Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Reprod Fertil Dev ; 27(2): 419-26, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24384042

ABSTRACT

Decreasing oocyte competence with maternal aging is a major factor in mammalian infertility. One of the factors contributing to this infertility is changes to chromatin modifications, such as histone acetylation in old MII stage oocytes. Recent studies indicate that changes in histone acetylation at MII arise at the germinal vesicle (GV) stage. We hypothesised that histone methylation could also change in old GV oocytes. To test this hypothesis, we examined mono-, di- and trimethylation of histone H3 lysine 4 (H3K4 me1, me2 and me3, respectively) in young and older oocytes from 6-8- and 42-44-week-old mice, respectively. We found that H3K4 me2 and me3 decreased in older compared with young GV oocytes (100% vs. 81% and 100% vs. 87%, respectively; P<0.05). H3K4 me2 later increased in older MII oocytes (21% vs. 56%; P<0.05). We also examined the expression of genes encoding the H3K4 demethylases lysine (K)-specific demethylase 1A (Kdm1a) and retinol binding protein 2 (Rbp2). Expression of Kdm1a increased at both the mRNA and protein levels in older GV oocytes, but decreased in older MII oocytes (P<0.05), and was negatively correlated with H3K4 me2 levels. Conversely, expression of Rbp2 mRNA and protein decreased in older GV oocytes (P<0.05), and this was not correlated with H3K4 me3 levels. Finally, we showed that inhibition of Kdm1a of older oocytes at the GV stage restored levels of H3K4 me2 at the MII stage to those seen in 'young' oocytes (41% vs. 38%; P>0.05). These results suggest that changes in expression of H3K4 me2 and Kdm1a in older GV oocytes may represent a molecular mechanism underlying human infertility caused by aging.


Subject(s)
Aging/physiology , Cell Nucleus/metabolism , DNA Methylation/physiology , Histones/metabolism , Infertility, Female/etiology , Oocytes/metabolism , Animals , DNA Primers/genetics , Female , Histone Demethylases/metabolism , Immunohistochemistry , In Vitro Oocyte Maturation Techniques/methods , Mice , Microscopy, Fluorescence , Real-Time Polymerase Chain Reaction , Retinol-Binding Proteins, Cellular/metabolism , Tranylcypromine
2.
In Vitro Cell Dev Biol Anim ; 50(7): 603-13, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24619213

ABSTRACT

Extensive and dynamic chromatin remodeling occurs after fertilization, including DNA methylation and histone modifications. These changes underlie the transition from gametic to embryonic chromatin and are thought to facilitate early embryonic development. Histone H3 lysine 4 methylation (H3K4me) is an important epigenetic mechanism that associates with gene-specific activation and functions in development. However, dynamic regulation of H3K4me during early embryonic development remains unclear. Herein, the authors examined the dynamic changes of H3K4me and its key regulators (Ash1l, Ash2l, Kmt2a, Kmt2b, Kmt2c, Setd1a, Setd7, Kdm1a, Kdm1b, Kdm5a, Kdm5b, Kdm5c, and Kdm5d) in mouse oocytes and preimplantation embryos. An increase in levels of H3K4me2 and me3 was observed at the one- to two-cell stages (P < 0.05), corresponding to the period of embryonic genome activation (EGA). Subsequently, the H3K4me2 level dramatically decreased at the four-cell stage and remained at low level until the blastocyst stage (P < 0.05), whereas the H3K4me3 level transiently decreased in the four-cell embryos but steadily increased to the peak in the blastocysts (P < 0.05). The high level of H3K4me2 during the EGA was coinciding with a peak expression of its methyltransferase, ASH2L, which may stabilize this methylation level during this period. Correspondingly, a concomitant decrease in levels of its demethylases, KDM5B and KDM1A, was observed. H3K4me3 was correlated to the expression of its methyltransferase (KMT2B) and demethylase (KDM5A). Thus, these enzymes may function for the EGA and the first lineage segregation in preimplantation mouse embryos.


