encyclopedia of Genes and Genomes analysis. Three circRNAs (hsa_circ_0136134, hsa_circ_0017650, and hsa­circRNA9227­1) were selected for quantitative PCR (qPCR) validation, and the qPCR results were consistent with the microarray results. Furthermore, MiRanda software was used to predict the microRNAs (miRNAs) potentially targeting the top 10 up­ and downregulated circRNAs, and 14 miRNAs with more than two miRNA response elements targeting these circRNAs. This is the first study of the expression profiles of circRNAs in HPA­v and adipocytes and may suggest potential therapeutic targets for the visceral obesity.


Subject(s)
Adipocytes/metabolism , Gene Expression Profiling , Gene Expression Regulation , RNA, Circular/genetics , Adipocytes/cytology , Cell Differentiation/genetics , Down-Regulation/genetics , Humans , Intra-Abdominal Fat/cytology , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/metabolism , Up-Regulation/genetics
8.
Environ Mol Mutagen ; 61(3): 316-328, 2020 03.
Article in English | MEDLINE | ID: mdl-31654581

ABSTRACT

Although health hazards of 4-nitrophenol (PNP) exposure have been reported, the adverse effects of PNP exposure on cancer biological features are still unknown. We investigated the effects of administration of PNP in T24 human bladder cancer cells. The results showed that PNP exposure promoted cellular proliferation, migration and invasion, inhibited adhesion and apoptosis in vitro. Using quantitative real-time PCR, we found that (1) the mRNA expression levels of cell-cycle regulators PCNA, cyclin D1 and COX-2 were increased in PNP-treated cells compared to controls, however, that of pro-apoptotic gene Bax was decreased; (2) the expression level of EMT-associated gene E-cadherin was decreased in PNP-treated cells, whereas those of N-cadherin, vimentin, snail, and slug were increased; (3) the expression levels of cancer-promoting genes HIF-1, IL-1ß, VEGFα and K-Ras were enhanced, but those of tumor suppressors p53, PTEN and BRCA were decreased. There was a positive association between PNP exposure times and the promotion effects. Finally, we found that the expression level of PPARγ (γ1 isoform) was increased in PNP-treated T24 cells. GW9662, a specific PPARγ antagonist, attenuated PNP-induced cell migration and invasion. These findings indicate that PNP exposure may promote bladder cancer growth and progression involving PPARγ signaling. PPARγ is a potential target for development of novel intervention study on environment pollution. Environ. Mol. Mutagen. 61:316-328, 2020. © 2019 Wiley Periodicals, Inc.


Subject(s)
Carcinogens, Environmental/toxicity , Cell Proliferation/drug effects , Endocrine Disruptors/toxicity , Epithelial-Mesenchymal Transition/drug effects , Nitrophenols/toxicity , Urinary Bladder Neoplasms/chemically induced , Cell Line, Tumor , Cell Movement/drug effects , Disease Progression , Humans , Neoplasm Invasiveness/pathology , Urinary Bladder Neoplasms/pathology
9.
Prostate ; 79(11): 1284-1293, 2019 08.
Article in English | MEDLINE | ID: mdl-31212374

ABSTRACT

BACKGROUND: The increasing incidence of prostate cancer (PCa) indicates an urgent need for the development of new effective drugs in PCa therapy. Triptonide has been reported to have a strong inhibition activity in cancers through screening of Chinese herbal medicine. This study aims to investigate the effects of triptonide on anti-PCa activity and its mechanisms. METHODS: Three human advanced PCa cell lines PC3, DU145, and LNCap, and a human normal prostate epithelial cell line RWPE were treated with a range (0, 1.25, 2.5, 5, 10, 20, 40, 80, 160, and 320 nM) of triptonide concentrations for 72 hours respectively. Then, cell viability was assessed by cell counting kit-8. PCa cells were treated with different doses (0-20 nM) of triptonide for 72 hours. Cell cycle and apoptosis were assessed by flow cytometry assays. Nude mice bearing human PCa xenografts were intraperitoneally injected daily with either triptonide (10 mg/kg/d) or phosphate-buffered saline as a control for 35 days. RNA-sequencing (RNA-seq) was performed by an Illumina Hiseq Sequencing platform and confirmed by a real-time polymerase chain reaction. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes pathway analysis, and ingenuity pathway analysis were used to analyze RNA-seq results. RESULTS: Triptonide effectively inhibits the proliferation of human PCa cells PC3, DU145, and LNCap in vitro with their IC50 values as 11.961, 10.259, and 12.012 nM, respectively. Triptonide (10 mg/kg) potently inhibits the growth of PCa cell xenografts in vivo at an inhibition rate of over 97.95%. Treatment with triptonide (5 nM) significantly promotes cell apoptosis and retaining cell-cycle arrest in the G2/M phase. RNA-seq data revealed that total of 936 genes were upregulated or downregulated in triptonide treated. Moreover, the phosphorylation of mechanistic target of rapamycin (mTOR) and the downstream protein p70S6K were both inhibited, most obviously in PCa cells. CONCLUSIONS: Our findings suggest that triptonide can efficaciously suppress PCa growth in vitro and in vivo via inhibiting the phosphorylation of mTOR and the activities of related downstream signaling pathways.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Prostate/drug effects , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Triterpenes/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Cell Cycle Checkpoints/drug effects , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Male , Mice , Mice, Nude , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Triterpenes/therapeutic use
10.
Cytotherapy ; 21(4): 416-427, 2019 04.
Article in English | MEDLINE | ID: mdl-30904330

