Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
BMC Musculoskelet Disord ; 25(1): 206, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38454404

ABSTRACT

BACKGROUND: Osteoporosis is a genetic disease caused by the imbalance between osteoblast-led bone formation and osteoclast-induced bone resorption. However, further gene-related pathogenesis remains to be elucidated. METHODS: The aberrant expressed genes in osteoporosis was identified by analyzing the microarray profile GSE100609. Serum samples of patients with osteoporosis and normal group were collected, and the mRNA expression of candidate genes was detected by quantitative real-time polymerase chain reaction (qRT-PCR). The mouse cranial osteoblast MC3T3-E1 cells were treated with dexamethasone (DEX) to mimic osteoporosis in vitro. Alizarin Red staining and alkaline phosphatase (ALP) staining methods were combined to measure matrix mineralization deposition of MC3T3-E1 cells. Meanwhile, the expression of osteogenesis related genes including alkaline phosphatase (ALP), osteocalcin (OCN), osteopontin (OPN), Osterix, and bone morphogenetic protein 2 (BMP2) were evaluated by qRT-PCR and western blotting methods. Then the effects of candidate genes on regulating impede bone loss caused by ovariectomy (OVX) in mice were studied. RESULTS: Cyclin A1 (CCNA1) was found to be significantly upregulated in serum of osteoporosis patients and the osteoporosis model cells, which was in line with the bioinformatic analysis. The osteogenic differentiation ability of MC3T3-E1 cells was inhibited by DEX treatment, which was manifested by decreased Alizarin Red staining intensity, ALP staining intensity, and expression levels of ALP, OCN, OPN, Osterix, and BMP2. The effects of CCNA1 inhibition on regulating osteogenesis were opposite to that of DEX. Then, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis demonstrated that genes negatively associated with CCNA1 were enriched in the TGF-beta signaling pathway. Inhibitor of TGF-beta signaling pathway partly reversed osteogenesis induced by suppressed CCNA1. Furthermore, suppressed CCNA1 relieved bone mass of OVX mice in vivo. CONCLUSION: Downregulation of CCNA1 could activate TGF-beta signaling pathway and promote bone formation, thus playing a role in treatment of osteoporosis.


Subject(s)
Anthraquinones , Osteoporosis , Transforming Growth Factor beta , Animals , Female , Humans , Mice , Alkaline Phosphatase/metabolism , Cell Differentiation , Cyclin A1/metabolism , Osteoblasts/metabolism , Osteogenesis , Osteoporosis/chemically induced , Transforming Growth Factor beta/metabolism , Transforming Growth Factors/adverse effects , Transforming Growth Factors/metabolism
2.
BMC Biol ; 21(1): 75, 2023 04 07.
Article in English | MEDLINE | ID: mdl-37024934

ABSTRACT

BACKGROUND: Angiogenesis plays important roles in physiological and pathologic conditions, but the mechanisms underlying this complex process often remain to be elucidated. In recent years, liquid-liquid phase separation (LLPS) has emerged as a new concept to explain many cellular functions and diseases. However, whether LLPS is involved in angiogenesis has not been studied until now. Here, we investigated the potential role of LLPS in angiogenesis and endothelial function. RESULTS: We found 1,6-hexanediol (1,6-HD), an inhibitor of LLPS, but not 2,5-hexanediol (2,5-HD) dramatically decreases neovascularization of Matrigel plug and angiogenesis response of murine corneal in vivo. Moreover, 1,6-HD but not 2,5-HD inhibits microvessel outgrowth of aortic ring and endothelial network formation. The endothelial function of migration, proliferation, and cell growth is suppressed by 1,6-HD. Global transcriptional analysis by RNA-sequencing reveals that 1,6-HD specifically blocks cell cycle and downregulates cell cycle-related genes including cyclin A1. Further experimental data show that 1,6-HD treatment greatly reduces the expression of cyclin A1 but with minimal effect on cyclin D1, cyclin E1, CDK2, and CDK4. The inhibitory effect of 1,6-HD on cyclin A1 is mainly through transcriptional regulation because proteasome inhibitors fail to rescue its expression. Furthermore, overexpression of cyclin A1 in HUVECs largely rescues the dysregulated tube formation upon 1,6-HD treatment. CONCLUSIONS: Our data reveal a critical role of LLPS inhibitor 1,6-HD in angiogenesis and endothelial function, which specifically affects endothelial G1/S transition through transcriptional suppression of CCNA1, implying LLPS as a possible novel player to modulate angiogenesis, and thus, it might represent an interesting therapeutic target to be investigated in clinic angiogenesis-related diseases in future.


Subject(s)
Cyclin A1 , Neovascularization, Pathologic , Humans , Mice , Animals , Cyclin A1/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Cell Movement , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Cell Proliferation
3.
Int Wound J ; 20(1): 131-139, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35606330

ABSTRACT

C-MYC-mediated keloid fibroblasts proliferation and collagen deposit may contribute to the development of keloids. F-box and leucine-rich repeat protein 6 (FBXL6) is reported to be involved in tumour progression, while the role of FBXL6 in keloid fibroblasts is not deciphered. Normal control skins, hypertrophic scars and keloid tissues were collected and prepared for FBXL6 detection. FBXL6 short hairpin RNAs (shRNAs) or FBXL6 over-expression plasmids were transfected into keloid fibroblasts, and then c-MYC plasmids were further transfected. Cell viability was assayed with a Cell-Counting Kit-8 kit. The relative expression of FBXL6, Cyclin A1, Cyclin D2, Cyclin E1 and Collagen I was detected with real-time PCR and Western blot. Elevated FBXL6 expression could be observed in keloid tissues and hypertrophic scars. FBXL6 shRNAs transfection could inhibit the viability of keloid fibroblasts with diminished c-MYC expression and down-regulated Cyclin A1, Cyclin D2, Cyclin E1 and Collagen I expression. At the same time, overexpressed FBXL6 could promote the proliferation of keloid fibroblasts. Overexpression of c-MYC could promote the proliferation of keloid fibroblasts reduced by FBXL6 shRNAs with up-regulated Cyclin A1 and Collagen I expression. FBXL6 could promote the growth of keloid fibroblasts by inducing c-MYC expression, which could be targeted in keloids treatment.


