Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Neurobiol Dis ; 89: 202-12, 2016 May.
Article in English | MEDLINE | ID: mdl-26875527

ABSTRACT

Perinatal hypoxic-ischemic encephalopathy (HIE) is associated with high neonatal mortality and severe long-term neurologic morbidity. Yet the mechanisms of brain injury in infants with HIE remain largely elusive. The present study determined a novel mechanism of microRNA-210 (miR-210) in silencing endogenous neuroprotection and increasing hypoxic-ischemic brain injury in neonatal rats. The study further revealed a potential therapeutic effect of miR-210 inhibition using complementary locked nucleic acid oligonucleotides (miR-210-LNA) in 10-day-old neonatal rats in the Rice-Vannucci model. The underlying mechanisms were investigated with intracerebroventricular injection (i.c.v) of miR-210 mimic, miR-210-LNA, glucocorticoid receptor (GR) agonist and antagonist. Luciferase reporter gene assay was conducted for identification of miR-210 targeting GR 3'untranslated region. The results showed that the HI treatment significantly increased miR-210 levels in the brain, and miR-210 mimic significantly decreased GR protein abundance and exacerbated HI brain injury in the pups. MiR-210-LNA administration via i.c.v. 4h after the HI insult significantly decreased brain miR-210 levels, increased GR protein abundance, reduced HI-induced neuronal death and brain infarct size, and improved long-term neurological function recovery. Of importance, the intranasal delivery of miR-210-LNA 4h after the HI insult produced similar effects in decreasing HI-induced neonatal brain injury and improving neurological function later in life. Altogether, the present study provides evidence of a novel mechanism of miR-210 in a neonatal HI brain injury model, and suggests a potential therapeutic approach of miR-210 inhibition in the treatment of neonatal HIE.


Subject(s)
Brain/metabolism , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/prevention & control , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Neuroprotective Agents/administration & dosage , Oligonucleotides/administration & dosage , 3' Untranslated Regions , Animals , Animals, Newborn , Rats , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism
2.
J Mol Cell Cardiol ; 91: 160-71, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26779948

ABSTRACT

Large studies in humans and animals have demonstrated a clear association of an adverse intrauterine environment with an increased risk of cardiovascular disease later in life. Yet mechanisms remain largely elusive. The present study tested the hypothesis that gestational hypoxia leads to promoter hypermethylation and epigenetic repression of the glucocorticoid receptor (GR) gene in the developing heart, resulting in increased heart susceptibility to ischemia and reperfusion injury in offspring. Hypoxic treatment of pregnant rats from day 15 to 21 of gestation resulted in a significant decrease of GR exon 14, 15, 16, and 17 transcripts, leading to down-regulation of GR mRNA and protein in the fetal heart. Functional cAMP-response elements (CREs) at -4408 and -3896 and Sp1 binding sites at -3425 and -3034 were identified at GR untranslated exon 1 promoters. Hypoxia significantly increased CpG methylation at the CREs and Sp1 binding sites and decreased transcription factor binding to GR exon 1 promoter, accounting for the repression of the GR gene in the developing heart. Of importance, treatment of newborn pups with 5-aza-2'-deoxycytidine reversed hypoxia-induced promoter methylation, restored GR expression and prevented hypoxia-mediated increase in ischemia and reperfusion injury of the heart in offspring. The findings demonstrate a novel mechanism of epigenetic repression of the GR gene in fetal stress-mediated programming of ischemic-sensitive phenotype in the heart.


Subject(s)
Epigenesis, Genetic , Hypoxia/genetics , Myocardial Reperfusion Injury/genetics , Oxygen/pharmacology , Receptors, Glucocorticoid/genetics , Sp1 Transcription Factor/genetics , Animals , Animals, Newborn , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Binding Sites , DNA Methylation/drug effects , Decitabine , Exons , Female , Hypoxia/drug therapy , Hypoxia/metabolism , Hypoxia/pathology , Male , Maternal Exposure , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Phenotype , Pregnancy , Promoter Regions, Genetic , Protein Binding , Rats , Rats, Sprague-Dawley , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/metabolism , Response Elements , Sp1 Transcription Factor/metabolism
3.
PLoS One ; 10(4): e0125033, 2015.
Article in English | MEDLINE | ID: mdl-25923220