ABSTRACT

Pulmonary arterial hypertension (PAH) is characterized by pulmonary arterial endothelial hyperproliferation and dysfunction. Restoration of endothelial function is a common goal of available treatments. In the present study, human adipose-derived mesenchymal stromal cells (ASCs) were co-cultured with monocrotaline pyrrole-treated human pulmonary arterial endothelial cells (HPAECs); increased proliferation of HPAECs and expression of vascular endothelial growth factor (VEGF) were observed. High throughput sequencing results showed that six microRNAs (miMNAs) of ASCs were significantly dysregulated. In monocrotaline-induced PAH rat models, ASC transplantation improved the right ventricle systolic pressure, right ventricle hypertrophy and pulmonary endothelium hyperproliferation, and four of the six miRNAs were validated in the lung tissue samples. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis showed that these dysregulated miRNAs were involved in the regulation of transcription, signal transduction, negative regulation of cell proliferation through mitogen-activated protein kinase (MAPK) signaling pathway, Wnt signaling pathway, VEGF signaling pathway, cytokine-cytokine receptor interaction, regulation of actin cytoskeleton, transforming growth factor (TGF)-beta signaling pathway and P53 signaling pathway. Our data indicates that the unique six miRNA expression signature could be involved in the PAH endothelial repair by ASCs.


Subject(s)
Adipose Tissue/cytology , Endothelium/metabolism , Endothelium/physiopathology , Hemodynamics , Mesenchymal Stem Cells/cytology , MicroRNAs/genetics , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Arterial Hypertension/therapy , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Coculture Techniques , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Ontology , Hemodynamics/drug effects , Humans , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/physiopathology , Male , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , MicroRNAs/metabolism , Monocrotaline/analogs & derivatives , Monocrotaline/pharmacology , Pulmonary Arterial Hypertension/genetics , Pulmonary Artery/pathology , Rats, Sprague-Dawley , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
11.
BMC Cancer ; 17(1): 805, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29191172

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) exhibit essential regulatory functions related to cell growth, apoptosis, development and differentiation. Dysregulated expression of miRNAs is associated with a wide variety of human diseases. As such miRNA signatures are valuable as biomarkers for disease and for making treatment decisions. Hepatitis B virus (HBV) is a major risk factor for hepatocellular carcinoma (HCC). Here we screened for miRNAs in chronic HBV associated HCC. METHODS: To determine the miRNAs in HCC occurrence associated with HBV infection, we analyzed global miRNA expression profiles in 12 pairs of HCC and adjacent matched non-HCC tissues from HBV-positive and HBV-negative patients using microarray analyses. The microarray result was validated by real-time PCR in 32 HBV-positive and 24 HBV-negative patient HCC samples. The potential candidate target genes of the miRNAs were predicted by miRWalk software. Genes simultaneously predicted as targets by two or more miRNAs were subjected to GO and KEGG pathway analysis. The miRNA regulatory network analysis was performed using the Ingenuity Pathway Analysis (IPA) software. RESULTS: Eight miRNAs (miR-223, miR-98, miR-15b, miR-199a-5p, miR-19b, miR-22, miR-451, and miR-101) were involved in HBV-unrelated HCC, 5 miRNAs (miR-98, miR-375, miR-335, miR-199a-5p, and miR-22) were involved in HBV infection, and 7 miRNAs (miR-150, miR-342-3p, miR-663, miR-20b, miR-92a-3p, miR-376c-3p and miR-92b) were specifically altered in HBV-related HCC. Gene Ontology and KEGG analyses predict that these HBV-related HCC miRNAs are involved in the regulation of: transcription, RNA polymerase II promoter, phosphorylation of proteins through MAPK signaling pathway, focal adhesion, and actin cytoskeleton. IPA analysis also suggest that these miRNAs act on AGO2, TP53, CCND1, and 11 other genes that significantly influence HCC occurrence and HBV infection. CONCLUSION: Our data indicates that the unique 7 miRNAs expression signature could be involved in the development HBV- related HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Profiling/methods , Hepatitis B/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Carcinoma, Hepatocellular/virology , Computational Biology , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Liver Neoplasms/virology , Oligonucleotide Array Sequence Analysis , Software
12.
Cell Physiol Biochem ; 44(5): 1923-1938, 2017.
Article in English | MEDLINE | ID: mdl-29224008

