Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
J Virol ; 95(24): e0139921, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34586865

ABSTRACT

Targeting host factors is a promising strategy to develop broad-spectrum antiviral drugs. Drugs targeting anti-apoptotic Bcl-2 family proteins that were originally developed as tumor suppressors have been reported to inhibit multiplication of different types of viruses. However, the mechanisms whereby Bcl-2 inhibitors exert their antiviral activity remain poorly understood. In this study, we have investigated the mechanisms by which obatoclax (OLX) and ABT-737 Bcl-2 inhibitors exhibited a potent antiviral activity against the mammarenavirus lymphocytic choriomeningitis virus (LCMV). OLX and ABT-737 potent anti-LCMV activity was not associated with their proapoptotic properties but rather with their ability to induce cell arrest at the G0/G1 phase. OLX- and ABT-737-mediated inhibition of Bcl-2 correlated with reduced expression levels of thymidine kinase 1 (TK1), cyclin A2 (CCNA2), and cyclin B1 (CCNB1) cell cycle regulators. In addition, small interfering RNA (siRNA)-mediated knockdown of TK1, CCNA2, and CCNB1 resulted in reduced levels of LCMV multiplication. The antiviral activity exerted by Bcl-2 inhibitors correlated with reduced levels of viral RNA synthesis at early times of infection. Importantly, ABT-737 exhibited moderate efficacy in a mouse model of LCMV infection, and Bcl-2 inhibitors displayed broad-spectrum antiviral activities against different mammarenaviruses and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Our results suggest that Bcl-2 inhibitors, actively being explored as anticancer therapeutics, might be repositioned as broad-spectrum antivirals. IMPORTANCE Antiapoptotic Bcl-2 inhibitors have been shown to exert potent antiviral activities against various types of viruses via mechanisms that are currently poorly understood. This study has revealed that Bcl-2 inhibitors' mediation of cell cycle arrest at the G0/G1 phase, rather than their proapoptotic activity, plays a critical role in blocking mammarenavirus multiplication in cultured cells. In addition, we show that Bcl-2 inhibitor ABT-737 exhibited moderate antimammarenavirus activity in vivo and that Bcl-2 inhibitors displayed broad-spectrum antiviral activities against different mammarenaviruses and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Our results suggest that Bcl-2 inhibitors, actively being explored as anticancer therapeutics, might be repositioned as broad-spectrum antivirals.


Subject(s)
Apoptosis , Arenaviridae/drug effects , COVID-19 Drug Treatment , Proto-Oncogene Proteins c-bcl-2/metabolism , A549 Cells , Animals , Antiviral Agents/pharmacology , Apoptosis Regulatory Proteins/pharmacology , Biphenyl Compounds/pharmacology , COVID-19/virology , Cell Cycle , Cell Cycle Checkpoints/drug effects , Cells, Cultured/drug effects , Cells, Cultured/virology , Chlorocebus aethiops , Cyclin A2/biosynthesis , Cyclin B1/biosynthesis , G1 Phase , Humans , Indoles/pharmacology , Mice , Mice, Inbred C57BL , Nitrophenols/pharmacology , Piperazines/pharmacology , Pyrroles/pharmacology , Resting Phase, Cell Cycle , SARS-CoV-2 , Sulfonamides/pharmacology , Thymidine Kinase/biosynthesis , Vero Cells
2.
Appl Immunohistochem Mol Morphol ; 29(1): 56-61, 2021 01.
Article in English | MEDLINE | ID: mdl-32134755

ABSTRACT

Helicobacter pylori infection is strongly associated with primary gastric diseases, such as extranodal mucosa-associated lymphoid tissue (MALT) lymphoma, diffuse large B-cell lymphoma (DLBCL) with histologic evidence of MALT origin, and gastric carcinoma. The cytotoxin-associated gene A (CagA) protein behaves as a bacterial oncoprotein, promoting tumorigenesis via dysregulation of the phosphatidylinositol 3-kinase/AKT pathway (PI3K/AKT). We investigated the molecular mechanisms of PI3K/AKT pathway dysregulation in H. pylori-induced MALT and DLBCL gastric lymphoma. Immunohistochemical assays for CagA, phospho(p)-S473-AKT, PTEN, SHIP, and cyclin A2 proteins were performed on samples from 23 patients with H. pylori-positive MALT lymphoma and 16 patients with H. pylori-positive gastric DLBCL. We showed that CagA localization is correlated with the activation of the AKT pathway in both MALT and DLBCL lymphoma cells. Interestingly, we found a close association between the loss of PTEN, the overexpression of cyclin A2, and the phosphorylation of AKT in gastric MALT and DLBCL tumor cells.