Subject(s)
Cicatrix, Hypertrophic , Keloid , Humans , Keloid/genetics , Keloid/pathology , Cicatrix, Hypertrophic/metabolism , Cyclin A1/metabolism , Cyclin D2/metabolism , Collagen/metabolism , Cell Proliferation/genetics , Fibroblasts/metabolism , Cells, Cultured
4.
Zhonghua Gan Zang Bing Za Zhi ; 31(10): 1043-1050, 2023 Oct 20.
Article in Zh | MEDLINE | ID: mdl-38016768

ABSTRACT

Objective: To investigate the effect of cyclin A1 on the invasion, metastasis, and prognosis of hepatocellular carcinoma (HCC). Methods: Immunohistochemistry (IHC) was used to detect the expressional condition of cyclin A1 in HCC and paraffin-embedded non-tumor adjacent tissues. Kaplan-Meier method was used for the survival analysis of patients with HCC. Western blot (WB) was used to detect the expression of cyclin A1 in HCCLM3 and QGY-7703 cells. Scratch wound healing assay, transwell migration, and invasion assay were used to detect the effect of cyclin A1 overexpression on cell migration and invasion ability. WB was used to detect changes in the expression of matrix metalloproteinase (MMP) 2, MMP9, and vascular endothelial growth factor (VEGF) after overexpression of cyclin A1. Measurement data were compared using a t-test and analysis of variance. Count data was measured using χ (2) test and the Log-rank method was performed for survival analysis. Results: Cyclin A1 expression rates were higher in the tissues of HCC patients with recurrent metastasis than in the tissues of patients without recurrent metastasis (60.42% vs. 46.81%, χ (2) = 4.711, P < 0.05). The overall postoperative survival time (OS) and disease-free survival (DFS) were shorter in patients with high cyclin A1 expression than those with low cyclin A1 expression (45.9 months vs. 53.1 months; 42.9 months vs. 51.3 months, and P < 0.01). The postoperative OS and DFS were shorter in patients with high cyclin A1 expression and recurrent metastasis than those with low cyclin A1 expression without recurrent metastasis (31.7 months vs. 43.9 months; 18.0 months vs. 31.5 months, and P < 0.05). HCCLM3 and QGY-7703 cells were higher in the cyclin A1-pEX group than in the empty vector (vector) group (1.56 ± 0.06 vs. 0.18 ± 0.01, t = 18.75, P < 0.001; 1.31 ± 0.05 vs.0.37 ± 0.02, t = 15.17, P < 0.001). The migrated distances of HCCLM3 cells in the cyclin A1-pEX group and the vector group were (536.7 ± 14.5) µm and (327.3 ± 9.3) µm, t = 11.84, P < 0.05, respectively, while the migrated distances of QGY-7703 cells in the two groups were (916.7 ± 35.3) µm and (320.0 ± 20.8) µm, t = 13.54, P < 0.01. The migrated numbers of HCCLM3 cells in the cyclin A1-pEX group and vector group were (37.3 ± 2.4) and (7.0 ± 1.2), t = 12.67, P < 0.001, and the number of invasive cells was (73.7 ± 4.1) and (12.6 ± 1.5), t = 12.36, P < 0.001, respectively. The migrated numbers of QGY-7703 cells in the two groups were (153.3 ± 6.0) and (17.7 ± 3.7), t = 17.59, P < 0.001, and the number of invasive cells was (45.0 ± 2.9) and (9.3 ± 1.5), t = 10.66, P < 0.001, respectively. The expression levels of MMP2, MMP9, and VEGF in HCCLM3 and QGY-7703 cells were significantly higher in the cyclin A1-pEX group than those in the vector group (P < 0.05). Conclusion: Cyclin A1 plays an important role in HCC invasion and metastasis, but HCC patients with high cyclin A1 expression have a poor prognosis. Hence, cyclin A1 has high guiding significance for evaluating patient prognosis.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cyclin A1/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/pathology , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness , Prognosis , Vascular Endothelial Growth Factor A/metabolism
5.
Glycobiology ; 32(1): 73-82, 2022 02 26.
Article in English | MEDLINE | ID: mdl-34791227