ABSTRACT

BACKGROUND/AIMS: Glial cell line-derived neurotrophic factor (GDNF) is an important factor promoting invasive glioma growth. This study was performed to reveal a unique mechanism of glioma cell proliferation and migration. METHODS: Human U251 glioma cells were used to screen the optimal GDNF concentration and treatment time to stimulate proliferation and migration. MicroRNA (MiRNA) expression profiles were detected by microarray and confirmed by real-time polymerase chain reaction (PCR). The target genes of differentially expressed miRNAs were predicted by miRWalk, and those targeted by multiple miRNAs were screened with Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. A regulatory miRNA network was constructed using ingenuity pathway analysis (IPA). Target gene expression of differentially expressed miRNAs was examined by real-time PCR or mRNA microarray. RESULTS: The results show that 50 ng/mL GDNF for 24 h significantly promotes U251 glioma cell proliferation and migration (P < 0.05). Seven miRNAs (hsa-miR-194-5p, hsa-miR-152-3p, hsa-miR-205-5p, hsa-miR-629-5p, hsa-miR-3609, hsa-miR-183-5p, and hsa-miR-487b-3p) were significantly up-regulated after GDNF treatment (P < 0.05). These miRNAs are primarily involved in signal transduction, cell adhesion and cell cycle through mitogen-activated protein kinase (MAPK) signaling, focal adhesion and glioma signal pathways. Five of these miRNAs (hsa-miR-194-5p, hsa-miR-152-3p, hsa-miR-205-5p, hsa-miR-183-5p, and hsa-miR-487b-3p) co-regulate TP53 and Akt. mRNA expression levels of four genes co-targeted by two or more up-regulated miRNAs were significantly decreased after GDNF treatment (P < 0.05). CONCLUSION: GDNF treatment of U251 glioma cells significantly increased the expression of seven miRNAs involved in cell adhesion and the cell cycle.


Subject(s)
Cell Proliferation/drug effects , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , MicroRNAs/metabolism , Cell Adhesion/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cluster Analysis , Glioma/metabolism , Glioma/pathology , Humans , MicroRNAs/genetics , Mitogen-Activated Protein Kinases/metabolism , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects
13.
Toxicol Lett ; 278: 9-17, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28666825