Subject(s)
Cyclin A2/biosynthesis , Gene Expression Regulation, Neoplastic , Helicobacter Infections/metabolism , Helicobacter pylori/metabolism , Lymphoma, B-Cell, Marginal Zone/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , PTEN Phosphohydrolase/deficiency , Signal Transduction , Stomach Neoplasms/metabolism , Up-Regulation , Cyclin A2/genetics , Female , Helicobacter Infections/genetics , Helicobacter Infections/pathology , Humans , Lymphoma, B-Cell, Marginal Zone/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Male , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Stomach Neoplasms/genetics
3.
Sci Rep ; 10(1): 1140, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31980707

ABSTRACT

PDZRN3 is a PDZ domain-containing RING-finger family protein that functions in various developmental processes. We previously showed that expression of PDZRN3 is induced together with that of MyoD during the early phase of skeletal muscle regeneration in vivo. We here show that PDZRN3 suppresses apoptosis and promotes proliferation in myoblasts in a manner dependent on cyclin A2. Depletion of PDZRN3 in mouse C2C12 myoblasts by RNA interference reduced the proportion of Ki-67-positive cells and the level of Akt phosphorylation, implicating PDZRN3 in regulation of both cell proliferation and apoptosis. Exposure of C2C12 cells as well as of C3H10T1/2 mesenchymal stem cells and NIH-3T3 fibroblasts to various inducers of apoptosis including serum deprivation resulted in a greater increase in the amount of cleaved caspase-3 in PDZRN3-depleted cells than in control cells. The abundance of cyclin A2 was reduced in PDZRN3-depleted C2C12 myoblasts, as was that of Mre11, which contributes to the repair of DNA damage. Overexpression of cyclin A2 restored the expression of Mre11 and Ki-67 as well as attenuated caspase-3 cleavage in PDZRN3-depleted cells deprived of serum. These results indicate that PDZRN3 suppresses apoptosis and promotes proliferation in myoblasts and other cell types by maintaining cyclin A2 expression.


Subject(s)
Apoptosis/physiology , Cyclin A2/biosynthesis , Myoblasts/metabolism , Ubiquitin-Protein Ligases/physiology , Animals , Cell Adhesion , Cell Cycle , Cell Division , Cells, Cultured , Cyclin A2/genetics , DNA Damage , Down-Regulation , Gene Expression Regulation , Genomic Instability , Mesenchymal Stem Cells/metabolism , Mice , Myoblasts/cytology , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology
4.
Pak J Pharm Sci ; 33(5): 2083-2088, 2020 Sep.
Article in English | MEDLINE | ID: mdl-33824116

ABSTRACT

The bioengineered luciferase reporter has been widely used for monitoring of a variety of molecular events in living cells because of their ability to provide highly sensitive quantitation with broad linearity. In the present study, we made a cyclin A2-luciferase (CYCA-Luc) fusion protein and examined the utility of this optical reporter for monitoring G2-phase cell cycle arrest in living animals. In vitro luciferase assay and in vivo bioluminescence imaging assay showed that the lithium chloride (LiCl), G2-phase-specific drug, induced G2-phase arrest of cell cycle and increased the activity of this reporter under in vitro or in vivo conditions, and this reporter can also be potentially used in high-throughput screening efforts aimed at discovering novel anti-cancer drugs that will cause cell cycle arrest at the G2-phase in cultivated cell lines and animal models.


Subject(s)
Cyclin A2/genetics , G2 Phase Cell Cycle Checkpoints , Genes, Reporter , Luciferases/genetics , Optical Imaging , Uterine Cervical Neoplasms/pathology , Animals , Cyclin A2/biosynthesis , Female , G2 Phase Cell Cycle Checkpoints/drug effects , HeLa Cells , High-Throughput Screening Assays , Humans , Lithium Chloride/pharmacology , Luciferases/biosynthesis , Luminescent Measurements , Mice, Inbred BALB C , Mice, Nude , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Xenograft Model Antitumor Assays
5.
Pathology ; 51(5): 481-486, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31230818

ABSTRACT

Carcinoid classification in the lung is still based on morphological criteria. Although there are many studies investigating the role of Ki-67 proliferation index in the classification of lung neuroendocrine tumours, it is still not used in routine diagnostics. Interestingly, cyclins, which have a crucial role in controlling the cell cycle, have not been thoroughly studied in lung neuroendocrine tumours. The aim of our study was to investigate the correlation of cyclin A2 and B1 expression with prognosis, Ki-67 proliferation index, and carcinoid morphology. A cohort of 134 resected typical and atypical carcinoids was stained with antibodies against Ki-67, cyclin A2 and B1. The positive nuclear reaction was assessed in hot spot areas and expressed as the percentage of tumour cells. Univariate analyses found the highest relative hazard between low and high cyclin A2 expression both with respect to overall survival [hazard ratio (HR)=16; 95% confidence interval (CI) 4.8-51; p=0.0000054], and relapse (HR=8; 95% CI 3.1-21; p=0.00002). In multivariate analysis for overall survival cyclin A2 (HR=10; 95% CI 2.5->100; p=0.0082) and B1 (HR=6.5; 95% CI 1.5-35; p=0.02) remained significant when adjusted for other risk factors, whereas Ki-67 was no longer significant (HR=0.64; 95% CI 0.003-5.5; p=0.65). This suggests that Ki-67 is closer to conventional risk factors for survival than cyclin A2 and B1. Furthermore, the analysis revealed 4 mitoses per 2 mm2 as a more powerful prognostic cut-off than currently accepted 2 mitoses. We have clearly demonstrated that application of cyclin A2 and cyclin B1 might bring additional value regarding the overall and progression-free survival of patients with carcinoids of the lung.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoid Tumor/pathology , Cyclin A2/biosynthesis , Cyclin B1/biosynthesis , Lung Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Carcinoid Tumor/mortality , Female , Humans , Lung Neoplasms/mortality , Male , Middle Aged , Prognosis , Progression-Free Survival , Young Adult
6.
Mol Carcinog ; 58(7): 1248-1259, 2019 07.
Article in English | MEDLINE | ID: mdl-31100197