ABSTRACT

Enhanced sebocyte proliferation is associated with the pathogenesis of human skin diseases related to sebaceous gland hyperfunction and androgens, which are known to induce sebocyte proliferation, are key mediators of this process. Galectin-12, a member of the ß-galactoside-binding lectin family that is preferentially expressed by adipocytes and functions as an intrinsic negative regulator of lipolysis, has been shown to be expressed by human sebocytes. In this study, we identified galectin-12 as an important intracellular regulator of sebocyte proliferation. Galectin-12 knockdown in the human SZ95 sebocyte line suppressed cell proliferation, and its overexpression promoted cell cycle progression. Inhibition of galectin-12 expression reduced the androgen-induced SZ95 sebocyte proliferation and growth of sebaceous glands in mice, respectively. The mRNA expression of the key cell cycle regulators cyclin A1 (CCNA1) and cyclin-dependent kinase 2CDK2 was reduced in galectin-12 knockdown SZ95 sebocytes, suggesting a pathway of galectin-12 regulation of sebocyte proliferation. Further, galectin-12 enhanced peroxisome proliferator-activated receptor gamma (PPARγ) expression and transcriptional activity in SZ95 sebocytes, consistent with our previous studies in adipocytes. Rosiglitazone, a PPARγ ligand, induced CCNA1 levels, suggesting that galectin-12 may upregulate CCNA1 expression via PPARγ. Our findings suggest the possibility of targeting galectin-12 to treat human sebaceous gland hyperfunction and androgen-associated skin diseases.


Subject(s)
Cyclin A1 , Sebaceous Glands , Animals , Cell Cycle/genetics , Cell Proliferation , Cyclin A1/metabolism , Cyclin-Dependent Kinase 2 , Galectins/genetics , Galectins/metabolism , Mice , Sebaceous Glands/metabolism
6.
J Enzyme Inhib Med Chem ; 37(1): 1884-1902, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35801486

ABSTRACT

A new series of 1H-pyrrole (6a-c, 8a-c), pyrrolo[3,2-d]pyrimidines (9a-c) and pyrrolo[3,2-e][1, 4]diazepines (11a-c) were designed and synthesised. These compounds were designed to have the essential pharmacophoric features of EGFR Inhibitors, they have shown anticancer activities against HCT116, MCF-7 and Hep3B cancer cells with IC50 values ranging from 0.009 to 2.195 µM. IC50 value of doxorubicin is 0.008 µM, compounds 9a and 9c showed IC50 values of 0.011 and 0.009 µM respectively against HCT-116 cells. Compound 8b exerted broad-spectrum activity against all tested cell lines with an IC50 value less than 0.05 µM. Compound 8b was evaluated against a panel of kinases. This compound potently inhibited CDK2/Cyclin A1, DYRK3 and GSK3 alpha kinases with 10-23% compared to imatinib (1-10%). It has also arrested the cell cycle of MCF-7 cells at the S phase. Its antiproliferative activity was further augmented by molecular docking into the active sites of EGFR and CDK2 cyclin A1.


Subject(s)
Antineoplastic Agents , Pyrimidines , Antineoplastic Agents/chemistry , Azepines/pharmacology , Cell Proliferation , Cyclin A1/metabolism , Cyclin-Dependent Kinase 2/metabolism , Drug Screening Assays, Antitumor , ErbB Receptors/metabolism , Glycogen Synthase Kinase 3/metabolism , Humans , Molecular Docking Simulation , Molecular Structure , Protein Kinase Inhibitors , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases , Pyrimidines/chemistry , Pyrroles/chemistry , Structure-Activity Relationship
7.
Cancer Sci ; 112(6): 2287-2298, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33738896

ABSTRACT

Drug resistance is a significant obstacle to effective cancer treatment. Drug resistance develops from initially reversible drug-tolerant cancer cells, which offer therapeutic opportunities to impede cancer relapse. The mechanisms of resistance to proteasome inhibitor (PI) therapy have been investigated intensively, however the ways by which drug-tolerant cancer cells orchestrate their adaptive responses to drug challenges remain largely unknown. Here, we demonstrated that cyclin A1 suppression elicited the development of transient PI tolerance in mixed-lineage leukemia (MLL) cells. This adaptive process involved reversible downregulation of cyclin A1, which promoted PI resistance through cell-cycle arrest. PI-tolerant MLL cells acquired cyclin A1 dependency, regulated directly by MLL protein. Loss of cyclin A1 function resulted in the emergence of drug tolerance, which was associated with patient relapse and reduced survival. Combination treatment with PI and deubiquitinating enzyme (DUB) inhibitors overcame this drug resistance by restoring cyclin A1 expression through chromatin crosstalk between histone H2B monoubiquitination and MLL-mediated histone H3 lysine 4 methylation. These results reveal the importance of cyclin A1-engaged cell-cycle regulation in PI resistance in MLL cells, and suggest that cell-cycle re-entry by DUB inhibitors may represent a promising epigenetic therapeutic strategy to prevent acquired drug resistance.


Subject(s)
Cyclin A1/metabolism , Deubiquitinating Enzymes/antagonists & inhibitors , Drug Tolerance , Leukemia, Biphenotypic, Acute/drug therapy , Proteasome Inhibitors/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Chromatin/metabolism , Cyclin A1/genetics , Drug Resistance, Neoplasm , Drug Tolerance/genetics , Gene Expression Regulation, Neoplastic , Histones/metabolism , Humans , Leukemia, Biphenotypic, Acute/genetics , Leukemia, Biphenotypic, Acute/metabolism , Leukemia, Biphenotypic, Acute/pathology , Methylation , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Prognosis , Proteasome Inhibitors/therapeutic use , Ubiquitination
8.
Br J Cancer ; 125(8): 1122-1134, 2021 10.
Article in English | MEDLINE | ID: mdl-34290392

