Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Lipids Health Dis ; 16(1): 66, 2017 Mar 31.
Article in English | MEDLINE | ID: mdl-28359281

ABSTRACT

BACKGROUND: We have previously demonstrated that estrogen could significantly enhance expression of apolipoprotein M (apoM), whereas the molecular basis of its mechanism is not fully elucidated yet. To further investigate the mechanism behind the estrogen induced up-regulation of apoM expression. RESULTS: Our results demonstrated either free 17ß-estradiol (E2) or membrane-impermeable bovine serum albumin-conjugated E2 (E2-BSA) could modulate human apoM gene expression via the estrogen receptor alpha (ER-α) pathway in the HepG2 cells. Moreover, experiments with the luciferase activity analysis of truncated apoM promoters could demonstrate that a regulatory region (from-1580 to -1575 bp (-GGTCA-)) upstream of the transcriptional start site of apoM gene was essential for the basal transcriptional activity that regulated by the ER-α. With the applications of an electrophoresis mobility shift assay and a chromatin immunoprecipitation assay, we could successfully identify a specific ER-α binding element in the apoM promoter region. CONCULSION: In summary, the present study indicates that 17ß-estradiol induced up-regulation of apoM in HepG2 cells is through an ER-α-dependent pathway involving ER-α binding element in the promoter of the apoM gene.


Subject(s)
Apolipoproteins/genetics , Estradiol/physiology , Estrogen Receptor alpha/physiology , Lipocalins/genetics , Transcriptional Activation , Apolipoproteins/metabolism , Apolipoproteins M , Base Sequence , Binding Sites , Hep G2 Cells , Humans , Lipocalins/metabolism , MCF-7 Cells , Promoter Regions, Genetic , Protein Binding , Sequence Analysis, DNA , Up-Regulation
2.
Lipids Health Dis ; 14: 110, 2015 Sep 16.
Article in English | MEDLINE | ID: mdl-26377577

ABSTRACT

BACKGROUND: We previously demonstrated that hyperglycemia could suppress apolipoprotein M (apoM) synthesis both in vivo and in vitro; however, the mechanism of hyperglycemia-induced downregulation of apoM expression is unknown yet. METHODS: In the present study we further examined if hexosamine pathway, one of the most important pathways of glucose turnover, being involved in modulating apoM expression in the hyperglycemia condition. We examined the effect of glucosamine, a prominent component of hexosamine pathway and intracellular mediator of insulin resistance, on apoM expression in HepG2 cells and in rat's models. In the present study we also determined apolipoprotein A1 (apoA1) as a control gene. RESULTS: Our results demonstrated that glucosamine could even up-regulate both apoM and apoA1 expressions in HepG2 cell cultures. The glucosamine induced upregulation of apoM expression could be blocked by addition of azaserine, an inhibitor of hexosamine pathway. Moreover, intravenous infusion of glucosamine could enhance hepatic apoM expression in rats, although serum apoM levels were not significantly influences. CONCLUSIONS: It is concluded that both exogenous and endogenous glucosamine were essential for the over-expression of apoM, which may suggest that the increased intracellular content of glucosamine does not be responsible for the depressed apoM expression at hyperglycemia condition.


Subject(s)
Apolipoprotein A-I/genetics , Apolipoproteins/genetics , Hyperglycemia/genetics , Lipocalins/genetics , Liver/metabolism , Animals , Antimetabolites, Antineoplastic/pharmacology , Apolipoprotein A-I/metabolism , Apolipoproteins/metabolism , Apolipoproteins M , Azaserine/pharmacology , Gene Expression Regulation , Glucosamine/administration & dosage , Glucosamine/metabolism , Hep G2 Cells , Humans , Hyperglycemia/metabolism , Hyperglycemia/pathology , Infusions, Intravenous , Lipocalins/metabolism , Liver/pathology , Male , Rats , Rats, Sprague-Dawley , Signal Transduction
3.
Int J Med Sci ; 11(10): 1015-21, 2014.
Article in English | MEDLINE | ID: mdl-25136257

ABSTRACT

Apolipoprotein M (APOM) has been suggested as a vasculoprotective constituent of high density lipoprotein (HDL), which plays a crucial role behind the mechanism of HDL-mediated anti-atherosclerosis. Previous studies demonstrated that insulin resistance could associate with decreased APOM expressions. In agreement with our previous reports, here, we further confirmed that the insulin sensitivity was also reduced in rats treated with high concentrations of glucose; such effect could be reversed by administration of rosiglitazone, a peroxisome proliferator-activated receptor-γ (PPARγ). The present study shows that Apom expression is significantly affected by either rosiglitazone or hyperglycemia alone without cross interaction with each other, which indicates that the pathway of Apom expression regulating by hyperglycemia might be differed from that by rosiglitazone. Further study indicated that hyperglycemia could significantly inhibit mRNA levels of Lxrb (P=0.0002), small heterodimer partner 1 (Shp1) (P<0.0001), liver receptor homologue-1 (Lrh1) (P=0.0012), ATP-binding cassette transporter 1 (Abca1) (P=0.0012) and Pparb/d (P=0.0043). Two-way ANOVA analysis demonstrated that the interactions between rosiglitazone and infusion of 25% glucose solution on Shp1 (P=0.0054) and Abca1 (4E, P=0.0004) mRNA expression was statistically significant. It is concluded that rosiglitazone could increase Apom expression, of which the detailed mechanism needs to be further investigated. The downregulation of Apom by hyperglycemia might be mainly through decreasing expression of Pparg and followed by inhibiting Lxrb in rats.