ABSTRACT

The potential adverse effect of synthetic glucocorticoid, dexamethasone therapy on the developing heart remains unknown. The present study investigated the effects of dexamethasone on cardiomyocyte proliferation and binucleation in the developing heart of newborn rats and evaluated DNA methylation as a potential mechanism. Dexamethasone was administered intraperitoneally in a three day tapered dose on postnatal day 1 (P1), 2 and 3 to rat pups in the absence or presence of a glucocorticoid receptor antagonist Ru486, given 30 minutes prior to dexamethasone. Cardiomyocytes from P4, P7 or P14 animals were analyzed for proliferation, binucleation and cell number. Dexamethasone treatment significantly increased the percentage of binucleated cardiomyocytes in the hearts of P4 pups, decreased myocyte proliferation in P4 and P7 pups, reduced cardiomyocyte number and increased the heart to body weight ratio in P14 pups. Ru486 abrogated the effects of dexamethasone. In addition, 5-aza-2'-deoxycytidine (5-AZA) blocked the effects of dexamethasone on binucleation in P4 animals and proliferation at P7, leading to recovered cardiomyocyte number in P14 hearts. 5-AZA alone promoted cardiomyocyte proliferation at P7 and resulted in a higher number of cardiomyocytes in P14 hearts. Dexamethasone significantly decreased cyclin D2, but not p27 expression in P4 hearts. 5-AZA inhibited global DNA methylation and blocked dexamethasone-mediated down-regulation of cyclin D2 in the heart of P4 pups. The findings suggest that dexamethasone acting on glucocorticoid receptors inhibits proliferation and stimulates premature terminal differentiation of cardiomyocytes in the developing heart via increased DNA methylation in a gene specific manner.


Subject(s)
Dexamethasone/administration & dosage , Epigenesis, Genetic , Heart/growth & development , Myocytes, Cardiac/pathology , Receptors, Glucocorticoid/biosynthesis , Animals , Animals, Newborn , Cell Proliferation/drug effects , Cyclin D2/biosynthesis , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , DNA Methylation/drug effects , Gene Expression Regulation, Developmental/drug effects , Heart/drug effects , Humans , Mifepristone/administration & dosage , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats
4.
Drug Discov Today ; 19(12): 1883-96, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25256780

ABSTRACT

Adverse environmental conditions faced by an individual early during its life, such as gestational hypoxia, can have a profound influence on the risk of diseases, such as neurological disorders, in later life. Clinical and preclinical studies suggest that epigenetic programming of gene expression patterns in response to maternal stress have a crucial role in the fetal origins of neurological diseases. Herein, we summarize recent studies regarding the role of epigenetic mechanisms in the developmental programming of neurological diseases in offspring, primarily focusing on DNA methylation/demethylation and miRNAs. Such information could increase our understanding of the fetal origins of adult diseases and help develop effective prevention and intervention against neurological diseases.


Subject(s)
Brain Diseases/genetics , Fetal Hypoxia/genetics , Animals , Brain Diseases/etiology , Epigenesis, Genetic , Fetal Hypoxia/complications , Humans
5.
Hypertension ; 64(3): 610-8, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24914190

ABSTRACT

Our previous study demonstrated that pregnancy increased large-conductance Ca(2+)-activated potassium channel ß1 subunit (BKß1) expression and large-conductance Ca(2+)-activated potassium channel activity in uterine arteries, which were abrogated by chronic hypoxia. The present study tested the hypothesis that promoter methylation/demethylation is a key mechanism in epigenetic reprogramming of BKß1 expression patterns in uterine arteries. Ovine BKß1 promoter of 2315 bp spanning from -2211 to +104 of the transcription start site was cloned, and an Sp1-380 binding site that contains CpG dinucleotide in its core binding sequences was identified. Site-directed deletion of the Sp1 site significantly decreased the BKß1 promoter activity. Estrogen receptor-α bound to the Sp1 site through tethering to Sp1 and upregulated the expression of BKß1. The Sp1 binding site at BKß1 promoter was highly methylated in uterine arteries of nonpregnant sheep, and methylation inhibited transcription factor binding and BKß1 promoter activity. Pregnancy caused a significant decrease in CpG methylation at the Sp1 binding site and increased Sp1 binding to the BKß1 promoter and BKß1 mRNA abundance. Chronic hypoxia during gestation abrogated this pregnancy-induced demethylation and upregulation of BKß1 expression. The results provide evidence of a novel mechanism of promoter demethylation in pregnancy-induced reprogramming of large-conductance Ca(2+)-activated potassium channel expression and function in uterine arteries and suggest new insights of epigenetic mechanisms linking gestational hypoxia to aberrant uteroplacental circulation and increased risk of preeclampsia.


Subject(s)
Adaptation, Physiological/physiology , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/physiology , Pregnancy, Animal/physiology , Uterine Artery/physiology , Animals , CpG Islands/genetics , CpG Islands/physiology , DNA Methylation , Epigenesis, Genetic/physiology , Female , Large-Conductance Calcium-Activated Potassium Channel beta Subunits/genetics , Models, Animal , Pregnancy , Pregnancy-Specific beta 1-Glycoproteins/physiology , Protein Binding/physiology , Sheep , Up-Regulation
6.
Neurobiol Dis ; 65: 172-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24513088