ABSTRACT

BACKGROUND: Large-scale genetic and epigenetic deregulations enable cancer cells to ectopically activate tissue-specific expression programmes. A specifically designed strategy was applied to oral squamous cell carcinomas (OSCC) in order to detect ectopic gene activations and develop a prognostic stratification test. METHODS: A dedicated original prognosis biomarker discovery approach was implemented using genome-wide transcriptomic data of OSCC, including training and validation cohorts. Abnormal expressions of silent genes were systematically detected, correlated with survival probabilities and evaluated as predictive biomarkers. The resulting stratification test was confirmed in an independent cohort using immunohistochemistry. RESULTS: A specific gene expression signature, including a combination of three genes, AREG, CCNA1 and DDX20, was found associated with high-risk OSCC in univariate and multivariate analyses. It was translated into an immunohistochemistry-based test, which successfully stratified patients of our own independent cohort. DISCUSSION: The exploration of the whole gene expression profile characterising aggressive OSCC tumours highlights their enhanced proliferative and poorly differentiated intrinsic nature. Experimental targeting of CCNA1 in OSCC cells is associated with a shift of transcriptomic signature towards the less aggressive form of OSCC, suggesting that CCNA1 could be a good target for therapeutic approaches.


Subject(s)
Amphiregulin/genetics , Cyclin A1/genetics , DEAD Box Protein 20/genetics , Gene Expression Profiling/methods , Mouth Neoplasms/pathology , Squamous Cell Carcinoma of Head and Neck/pathology , Amphiregulin/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cyclin A1/metabolism , DEAD Box Protein 20/metabolism , Data Mining , Female , Humans , Male , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Neoplasm Invasiveness , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Retrospective Studies , Sequence Analysis, RNA , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism
9.
J Cell Physiol ; 235(10): 7136-7145, 2020 10.
Article in English | MEDLINE | ID: mdl-32030765

ABSTRACT

Mammalian cyclin A1 is prominently expressed in testis and essential for meiosis in the male mouse, however, it shows weak expression in ovary, especially during oocyte maturation. To understand why cyclin A1 behaves in this way in the oocyte, we investigated the effect of cyclin A1 overexpression on mouse oocyte meiotic maturation. Our results revealed that cyclin A1 overexpression triggered meiotic resumption even in the presence of germinal vesicle breakdown inhibitor, milrinone. Nevertheless, the cyclin A1-overexpressed oocytes failed to extrude the first polar body but were completely arrested at metaphase I. Consequently, cyclin A1 overexpression destroyed the spindle morphology and chromosome alignment by inducing premature separation of chromosomes and sister chromatids. Therefore, cyclin A1 overexpression will prevent oocyte maturation although it can promote meiotic resumption. All these results show that decreased expression of cyclin A1 in oocytes may have an evolutional significance to keep long-lasting prophase arrest and orderly chromosome separation during oocyte meiotic maturation.


Subject(s)
Chromosome Segregation/genetics , Chromosome Segregation/physiology , Cyclin A1/genetics , Cyclin A1/metabolism , Meiosis/genetics , Meiosis/physiology , Oocytes/metabolism , Animals , Chromosome Segregation/drug effects , Female , Meiosis/drug effects , Mice , Mice, Inbred ICR , Milrinone/pharmacology , Oocytes/cytology , Oocytes/drug effects , Oogenesis/drug effects , Oogenesis/genetics , Oogenesis/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Separase/metabolism , Up-Regulation
10.
J Cell Physiol ; 234(4): 3555-3569, 2019 04.
Article in English | MEDLINE | ID: mdl-30565670

ABSTRACT

Hepatocellular carcinoma (HCC) is mainly associated with hepatitis B virus (HBV) infection and characterized by metastasizing and infiltrating adjacent and distant tissues. Notably, microRNA-1271 (miR-1271) is a tumor suppressor in various cancers. Therefore, we evaluate the ability of miR-1271 to influence cell proliferation, migration, invasion, and apoptosis in HBV-associated HCC through the Adenosine monophosphate-activated protein kinase (AMPK) signaling pathway via targeting CCNA1. HBV-associated HCC and adjacent normal tissues were collected to identify the expression of miR-1271 and CCNA1. To verify the relationship between miR-1271 and CCNA1, we used bioinformatics prediction and the dual-luciferase reporter gene assay. The effects of miR-1271 on HBV-associated HCC cell behaviors were investigated by treatment of the miR-1271 mimic, the miR-1271 inhibitor, or small interfering RNA against CCNA1. The HBV-DNA quantitative assay, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromid assay, scratch test, transwell assay, and flow cytometry were used to detect HBV-DNA replication, cell proliferation, invasion, migration, and apoptosis. MiR-1271 showed a low expression, whereas CCNA1 showed a high expression in HBV-associated HCC tissues. We identified that miR-1271 targeted and negatively regulated CCNA1. Upregulated miR-1271 and downregulated CCNA1 inhibited the HBV-associated HCC cell HBV-DNA replication, proliferation, migration, and invasion, while accelerating apoptosis by activating the AMPK signaling pathway. MiR-1271 promotes the activation of the AMPK signaling pathway by binding to CCNA1, whereby miR-1271 suppresses HBV-associated HCC progression. This study points to a potential therapeutic approach of downregulation of miR-1271 in HBV-associated HCC treatment.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Carcinoma, Hepatocellular/enzymology , Cyclin A1/metabolism , Hepatitis B virus/growth & development , Hepatitis B/virology , Liver Neoplasms/enzymology , MicroRNAs/metabolism , Apoptosis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/virology , Cell Movement , Cell Proliferation , Cyclin A1/genetics , DNA Replication , DNA, Viral/biosynthesis , DNA, Viral/genetics , Disease Progression , Female , Hep G2 Cells , Hepatitis B/complications , Hepatitis B virus/genetics , Host-Pathogen Interactions , Humans , Liver Neoplasms/genetics , Liver Neoplasms/virology , Male , MicroRNAs/genetics , Middle Aged , Neoplasm Invasiveness , Signal Transduction , Virus Replication
11.
Am J Physiol Endocrinol Metab ; 316(3): E443-E452, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30576241