Subject(s)
Apolipoproteins/metabolism , Gene Expression Regulation/drug effects , Lipocalins/metabolism , Liver/drug effects , Liver/metabolism , Thiazolidinediones/pharmacology , Animals , Apolipoproteins M , Hyperglycemia/metabolism , Hypoglycemic Agents/pharmacology , Male , Rats , Rats, Sprague-Dawley , Rosiglitazone
4.
PLoS One ; 9(8): e105681, 2014.
Article in English | MEDLINE | ID: mdl-25144649

ABSTRACT

BACKGROUND: Apolipoprotein M (ApoM) is a constituent of high-density lipoproteins (HDL). It plays a crucial role in HDL-mediated reverse cholesterol transport. Insulin resistance is associated with decreased ApoM levels. AIMS: To assess the effects of increased free fatty acids (FFAs) levels after short-term Intralipid infusion on insulin sensitivity and hepatic ApoM gene expression. METHODS: Adult male Sprague-Dawley (SD) rats infused with 20% Intralipid solution for 6 h. Glucose infusion rates (GIR) were determined by hyperinsulinemic-euglycemic clamp during Intralipid infusion and plasma FFA levels were measured by colorimetry. Rats were sacrificed after Intralipid treatment and livers were sampled. Human embryonic kidney 293T cells were transfected with a lentivirus mediated human apoM overexpression system. Goto-Kakizaki (GK) rats were injected with the lentiviral vector and insulin tolerance was assessed. Gene expression was assessed by real-time RT-PCR and PCR array. RESULTS: Intralipid increased FFAs by 17.6 folds and GIR was decreased by 27.1% compared to the control group. ApoM gene expression was decreased by 40.4% after Intralipid infusion. PPARß/δ expression was not changed by Intralipid. Whereas the mRNA levels of Acaca, Acox1, Akt1, V-raf murine sarcoma 3611 viral oncogene homolog, G6pc, Irs2, Ldlr, Map2k1, pyruvate kinase and RBC were significantly increased in rat liver after Intralipid infusion. The Mitogen-activated protein kinase 8 (MAPK8) was significantly down-regulated in 293T cells overexpressing ApoM. Overexpression of human ApoM in GK rats could enhance the glucose-lowering effect of exogenous insulin. CONCLUSION: These results suggest that Intralipid could decrease hepatic ApoM levels. ApoM overexpression may have a potential role in improving insulin resistance in vivo and modulating apoM expression might be a future therapeutic strategy against insulin resistance in type 2 diabetes.


Subject(s)
Apolipoproteins/genetics , Apolipoproteins/metabolism , Insulin Resistance , Lipocalins/genetics , Lipocalins/metabolism , Animals , Apolipoproteins M , Fatty Acids, Nonesterified/blood , Gene Expression , HEK293 Cells , Humans , Insulin/blood , Insulin/metabolism , Insulin Resistance/genetics , Liver/metabolism , Male , PPAR delta/genetics , PPAR delta/metabolism , PPAR-beta/genetics , PPAR-beta/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Signal Transduction
5.
Int J Med Sci ; 11(4): 365-72, 2014.
Article in English | MEDLINE | ID: mdl-24578614

ABSTRACT

The present study investigated the correlation among genetic polymorphisms of the proximal promoter region of apolipoprotein M (apoM) gene, the polymorphisms in relation to apoM expressions and the susceptibility to coronary artery diseases (CAD) in a Han Chinese population. Four common polymorphic sites, i.e., T-1628G, C-1065A, T-855C and T-778C, were confirmed, and a new deletion mutation C-724del was found, in 206 CAD patients and 209 non-CAD patients using direct DNA sequencing analyses. Occurrences of alleles T-1628G, T-855C and C-724del were significantly higher in CAD patients compared to non-CAD patients. Moreover we examined all these polymorphisms in relation to apoM expression by applying luciferase reporter assay. It demonstrated that constructs -855C and 724del showed obvious decreased luciferase activities, i.e., (0.93±0.15 vs. 2.11±0.15; P=0.012) and (1.13±0.25 vs. 2.11±0.15; P=0.009) respectively, which indicates these two polymorphisms could confer decreased apoM expressions. Meanwhile the occurrences of these two SNP were also significantly higher in the CAD patients than in non-CAD patients. It is therefore reasonable to speculate that down-regulated apoM expressions in relation to these polymorphisms may affect HDL and cholesterol metabolism in vivo and further influence the susceptibility to CAD, although the underlying mechanisms need further investigation.