ABSTRACT

Gestational hypoxia is a common stress to the fetal development and increases the risk of neonatal morbidity. The present study tested the hypothesis that fetal hypoxia results in heightened brain vulnerability to hypoxic-ischemic (HI) injury in neonatal rats via down-regulation of glucocorticoid receptor (GR) in the developing brain. Time-dated pregnant rats were exposed to hypoxia (10.5% O2) from days 15 to 21 of gestation. Brain HI injury was determined in day 10 pups. Maternal hypoxia resulted in asymmetric intrauterine growth restriction in the fetus. The brain HI injury was significantly increased in maternal hypoxia-treated pups as compared with the normoxia control in both males and females. Activation of brain GR by dexamethasone injection into the right lateral ventricle produced a concentration-dependent reduction of HI-induced brain injury in control pups. Maternal hypoxia significantly decreased GR mRNA and protein abundance in the fetal brain and neonatal hippocampus and abolished the dexamethasone-mediated neuroprotective effect in pup brains. This decreased GR expression was resulted from increased DNA methylation, decreased binding of transcription factors Egr-1 and Sp1 to GR gene exon 17 and 111 promoters, and reduced expression of GR exon 17 and 111 mRNA variants. The results demonstrate that gestational hypoxia causes epigenetic repression of GR gene expression in the developing brain resulting in the heightened brain vulnerability to HI injury in neonatal rats.


Subject(s)
Fetal Hypoxia/complications , Gene Expression Regulation, Developmental/physiology , Hypoxia-Ischemia, Brain/etiology , Hypoxia-Ischemia, Brain/metabolism , Receptors, Glucocorticoid/metabolism , Age Factors , Animals , Animals, Newborn , Body Weight , Brain/growth & development , Brain/metabolism , Brain Infarction/etiology , Brain Infarction/pathology , DNA Methylation , Dexamethasone/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Fetal Hypoxia/pathology , Gene Expression Regulation, Developmental/drug effects , Glucocorticoids/therapeutic use , Hypoxia-Ischemia, Brain/drug therapy , Male , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Glucocorticoid/genetics , Sex Factors
7.
Mol Genet Metab ; 111(4): 477-83, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24529979

ABSTRACT

Glucose is the primary energy source for eukaryotic cells and the predominant substrate for the brain. GLUT3 is essential for trans-placental glucose transport and highly expressed in the mammalian brain. To further elucidate the role of GLUT3 in embryonic development, we utilized the vertebrate whole animal model system of Danio rerio as a tractable system for defining the cellular and molecular mechanisms altered by impaired glucose transport and metabolism related to perturbed expression of GLUT3. The comparable orthologue of human GLUT3 was identified and the expression of this gene abrogated during early embryonic development. In a dose-dependent manner embryonic brain development was disrupted resulting in a phenotype of aberrant brain organogenesis, associated with embryonic growth restriction and increased cellular apoptosis. Rescue of the morphant phenotype was achieved by providing exogenous GLUT3 mRNA. We conclude that GLUT3 is critically important for brain organogenesis and embryonic growth. Disruption of GLUT3 is responsible for the phenotypic spectrum of embryonic growth restriction to demise and neural apoptosis with microcephaly.


Subject(s)
Brain/embryology , Brain/metabolism , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Glucose Transporter Type 3/genetics , Zebrafish Proteins/genetics , Zebrafish/embryology , Zebrafish/genetics , Animals , Apoptosis/genetics , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Gene Knockdown Techniques , Glucose Transporter Type 3/metabolism , Humans , In Situ Hybridization , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Survival Analysis , Zebrafish Proteins/metabolism
8.
Am J Physiol Regul Integr Comp Physiol ; 304(8): R613-20, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23427085

ABSTRACT

Maternal hypoxia inhibits cardiomyocyte proliferation in the heart of fetal and neonatal rats. The present study tested the hypothesis that hypoxia has a direct effect inhibiting cardiomyocyte proliferation via upregulating tissue inhibitors of metalloproteinases (TIMP) in fetal rat hearts. Isolated fetal rat hearts and rat embryonic ventricular myocyte H9c2 cells were treated ex vivo with 20% or 1% O(2) for 48 or 24 h, respectively. Hypoxia caused a significant reduction in cardiomyocyte Ki-67 expression and bromodeoxyuridine incorporation in fetal hearts and H9c2 cells. In both fetal hearts and H9c2 cells, hypoxia resulted in a significant decrease in a cell division marker cyclin D2 but an increase in a cell division inhibitor p27. Additionally, hypoxia caused an upregulation of TIMP-3 and TIMP-4 in fetal hearts and H9c2 cells. Knockdown of TIMP-3 in H9c2 cells significantly increased cyclin D2 and Ki-67 and partially blocked the hypoxia-induced inhibition of cyclin D2 and Ki-67 in H9c2 cells. Unlike TIMP-3, TIMP-4 knockdown had no significant effects on the basal levels of cell proliferation but completely abrogated the hypoxia-mediated effects. These findings provide evidence of a novel causal role of TIMP-4 and TIMP-3 in the direct inhibitory effect of hypoxia on cardiomyocyte proliferation in the developing heart.