ABSTRACT

The purpose of this study was to explore the role of microRNA-451a (miR-451a) in diabetic retinopathy through activating transcription factor 2 (ATF2). The epiretinal membrane samples from patients with proliferative diabetic retinopathy (PDR) were immunolabeled with an antibody for Ki-67 to identify the proliferative cells. The expression of miR-451a was measured by qRT-PCR in the retina of Akita mice and in RPE cells under diabetic conditions. The potential downstream targets of miR-451a were predicted by bioinformatics and confirmed by dual luciferase assay, qRT-PCR, and Western blotting. Mitochondrial function, cell proliferation, and migration assays were used to detect the functional change after transfection of miR-451a mimic and inhibitor. Proliferative RPE cells were identified in the epiretinal membrane from PDR patients. The expression of miR-451a was downregulated both in the retina of Akita mice and 4-hydroxynonenal (4-HNE)-treated RPE cells. Bioinformatic analysis and luciferase assay identified ATF2 as a potential target of miR-451a. miR-451a inhibited proliferation and migration of RPE cells. The mitochondrial function was enhanced by miR-451a mimic, but suppressed by miR-451a inhibitor. In diabetic conditions, miR-451a showed a protective effect on mitochondrial function. The results of qRT-PCR and Western blotting revealed that overexpression of miR-451a downregulated the expression of ATF2 and its downstream target genes CyclinA1, CyclinD1, and MMP2. In conclusion, miR-451a/ATF2 plays a vital role in the regulation of proliferation and migration in RPE cells through regulation of mitochondrial function, which may provide new perspectives for developing effective therapies for PDR.


Subject(s)
Activating Transcription Factor 2/genetics , Diabetic Retinopathy/genetics , MicroRNAs/genetics , Mitochondria/metabolism , Activating Transcription Factor 2/metabolism , Adult , Aged , Animals , Cell Movement , Cell Proliferation , Cyclin A1/genetics , Cyclin A1/metabolism , Cyclin D1/genetics , Cyclin D1/metabolism , Diabetes Mellitus, Type 1/metabolism , Diabetic Retinopathy/metabolism , Disease Models, Animal , Female , Humans , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice , MicroRNAs/metabolism , Middle Aged , Retinal Pigment Epithelium
12.
Biochem Biophys Res Commun ; 498(1): 45-51, 2018 03 25.
Article in English | MEDLINE | ID: mdl-29499194

ABSTRACT

Histone acetylation and deacetylation correlate with diverse biological phenomena through gene transcription. Histone deacetylases (HDACs) regulate deacetylation of histones and other proteins. However, as a member of the HDAC family, HDAC8 function during neurodevelopment is currently unknown. Therefore, we investigated HDAC8 function during neurodevelopment by examining embryoid body (EB) formation in P19 cells. HDAC8-selective inhibitor (NCC-149) (HDAC8i)-treated cells showed smaller EBs than non-treated cells, as well as reduced expression levels of the neuronal marker, NeuN. Additionally, HDAC8i treatment led to inhibition of cellular proliferation by G2/M phase accumulation and downregulated cyclin A2 and cyclin B1 gene expression. Furthermore, two independent HDAC8 knockout cell lines were established by CRISPR-Cas9, which resulted in smaller EBs, similar to HDAC8i-treated cells. These results suggest that HDAC8 regulates neural differentiation by exerting control of EB formation.


Subject(s)
Cell Differentiation , Embryoid Bodies/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Neurons/cytology , Animals , Base Sequence , Cell Cycle Checkpoints/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin A1/metabolism , Cyclin B2/metabolism , Down-Regulation/drug effects , Embryoid Bodies/drug effects , G2 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockout Techniques , Humans , Mice , Mitosis/drug effects , Neurons/drug effects , Neurons/metabolism
13.
Zhonghua Nan Ke Xue ; 24(1): 19-26, 2018.
Article in Zh | MEDLINE | ID: mdl-30157355

ABSTRACT

OBJECTIVE: To explore the change in the proliferation of spermatogenic cells in the male mouse with infertility induced by exogenous estradiol benzoate (EB). METHODS: Sixty male mice aged 4 weeks were randomly divided into a control, a low-dose EB, and a high-dose EB group to be injected intramuscularly with corn oil at 150 µl or EB at 5 or 10 mg/kg, respectively, every other day for 4 weeks. Then, we obtained the weight and indexes of the testis, performed HE staining of the paraffin sections of the testis tissue and epididymal cauda, counted the spermatozoa in the epididymal sperm suspension, and determined the expression of the proliferating cell nuclear antigen (PCNA), the mRNA expressions of CyclinA1, CyclinB1, VASA and p53, and the protein expressions of p53 and phosphorylated p53 in the testis by immunohistochemistry, qRT-PCR and Western blot, respectively. RESULTS: In comparison with the controls, the mice treated with EB showed significantly decreased testicular indexes (P <0.05), no sperm in the sperm suspension or epididymal tubes, remarkably reduced numbers of spermatogonia, primary spermatocytes and Sertoli cells (P <0.05), down-regulated expression of PCNA (P <0.05) and mRNA expressions of CyclinA1, CyclinB1, PCNA and VASA in the seminiferous tubules (P <0.05), but a dose-dependent increase of the p53 level (P <0.05). Western blot revealed markedly higher levels of p53 protein expression and phosphorylation in the EB than in the control group (P <0.05) and even higher in the 10 mg/kg than in the 5 mg/kg EB group (P <0.05). CONCLUSIONS: EB inhibited the proliferation of spermatogenic cells by down-regulating the expressions of cell cycle-related factors in a dose-dependent manner, which might contribute to abnormal proliferation of spermatogenic cells in the testis of infertile male mice.