Subject(s)
Apolipoproteins/genetics , Coronary Artery Disease/genetics , Lipocalins/genetics , Promoter Regions, Genetic/genetics , Aged , Apolipoproteins M , Cell Line , Female , Genetic Predisposition to Disease/genetics , Genotype , Haplotypes/genetics , Humans , Male , Middle Aged , Mutagenesis, Site-Directed , Polymorphism, Single Nucleotide/genetics , Real-Time Polymerase Chain Reaction
6.
Biochem Biophys Res Commun ; 445(1): 203-7, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24508264

ABSTRACT

It has been demonstrated that apolipoprotein M (APOM) is a vasculoprotective constituent of high density lipoprotein (HDL), which could be related to the anti-atherosclerotic property of HDL. Investigation of regulation of APOM expression is of important for further exploring its pathophysiological function in vivo. Our previous studies indicated that expression of APOM could be regulated by platelet activating factor (PAF), transforming growth factors (TGF), insulin-like growth factor (IGF), leptin, hyperglycemia and etc., in vivo and/or in vitro. In the present study, we demonstrated that palmitic acid could significantly inhibit APOM gene expression in HepG2 cells. Further study indicated neither PI-3 kinase (PI3K) inhibitor LY294002 nor protein kinase C (PKC) inhibitor GFX could abolish palmitic acid induced down-regulation of APOM expression. In contrast, the peroxisome proliferator-activated receptor beta/delta (PPARß/δ) antagonist GSK3787 could totally reverse the palmitic acid-induced down-regulation of APOM expression, which clearly demonstrates that down-regulation of APOM expression induced by palmitic acid is mediated via the PPARß/δ pathway.


Subject(s)
Apolipoproteins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Lipocalins/genetics , PPAR delta/genetics , PPAR-beta/genetics , Palmitic Acid/pharmacology , Apolipoproteins M , Benzamides/pharmacology , Chromones/pharmacology , Dose-Response Relationship, Drug , Hep G2 Cells , Humans , Indoles/pharmacology , Maleimides/pharmacology , Morpholines/pharmacology , PPAR delta/antagonists & inhibitors , PPAR delta/metabolism , PPAR-beta/antagonists & inhibitors , PPAR-beta/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase C/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Sulfones/pharmacology
7.
Biochem Biophys Res Commun ; 421(1): 152-6, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22516753

ABSTRACT

We have previously reported that liver X receptor (LXR) agonist, TO901317, could significantly inhibit hepatic apolipoprotein M (apoM) expression. It has been reported that TO901317 could activate the ATP-binding cassette transporter A1 (ABCA1) that mediates cholesterol efflux to the lipid-poor apoAI, which is an essential step for the high-density lipoprotein (HDL) formation. It is unknown if ABCA1 may regulate hepatic apoM expression. In the present study, HepG2 cells were cultured with the synthetic LXR agonists, TO901317 or GW3965 in the presence or absence of ABCA1 antagonist, disodium 4,4'-diisothiocyanatostilbene- 2,2'-disulfonate (DIDS). The mRNA levels of ABCA1, apoM and liver receptor homolog-1 (LRH-1) determined by the real-time RT-PCR. It demonstrated that both TO901317 and GW3965 could significantly enhance ABCA1 expression, and simultaneously, inhibit LRH1 expression. However, TO901317 alone could significantly inhibit apoM expression, while GW3965 alone did not influence apoM expression. ABCA1 antagonist, DIDS, have no effects on GW3965 induced upregulation of ABCA1 and downregulation of LRH1. However, apoM mRNA level was significantly decreased when the cells cultured with GW3965 together with DIDS. The present study demonstrated that apoM expression could be elevated by ABCA1 via the RXR/LXR pathway and LRH1 does not involve in the regulation of apoM by the activation of ABCA1, although the direct regulative pathway(s) between ABCA1 and apoM gene is still unknown yet. The detailed mechanism needs further investigation.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Apolipoproteins/biosynthesis , Lipocalins/biosynthesis , Orphan Nuclear Receptors/metabolism , Retinoid X Receptor alpha/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/antagonists & inhibitors , Apolipoproteins/genetics , Apolipoproteins M , Benzoates/pharmacology , Benzylamines/pharmacology , Hep G2 Cells , Humans , Hydrocarbons, Fluorinated/pharmacology , Ligands , Lipocalins/genetics , Liver X Receptors , Orphan Nuclear Receptors/agonists , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Sulfonamides/pharmacology , Up-Regulation
8.
Lipids Health Dis ; 10: 199, 2011 Nov 04.
Article in English | MEDLINE | ID: mdl-22054074

ABSTRACT

BACKGROUND: Apolipoprotein M (apoM) may have potential antiatherosclerotic properties. It has been reported that apoM expression could be regulated by many intracellar and extracellar factors. In the present study we further investigated regulation of apoM expression in Caco-2 cells stimulated by a liver X receptor (LXR) agonist, TO901317. MATERIALS AND METHODS: Caco-2 cells were cultured in the presence of either TO901317, farnesoid X receptor (FXR) antagonist guggulsterone or TO901317 together with guggulsterone at different concentrations for 24 hrs. The mRNA levels of ATP-binding cassette transporter A1 (ABCA1), apoA1, apoM, liver receptor homologue-1 (LRH-1) and short heterodimer partner 1 (SHP1) were determined by real-time RT-PCR. RESULTS: When Caco-2 cell cultured with TO901317 alone, the mRNA levels of ABCA1, apoA1, apoM, LRH-1 and SHP1 were significantly increased with dose-dependent manners (p < 0.05), whereas when the cells cultured with guggulsterone alone, the mRNA levels of apoM, SHP1 and LRH-1 (p < 0.05) were strongly inhibited. Moreover, guggulsterone could abolish the TO901317 enhanced mRNA levels of apoA1 apoM, SHP1 and LRH-1. CONCLUSION: The present study demonstrated that LXR agonist TO901317 induced apoM expression in Caco-2 cells might be mediated via the LXR/FXR pathway.