Subject(s)
Cell Proliferation , Fetal Heart/physiology , Fetal Hypoxia/pathology , Myocytes, Cardiac/physiology , Tissue Inhibitor of Metalloproteinases/physiology , Animals , Antimetabolites , Blotting, Western , Bromodeoxyuridine , Cell Line , Cell Size , Cyclin D2/biosynthesis , Cyclin D2/physiology , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , Female , Fluorescent Antibody Technique , Ki-67 Antigen/biosynthesis , Pregnancy , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Tissue Inhibitor of Metalloproteinase-3/genetics , Tissue Inhibitor of Metalloproteinase-3/physiology , Tissue Inhibitor of Metalloproteinases/genetics , Transfection , Up-Regulation/physiology , Tissue Inhibitor of Metalloproteinase-4
9.
Am J Physiol Regul Integr Comp Physiol ; 304(9): R683-9, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23427086

ABSTRACT

Fetal hypoxia causes protein kinase Cε (PKCε) gene repression in the heart resulting in heightened ischemic injury in male offspring in a sex-dependent manner. The present study tested the hypothesis that heightened methylation of the early growth response factor-1 (Egr-1) binding site at PKCε gene promoter contributes to sex dimorphism of hypoxia-induced programming of PKCε gene repression in the developing heart. Pregnant rats were divided into normoxic and hypoxic (10.5% O2 from day 15 to 21 of gestation) groups. Hypoxia selectively decreased PKCε mRNA and protein abundance in the heart of male, but not female, near-term (21 days) fetuses. Methylation of the CpG site at the Egr-1 binding site of PKCε promoter was significantly increased in the male hearts by hypoxia, resulting in decreased Egr-1 binding affinity and reduced Egr-1 binding to the PKCε promoter. Nuclear Egr-1 levels were not affected by hypoxia. There was significantly higher abundance of estrogen receptor α (ERα) and ß (ERß) isoforms in female than in male fetal hearts, which were not significantly altered by hypoxia. Both ERα and ERß bind to the Egr-1 binding site with significant greater levels in the female fetal hearts. The increased methylation with reduced Egr-1 binding and PKCε gene repression persisted in 3-mo-old adult male hearts in a sex-dependent manner. The results indicate a key role for heightened methylation of the Egr-1 binding site in hypoxia-mediated programming of PKCε gene repression in the developing heart and suggest a novel protective mechanism of ER by binding to the Egr-1 binding site in epigenetic regulation of PKCε gene expression patterns in the early developmental stage.


Subject(s)
Early Growth Response Protein 1/genetics , Epigenesis, Genetic/physiology , Fetal Hypoxia/genetics , Heart/growth & development , Myocardium/enzymology , Protein Kinase C-epsilon/genetics , Animals , Animals, Newborn , Antimetabolites/pharmacology , Azacitidine/pharmacology , Binding Sites , Blotting, Western , Chromatin Immunoprecipitation , DNA Methylation , Electrophoretic Mobility Shift Assay , Epigenesis, Genetic/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Female , Male , Pregnancy , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Sex Characteristics
10.
Front Neuroendocrinol ; 34(1): 27-46, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23200813

ABSTRACT

Adverse environments during the fetal and neonatal development period may permanently program physiology and metabolism, and lead to increased risk of diseases in later life. Programming of the hypothalamic-pituitary-adrenal (HPA) axis is one of the key mechanisms that contribute to altered metabolism and response to stress. Programming of the HPA axis often involves epigenetic modification of the glucocorticoid receptor (GR) gene promoter, which influences tissue-specific GR expression patterns and response to stimuli. This review summarizes the current state of research on the HPA axis and programming of health and disease in the adult, focusing on the epigenetic regulation of GR gene expression patterns in response to fetal and neonatal stress. Aberrant GR gene expression patterns in the developing brain may have a significant negative impact on protection of the immature brain against hypoxic-ischemic encephalopathy in the critical period of development during and immediately after birth.


Subject(s)
Epigenesis, Genetic/physiology , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Stress, Physiological/physiology , Animals , Epigenesis, Genetic/genetics , Humans , Hypothalamo-Hypophyseal System/embryology , Hypothalamo-Hypophyseal System/growth & development , Pituitary-Adrenal System/embryology , Pituitary-Adrenal System/growth & development , Receptors, Glucocorticoid/genetics
11.
PLoS One ; 7(9): e45290, 2012.
Article in English | MEDLINE | ID: mdl-22984631

ABSTRACT

BACKGROUND: Lysosomal protein transmembrane 4 beta (LAPTM4B) is a novel cancer-related gene which has two alleles designated LAPTM4B*1 and LAPTM4B*2. In this study we investigated the correlation of LAPTM4B genotype with prognosis and clinicopathologic features in patients who had undergone curative resection for gallbladder carcinoma (GBC). METHODOLOGY/PRINCIPAL FINDINGS: PCR assay was performed to determine the LAPTM4B genotype in 85 patients. The correlation of LAPTM4B genotype with clinicopathologic parameters was assessed with the Chi-squared test. Differences in patient survival were determined by the Kaplan-Meier method. Multivariate analysis of prognostic factors was carried out with Cox regression analysis. Patients with LAPTM4B *2 had both significantly shorter overall survival (OS) and shorter disease-free survival (DFS) (both P<0.001). Multivariate analysis showed that LAPTM4B genotype is a prognostic factor for OS and DFS (both P<0.001). CONCLUSIONS/SIGNIFICANCE: LAPTM4B allele *2 is a risk factor associated with poor prognosis in patients with resected GBC, and LAPTM4B status may be therefore be useful preoperatively as an adjunct in evaluation of the operability of GBC.