Subject(s)
Chromatin Assembly and Disassembly/physiology , DNA Methylation/physiology , Embryonic Development/physiology , Epigenesis, Genetic/physiology , Gene Expression Regulation, Developmental/physiology , Histone-Lysine N-Methyltransferase/metabolism , Oxidoreductases, N-Demethylating/metabolism , Animals , DNA Primers/genetics , Female , Fluorescent Antibody Technique , Histone Methyltransferases , Histones/metabolism , Male , Mice , Real-Time Polymerase Chain Reaction
3.
Mol Med Rep ; 7(4): 1229-34, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23404188

ABSTRACT

Previous data have shown that the type II cGMP­dependent protein kinase (PKG II) inhibits the EGF­induced MAPK signaling pathway. In order to thoroughly investigate PKG, it is necessary to elucidate the function of another type of PKG, PKG I. The aim of this study was to investigate the possible inhibitory effect of PKG II and PKG I activity on the basic fibroblast growth factor (bFGF)­induced proliferation and migration of U251 human glioma cells and the possible underlying mechanisms. U251 cells were infected with adenoviral constructs encoding cDNA of PKG I (Ad­PKG I) or PKG II (Ad­PKG II) to increase the expression levels of PKG I or PKG II and then treated with 8­Br­cGMP and 8­pCPT­cGMP, respectively, to activate the enzyme. An MTT assay was used to detect the proliferation of the U251 cells. The migration of the U251 cells was analyzed using a Transwell migration assay. Western blot analysis was used to detect the phosphorylation/activation of the fibroblast growth factor receptor (FGFR), MEK and ERK and the nuclear distribution of p-ERK. The results showed that bFGF treatment increased the proliferation and migration of U251 cells, accompanied by increased phosphorylation of FGFR, MEK and ERK. Furthermore, the nuclear distribution of p-ERK increased following bFGF treatment. Increasing the activity of PKG II through infection with Ad-PKG II and stimulation with 8-pCPT-cGMP significantly attenuated the aforementioned effects of the bFGF treatment, while increased PKG I activity did not inhibit the effects of bFGF treatment. These data suggest that increased PKG II activity attenuates bFGF­induced proliferation and migration by inhibiting the MAPK/ERK signaling pathway, whereas PKG I does not.


Subject(s)
Cyclic GMP-Dependent Protein Kinase Type II/genetics , Cyclic GMP-Dependent Protein Kinase Type I/genetics , Fibroblast Growth Factor 2/genetics , Glioma/genetics , Adenoviridae , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Cyclic GMP-Dependent Protein Kinase Type I/metabolism , Cyclic GMP-Dependent Protein Kinase Type II/metabolism , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Glioma/pathology , Humans , Phosphorylation/drug effects , Signal Transduction/drug effects
4.
Oncol Lett ; 4(5): 1104-1108, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23162660

ABSTRACT

In our previous study, we demonstrated that type II cGMP-dependent protein kinase (PKG II) was expressed at lower levels in different human cancer cell lines and that exogenous PKG II inhibited epidermal growth factor (EGF)-induced MAPK/ERK signaling. In order to investigate its functions further in this signaling pathway, it is necessary to elucidate whether endogenous PKG has the same effect or not. This study aimed to investigate the possible inhibitory effect of endogenous PKG activity on EGF-induced MAPK/ERK signal transduction in human lung cancer cells and its mechanism. Human small cell lung carcinoma cells (SCLCs) were treated with the PKG-selective cGMP analog 8-pCPT-cGMP to activate endogenous PKG, EGF and cGMP followed by EGF, respectively. The results showed that increased endogenous PKG activity inhibited the EGF-induced phosphorylation of the epidermal growth factor receptor (EGFR) and the binding between Sos1 and Grb2. In addition, EGF-triggered Ras activation was reversed by increased endogenous PKG activity. While the EGF-induced phosphorylation of MEK and ERK were inhibited by increased endogenous PKG activity, there was a significant increase of phosphorylated vasodilator-stimulated phosphoprotein (p-VASP) at Ser239. Furthermore, we investigated whether endogenous PKG exerted its effects on EGF-induced MAPK/ERK signaling through phosphorylation of VASP at Ser239. Downregulation of the levels of p-VASP Ser239 by point mutation blocked the effects of endogenous PKG on EGF-induced MAPK/ERK signal transduction. The data shown here suggest that endogenous PKG reverses the EGF-induced MAPK/ERK signaling pathway by phosphorylating VASP at Ser239.