ABSTRACT

Purpurogallin is a natural compound that is extracted from nutgalls and oak bark and it possesses antioxidant, anticancer, and anti-inflammatory properties. However, the anticancer capacity of purpurogallin and its molecular target have not been investigated in esophageal squamous cell carcinoma (ESCC). Herein, we report that purpurogallin suppresses ESCC cell growth by directly targeting the mitogen-activated protein kinase kinase 1/2 (MEK1/2) signaling pathway. We found that purpurogallin inhibits anchorage-dependent and -independent ESCC growth. The results of in vitro kinase assays and cell-based assays indicated that purpurogallin also strongly attenuates the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway and also directly binds to and inhibits MEK1 and MEK2 activity. Furthermore, purpurogallin contributed to S and G2 phase cell cycle arrest by reducing cyclin A2 and cyclin B1 expression and also induced apoptosis by activating poly (ADP ribose) polymerase (PARP). Notably, purpurogallin suppressed patient-derived ESCC tumor growth in an in vivo mouse model. These findings indicated that purpurogallin is a novel MEK1/2 inhibitor that could be useful for treating ESCC.


Subject(s)
Antineoplastic Agents/pharmacology , Benzocycloheptenes/pharmacology , Esophageal Neoplasms/drug therapy , Esophageal Squamous Cell Carcinoma/drug therapy , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cyclin A2/biosynthesis , Cyclin B1/biosynthesis , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Mice , Plant Preparations/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , S Phase Cell Cycle Checkpoints/drug effects , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
7.
Int J Cancer ; 142(9): 1938-1951, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29238973

ABSTRACT

The overall 5-year survival rate of patients with human pancreatic cancer remains less than 8% because of its aggressive growth, early metastasis and resistance to conventional chemoradiotherapy. It is essential to develop innovative and effective therapeutic agents to improve its prognosis. Demethylzeylasteral (ZST93) is a novel triterpenoid monomer extracted from the xylem of Tripterygium roots. Our study aimed to assess the effects of ZST93 on cell proliferation and its role in the chemosensitivity to gemcitabine in human pancreatic cancer cells. The effects of ZST93 on cancer cell proliferation, cell cycle distribution, apoptosis and autophagy were evaluated in various human pancreatic cancer cell lines, and the antitumor effects of ZST93 alone and in combination with gemcitabine were identified in a xenograft mouse model. The results showed that ZST93 could inhibit the proliferation of pancreatic cancer cells and arrest cell cycle at G0/G1 phase by regulating the expression of Cyclin D1 and Cyclin A2. Moreover, ZST93 killed pancreatic cancer cells through two different mechanisms: inducing autophagic cell death at low concentrations and apoptotic cell death at high concentrations. Furthermore, ZST93 could enhance the chemosensitivity of pancreatic cancer cells to gemcitabine both in vitro and in vivo through modulation of the cross talk between autophagy and apoptosis. ZST93 is a potential therapeutic agent for developing novel therapeutic strategies in human pancreatic cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/drug therapy , Triterpenes/pharmacology , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Cyclin A2/biosynthesis , Cyclin A2/genetics , Cyclin D1/biosynthesis , Cyclin D1/genetics , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Drug Synergism , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Random Allocation , Triterpenes/administration & dosage , Xenograft Model Antitumor Assays , Gemcitabine
8.
Int J Mol Med ; 40(3): 623-630, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28713904