Subject(s)
Biomarkers, Tumor/genetics , Gallbladder Neoplasms/genetics , Membrane Proteins/genetics , Oncogene Proteins/genetics , Adult , Aged , Alleles , Chi-Square Distribution , Female , Gallbladder Neoplasms/pathology , Gallbladder Neoplasms/surgery , Genotype , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Polymerase Chain Reaction , Prognosis , Proportional Hazards Models , Risk Assessment/statistics & numerical data , Risk Factors
12.
Stroke ; 43(9): 2483-90, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22738920

ABSTRACT

BACKGROUND AND PURPOSE: Maternal cigarette smoking increases the risk of neonatal morbidity. We tested the hypothesis that perinatal nicotine exposure causes heightened brain vulnerability to hypoxic-ischemic (HI) injury in neonatal rats through aberrant expression patterns of angiotensin II type 1 (AT(1)R) and type 2 (AT(2)R) receptors in the developing brain. METHODS: Nicotine was administered to pregnant rats through subcutaneous osmotic minipumps. HI brain injury was determined in 10-day-old pups. AT(1)R and AT(2)R expression patterns were assessed through Western blotting, quantitative polymerase chain reaction, immunofluorescence, and confocal imaging. RESULTS: Perinatal nicotine exposure significantly increased HI brain infarct size in male, but not female, pups. In fetal brains, nicotine caused a decrease in mRNA and protein abundance of AT(2)R but not AT(1)R. The downregulation of AT(2)R persisted in brains of male pups, and nicotine treatment resulted in a significant increase in methylation of CpG locus 3 bases upstream of TATA-box at the AT(2)R gene promoter. In female brains, there was an increase in AT(2)R but a decrease in AT(1)R expression. Both AT(1)R and AT(2)R expressed in neurons but not in astrocytes in the cortex and hippocampus. Central application of AT(1)R antagonist losartan or AT(2)R antagonist PD123319 increased HI brain infarct size in both male and female pups. In male pups, AT(2)R agonist CGP42112 abrogated nicotine-induced increase in HI brain infarction. In females, PD123319 uncovered the nicotine's effect on HI brain infarction. CONCLUSIONS: Perinatal nicotine exposure causes epigenetic repression of the AT(2)R gene in the developing brain resulting in heightened brain vulnerability to HI injury in neonatal male rats in a sex-dependent manner.


Subject(s)
Brain Injury, Chronic/pathology , Brain Ischemia/pathology , Hypoxia, Brain/pathology , Nicotine/toxicity , Nicotinic Agonists/toxicity , Receptors, Angiotensin/physiology , Angiotensin II/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Animals, Newborn , Blotting, Western , Brain/pathology , DNA Methylation/drug effects , Female , Fetal Growth Retardation/chemically induced , Fetal Growth Retardation/pathology , Imidazoles/pharmacology , Imidazoles/therapeutic use , Immunohistochemistry , Male , Microscopy, Confocal , Pregnancy , Pyridines/pharmacology , Pyridines/therapeutic use , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptor, Angiotensin, Type 1/biosynthesis , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 2/biosynthesis , Receptor, Angiotensin, Type 2/genetics
13.
PLoS One ; 7(4): e34984, 2012.
Article in English | MEDLINE | ID: mdl-22509374

ABSTRACT

BACKGROUND: Lysosomal protein transmembrane 4 beta (LAPTM4B) is a gene related to hepatocellular carcinoma that has two alleles designated LAPTM4B*1 and LAPTM4B*2. This study aimed to investigate the correlation of LAPTM4B genotype with prognosis and clinicopathologic features in patients who have undergone resection for hepatocellular carcinoma (HCC). METHODOLOGY/PRINCIPAL FINDINGS: The LAPTM4B genotype was analyzed by PCR in 68 patients who had undergone curative hepatic resection for hepatocellular carcinoma. The correlation of LAPTM4B genotype with clinicopathologic parameters was assessed with the Chi-squared test. Differences in patient survival were determined by the Kaplan-Meier method. Multivariate analysis of prognostic factors was carried out with Cox regression analysis. Patients with LAPTM4B *2 had both significantly shorter overall survival (OS) and shorter disease-free survival (DFS) (both P<0.001). Multivariate analysis showed that LAPTM4B genotype is an independent prognostic factor for OS and DFS (both P<0.001). CONCLUSIONS/SIGNIFICANCE: Allele *2 of LAPTM4B is a risk factor associated with poor prognosis in patients with resected HCC. LAPTM4B status may be useful preoperatively as an adjunct in evaluation of the operability of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Membrane Proteins/genetics , Oncogene Proteins/genetics , Prognosis , Adult , Aged , Aged, 80 and over , Alleles , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/surgery , Disease-Free Survival , Female , Genetic Association Studies , Humans , Kaplan-Meier Estimate , Liver Neoplasms/blood , Liver Neoplasms/surgery , Male , Membrane Proteins/blood , Middle Aged , Oncogene Proteins/blood
14.
FASEB J ; 26(7): 2753-63, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22441984