5.
Mol Med Rep ; 4(4): 697-703, 2011.
Article in English | MEDLINE | ID: mdl-21537845

ABSTRACT

The scaffold protein IQGAP1 is closely related to certain Rho GTPases. Research has revealed that IQGAP1 acts as an effector of Cdc42 and Rac1 in the regulation of cell activity such as proliferation and migration. However, whether IQGAP1 is associated with RhoC, another important Rho GTPase, is unclear. Previous results from our laboratory indicated that IQGAP1 and RhoC are highly expressed in gastric cancer tissues and cells. This study was designed to investigate the possible interaction between IQGAP1 and RhoC in the regulation of the migration of cancer cells. The expression of IQGAP1 and RhoC in gastric cancer tissues and cell lines was detected by Western blotting. siRNAs targeting IQGAP1 or RhoC were transfected into gastric cancer cells to knock down the expression of the proteins. Adenoviral constructs encoding full length IQGAP1, the C­terminal fragment of IQGAP1, and the constitutively active RhoC gene were used to infect gastric cancer cells to increase the expression of the proteins. The migratory activity of a gastric cancer cell line was measured by a transwell migration assay. Western blotting revealed that the IQGAP1 and RhoC proteins were highly expressed in gastric cancer tissues and cells. Spearman's rank correlation analysis indicated that the increases in the expression of IQGAP1 and RhoC were closely correlated. The transwell migration assay revealed that both IQGAP1 and RhoC stimulated the migration activity of the gastric cancer cell line AGS. The knockdown of IQGAP1 expression by siRNA blocked the migration­stimulating activity of RhoC, while the knockdown of RhoC expression had no effect on the migration-stimulatory activity of IQGAP1. Co-IP results showed that RhoC and IQGAP1 bound to each other. These results reveal a previously unrecognized interaction between IQGAP1 and RhoC, and demonstrate that IQGAP1 is a downstream effector of RhoC in the regulation of the migration activity of gastric cancer cells.


Subject(s)
Stomach Neoplasms/metabolism , ras GTPase-Activating Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Cell Line, Tumor , Cell Movement , Humans , RNA Interference , RNA, Small Interfering/metabolism , Stomach Neoplasms/pathology , ras GTPase-Activating Proteins/antagonists & inhibitors , ras GTPase-Activating Proteins/genetics , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/genetics , rhoC GTP-Binding Protein
6.
Yi Chuan ; 32(4): 331-8, 2010 Apr.
Article in Chinese | MEDLINE | ID: mdl-20423886

ABSTRACT

Discovery of histone lysine specific demethylase 1 (LSD1) indicates that even histone methylation is reversible. Structural analysis shows that LSD1 is a flavin-dependent amine oxidase, which is able to catalyze the specific removal of methyl groups from mono- and dimethylated Lys4 and Lys9 of histone H3. Functional studies demonstrate that LSD1 regulates activation and inhibition of gene transcription in the nucleus, which is known as the innermost gene switch of cells. LSD1 plays important roles in embryonic development and tumorigenesis. Here, we review recent insights into the structure and chemical mechanism of LSD1, and its regulatory roles in development and cancer.