ABSTRACT

Nitrogen-containing bisphosphonates (N-BPs) are the most widely used anti-resorptive agents in the treatment of bone-related diseases. N-BPs inhibit bone resorption by specifically targeting osteoclasts, bone-resorbing cells. However, soft tissue toxicity, such as oral or gastrointestinal (GI) ulcerations has frequently been reported in N-BP users, suggesting that N-BPs may also directly target cells other than osteoclasts. Previously, we reported that BPs inhibit proliferation without inducing the apoptosis of normal human oral keratinocytes (NHOKs). However, the molecular mechanisms through which N-BPs inhibit the proliferation of NHOKs are not yet fully understood. In this study, we performed gene expression profiling in N-BP-treated NHOKs and identified cyclin A2 as one of the most commonly downregulated genes. When the NHOKs were treated with N-BPs, we found that the level of cyclin A2 was suppressed in a dose- and time-dependent manner. In addition, the protein level of cyclin A2 was also significantly lower in oral epithelial cells in N-BP-treated oral mucosal tissue constructs. Cyclin A2 promoter reporter assay revealed that N-BPs inhibited the luciferase activity, indicating that the inhibition of cyclin A2 expression occurs at the transcriptional level. Furthermore, N-BPs did not alter the expression of cyclin A2 in normal human oral fibroblasts (NHOFs), suggesting that the effect of N-BPs on cyclin A2 expression may be cell-type specific. Thus, the findings of our study demonstrate that the inhibition of NHOK proliferation by N-BPs is mediated, at least in part, by the suppression of cyclin A2 expression at the transcriptional level, which may explain the underlying mechanisms of soft tissue toxicity by N-BPs.


Subject(s)
Cell Proliferation/drug effects , Cyclin A2/biosynthesis , Diphosphonates/pharmacology , Gene Expression Regulation/drug effects , Keratinocytes/metabolism , Transcription, Genetic/drug effects , Humans , Keratinocytes/cytology , Mouth Mucosa
9.
PLoS Genet ; 13(4): e1006704, 2017 04.
Article in English | MEDLINE | ID: mdl-28380054

ABSTRACT

The meiosis-specific chromosomal events of homolog pairing, synapsis, and recombination occur over an extended meiotic prophase I that is many times longer than prophase of mitosis. Here we show that, in mice, maintenance of an extended meiotic prophase I requires the gene Meioc, a germ-cell specific factor conserved in most metazoans. In mice, Meioc is expressed in male and female germ cells upon initiation of and throughout meiotic prophase I. Mouse germ cells lacking Meioc initiate meiosis: they undergo pre-meiotic DNA replication, they express proteins involved in synapsis and recombination, and a subset of cells progress as far as the zygotene stage of prophase I. However, cells in early meiotic prophase-as early as the preleptotene stage-proceed to condense their chromosomes and assemble a spindle, as if having progressed to metaphase. Meioc-deficient spermatocytes that have initiated synapsis mis-express CYCLIN A2, which is normally expressed in mitotic spermatogonia, suggesting a failure to properly transition to a meiotic cell cycle program. MEIOC interacts with YTHDC2, and the two proteins pull-down an overlapping set of mitosis-associated transcripts. We conclude that when the meiotic chromosomal program is initiated, Meioc is simultaneously induced so as to extend meiotic prophase. Specifically, MEIOC, together with YTHDC2, promotes a meiotic (as opposed to mitotic) cell cycle program via post-transcriptional control of their target transcripts.


Subject(s)
Cell Cycle Proteins/genetics , Cyclin A2/biosynthesis , Meiosis/genetics , Prophase/genetics , RNA-Binding Proteins/genetics , Animals , Cell Cycle Proteins/biosynthesis , Chromosome Pairing/genetics , Cyclin A2/genetics , Gene Expression Regulation, Developmental , Male , Mice , Mitosis/genetics , RNA-Binding Proteins/metabolism , Spermatocytes , Spermatogenesis/genetics , Spermatogonia/growth & development , Spermatogonia/metabolism
10.
Platelets ; 28(5): 491-497, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27780401

ABSTRACT

Platelet rich plasma (PRP) contains various cytokines and growth factors which may be beneficial to the healing process of injured muscle. The purpose of this study is to investigate the effect and molecular mechanism of PRP releasate on proliferation of skeletal muscle cells. Skeletal muscle cells intrinsic to Sprague-Dawley rats were treated with PRP releasate. Cell proliferation was evaluated by 3-[4,5-Dimethylthiazol- 2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay and immunocytochemistry with Ki-67 stain. Flow cytometric analysis was used to evaluate the cell cycle progression. Western blot analysis was used to evaluate the protein expressions of PCNA, cyclin E1, cyclin A2, cyclin B1, cyclin dependent kinase (cdk)1 and cdk2. The results revealed that PRP releasate enhanced proliferation of skeletal muscle cells by shifting cells from G1 phase to S phase and G2/M phases. Ki-67 stain revealed the increase of proliferative capability after PRP releasate treatment. Protein expressions including cyclin A2, cyclin B1, cdk1, cdk2 and PCNA were up-regulated by PRP releasate in a dose-dependent manner. It was concluded that PRP releasate promoted proliferation of skeletal muscle cells in association with the up-regulated protein expressions of PCNA, cyclin A2, cyclin B1, cdk1 and cdk2.