ABSTRACT

Heart disease is the leading cause of death in the United States. Recent studies demonstrate that fetal programming of PKCε gene repression results in ischemia-sensitive phenotype in the heart. The present study tests the hypothesis that increased norepinephrine causes epigenetic repression of PKCε gene in the heart via Nox1-dependent reactive oxygen species (ROS) production. Prolonged norepinephrine treatment increased ROS production in fetal rat hearts and embryonic ventricular myocyte H9c2 cells via a selective increase in Nox1 expression. Norepinephrine-induced ROS resulted in an increase in PKCε promoter methylation at Egr-1 and Sp-1 binding sites, leading to PKCε gene repression. N-acetylcysteine, diphenyleneiodonium, and apocynin blocked norepinephrine-induced ROS production and the promoter methylation, and also restored PKCε mRNA and protein to control levels in vivo in fetal hearts and in vitro in embryonic myocyte cells. Accordingly, norepinephrine-induced ROS production, promoter methylation, and PKCε gene repression were completely abrogated by knockdown of Nox1 in cardiomyocytes. These findings provide evidence of a novel interaction between elevated norepinephrine and epigenetic repression of PKCε gene in the heart mediated by Nox1-dependent oxidative stress and suggest new insights of molecular mechanisms linking the heightened sympathetic activity to aberrant cardioprotection and increased ischemic vulnerability in the heart.


Subject(s)
Fetal Heart/drug effects , Fetal Heart/metabolism , NADH, NADPH Oxidoreductases/metabolism , Norepinephrine/pharmacology , Protein Kinase C-epsilon/genetics , Animals , Binding Sites/genetics , Cell Line , CpG Islands , DNA Methylation/drug effects , Early Growth Response Protein 1/metabolism , Epigenesis, Genetic/drug effects , Gene Knockdown Techniques , Models, Cardiovascular , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NADH, NADPH Oxidoreductases/antagonists & inhibitors , NADH, NADPH Oxidoreductases/genetics , NADPH Oxidase 1 , Promoter Regions, Genetic , Protein Kinase C-epsilon/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Rats , Reactive Oxygen Species/metabolism , Sp1 Transcription Factor/metabolism
15.
Med Oncol ; 29(4): 2809-13, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22302286

ABSTRACT

Gallbladder carcinoma (GBC) is a malignancy with an extremely poor prognosis. In order to improve the survival rate, identification of new susceptibility risk factors is of importance. Here, we report findings on the novel cancer-related gene lysosomal protein transmembrane 4 beta (LAPTM4B) that has two alleles designated LAPTM4B*1 and LAPTM4B*2. Allele *1 differs from allele *2 in that it contains one copy of a 19-bp sequence, whereas this sequence is duplicated in exon 1 of allele *2. This study aimed to investigate the relationship of LAPTM4B allelic variation and GBC susceptibility. LAPTM4B genotype was analyzed in 155 healthy individuals and 91 GBC patients by PCR, and the genotypic distribution of LAPTM4B was analyzed with the chi-squared test. The frequency of allele *2 was 37.9 and 24.8% in the GBC and the control groups, respectively, representing a significant difference between these two groups (P<0.001). LAPTM4B allele *2 may be a risk factor associated with genetic susceptibility to GBC.


Subject(s)
Asian People/genetics , Gallbladder Neoplasms/genetics , Genetic Predisposition to Disease , Membrane Proteins/genetics , Oncogene Proteins/genetics , Polymorphism, Genetic , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Base Sequence , Female , Genotype , Humans , Male , Middle Aged , Molecular Sequence Data
16.
Cardiovasc Res ; 93(2): 302-10, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22139554