Subject(s)
Histone Demethylases/metabolism , Animals , Embryonic Development , Histone Demethylases/chemistry , Humans , Neoplasms/metabolism , Neoplasms/pathology
7.
Chin J Cancer ; 29(2): 158-62, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20109343

ABSTRACT

BACKGROUND AND OBJECTIVE: Nitric oxide (NO) is involved in many physiologic and pathologic processes. As an important biologic mediator, NO has been the focus of cancer study for its function in tumorigenesis, tumor progression, and death. This study investigated the effect of NO donor sodium nitroprusside (SNP) on the growth and proliferation of gastric cancer cell line AGS. METHODS: The growth inhibition of AGS cells was analyzed using MTT assay. The cell cycle was measured using flow cytometry. The changes of mRNA expression of proliferating cell nuclear antigen (PCNA) and caspase-3 were examined using reverse transcriptase polymerase chain reaction (RT-PCR), and the protein expressions of PCNA and caspase-3 were analyzed using Western blot. RESULTS: Dose-dependent SNP inhibited cell growth and proliferation. When the AGS cells were treated with SNP at 100, 500, 1000, 1500, and 2000 mumol/L for 24 h, the growth inhibition rates were (2.02 +/- 2.96)%, (10.82 +/- 2.21)%, (18.95 +/- 3.35)%, (26.88 +/- 2.54)%, and (42.57 +/- 1.27)%, respectively (P < 0.05). SNP altered the cell cycle in AGS cells. Compared with the control group, treatment with SNP at 100, 500, 1000, 1500, and 2000 mumol/L for 24 h reduced the number of cells in the S phase by 2.29%, 7.8%, 11.34%, 20.49%, and 23.6%, respectively, and enhanced the number of cells in the G1/G0 phases by 3.33%, 9.3%, 13.46%, 21.37%, and 24.73%, respectively (P < 0.05). With the increasing concentration and action time of SNP, the expressions of PCNA mRNA and protein decreased. The expression of caspase-3 mRNA remained unchanged, but procaspase-3 was activated. CONCLUSION: NO not only inhibits cell growth and proliferation, but also induces apoptosis in gastric cancer cells, and such effects of NO showed significant dose-dependent activity.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Nitric Oxide Donors/pharmacology , Nitroprusside/pharmacology , Stomach Neoplasms/pathology , Caspase 3/genetics , Caspase 3/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic , Humans , Proliferating Cell Nuclear Antigen/genetics , Proliferating Cell Nuclear Antigen/metabolism , RNA, Messenger/metabolism , Stomach Neoplasms/metabolism
8.
Mol Med Rep ; 3(2): 361-6, 2010.
Article in English | MEDLINE | ID: mdl-21472248

ABSTRACT

Our previous studies have demonstrated that the expression and activity of protein kinase G (PKG) II are significantly lower in human gastric cancer cell lines than in normal cells. This study was designed to investigate the effect of PKG II activation on the proliferation of human cultured BGC-823 gastric cancer cells. An adenoviral construct encoding the PKG II gene (Ad-PKG II) was used to infect BGC-823 cells, and the activity of the enzyme was induced by cGMP analogue 8-pCPT-cGMP. The proliferation-inhibitory effect of PKG II was analyzed by the MTT assay, BrdU incorporation assay and detection of proliferating cell nuclear antigen (PCNA) expression. Colony formation in soft agarose was performed to analyze the effect of PKG II on the anchorage-independent growth of the cells. The effect of PKG II in vivo was investigated in an immunocompromised nude mice model, and its effect on the cell cycle was analyzed by flow cytometry. The results showed that Ad-PKG II infection increased the expression of PKG II in BGC-823 cells. The activation of PKG II by 8-pCPT-cGMP caused a significant decrease in the number of live cells and inhibited DNA synthesis in individual cells. PKG II activation inhibited the EGF-induced increase in PCNA expression. The activation of PKG II also caused a significant inhibition of colony formation in soft agarose and significantly suppressed the in vivo growth of BGC-823 cells in immunocompromised nude mice. There was substantial cell arrest at the G1 phase and a decrease in the number of S phase cells in the Ad-PKG II/8-pCPT-cGMP-treated cells. These data indicate that the activation of PKG II by 8-pCPT-cGMP inhibits the proliferation of human gastric cancer cells.