ABSTRACT

Lyn is a proto-oncogene overexpressed and constitutively activated in lymphoma, and plays an important role in lymphoma initiation and malignant progression. Hence, the oncogenic Lyn has recently been targeted for novel anti-lymphoma drug discovery; however, the effective Lyn-targeted drug for lymphoma treatment with low toxicity is absent in the clinical setting. The goal of this study is to explore powerful and low toxic Lyn-targeted anti-lymphoma agent. Here we show that triptonide, a small molecule purified from the herb Tripterygium wilfordii Hook F, potently inhibits the proliferation of human B-lymphoma Raji and T-lymphoma Jurkat cells with IC50 of 5.7nM and 4.8nM, respectively. Strikingly, triptonide at a dose of 5mg/kg/day almost completely inhibited the lymphoma growth in human lymphoma cells-xenografted mice without obvious side effects, particularly; the tumors in 6 mice among the 8 xenografted mice were completely eradicated in vivo. Cell biological studies showed that triptonide at the doses of 2.5-10nM notably suppressed B-lymphoma cell colony-forming capability, and that triptonide at the dose of 20nM promoted apoptosis through activation of PARP and caspase 3, but reduction of BCL2 protein levels in the lymphoma cells. Molecular studies revealed that triptonide markedly inhibited oncogenic Lyn transcription through suppressing the promoter activity of the gene, and that it remarkably reduced both total and phosphorylated Lyn proteins, and diminished Lyn downstream ERK and ATK signal pathways. Additionally, triptonide significantly enhanced p38 phosphorylation. Together, triptonide exerts potent anti-lymphoma effect with low toxicity mainly through inhibition of proto-oncogene Lyn transcription and suppression of Lyn downstream ERK and ATK signal pathways, providing an attractive drug candidate for development of novel anti-lymphoma therapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Lymphoma/drug therapy , Transcription, Genetic/drug effects , Triterpenes/pharmacology , src-Family Kinases/metabolism , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Inhibitory Concentration 50 , Jurkat Cells , Lymphoma/enzymology , Lymphoma/genetics , Lymphoma/pathology , Mice, Inbred NOD , Mice, SCID , Phosphorylation , Proto-Oncogene Mas , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Time Factors , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/metabolism , src-Family Kinases/genetics
14.
Regul Toxicol Pharmacol ; 87: 88-94, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28476554

ABSTRACT

4-Nitrophenol (PNP), a well-established human carcinogen, has been proven to have detrimental effects on reproductive system of male rats in previous studies. The molecular mechanisms involved PNP-induced damage remain to be established. Autophagy can exert protective effects on various cytotoxic factors that induce injury. In the present study, we aim to investigate whether autophagy is induced by PNP and the function of autophagy in PNP-induced injury in NHPrE1, a normal human prostate epithelial progenitor cell line. Our results indicate that PNP induced oxidative stress as evidenced by increased MDA levels and decreased activity of SOD and GSH-Px. PNP also increased apoptosis of NHPrE1 cells as evidenced by western blot and Hoechst 33258 staining and activated autophagy in NHPrE1 cells detected by RT-PCR and western blot. Inhibition of autophagy by 3-MA further increased PNP-induced oxidative stress and apoptosis of NHPrE1 cells. We also found that PNP-induced apoptosis was suppressed by N-acetylcysteine, suggesting oxidative stress may play an important role in PNP cytotoxicity. Furthermore, phosphorylation of mTOR protein was inhibited by PNP, indicating that PNP might induce autophagy in NHPrE1 cells via inhibiting mTOR pathway. In conclusion, these results suggest that activation of autophagy should play a protective role in PNP-induced oxidative stress and apoptosis of NHPrE1 cells, which might be mediated through mTOR pathway.


Subject(s)
Autophagy/drug effects , Nitrophenols/toxicity , Oxidative Stress/drug effects , Prostate/cytology , Stem Cells/drug effects , Animals , Apoptosis , Humans , Male , Rats
15.
Stem Cells Int ; 2016: 3162363, 2016.
Article in English | MEDLINE | ID: mdl-26783401

ABSTRACT

The development of induced pluripotent stem cells (iPSCs) has enabled study of the mechanisms underlying cellular reprogramming. Here, we have studied the dynamic distribution of H2A.Z during induced reprogramming with chromatin immunoprecipitation deep sequencing (ChIP-Seq). We found that H2A.Z tended to accumulate around transcription start site (TSS) and incorporate in genes with a high transcriptional activity. GO analysis with H2A.Z incorporated genes indicated that most genes are involved in chromatin assembly or disassembly and chromatin modification both in MEF and Day 7 samples, not in iPSCs. Furthermore, we detected the highest level of incorporation of H2A.Z around TSS in Day 7 samples compared to MEFs and iPSCs. GO analysis with only incorporated genes in Day 7 also displayed the function of chromatin remodeling. So, we speculate H2A.Z may be responsible for chromatin remodeling to enhance the access of transcription factors to genes important for pluripotency. This study therefore provides a deeper understanding of the mechanisms underlying induced reprogramming.