ABSTRACT

AIMS: Hypoxia causes protein kinase C epsilon (PKCε) gene repression in foetal hearts, resulting in heightened cardiac susceptibility to ischaemic injury in offspring. We tested the hypothesis that hypoxia inducible factor 1 (HIF-1) and/or reactive oxygen species (ROS) mediate hypoxia-induced PKCε gene repression. METHODS AND RESULTS: Hypoxia induced in vivo to pregnant rats, ex vivo to isolated foetal rat hearts, and in vitro in the rat embryonic ventricular myocyte cell line H9c2 resulted in a comparable decrease in PKCε protein and mRNA abundance in foetal hearts and H9c2 cells, which was associated with a significant increase in CpG methylation of the SP1-binding sites at the PKCε promoter. In H9c2 cells and foetal hearts, hypoxia caused nuclear accumulation of HIF-1α, which was inhibited by 3-(5'-hydroxymethyl-2'-furyl)-1-benzylindazole and 2-methoxy estradiol. The HIF-1α inhibitors had no significant effect on hypoxia-induced PKCε mRNA repression. Hypoxia produced a time-dependent increase in ROS production in H9c2 cells and foetal hearts that was blocked by ROS scavengers N-acetyl-cysteine or tempol. In accordance, N-acetyl-cysteine and tempol, but not apocynin, inhibited the hypoxic effect and restored PKCε protein and mRNA expression to the control values in foetal hearts and H9c2 cells. The ROS scavengers blocked hypoxia-induced CpG methylation of the SP1-binding sites, restored SP1 binding to the PKCε promoter, and abrogated the hypoxia-induced increase in the susceptibility of the heart to ischaemic injury in offspring. CONCLUSIONS: The results demonstrate that hypoxia induces epigenetic repression of the PKCε gene through a NADPH oxidase-independent ROS-mediated pathway in the foetal heart, leading to heightened heart vulnerability to ischaemic injury in offspring.


Subject(s)
Epigenesis, Genetic , Fetal Heart/metabolism , Gene Expression Regulation, Developmental , Hypoxia/metabolism , Oxidative Stress , Protein Kinase C-epsilon/genetics , Acetylcysteine/pharmacology , Animals , Cyclic N-Oxides/pharmacology , DNA Methylation , Female , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Myocardial Reperfusion Injury/etiology , NADPH Oxidases/physiology , Pregnancy , Promoter Regions, Genetic , Protein Kinase C-epsilon/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Spin Labels
17.
Med Oncol ; 29(4): 2744-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22207410

ABSTRACT

Hepatocellular carcinoma (HCC) is the leading cause of cancer mortality in many countries. Evaluation of new susceptibility risk factors is therefore warranted in order to explore means to improve the survival rate. Here, we report on a novel HCC-related gene known as lysosomal protein transmembrane 4 beta (LAPTM4B) that has two alleles designated LAPTM4B*1 and LAPTM4B*2. Allele *1 differs from allele *2 in that it contains one copy of a 19-bp sequence, whereas this sequence is duplicated in allele *2 in exon 1 of LAPTM4B. In this study, we aimed to investigate the relationship between LAPTM4B allelic variation and HCC susceptibility. The LAPTM4B genotype was analyzed in the blood samples from 102 HCC patients and 135 healthy individuals by PCR. The genotypic distribution of LAPTM4B was analyzed using the chi-squared test. The frequencies of allele *2 were 38.24 and 24.07% in the HCC group and control group, respectively, representing a significant difference between these two groups (P<0.001). Thus, allele *2 of LAPTM4B appears to be associated with genetic susceptibility of HCC and may therefore be considered as a risk factor.


Subject(s)
Asian People/genetics , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Membrane Proteins/genetics , Oncogene Proteins/genetics , Polymorphism, Genetic , Adult , Aged , Aged, 80 and over , Alleles , Amino Acid Sequence , Base Sequence , Carcinoma, Hepatocellular/etiology , Female , Genetic Predisposition to Disease , Genotype , Humans , Liver Neoplasms/etiology , Male , Middle Aged , Molecular Sequence Data
18.
Surg Today ; 41(6): 810-7, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21626328

ABSTRACT

PURPOSE: The newly-identified lysosomal protein transmembrane 4 beta-35 (LAPTM4B-35) plays important roles in tumor progression and metastasis, while argininosuccinate synthetase (ASS) provides arginine as an indispensable nutrient for hepatocellular carcinoma (HCC). The present study investigated the clinical significance of the coexpression of LAPTM4B-35 and ASS in HCC patients on determining the prognosis. METHODS: Immunohistochemistry was used to evaluate the expression of LAPTM4B-35 and ASS in HCC tissues and paired noncancerous liver samples from 71 patients. The correlation of combined LAPTM4B-35 and ASS expression with selected clinicopathologic parameters was assessed with the chi-squared test. Patient survival and differences in survival were determined by the Kaplan-Meier method and the log-rank test. A Cox regression analysis was adopted for a multivariate analysis of the prognostic factors. RESULTS: Combined LAPTM4B-35 and ASS expression was significantly associated with TNM stage and portal vein invasion. In addition, patients with HCCs expressing both LAPTM4B-35 and ASS exhibited both markedly poorer overall survival (OS) and disease-free survival (DFS) (both P < 0.001). According to the multivariate analyses, combined LAPTM4B-35 and ASS expression was found to be an independent prognostic factor for OS and DFS (P = 0.039 and P = 0.035, respectively). CONCLUSION: The overexpression of LAPTM4B-35 in combination with positive ASS expression is a negative prognostic marker for HCC.