Subject(s)
Cell Proliferation/drug effects , Cyclin-Dependent Kinases/biosynthesis , Cyclins/biosynthesis , Muscle, Skeletal/metabolism , Platelet-Rich Plasma , Proliferating Cell Nuclear Antigen/biosynthesis , Animals , CDC2 Protein Kinase/biosynthesis , Cyclin A2/biosynthesis , Cyclin B1/biosynthesis , Cyclin-Dependent Kinase 2/biosynthesis , Dose-Response Relationship, Drug , Muscle Proteins/biosynthesis , Muscle, Skeletal/cytology , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects
12.
BMC Cancer ; 16: 252, 2016 Mar 28.
Article in English | MEDLINE | ID: mdl-27020049

ABSTRACT

BACKGROUND: MicroRNAs have been shown to be important regulators of the immune response and the development of the immune system. It was reported that microRNA-125b (miR-125b) was down-regulated in macrophages challenged with endotoxin. However, little is known about the function and mechanism of action of miR-125b in macrophage activation. Macrophages use L-arginine to synthesize nitric oxide (NO) through inducible NO synthase (iNOS), and the released NO contributes to the tumoricidal activity of macrophages. METHODS: Luciferase reporter assays were employed to validate regulation of a putative target of miR-125b. The effect of miR-125b on endogenous levels of this target were subsequently confirmed via Western blot. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was performed to determine the expression level of miR-125b in macrophage. MTS assays were conducted to explore the impact of miR-125b overexpression on the cell viability of 4T1 cells. RESULTS: Here, we demonstrate that mmu-miR-125b overexpression suppresses NO production in activated macrophages and that LPS-activated macrophages with overexpressed mmu-miR-125b promote 4T1 tumor cell proliferation in vitro and 4T1 tumor growth in vivo. CCNA2 and eEF2K are the direct and functional targets of mmu-miR-125b in macrophages; CCNA2 and eEF2K expression was knocked down, which mimicked the mmu-miR-125b overexpression phenotype. CONCLUSIONS: These data suggest that mmu-miR-125b decreases NO production in activated macrophages at least partially by suppressing eEF2K and CCNA2 expression.


Subject(s)
Cyclin A2/genetics , Elongation Factor 2 Kinase/genetics , MicroRNAs/biosynthesis , Nitric Oxide/biosynthesis , Cell Line, Tumor , Cell Proliferation/genetics , Cyclin A2/biosynthesis , Elongation Factor 2 Kinase/biosynthesis , Endotoxins/toxicity , Gene Expression Regulation , Humans , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , MicroRNAs/immunology , Nitric Oxide/immunology , Nitric Oxide Synthase Type II/genetics
13.
Mol Med Rep ; 13(2): 1275-80, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26676380

ABSTRACT

Osteosarcoma is the most common type of primary malignant tumor of the bone. However, mechanisms underlying osteosarcoma cell proliferation are poorly understood. The present study shows that RBEL1, a newly identified Rab-like GTPase, may be a key regulator of osteosarcoma cell proliferation. Knockdown of RBEL1 in osteosarcoma cells resulted in impaired colony formation and cell proliferation. Cell cycle analysis suggested that RBEL1 depletion induced G1-S arrest in osteosarcoma cells. Furthermore, it was demonstrated that retinoblastoma 1 (Rb) was upregulated and activated following RBEL1 knockdown. In addition, Rb inhibitory downstream targets, such as cyclin A2, cyclin D1, c-Myc and cyclin-dependent kinase 2, were downregulated. Rb knockdown reversed RBEL1 depletion-induced tumor suppressive effects. In conclusion, the present results suggest that RBEL1 modulates cell proliferation and G1­S transition by inhibiting Rb in osteosarcoma. These results suggest a potential therapeutic target in osteosarcoma.


Subject(s)
Cell Proliferation/genetics , Osteosarcoma/genetics , Retinoblastoma Protein/biosynthesis , ras Proteins/genetics , Cell Line, Tumor , Cyclin A2/biosynthesis , Cyclin D1/biosynthesis , Cyclin-Dependent Kinase 2/biosynthesis , Gene Knockdown Techniques , Humans , Osteosarcoma/pathology , Phosphorylation , Proto-Oncogene Proteins c-myc/biosynthesis , Retinoblastoma Protein/genetics , ras Proteins/biosynthesis
14.
PLoS One ; 10(8): e0135845, 2015.
Article in English | MEDLINE | ID: mdl-26274615