9.
Mol Med Rep ; 3(4): 657-62, 2010.
Article in English | MEDLINE | ID: mdl-21472294

ABSTRACT

Vasodilator stimulated phosphoprotein (VASP) is known as an actin-binding protein. The phosphorylation of VASP plays an important role in its function. In a previous study, serine 157 phosphorylated VASP (p-VASP S157) was shown to be co-localized with α-tubulin on the spindle of SGC-7901 cells. In the present study, we demonstrated that the level of p-VASP S157 increases and has a peak which coincides with serine 10 phosphorylated histone 3 (p-H3 S10) during mitotic progression in a human cervical cancer cell line (HeLa cells). Application of protein kinase A inhibitor H89, protein kinase G inhibitor KT5823 and protein kinase C inhibitor Go6983, or a combination of these inhibitors, caused a partial decrease in p-VASP S157 and a delay in G2/M progression. Depletion of p-VASP S157 by VASP siRNA resulted in an increase in binucleated cells and x4n cells, a further delay in G2/M progression and the inhibition of HeLa cell proliferation. These results suggest that p-VASP S157 may play an important role in the G2/M transition and the completion of cytokinesis in HeLa cells.

10.
Article in English | MEDLINE | ID: mdl-18349510

ABSTRACT

The sources of nitric oxide (NO) production in response to abscisic acid (ABA) and the role of NO in ABA-induced hydrogen peroxide (H(2)O(2)) accumulation and subcellular antioxidant defense in leaves of maize (Zea mays L.) plants were investigated. ABA induced increases in generation of NO and activity of nitric oxide synthase (NOS) in maize leaves. Such increases were blocked by pretreatment with each of the two NOS inhibitors. Pretreatments with a NO scavenger or NR inhibitors inhibited ABA-induced increase in production of NO, but did not affect the ABA-induced increases in activity of NOS, indicating that ABA-induced NO production originated from sources of NOS and NR. ABA- and H(2)O(2)-induced increases in expression of the antioxidant genes superoxide dismutase 4 (SOD4), cytosolic ascorbate peroxidase (cAPX), and glutathione reductase 1 (GR1) and the activities of the chloroplastic and cytosolic antioxidant enzymes were arrested by pretreatments with the NO scavenger, inhibitors of NOS and NR, indicating that NO is involved in the ABA- and H(2)O(2)-induced subcellular antioxidant defense reactions. On the other hand, NO donor sodium nitroprusside (SNP) reduced accumulation of H(2)O(2) induced by ABA, and c-PTIO reversed the effect of SNP in decreasing the accumulation of H(2)O(2). SNP induced increases in activities of subcellular antioxidant enzymes, and the increases were substantially prevented from occurring by the pretreatment with c-PTIO. These results suggest that ABA induces production of H(2)O(2) and NO, which can up-regulate activities of the subcellular antioxidant enzymes, to prevent overproduction of H(2)O(2) in maize plants. There is a negative feedback loop between NO and H(2)O(2) in ABA signal transduction in maize plants.


Subject(s)
Abscisic Acid/pharmacology , Nitric Oxide/metabolism , Nitric Oxide/physiology , Plant Leaves/metabolism , Zea mays/metabolism , Antioxidants/metabolism , Ascorbate Peroxidases , Glutathione Reductase/metabolism , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Microscopy, Confocal , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitroprusside/pharmacology , Peroxidases/metabolism , Plant Leaves/drug effects , Superoxide Dismutase/metabolism , Zea mays/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...