Subject(s)
Argininosuccinate Synthase/biosynthesis , Biomarkers, Tumor/biosynthesis , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Membrane Proteins/biosynthesis , Oncogene Proteins/biosynthesis , Adult , Aged , Carcinoma, Hepatocellular/pathology , Female , Humans , Immunohistochemistry , Liver Neoplasms/pathology , Male , Middle Aged , Prognosis , Proportional Hazards Models , Survival Analysis
19.
Cardiovasc Res ; 89(1): 89-97, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20733009

ABSTRACT

AIMS: foetal nicotine exposure results in decreased protein kinase C epsilon (PKCε) expression and increased cardiac vulnerability to ischaemia and reperfusion injury in adult rat offspring. The present study tested the hypothesis that maternal nicotine administration causes increased promoter methylation of the PKCε gene resulting in PKCε repression in the heart. METHODS AND RESULTS: nicotine treatment of pregnant rats starting at day 4 of gestation increased the methylation of the Egr-1 binding site at the PKCε gene promoter and decreased PKCε protein and mRNA abundance in near-term foetal hearts. Methylation of the Egr-1 binding site reduced Egr-1 binding to the PKCε promoter in the heart. Site-specific deletion of the Egr-1 binding site significantly decreased PKCε promoter activity. The effects of nicotine were sustained in the heart of adult offspring. Ex vivo studies found no direct effect of nicotine on PKCε gene expression. However, maternal nicotine administration increased norepinephrine content in the foetal heart. Treatment of isolated foetal hearts with norepinephrine resulted in the same effects of increased methylation of the Egr-1 binding site and PKCε gene repression in the heart. 5-Aza-2'-deoxycytidine inhibited the norepinephrine-induced increase in methylation of the Egr-1 binding site and restored Egr-1 binding and PKCε gene expression to the control levels. CONCLUSION: this study demonstrates that prolonged nicotine exposure increases the sympathetic neurotransmitter release in the foetal heart and causes programming of PKCε gene repression through promoter methylation, linking maternal smoking to pathophysiological consequences in the offspring heart.


Subject(s)
Heart/drug effects , Myocardium/enzymology , Nicotine/toxicity , Prenatal Exposure Delayed Effects/enzymology , Prenatal Exposure Delayed Effects/genetics , Protein Kinase C-epsilon/genetics , Animals , Base Sequence , Binding Sites/drug effects , Binding Sites/genetics , Cell Line , DNA Methylation/drug effects , DNA Primers/genetics , Early Growth Response Protein 1/metabolism , Female , Gene Expression/drug effects , In Vitro Techniques , Nicotine/administration & dosage , Norepinephrine/metabolism , Norepinephrine/pharmacology , Pregnancy , Promoter Regions, Genetic/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
20.
Hepatogastroenterology ; 57(98): 207-11, 2010.
Article in English | MEDLINE | ID: mdl-20583413

ABSTRACT

BACKGROUND/AIMS: It was previously established that LAPTM4B-35 highly expressed in gallbladder carcinoma and being of clinicopathological and prognostic significances. However, expression of LAPTM4B gene in gallbladder carcinoma (GBC-SD), a gallbladder carcinoma cell line, and its role in invasive potential remain unclear. METHODOLOGY: Expression of LAPTM4B in GBC-SD cells was first detected. Plasmids, pcDNA3-AE (containing complete open reading frame of LAPTM4B) and Mock (pcDNA3), were transiently transfected into GBC-SD cells. Invasive phenotypes (migration and invasion) and relative molecules were then shown by transwell assay, crossing river test and Western blot analysis. RESULTS: Immunocytochemical staining revealed that LAPTM4B-35 positively expressed in cytoplasm of GBC-SD cells. But LAPTM4B-35 expression was obviously weaker in GBC-SD cells than that in BEL-7402 cells (positive control). Besides, cells transfected with pcDNA3-AE presented shorter crossing river time, less migrated and invaded cell numbers, compared with cells transfected with the Mock plasmid and parent cells. Finally, increased expressions of active uPA, MMP-9, pro MMP-2 and active MMP-2 were also observed in cells transfected with pcDNA3-AE. CONCLUSIONS: Our data suggested that LAPTM4B expressed in GBC-SD cells at a relatively low level. Forced overexpression of LAPTM4B increased invasive potential of GBC-SD cells, through modulating molecules associated with degradation of extracellular matrix.


Subject(s)
Gallbladder Neoplasms/genetics , Membrane Proteins/genetics , Oncogene Proteins/genetics , Blotting, Western , Cell Line, Tumor , Cell Movement , Dinucleoside Phosphates/metabolism , Extracellular Matrix/metabolism , Gallbladder Neoplasms/metabolism , Humans , Immunoenzyme Techniques , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Membrane Proteins/metabolism , Oncogene Proteins/metabolism , Open Reading Frames , Phenotype , Plasmids , Statistics, Nonparametric , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...