Subject(s)
Apolipoproteins/metabolism , Gene Expression Regulation/drug effects , Hydrocarbons, Fluorinated/pharmacology , Lipocalins/metabolism , Orphan Nuclear Receptors/agonists , Pregnenediones/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Sulfonamides/pharmacology , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Apolipoproteins/genetics , Apolipoproteins M , Caco-2 Cells , Gene Expression , Humans , Lipocalins/genetics , Liver X Receptors , Orphan Nuclear Receptors/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism
9.
Biochim Biophys Acta ; 1811(12): 1146-51, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21816233

ABSTRACT

Apolipoprotein M (apoM) is present predominantly in high-density lipoprotein (HDL) in human plasma, thus possibly involved in the regulation of HDL metabolism and the process of atherosclerosis. Although estrogen replacement therapy increases serum levels of apoAI and HDL, it does not seem to reduce the cardiovascular risk in postmenopausal women. Therefore, we investigated the effects of estrogen on apoM expression in vitro and in vivo. HepG2 cells were incubated with different concentrations of estrogen with or without the estrogen receptor antagonist, fulvestrant, and apoM expression in the cells was determined. Hepatic apoM expression and serum levels of apoM were also determined in normal and in ovariectomized rats treated with either placebo or estradiol benzoate, using sham operated rats as controls. Estrogen significantly increased mRNA levels of apoM and apoAI in HepG2 cell cultures in a dose- and time-dependent manner; the upregulation of both apolipoproteins was fully abolished by addition of estrogen receptor antagonist. In normal rats, estrogen treatment led to an increase in plasma lipid levels including HDL cholesterol, a marked upregulation of apoM mRNA and a significant increase in serum levels of apoM. The same pattern of regulation was found in ovariectomized rats treated with estrogen. Thus, estrogen upregulates apoM expression both in vivo and in vitro by mechanism(s) involving the estrogen receptor.


Subject(s)
Apolipoproteins/blood , Atherosclerosis/blood , Estradiol/analogs & derivatives , Lipocalins/blood , Liver/metabolism , Postmenopause/blood , Receptors, Estrogen/metabolism , Animals , Apolipoprotein A-I/antagonists & inhibitors , Apolipoprotein A-I/blood , Apolipoproteins/antagonists & inhibitors , Apolipoproteins M , Atherosclerosis/genetics , Cholesterol, HDL/blood , Dose-Response Relationship, Drug , Estradiol/administration & dosage , Estradiol/adverse effects , Estradiol/pharmacology , Estradiol/therapeutic use , Estrogen Antagonists/pharmacology , Estrogen Antagonists/therapeutic use , Estrogens/administration & dosage , Estrogens/adverse effects , Female , Fulvestrant , Gene Expression/drug effects , Hep G2 Cells , Humans , Lipocalins/antagonists & inhibitors , Liver/drug effects , Ovariectomy , Postmenopause/drug effects , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Receptors, Estrogen/genetics , Up-Regulation
10.
Lipids Health Dis ; 10: 112, 2011 Jul 05.
Article in English | MEDLINE | ID: mdl-21729290

ABSTRACT

OBJECTIVE: To investigate effects of lipid lowering drug, simvastatin, on apolipoprotein M expression in the hyperlipidemic mice and in hepatic cell line, HepG2 cells. METHODS: Swiss male mice were randomly divided into the high fat group and control group, and were intragastrically fed with 0.9% saline (control group) or lipid emulsion (high fat group) at the daily dosage of 15 ml/kg body weight, respectively. After 8 weeks feeding, the hyperlipidemic model was successfully induced and these hyperlipidemic mice were then randomly divided into three experimental groups: vehicle control group, high-dose simvastatin-treated group (100 mg/kg body weight), and low-dose simvastatin-treated group (10 mg/kg body weight). Mice were dosed daily for 6 weeks of simvastatin before mice were sacrificed for determining serum lipid profile and apoM protein levels that was determined by using dot blotting analysis. Effects of simvastatin on apoM mRNA expression in the HepG2 cells were determined by real-time RT-PCR. RESULTS: Comparing to high fat model mice without simvastatin treatment, 100 mg/kg simvastatin could significantly increase serum total cholesterol (P < 0.05). Serum apoM levels, in all mice, were significantly lower in the mice at the age of 26 weeks than the mice at 12 weeks old (P < 0.05), which indicated that serum apoM levels were significantly correlated to the mice age. It demonstrated also that treatment of simvastatin did not influence serum apoM levels in these mouse model, although serum apoM levels were increased by about 13% in the 10 mg/kg simvastatin group than in the vehicle control group without simvastatin. In HepG2 cell cultures, simvastatin could significantly decrease apoM mRNA levels with dose- and time-dependent manners. At 10 µM simvastatin treatment, apoM mRNA decreased by 52% compared to the controls. CONCLUSION: The present study suggested that simvastatin, in vivo, had no effect on apoM levels in the hyperlipidemic mouse model. ApoM serum levels in mice were significantly correlated to the animal's age, whereas in cell cultures simvastatin does inhibit apoM expression in the HepG2 cells. The mechanism behind it is not known yet.