Subject(s)
Cell Proliferation/drug effects , Contraceptive Agents/pharmacology , Estradiol/analogs & derivatives , Infertility, Male/pathology , Spermatozoa/drug effects , Testis/drug effects , Animals , Cell Proliferation/physiology , Contraceptive Agents/administration & dosage , Cyclin A1/metabolism , Cyclin B1/metabolism , DEAD-box RNA Helicases/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Epididymis/cytology , Epididymis/drug effects , Estradiol/administration & dosage , Estradiol/pharmacology , Male , Mice , Proliferating Cell Nuclear Antigen/metabolism , RNA, Messenger/metabolism , Random Allocation , Seminiferous Tubules/cytology , Seminiferous Tubules/drug effects , Sertoli Cells/cytology , Sertoli Cells/drug effects , Sperm Count , Spermatocytes/cytology , Spermatocytes/drug effects , Spermatogonia/cytology , Spermatogonia/drug effects , Spermatozoa/cytology , Testis/cytology , Testis/metabolism , Tumor Suppressor Protein p53/metabolism
14.
Exp Physiol ; 102(11): 1474-1485, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28786140

ABSTRACT

NEW FINDINGS: What is the central question of this study? We investigated whether 5-aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside (AICAR) could prevent acute increases in body fat and changes in omental and subcutaneous adipose tissue following the sudden transition from physical activity to physical inactivity. What is the main finding and its importance? AICAR prevented fat gains following the transition from physical activity to inactivity to levels comparable to rats that remained physically active. AICAR and continuous physical activity produced depot-specific changes in cyclin A1 mRNA and protein that were associated with the prevention of fat gain. These findings suggest that targeting AMP-activated protein kinase signalling could oppose rapid adipose mass growth. The transition from physical activity to inactivity is associated with drastic increases in 'catch-up' fat that in turn foster the development of many obesity-associated maladies. We tested whether 5-aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside (AICAR) treatment would prevent gains in body fat following the sudden transition from a physically active state to an inactive state by locking a voluntary running wheel. Male Wistar rats were either sedentary (SED) or given wheel access for 4 weeks, at which time rats with wheels continued running (RUN), had their wheel locked (WL) or had WL with daily AICAR injection (WL + AICAR) for 1 week. RUN and WL + AICAR prevented gains in body fat compared with SED and WL (P < 0.001). Cyclin A1 mRNA, a marker of cell proliferation, was decreased in omental, but not subcutaneous adipose tissue, in RUN and WL + AICAR compared with SED and WL groups (P < 0.05). Both cyclin A1 mRNA and protein were positively associated with gains in fat mass (P < 0.05). Cyclin A1 mRNA in omental, but not subcutaneous, adipose tissue was negatively correlated with p-AMPK levels (P < 0.05). Differences in fat gain and omental mRNA and protein levels were independent of changes in food intake and in differences in select hypothalamic mRNAs. These findings suggest that AICAR treatment prevents acute gains in adipose tissue following physical inactivity to levels of rats that continuously run, and that together, continuous physical activity and AICAR could, at least initially in these conditions, exert similar inhibitory effects on adipogenesis in a depot-specific manner.


Subject(s)
Abdominal Fat/drug effects , Adipogenesis/drug effects , Adiposity/drug effects , Aminoimidazole Carboxamide/analogs & derivatives , Anti-Obesity Agents/pharmacology , Physical Conditioning, Animal/methods , Ribonucleotides/pharmacology , Sedentary Behavior , Subcutaneous Fat/drug effects , Weight Gain/drug effects , AMP-Activated Protein Kinases/metabolism , Abdominal Fat/metabolism , Aminoimidazole Carboxamide/pharmacology , Animals , Cyclin A1/genetics , Cyclin A1/metabolism , Enzyme Activation , Enzyme Activators/pharmacology , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Phosphorylation , Physical Exertion , Rats, Wistar , Running , Subcutaneous Fat/metabolism , Time Factors , Volition
15.
Biochem J ; 473(3): 257-66, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26574435