ABSTRACT

GNL3L is an evolutionarily conserved high molecular weight GTP binding nucleolar protein belonging to HSR1-MMR1 subfamily of GTPases. The present investigation reveals that GNL3L is a nucleo-cytoplasmic shuttling protein and its export from the nucleus is sensitive to Leptomycin B. Deletion mutagenesis reveals that the C-terminal domain (amino acids 501-582) is necessary and sufficient for the export of GNL3L from the nucleus and the exchange of hydrophobic residues (M567, L570 and 572) within the C-terminal domain impairs this process. Results from the protein-protein interaction analysis indicate that GNL3L interaction with CRM1 is critical for its export from the nucleus. Ectopic expression of GNL3L leads to lesser accumulation of cells in the 'G2/M' phase of cell cycle whereas depletion of endogenous GNL3L results in 'G2/M' arrest. Interestingly, cell cycle analysis followed by BrdU labeling assay indicates that significantly increased DNA synthesis occurs in cells expressing nuclear export defective mutant (GNL3L∆NES) compared to the wild type or nuclear import defective GNL3L. Furthermore, increased hyperphosphorylation of Rb at Serine 780 and the upregulation of E2F1, cyclins A2 and E1 upon ectopic expression of GNL3L∆NES results in faster 'S' phase progression. Collectively, the present study provides evidence that GNL3L is exported from the nucleus in CRM1 dependent manner and the nuclear localization of GNL3L is important to promote 'S' phase progression during cell proliferation.


Subject(s)
Cell Division/physiology , Cell Nucleus/metabolism , G2 Phase/physiology , GTP-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Active Transport, Cell Nucleus/physiology , Animals , COS Cells , Cell Nucleus/genetics , Chlorocebus aethiops , Cyclin A2/biosynthesis , Cyclin A2/genetics , Cyclin E/biosynthesis , Cyclin E/genetics , E2F1 Transcription Factor/biosynthesis , E2F1 Transcription Factor/genetics , GTP-Binding Proteins/genetics , HEK293 Cells , HeLa Cells , Hep G2 Cells , Humans , MCF-7 Cells , Mice , Mutagenesis , NIH 3T3 Cells , Nuclear Proteins/genetics , Oncogene Proteins/biosynthesis , Oncogene Proteins/genetics , Up-Regulation/physiology
15.
Pediatr Blood Cancer ; 62(8): 1414-20, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25728347

ABSTRACT

BACKGROUND: Infantile hemangioma (IH) is a benign vascular neoplasm resulting from the abnormal proliferation of endothelial cells and pericytes in infants. Propranolol, a non-selective ß-adrenergic blocker, has recently emerged as an effective therapy for IH, causing regression. However, its potential therapeutic mechanism remains largely unknown. PROCEDURE: An XPTS-1 cell line was established by isolating hemangioma-derived endothelial cells (HemECs) from a specimen of human proliferating IH. Flow cytometer assay was performed to assess the effect of propranolol on cell cycle distribution. Western blot was employed to determine changes of protein expression. Matrigel invasion and tube formation assays were used to measure invasion ability and tube formation ability, respectively. Commercial kits were employed to quantify NO and VEGF levels. RESULTS: Propranolol blocked norepinephrine-induced HemECs cell cycle progression as well as the expression of cyclin A2 and cyclin D2; whereas p21 and p27 proteins were altered conversely. Propranolol inhibited norepinephrine-induced cell invasion by reducing the expression of MMP-9, VEGF, and p-cofilin. NO and VEGF release induced by norepinephrine was decreased by propranolol pretreatment, coincident with alterations in the phosphorylation of Akt, eNOS, and VEGFR-2. Tube formation ability and subsequent levels of NO and VEGF elevated by norepinephrine were distinctively counteracted in HemECs. CONCLUSIONS: The current study demonstrated the antiangiogenic properties of propranolol in vitro and that the drug was able to induce the regression of hemangioma cells via the inhibition of cell cycle progression, invasion, and tube formation, concomitantly with decreased NO and VEGF levels through the down-regulation of the PI3K/Akt/eNOS/VEGF pathway.


Subject(s)
Adrenergic beta-Antagonists/therapeutic use , Angiogenesis Inhibitors/therapeutic use , Hemangioma/drug therapy , Neovascularization, Pathologic/drug therapy , Propranolol/therapeutic use , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chromones/pharmacology , Cinnamates/pharmacology , Cyclin A2/biosynthesis , Cyclin D2/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Endothelial Cells/metabolism , Humans , Infant , Infant, Newborn , Morpholines/pharmacology , NG-Nitroarginine Methyl Ester/pharmacology , Neoplasm Invasiveness/pathology , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type III/metabolism , Norepinephrine/pharmacology , Pericytes/metabolism , Phosphatidylinositol 3-Kinases/biosynthesis , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proliferating Cell Nuclear Antigen/biosynthesis , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor Receptor-1/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
16.
Mol Endocrinol ; 29(1): 108-20, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25387052