Subject(s)
Apolipoproteins/blood , Gene Expression/drug effects , Hypolipidemic Agents/pharmacology , Simvastatin/pharmacology , Animals , Apolipoproteins/genetics , Apolipoproteins M , Dietary Fats , Hep G2 Cells , Humans , Hyperlipidemias/blood , Hyperlipidemias/chemically induced , Hyperlipidemias/drug therapy , Hypolipidemic Agents/therapeutic use , Lipids/blood , Male , Mice , Simvastatin/therapeutic use
11.
Lipids Health Dis ; 9: 102, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20846402

ABSTRACT

BACKGROUND: It has been well documented that apolipoprotein M (apoM) is principally expressed in the liver and kidney. However we found that there was weak apoM expression in other tissues or organs too, which could not be ignored. In the present study, we therefore examined apoM expression in human colorectal tissues including cancer tissues, cancer adjacent normal tissues, polyp tissues and normal mucosa as well as inflammatory mucosa. METHODS: Tissue samples were collected from patients who underwent surgical resection or endoscopic examination. ApoM mRNA levels were determined by the real-time RT-PCR and apoM protein mass were examined by the immunohistochemistry. RESULTS: ApoM protein can be detected in all colorectal tissues. However, apoM protein mass were significantly lower in the cancer tissues than its matched adjacent normal tissues, polyp tissues, normal mucosa and inflammatory mucosa. In parallel, apoM mRNA levels in the colorectal cancer tissues (0.0536 ± 0.0131) were also significantly lower than those in their adjacent normal tissues (0.1907 ± 0.0563) (P = 0.033). Interestingly, apoM mRNA levels in colorectal cancer tissues were statistic significant higher in the patients with lymph node metastasis than the patients without lymph node metastasis (P = 0.008). Patients under Dukes' C and D stages had much higher apoM mRNA levels than patients under Dukes' A and B stages (P = 0.034). CONCLUSION: It is concluded that apoM could also be expressed in human colorectal tissues besides liver and kidney. ApoM mRNA levels in the colorectal cancer tissues were significantly increased in the patients with lymph node metastasis. Whether increased apoM expression in the patients with lymph node metastasis being related to patients' prognosis and the physiopathological importance of apoM expression in colorectal tissues need further investigation.


Subject(s)
Apolipoproteins/metabolism , Colon/metabolism , Gene Expression Regulation , Rectum/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/secondary , Adenocarcinoma/surgery , Adult , Aged , Aged, 80 and over , Apolipoproteins/genetics , Apolipoproteins M , Colon/cytology , Colon/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Female , Humans , Immunohistochemistry , Inflammation/metabolism , Inflammation/pathology , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Polyps/metabolism , Intestinal Polyps/pathology , Lipocalins , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Staging , RNA, Messenger/metabolism , Rectum/cytology , Rectum/pathology
12.
Lipids Health Dis ; 9: 48, 2010 May 17.
Article in English | MEDLINE | ID: mdl-20478061

ABSTRACT

Apolipoprotein B (apoB) containing lipoproteins, i.e. VLDL, LDL and Lp(a), are consequently lowered by ACTH treatment in humans. This is also seen as reduced plasma apoB by 20-30% and total cholesterol by 30-40%, mostly accounted for by a decrease in LDL-cholesterol. Studies in hepatic cell line (HepG2) cells showed that apoB mRNA expression is reduced in response to ACTH incubation and is followed by a reduced apoB secretion, which may hypothesize that ACTH lowering apoB containing lipoproteins in humans may be mediated by the inhibition of hepatic apoB synthesis. This was recently confirmed in vivo in a human postprandial study, where ACTH reduced transient apoB48 elevation from the small intestine, however, the exogenic lipid turnover seemed unimpaired. In the present study we investigated if lipid synthesis and/or secretion in HepG2 cells were also affected by pharmacological levels of ACTH to accompany the reduced apoB output. HepG2 cells were incubated with radiolabelled precursors ([14C]acetate and [3H]glycerol) either before or during ACTH stimuli. Cellular and secreted lipids were extracted with chloroform:methanol and separated by the thin layer chromatography (TLC), and [14C]labelled cholesterol and cholesteryl ester and [3H]labelled triglycerides and phospholipids were quantitated by the liquid scintillation counting. It demonstrated that ACTH administration did not result in any significant change in neither synthesis nor secretion of the studied lipids, this regardless of presence or absence of oleic acid, which is known to stabilize apoB and enhance apoB production. The present study suggests that ACTH lowers plasma lipids in humans mainly mediated by the inhibition of apoB synthesis and did not via the reduced lipid synthesis.