ABSTRACT

Growth factor-mediated hepatocyte proliferation is crucial in liver regeneration and the recovery of liver function after injury. The nuclear receptor, pregnane X receptor (PXR), is a key transcription factor for the xenobiotic-induced expression of genes associated with various liver functions. Recently, we reported that PXR activation stimulates xenobiotic-induced hepatocyte proliferation. In the present study, we investigated whether PXR activation also stimulates growth factor-mediated hepatocyte proliferation. In G0 phase-synchronized, immortalized mouse hepatocytes, serum or epidermal growth factor treatment increased cell growth and this growth was augmented by the expression of mouse PXR and co-treatment with pregnenolone 16α-carbonitrile (PCN), a PXR ligand. In a liver regeneration model using carbon tetrachloride, PCN treatment enhanced the injury-induced increase in the number of Ki-67-positive nuclei as well as Ccna2 and Ccnb1 mRNA levels in wild-type (WT) but not Pxr-null mice. Chronological analysis of this model demonstrated that PCN treatment shifted the maximum cell proliferation to an earlier time point and increased the number of M-phase cells at those time points. In WT but not Pxr-null mice, PCN treatment reduced hepatic mRNA levels of genes involved in the suppression of G0/G1- and G1/S-phase transition, e.g. Rbl2, Cdkn1a and Cdkn1b. Analysis of the Rbl2 promoter revealed that PXR activation inhibited its Forkhead box O3 (FOXO3)-mediated transcription. Finally, the PXR-mediated enhancement of hepatocyte proliferation was inhibited by the expression of dominant active FOXO3 in vitro. The results of the present study suggest that PXR activation stimulates growth factor-mediated hepatocyte proliferation in mice, at least in part, through inhibiting FOXO3 from accelerating cell-cycle progression.


Subject(s)
Cell Proliferation , Epidermal Growth Factor/metabolism , Forkhead Transcription Factors/metabolism , Hepatocytes/cytology , Receptors, Steroid/metabolism , Animals , Cell Cycle , Cells, Cultured , Cyclin A1/genetics , Cyclin A1/metabolism , Cyclin A2/genetics , Cyclin A2/metabolism , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Hepatocytes/metabolism , Liver/cytology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Pregnane X Receptor , Pregnenolone Carbonitrile/metabolism , Receptors, Steroid/genetics
16.
Bull Exp Biol Med ; 163(5): 623-626, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28948561

ABSTRACT

The contents of cyclins A1, B1, E1, and D1, cyclin-dependent kinases 2 and 4 (CDK2 and CDK4), cyclin-dependent kinase 2C inhibitor (p18), and cyclin-dependent kinase 1C inhibitor (p57) in whole blood mononuclear cells of practically healthy donors and convalescents from community-acquired pneumonia were measured after cell exposure to low-intensity electromagnetic radiation of 1 GHz. The level of cyclins A1, D1, and B1, p57, and p18 proteins, and CDK2 was reduced in convalescents, while the content of cyclin B1 was elevated in both groups. A strong correlation was found between the content of cyclin B1 and the levels of cyclin A1 and CDK2 in convalescents. Single exposure of whole blood cells to 1-GHz microwaves was accompanied by an increase in the content of the analyzed factors (particularly, cyclins A1 and E1). These data indicate that radiation induced activation of the synthetic phase of the cell cycle.


Subject(s)
Community-Acquired Infections/metabolism , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Leukocytes, Mononuclear/metabolism , Microwaves/adverse effects , Pneumonia/metabolism , Adult , Cell Cycle/radiation effects , Cell Cycle Proteins/metabolism , Cells, Cultured , Community-Acquired Infections/complications , Cyclin A1/metabolism , Cyclin-Dependent Kinase 2/metabolism , Humans , Leukocytes, Mononuclear/radiation effects , Male , Pneumonia/complications , Young Adult
17.
Molecules ; 21(7)2016 Jul 19.
Article in English | MEDLINE | ID: mdl-27447597

ABSTRACT

Protocatechualdehyde (PCA) extracted from Phellinus gilvus exhibits anti-cancer activity in human colorectal carcinoma cells (HT-29). However, the underlying mechanisms remain poorly understood. We performed an in vitro study involving MTT, flow cytometry, RT-PCR, and western blot analyses to investigate the effects of PCA treatment on cell proliferation, cell cycle distribution, apoptosis, and expression of several cell cycle-related genes in HT-29 cells. The treatment enhanced S-phase cell cycle and apoptosis in HT-29 cells in a dose-dependent manner. Western blot results showed that PCA treatment decreased the expression levels of cyclin A, cyclin D1, and p27(KIP1) but increased those of cyclin-dependent kinase 2 (CDK2) in HT-29 cells. Furthermore, the expression levels of B-cell lymphoma/leukemia-2 (Bcl-2) and B-cell lymphoma/leukemia-xL (Bcl-xL) were down-regulated, whereas the levels of BH3-interacting domain death agonist (Bid), Bcl-2 homologous antagonist/killer (Bak), and cytosolic cytochrome c were significantly upregulated. Thus, the enzymes caspases-9, -3, -8, and -6 were found to be activated in HT-29 cells with PCA treatment. These results indicate that PCA-induced S-phase cell cycle arrest and apoptosis involve p27(KIP1)-mediated activation of the cyclin-A/D1-Cdk2 signaling pathway and the mitochondrial apoptotic pathway.