ABSTRACT

Type 2 diabetes mellitus (T2DM) is caused by relative insulin deficiency, subsequent to both reduced ß-cell mass and insufficient insulin secretion, and both augmenting ß-cell mass and ß-cell function are therapeutic strategies for treating T2DM. However, the relative significance of increasing ß-cell mass vs improving ß-cell stimulus secretion coupling remains unclear. We have developed a mouse model that allows proliferation of ß-cells in adult mice without affecting ß-cell function by inducible expression of the positive cell cycle regulator cyclin A2 specifically in ß-cells. In these mice, when kept on a standard diet, doubling of ß-cell mass does not result in altered glucose tolerance or glucose-stimulated circulating insulin levels. Notably, a doubling of ß-cell mass also does not confer improved glycemic control and ability of ß-cells to respond to diabetogenic high-fat diet-induced glucose intolerance. However, in high-fat diet-exposed mice, an increase in endogenous ß-cell mass confers increased potentiation of in vivo glucose-stimulated rise in circulating insulin in response to acute pharmacologic treatment with the incretin glucagon-like peptide-1 receptor agonist exendin-4. These observations indicate that increasing endogenous ß-cell mass may not be sufficient to improve glycemic control in T2DM without additional strategies to increase ß-cell stimulus secretion coupling.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Insulin/deficiency , Animals , Cell Count , Cell Proliferation , Cyclin A2/biosynthesis , Cyclin A2/genetics , Diet, High-Fat , Glucagon-Like Peptide-1 Receptor/metabolism , Glucose Intolerance/metabolism , Glucose Tolerance Test , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pancreas/cytology , Pancreas/metabolism
17.
Am J Physiol Renal Physiol ; 307(4): F396-406, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24573389

ABSTRACT

Hyperuricemia is associated with kidney complications including glomerulosclerosis and mesangial cell (MC) proliferation by poorly understood mechanisms. The present study investigated the underlying mechanisms that mediate uric acid (UA)-induced MC proliferation. A rat MC line, HBZY-1, was treated with various concentrations of UA in the presence or absence of a specific extracellular-regulated protein kinase 1/2 (ERK1/2) inhibitor (U0126), apocynin. UA dose dependently stimulated MC proliferation as shown by increased DNA synthesis and number of cells in the S and G2 phases in parallel with the upregulation of cyclin A2 and cyclin D1. In addition, UA time dependently promoted MC proliferation and significantly increased phosphorylation of ERK1/2 but not c-Jun NH2-terminal kinase and p38 MAPK in MCs as assessed by immunoblotting. Inhibition of ERK1/2 signaling via U0126 markedly blocked UA-induced MC proliferation. More importantly, UA induced intracellular reactive oxygen species (ROS) production of MCs dose dependently, which was completely blocked by apocynin, a specific NADPH oxidase inhibitor. Toll-like receptor (TLR)2 and TLR4 signaling had no effect on NADPH-derived ROS and UA-induced MC proliferation. Interestingly, pretreatment with apocynin inhibited ERK1/2 activation, the upregulation of cyclin A2 and cyclin D1, and MC proliferation. In conclusion, UA-induced MC proliferation was mediated by NADPH/ROS/ERK1/2 signaling pathway. This novel finding not only reveals the mechanism of UA-induced MC cell proliferation but also provides some potential targets for future treatment of UA-related glomerular injury.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Uric Acid/pharmacology , Acetophenones/pharmacology , Animals , Butadienes/pharmacology , Cell Proliferation/drug effects , Cyclin A2/biosynthesis , Cyclin D1/biosynthesis , Flavonoids/pharmacology , Mesangial Cells/drug effects , Mesangial Cells/metabolism , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , NADPH Oxidases/antagonists & inhibitors , Nitriles/pharmacology , Rats , Signal Transduction/drug effects , Signal Transduction/physiology , Toll-Like Receptor 2/physiology , Toll-Like Receptor 4/physiology
18.
Mol Reprod Dev ; 80(12): 1018-34, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24123525

ABSTRACT

In mammals, resting female oocytes reside in primordial ovarian follicles. An individual primordial follicle may stay quiescent for a protracted period of time before initiating follicular growth, which is also termed "activation." Female reproductive capacity is sustained by the gradual, streamlined activation of the entire population of primordial follicles, but this process also results in reproductive senescence in older animals. Based on the recent findings that genetically triggered, excessive mammalian target of rapamycin complex 1 (mTORC1) activation in mouse oocytes leads to accelerated primordial follicle activation, we examined the necessity of mTORC1 signaling in physiological primordial follicle activation. We found that induction of oocyte mTORC1 activity is associated with early follicular growth in neonatal mouse ovaries. Pharmacological inhibition of mTORC1 activity in vivo by rapamycin treatment leads to a marked, but partial, suppression of primordial follicle activation. The suppressive effect of rapamycin on primordial follicle activation was reproduced in cultured ovaries. While rapamycin did not apparently affect several plausible cellular targets in neonatal mouse ovaries, such as mTORC2, AKT, or cyclin-dependent kinase (CDK) inhibitor p27-KIP1, its inhibitory effect on Cyclin A2 gene expression implies that mTORC1 signaling in oocytes may engage a Cyclin A/CDK regulatory network that promotes primordial follicle activation. The current work strengthens the concept that mTORC1-dependent events in the oocytes of primordial follicles may represent potential targets for intervention in humans to slow the depletion of the ovarian reserve.


Subject(s)
Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/metabolism , Oocytes/cytology , Ovarian Follicle/growth & development , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Animals , Cyclin A2/biosynthesis , Cyclin A2/genetics , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , Female , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred C57BL , Multiprotein Complexes/biosynthesis , Organ Culture Techniques , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/biosynthesis , Signal Transduction , TOR Serine-Threonine Kinases/biosynthesis
19.
J Biol Chem ; 288(46): 33292-302, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24100041

ABSTRACT

Wilms' tumor 1-associating protein (WTAP) is a putative splicing regulator that is thought to be required for cell cycle progression through the stabilization of cyclin A2 mRNA and mammalian early embryo development. To further understand how WTAP acts in the context of the cellular machinery, we identified its interacting proteins in human umbilical vein endothelial cells and HeLa cells using shotgun proteomics. Here we show that WTAP forms a novel protein complex including Hakai, Virilizer homolog, KIAA0853, RBM15, the arginine/serine-rich domain-containing proteins BCLAF1 and THRAP3, and certain general splicing regulators, most of which have reported roles in post-transcriptional regulation. The depletion of these respective components of the complex resulted in reduced cell proliferation along with G2/M accumulation. Double knockdown of the serine/arginine-rich (SR)-like proteins BCLAF1 and THRAP3 by siRNA resulted in a decrease in the nuclear speckle localization of WTAP, whereas the nuclear speckles were intact. Furthermore, we found that the WTAP complex regulates alternative splicing of the WTAP pre-mRNA by promoting the production of a truncated isoform, leading to a change in WTAP protein expression. Collectively, these findings show that the WTAP complex is a novel component of the RNA processing machinery, implying an important role in both posttranscriptional control and cell cycle regulation.


Subject(s)
Alternative Splicing/physiology , Cell Division/physiology , G2 Phase/physiology , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , RNA Precursors/metabolism , Animals , Cell Cycle Proteins , Cyclin A2/biosynthesis , Cyclin A2/genetics , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred BALB C , Multiprotein Complexes/genetics , Nuclear Proteins/genetics , RNA Precursors/genetics , RNA Splicing Factors , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
20.
Carcinogenesis ; 34(6): 1304-14, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23385061

ABSTRACT

Tylophorine, a phenanthroindolizidine alkaloid, is the major medicinal constituent of herb Tylophora indica. Tylophorine treatment increased the accumulation of c-Jun protein, a component of activator protein 1 (AP1), in carcinoma cells. An in vitro kinase assay revealed that the resultant c-Jun phosphorylation was primarily mediated via activated c-Jun N-terminal protein kinase (JNK). Moreover, flow cytometry indicated that ectopically overexpressed c-Jun in conjunction with tylophorine significantly increased the number of carcinoma cells that were arrested at the G1 phase. The tylophorine-mediated downregulation of cyclin A2 protein levels is known to be involved in the primary G1 arrest. Chromatin immunoprecipitation and reporter assays revealed that tylophorine enhanced the c-Jun downregulation of the cyclin A2 promoter activity upon increased binding of c-Jun to the deregulation AP1 site and decreased binding to the upregulation activating transcription factor (ATF) site in the cyclin A2 promoter, thereby reducing cyclin A2 expression. Further, biochemical studies using pharmacological inhibitors and RNA silencing approaches demonstrated that tylophorine-mediated elevation of the c-Jun protein level occurs primarily via two discrete prolonged signaling pathways: (i) the NF-κB/PKCδ_(MKK4)_JNK cascade, which phosphorylates c-Jun and increases its stability by slowing its ubiquitination, and (ii) the PI3K_PDK1_PP2A_eEF2 cascade, which sustains eukaryotic elongation factor 2 (eEF2) activity and thus c-Jun protein translation. To the best of our knowledge, this report is the first to demonstrate the involvement of c-Jun in the anticancer activity of tylophorine and the release of c-Jun translation from a global translational blockade via the PI3K_PDK1_eEF2 signaling cascade.


Subject(s)
Alkaloids/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma/drug therapy , Indolizines/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Phenanthrenes/pharmacology , Proto-Oncogene Proteins c-jun/metabolism , Activating Transcription Factor 1/metabolism , Cell Line, Tumor , Cyclin A2/biosynthesis , Cyclin A2/genetics , Down-Regulation , Elongation Factor 2 Kinase/genetics , Elongation Factor 2 Kinase/metabolism , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , G1 Phase Cell Cycle Checkpoints , Hep G2 Cells , Humans , JNK Mitogen-Activated Protein Kinases/biosynthesis , MAP Kinase Signaling System , NF-kappa B/metabolism , Phenanthrolines , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Promoter Regions, Genetic/drug effects , Protein Kinase C-delta/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-jun/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , RNA Interference , RNA, Small Interfering , Transcription Factor AP-1/metabolism , Tylophora
SELECTION OF CITATIONS
SEARCH DETAIL
...