Subject(s)
Adrenocorticotropic Hormone/pharmacology , Lipids/biosynthesis , Apolipoproteins B/biosynthesis , Cholesterol/analysis , Cholesterol Esters/analysis , Hep G2 Cells , Humans , Lipid Metabolism , Radioactive Tracers , Triglycerides/analysis
13.
Anticancer Res ; 30(3): 981-5, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20393024

ABSTRACT

The present study determined mutations of phosphatase and tensin homolog (PTEN) gene in patients suffering from high-grade gliomas (WHO grades III and IV) and further investigated the mutations in correlation to patients' histopathological classification and short-term survival. Total RNA and genomic DNA were extracted from tumor tissues. Full-length PTEN cDNA sequences were amplified by polymerase chain reaction (PCR). The PCR products were directly sequenced, and the PTEN mutations were analyzed. It demonstrated that the incidence of PTEN mutations was 8/22 in these patients: one patient with WHO grade III glioma (1/11) and 7 patients with WHO grade IV glioma (7/11). Most patients had three or more mutations in the PTEN gene, with exons 2, 3, 4, 5, 6 and 7 as hot mutation regions, with mutation incidence from 62.5% to 75%. About 68.4% of mutations were missense, 26.3% same-sense and 5.3% nonsense mutations. The median survival times of the WHO grade III and IV groups were 250 and 53 weeks after surgery, respectively (p=0.016). The 36-week survival rate of patients with and without PTEN mutations was 62.5% and 92.9% (p=0.038, odds ratio=7.80), respectively. The present study suggests that PTEN mutations are late events in the malignant progression of glioma and the occurrence of PTEN mutations are significantly correlated to patients' short-term survival.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , Mutation , PTEN Phosphohydrolase/genetics , Adult , Aged , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Cell Differentiation/genetics , Codon, Nonsense , Female , Genotype , Glioma/enzymology , Glioma/pathology , Humans , Male , Middle Aged , Mutation, Missense , Neoplasm Staging , Survival Rate
14.
Int J Gen Med ; 2: 177-82, 2009 Dec 29.
Article in English | MEDLINE | ID: mdl-20360902

ABSTRACT

OBJECTIVE: It has been reported that single-nucleotide polymorphism (SNP) in the proximal promoter region of apolipoprotein M (apoM) gene may confer the risk in the development of type 2 diabetes (T2D) and coronary artery disease (CAD) in the Han Chinese. However, in a recent study demonstrated that plasma apoM level did not correlated to the coronary heart disease. In the present studies, we investigated the SNP T-778C of apoM gene in CAD patients and controls in the Han Chinese population. Moreover we examined whether serum apoM levels could be influenced by this promoter mutation. MATERIAL AND METHODS: One hundred twenty-six CAD patients and 118 non-CAD patients were subjected in the present study. All patients were confirmed by the angiography. The genotyping of polymorphisms T-778C in apoM promoter was determined by real-time polymerase chain reaction. Serum apoM levels were semi-quantitatively determined by the dot-blotting analysis. RESULTS: Distribution of apoM T-778C genotype in non-CAD patients was as following: 84.7% were T/T, 15.3% were T/C and 0.0% was C/C. T allele frequencies were 92.4% and C allele, 7.6%. In the CAD patients, 99 patients (78.6%) had the T/T genotype, 25 patients (19.8%) with T/C genotype and 2 patients (1.6%) with C/C genotype. The allele frequency was 88.5% for the T allele and 11.5% for the C allele. There was no statistical significant difference of serum apoM levels found in these three genotypes. CONCLUSIONS: There was no significant difference in allele or genotype frequencies between CAD patients and non-CAD patients. Binary logistic regression analysis with adjustments for age, gender, triglycerides, total cholesterol, low-density lipoprotein, high-density lipoprotein, apoAI, apoB, and LP(a) indicated that the TC and CC genotypes in SNP T-778C were not significantly associated with the development of CAD (odds ratio = 1.510, 95% confidence interval: 0.756-3.017; p = 0.243).

15.
Anal Biochem ; 386(2): 161-6, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19109921

ABSTRACT

Most available methods for detecting single nucleotide polymorphisms (SNPs) are based principally on the system that can produce an increased fluorescence signal during hybridization. In the current study, we demonstrate a method of base-quenched probe for polymerase chain reaction (PCR) genotyping that requires only a pair of primers and one fluorescent probe and does not invariably depend on the deoxyguanosine nucleotide. This method further exploits the phenomenon of fluorescence quenching of fluorescent-labeled probe during hybridization to its complementary target gene's sequence. 6-Carboxyfluorescein (FAM) can be directly conjugated to a base of either adenine (A), thymine (T), cytosine (C), or guanine (G), referred to as A-, T-, C-, or G-quenched probe, respectively, at either the 5' or 3' end. For describing the method in detail, we chose apolipoprotein M (apoM) as a target gene in the current study. DNA sequencing analyses validated that all four types of base-quenched probes could provide unbiased genotyping results (K = 1, P = 0.000), although the maximum speed of fluorescence increase, max(dF/dT), when using the G-quenched probe method, was approximately twofold lower than the others (P < 0.0001). Moreover, we applied this method to detect another seven SNPs in the genomes of phospholipase A2, monocyte chemoattractant protein 1 (MCP1), and l-ficolin, further confirming our method. It is concluded that this method is precise, simple, and economic as well as suitable for large-scale genotyping studies.