Subject(s)
Apoptosis/drug effects , Benzaldehydes/pharmacology , Catechols/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , S Phase Cell Cycle Checkpoints/drug effects , Signal Transduction/drug effects , Benzaldehydes/chemistry , Catechols/chemistry , Cell Proliferation/drug effects , Cyclin A1/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , HT29 Cells , Humans
18.
BMC Cancer ; 15: 784, 2015 Oct 24.
Article in English | MEDLINE | ID: mdl-26499264

ABSTRACT

BACKGROUND: Cyclin A1 is essential for male gametopoiesis. In acute myeloid leukemia, it acts as a leukemia-associated antigen. Cyclin A1 expression has been reported in several epithelial malignancies, including testicular, endometrial, and epithelial ovarian cancer (EOC). We analyzed Cyclin A1 expression in EOC and its correlation with clinical features to evaluate Cyclin A1 as a T-cell target in EOC. METHODS: Cyclin A1 mRNA expression in EOC and healthy tissues was quantified by microarray analysis and quantitative real-time PCR (qRT-PCR). Protein expression in clinical samples was assessed by immunohistochemistry (IHC) and was correlated to clinical features. RESULTS: Cyclin A1 protein was homogeneously expressed in 43 of 62 grade 3 tumor samples and in 1 of 10 grade 2 specimens (p < 0.001). Survival analysis showed longer time to progression (TTP) among patients with at least moderate Cyclin A1 expression (univariate: p = 0.018, multivariate: p = 0.035). FIGO stage, grading, age, macroscopic residual tumor after debulking, and peritoneal carcinomatosis / distant metastasis had no impact on TTP or overall survival (OS). CONCLUSION: Cyclin A1 is highly expressed in most EOCs. The mechanism behind the prolonged TTP in patients with high Cyclin A1 expression warrants further investigation. The frequent, selectively high expression of Cyclin A1 in EOC makes it a promising target for T-cell therapies.


Subject(s)
Biomarkers, Tumor/metabolism , Cyclin A1/metabolism , Ovarian Neoplasms/metabolism , RNA, Messenger/metabolism , Adult , Aged , Antineoplastic Agents/therapeutic use , Carboplatin/therapeutic use , Disease Progression , Female , Humans , Immunohistochemistry , Microarray Analysis , Middle Aged , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Paclitaxel/therapeutic use , Real-Time Polymerase Chain Reaction , Survival Analysis
19.
PLoS Genet ; 8(7): e1002865, 2012.
Article in English | MEDLINE | ID: mdl-22844260

ABSTRACT

Cell cycle control is modified at meiosis compared to mitosis, because two divisions follow a single DNA replication event. Cyclin-dependent kinases (CDKs) promote progression through both meiosis and mitosis, and a central regulator of their activity is the APC/C (Anaphase Promoting Complex/Cyclosome) that is especially required for exit from mitosis. We have shown previously that OSD1 is involved in entry into both meiosis I and meiosis II in Arabidopsis thaliana; however, the molecular mechanism by which OSD1 controls these transitions has remained unclear. Here we show that OSD1 promotes meiotic progression through APC/C inhibition. Next, we explored the functional relationships between OSD1 and the genes known to control meiotic cell cycle transitions in Arabidopsis. Like osd1, cyca1;2/tam mutation leads to a premature exit from meiosis after the first division, while tdm mutants perform an aberrant third meiotic division after normal meiosis I and II. Remarkably, while tdm is epistatic to tam, osd1 is epistatic to tdm. We further show that the expression of a non-destructible CYCA1;2/TAM provokes, like tdm, the entry into a third meiotic division. Finally, we show that CYCA1;2/TAM forms an active complex with CDKA;1 that can phosphorylate OSD1 in vitro. We thus propose that a functional network composed of OSD1, CYCA1;2/TAM, and TDM controls three key steps of meiotic progression, in which OSD1 is a meiotic APC/C inhibitor.


Subject(s)
Arabidopsis Proteins/genetics , Arabidopsis , Cyclin A1/genetics , Cyclins/genetics , Meiosis/genetics , Ubiquitin-Protein Ligase Complexes/genetics , Anaphase-Promoting Complex-Cyclosome , Animals , Arabidopsis/genetics , Arabidopsis/growth & development , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Cell Cycle Checkpoints/genetics , Cell Cycle Proteins/genetics , Cyclin A1/metabolism , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Epistasis, Genetic , Gametogenesis/genetics , Gene Expression Regulation, Plant , Gene Regulatory Networks , Mice , Mitosis/genetics , Mutation , Oocytes/metabolism , Phosphorylation , Plants, Genetically Modified , Signal Transduction , Ubiquitin-Protein Ligase Complexes/antagonists & inhibitors
20.
J Biol Chem ; 288(29): 21096-21104, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23760262

ABSTRACT

PCAF and GCN5 acetylate cyclin A at specific lysine residues targeting it for degradation at mitosis. We report here that histone deacetylase 3 (HDAC3) directly interacts with and deacetylates cyclin A. HDAC3 interacts with a domain included in the first 171 aa of cyclin A, a region involved in the regulation of its stability. In cells, overexpression of HDAC3 reduced cyclin A acetylation whereas the knocking down of HDAC3 increased its acetylation. Moreover, reduction of HDAC3 levels induced a decrease of cyclin A that can be reversed by proteasome inhibitors. These results indicate that HDAC3 is able to regulate cyclin A degradation during mitosis via proteasome. Interestingly, HDAC3 is abruptly degraded at mitosis also via proteasome thus facilitating cyclin A acetylation by PCAF/GCN5, which will target cyclin A for degradation. Because cyclin A is crucial for S phase progression and mitosis entry, the knock down of HDAC3 affects cell cycle progression specifically at both, S phase and G2/M transition. In summary we propose here that HDAC3 regulates cyclin A stability by counteracting the action of the acetylases PCAF/GCN5.


Subject(s)
Cyclin A1/metabolism , Histone Deacetylases/metabolism , Acetylation , HeLa Cells , Humans , Mitosis , Models, Biological , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Stability , Proteolysis , S Phase
SELECTION OF CITATIONS
SEARCH DETAIL