16.
PLoS One ; 9(7): e101588, 2014.
Article in English | MEDLINE | ID: mdl-25003630

ABSTRACT

BACKGROUND: Recent evidence shows that long non-coding RNA (LncRNA) play important regulatory roles in many biology process, including cell development, activation and oncogenesis. However, the roles of these LncRNAs in the development and activation of CD4+ T cells, which is an important component of immune response, remain unknown. RESULTS: To predict the function of LncRNA in the development and activation of CD4+ T cells, first, we examined the expression profiles of LncRNAs and mRNAs in CD4-CD8- (DN), CD4+CD8+ (DP), CD4+CD8-, and activated CD4+CD8- T cells in a microarray analysis and verified these results by real time PCRs (qPCR). We found that the expression of hundreds of LncRNAs significantly changed in each process of developmental transition, including DN into DP, DP into CD4+CD8-, and CD4+CD8- into activated CD4+ T cells. A Kendall distance analysis suggested that the expression of LncRNAs in DN, DP, CD4+CD8- T cells and activated CD4+ T cells were correlated with the expression of mRNAs in these T cells. The Blat algorithm and GO analysis suggested that LncRNAs may exert important roles in the development and activation of CD4+ T cells. These roles included proliferation, homeostasis, maturation, activation, migration, apoptosis and calcium ion transportation. CONCLUSION: The present study found that the expression profiles of LncRNAs in different stages of CD4+ T cells are distinguishable. LncRNAs are involved in the key biological process in CD4+ T cell development and activation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Lymphocyte Activation/genetics , RNA, Long Noncoding/genetics , Transcription, Genetic , Animals , CD4-Positive T-Lymphocytes/cytology , Cell Differentiation/genetics , Cluster Analysis , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Mice , RNA, Messenger/genetics
17.
Cell Reprogram ; 16(3): 196-205, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24802333

ABSTRACT

Induced pluripotent stem cells (iPSCs) have potential applications in the restoration of fertility, regenerative medicine, and animal biotechnology. In this study, we present the induction of iPSCs from mouse Sertoli cells (SCs) by introducing four factors--Oct4, Sox2, Klf4, and c-Myc. As early as day 3 after induction, expression of these factors was detected and typical embryonic stem-like cells began to form. On day 18, these exogenous genes were silenced and colonies were selected according to morphological characteristics. The iPSCs induced from SCs, termed SCiPSCs, strongly expressed pluripotent markers, showed a normal karyotype, and had proliferation and differentiation characteristics similar to those of embryonic stem cells (ESCs), both in vitro and in vivo. Furthermore, exposure of SCiPSCs to nitric oxide (NO) allowed them to maintain pluripotency through the activation of the pluripotent genes Oct4 and Sox2 and upregulation of Nanog expression. Moreover, NO prevented SCiPSCs from undergoing apoptosis by activating the antiapoptotic genes Bcl2 and Bcl2lll, downregulating the proapoptotic genes Bak1 and Casp7, and blocking the activation of the proapoptotic gene Bac. These effects were reversed by exposure to l-NG-monomethylarginine (l-NMMA), a NO inhibitor. These data demonstrate that iPSCs can be generated from SCs and that the self-renewal and pluripotency of SCiPS cells can be maintained in the presence of NO.


Subject(s)
Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , Sertoli Cells/cytology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Base Sequence , DNA Primers , Induced Pluripotent Stem Cells/drug effects , Kruppel-Like Factor 4 , Male , Mice , Nitric Oxide/pharmacology , Real-Time Polymerase Chain Reaction
18.
Exp Cell Res ; 326(2): 259-66, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24780820

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population and show significant expansion under pathological conditions. microRNA plays important roles in many biological processes, whether microRNAs have a function in the expansion of MDSCs is still not very clear. In this study, miR-34a overexpression can induce the expansion of MDSCs in bone marrow chimera and transgenic mice model. The experimental results suggest that miR-34a inhibited the apoptosis of MDSCs but did not affect the proliferation of MDSCs. The distinct mRNA microarray profiles of MDSCs of wild type and miR-34a over-expressing MDSCs combined with the target prediction of miR-34a suggest that miR-34a may target genes such as p2rx7, Tia1, and plekhf1 to inhibit the apoptosis of MDSCs. Taken together, miR-34a contributes to the expansion of MDSCs by inhibiting the apoptosis of MDSCs.


Subject(s)
Apoptosis/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Myeloid Progenitor Cells/cytology , Myeloid Progenitor Cells/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelopoiesis/genetics , Oligonucleotide Array Sequence Analysis , RNA-Binding Proteins/genetics , Receptors, Purinergic P2X7/genetics , Spleen/cytology , Spleen/metabolism , T-Cell Intracellular Antigen-1 , Thymus Gland/cytology , Thymus Gland/metabolism , Up-Regulation
19.
Reproduction ; 148(1): 33-41, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24686457

ABSTRACT

MicroRNAs (miRNAs) are non-coding RNA molecules of about 22 nucleotides that involved in post-transcriptional gene regulation. Evidence indicates that miRNAs play essential roles in endometriosis, pre-eclampsia, infertility and other reproductive system diseases. However, whether miRNAs are involved in recurrent spontaneous abortion (RSA) is unclear. In this work, we analysed the miRNA expression profiles in six pairs of villus or decidua from RSA patients and normal pregnancy (NP) women using a human miRNA microarray. Some of the chip results were confirmed by RT-qPCR. In the villi of RSA patients, expression of hsa-miR-184, hsa-miR-187 and hsa-miR-125b-2 was significantly higher, while expression of hsa-miR-520f, hsa-miR-3175 and hsa-miR-4672 was significantly lower, comparing with those of NP control. As well, a total of five miRNAs (hsa-miR-517c, hsa-miR-519a-1, hsa-miR-522, hsa-miR-520h and hsa-miR-184) were upregulated in the decidua of RSA patients. The target genes of these differentially expressed miRNAs were predicted by miRWalk, and we speculate a network of miRNA regulating RSA by target genes function on adhesion, apoptosis and angiogenesis. Our study may help clarify the molecular mechanisms which are involved in the progression of RSA, and provide a reference for future research.


Subject(s)
Abortion, Spontaneous/genetics , Chorionic Villi/chemistry , Gene Expression Profiling/methods , MicroRNAs/analysis , Oligonucleotide Array Sequence Analysis , Placenta/chemistry , Abortion, Spontaneous/pathology , Abortion, Spontaneous/physiopathology , Adult , Case-Control Studies , Chorionic Villi/pathology , Chorionic Villi/physiopathology , Cluster Analysis , Female , Gene Expression Regulation , Genetic Markers , Gestational Age , Humans , Placenta/pathology , Placenta/physiopathology , Pregnancy
20.
Mol Reprod Dev ; 80(12): 1009-17, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24123501

ABSTRACT

Embryonic stem cells (ESCs) are a powerful model for genetic engineering, studying developmental biology, and modeling disease. To date, ESCs have been established from the mouse (Evans and Kaufman, 1981, Nature 292:154-156), non-human primates (Thomson et al., , Proc Nat Acad Sci USA 92:7844-7848), humans (Thomson et al., 1998, Science 282:1145-1147), and rats (Buehr et al., , Cell 135:1287-1298); however, the derivation of ESCs from domesticated ungulates such as goats, sheep, cattle, and pigs have not been successful. Alternatively, induced pluripotent stem cells (iPSCs) can be generated by reprogramming somatic cells with several combinations of genes encoding transcription factors (OCT3/4, SOX2, KLF4, cMYC, LIN28, and NANOG). To date, iPSCs have been isolated from various species, but only limited information is available regarding goat iPSCs (Ren et al., 2011, Cell Res 21:849-853). The objectives of this study were to generate goat iPSCs from fetal goat primary ear fibroblasts using lentiviral transduction of four human transcription factors: OCT4, SOX2, KLF4, and cMYC. The goat iPSCs were successfully generated by co-culture with mitomycin C-treated mouse embryonic fibroblasts using medium supplemented with knockout serum replacement and human basic fibroblast growth factor. The goat iPSCs colonies are flat, compact, and closely resemble human iPSCs. They have a normal karyotype; stain positive for alkaline phosphatase, OCT4, and NANOG; express endogenous pluripotency genes (OCT4, SOX2, cMYC, and NANOG); and can spontaneously differentiate into three germ layers in vitro and in vivo.


Subject(s)
Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/cytology , Alkaline Phosphatase/biosynthesis , Animals , Cell Differentiation , Cells, Cultured , Coculture Techniques , Fibroblast Growth Factor 2/pharmacology , Fibroblasts/cytology , Goats , HEK293 Cells , Homeodomain Proteins/biosynthesis , Humans , Karyotype , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Inbred C57BL , Mitomycin/pharmacology , Nanog Homeobox Protein , Octamer Transcription Factor-3/biosynthesis , Octamer Transcription Factor-3/genetics , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , SOXB1 Transcription Factors/biosynthesis , SOXB1 Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...