Subject(s)
DNA Primers/chemistry , Deoxyguanosine/chemistry , Genotype , Polymerase Chain Reaction/methods , Polymorphism, Single Nucleotide , Fluorescence , Sequence Analysis, DNA/methods
16.
Biochem Biophys Res Commun ; 371(1): 114-7, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18413148

ABSTRACT

It has been demonstrated that apolipoprotein M (apoM), a recently discovered HDL apolipoprotein, has antiatherosclerotic properties, which may be mediated by the enhancement of reversed cholesterol transportation and/or hepatic cholesterol catabolism. The detailed mechanisms are unknown yet. Liver X receptor (LXR) belongs to the nuclear receptor superfamily and is a ligand-activated transcription factor involved in the regulation of lipid metabolism and inflammation. Activation of LXR in the cell cultures results in an enhancement of cholesterol efflux to apoAI. In the present study, we investigated effects of the LXR agonist, T0901317 on hepatic apoM expression in vivo and in vitro. Serum apoM levels in mice given T0901317 at 10 mg or 100 mg/kg for 7 days were reduced by 12-17% (P<0.05). In HepG2 cell cultures, apoM mRNA levels were significantly lower in presence of 25 microM T0901317 (37.1%) than in control cells (P<0.001). A similar reduction was found by the addition of 9-cis retinoic acid (RA). Twenty-five micromolar T0901317 together with 100 nM RA decreased apoM mRNA expression by 65% (P<0.001). Thus, the LXR agonist T0901317 significantly downregulates apoM mRNA expression in vivo and in vitro, which indicates that apoM is another novel target gene regulated by the LXR. The combination of RA and T0901317 showed additive effects, which suggests that apoM expression can be modulated by LXR/RXR pathway.


Subject(s)
Apolipoproteins/metabolism , DNA-Binding Proteins/agonists , Hypolipidemic Agents/administration & dosage , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Sulfonamides/administration & dosage , Animals , Apolipoproteins/blood , Apolipoproteins/genetics , Apolipoproteins B/blood , Apolipoproteins B/genetics , Apolipoproteins B/metabolism , Apolipoproteins M , Cell Line , Down-Regulation , Humans , Hydrocarbons, Fluorinated , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Orphan Nuclear Receptors , RNA, Messenger/metabolism
17.
Biochim Biophys Acta ; 1771(7): 879-82, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17556016

ABSTRACT

Diabetes is associated with low concentrations of apoM in plasma. In db/db mice, ob/ob mice as well as in the alloxan-induced diabetic mouse, the low apoM levels are paralleled by decreased expression of the apoM gene. In the latter model, insulin substitution tended to reverse the low apoM expression. It is not known whether the impairment in apoM expression can be ascribed to hyperglycemia, insulin deficiency or insulin resistance. In the present study, we investigated apoM levels and expression in rats rendered hyperglycemic by short-term glucose infusion. As expected, serum insulin concentrations rose moderately during the infusions. Serum apoM concentrations and hepatic apoM mRNA levels were significantly reduced in the hyperglycemic rats, indicating that the low expression of apoM in these diabetic models can be ascribed to hyperglycemia rather than to insulin deficiency or insulin resistance. However, in HepG2 cells both glucose and insulin markedly inhibited apoM expression these effects were additive. Thus, the possible effects of insulin in vivo seem to be mediated indirectly.


Subject(s)
Apolipoproteins/genetics , Down-Regulation/genetics , Hyperglycemia/pathology , Animals , Apolipoproteins/blood , Apolipoproteins M , Cell Line, Tumor , Down-Regulation/drug effects , Female , Glucose/pharmacology , Humans , Hyperglycemia/chemically induced , Insulin/pharmacology , Lipocalins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
18.
Atherosclerosis ; 191(2): 433-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-16839559

ABSTRACT

It has been repeatedly demonstrated that ACTH administration lowers plasma lipid concentrations in man. The present study was designed to test the hypothesis, based on observations of decreased apolipoprotein B (ApoB) synthesis and secretion in vitro, that ACTH administration inhibits the postprandial output of ApoB in man. Therefore, we studied the response to a fat-rich meal supplemented with Vitamin A in eight healthy volunteers, who underwent this test without premedication, after 4 days administration of ACTH, and after 4 days administration of a glucocorticoid (betamethasone). As expected, fasting plasma levels of low-density lipoproteins (LDL)-cholesterol (-25%) and ApoB (-17%) decreased after ACTH, but not after betamethasone administration. Also, the elevation of plasma ApoB-48 in response to fat intake (to twice the basal levels) was markedly reduced after ACTH administration. However, the postprandial rise in plasma triglycerides and retinyl palmitate was unimpaired, suggesting that ACTH administration induced the secretion of fewer but larger chylomicrons. The effect of betamethasone on the postprandial response was similar but less pronounced. This study confirms earlier reports on the lipid-lowering effects of ACTH and supports our theory, based on in vitro studies, that the lipid-lowering effects of ACTH administration in man involves an inhibition of ApoB production.


Subject(s)
Adrenocorticotropic Hormone/physiology , Apolipoprotein B-48/blood , Dietary Fats/administration & dosage , Lipid Metabolism/drug effects , Adrenocorticotropic Hormone/administration & dosage , Adult , Betamethasone/administration & dosage , Blood Glucose/drug effects , Cholesterol, LDL/blood , Chylomicrons/metabolism , Drug Administration Schedule , Fasting/blood , Female , Glucocorticoids/administration & dosage , Humans , Hydrocortisone/blood , Male , Middle Aged , Postprandial Period , Reference Values , Time Factors
19.
Anticancer Res ; 26(3B): 2237-42, 2006.
Article in English | MEDLINE | ID: mdl-16821594

ABSTRACT

The effects of autologous cytokine-induced killer (CIK) (CD3+CD56+) cells together with chemotherapy were investigated in patients who suffered from advanced gastric cancers (stage IV). Fifty-seven patients were divided into two groups: chemotherapy plus CIK biotherapy and chemotherapy alone. CIK cells were induced from autologous peripheral blood mononuclear cells in vitro and separated by flow cytometry and then transfused into the patients. The T-lymphocyte subgroups (CD3+, CD4+ or CD8+), CIK cells and NK cells (CD3-CD56+) were separated and determined by flow cytometry and the serum levels of MG7-Ag, CA72-4, CA19-9 and CEA were determined by ELISA or ECLIA. It was demonstrated that the cytotoxic activity of CIK cells reached a maximum between days 14 to 21 (68.7+/-10.9% and 65.3+/-10.4%, respectively). The amounts of CIK cells were gradually increased from day 0 to day 21 and slightly decreased in the further incubations. Thereafter, the CIK cells on days 14 to 21 (with the highest population of CIK cells) transfused back to the patients. The serum levels of the tumor markers were significantly decreased, the host immune function was increased and the short-term curative effect as well as the quality of life (QOL) were improved in the patients treated by chemotherapy plus CIK cells compared to the patients treated by chemotherapy alone. Moreover, the 2-year life-span was prolonged in the group treated by chemotherapy plus CIK cells compared to the group treated with chemotherapy alone. It is concluded that chemotherapy plus CIK cells has obvious benefits for patients who suffer from advanced gastric cancers.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Immunotherapy, Adoptive/methods , Killer Cells, Lymphokine-Activated/immunology , Stomach Neoplasms/immunology , Stomach Neoplasms/therapy , Adult , Aged , Antigens, Neoplasm/blood , Antigens, Tumor-Associated, Carbohydrate/blood , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , CA-19-9 Antigen/blood , Carcinoembryonic Antigen/blood , Combined Modality Therapy , Female , Fluorouracil/administration & dosage , Humans , Immunotherapy, Adoptive/adverse effects , Interferon-gamma/immunology , Interferon-gamma/pharmacology , Interleukin-2/immunology , Interleukin-2/pharmacology , Leucovorin/administration & dosage , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Male , Middle Aged , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Stomach Neoplasms/blood , Stomach Neoplasms/drug therapy
20.
Biochim Biophys Acta ; 1761(2): 256-60, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16542871

ABSTRACT

Apolipoprotein M (apoM) is a novel apolipoprotein present mostly in high-density lipoprotein (HDL) in human plasma. In the present study, we demonstrate that insulin, insulin-like growth factor I (IGF-I), and IGF-I potential peptide (IGF-IPP) significantly inhibits apoM expression, in a dose- and a time-dependent manner, in the human hepatoma cell line, HepG2 cells. Insulin-induced down-regulation of apoM was blocked by AG1024 (a specific insulin receptor inhibitor) and LY294002 (a phosphatidylinositol 3-kinase (PI3K) inhibitor), which indicates that it is mediated via the activation of PI3K pathway. In contrast, PD98059 (a MAP kinase inhibitor) did not influence insulin-induced down-regulation of apoM expression, and activation of neither PPAR-alpha agonist (GW7647) nor PPAR-gamma agonist (GW1929) influences apoM expression in HepG2 cells, which indicates that regulation of apoM expression is not related to the activation of PPAR-alpha and PPAR-gamma in hepatic cells, whereas, both PPAR-alpha and PPAR-gamma agonists could inhibit apoB expression. Moreover, in the present study, we demonstrated that PPAR beta/delta agonist (GW501516) could inhibit both apoM and apoB expression in the HepG2 cells. In conclusion, this study shows that apoM expression is regulated by PI3-kinase in HepG2-cells.


Subject(s)
Apolipoproteins/genetics , Phosphatidylinositol 3-Kinases/metabolism , Apolipoproteins B/genetics , Apolipoproteins M , Cell Line , Down-Regulation/drug effects , Gene Expression/drug effects , Humans , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Lipocalins , Peroxisome Proliferator-Activated Receptors/agonists , RNA, Messenger/genetics , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL