Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Biochemistry ; 59(45): 4321-4335, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33153264

ABSTRACT

ApoB lipoproteins (apo B-Lp) are produced in hepatocytes, and their secretion requires the cargo receptor sortilin. We examined the secretion of apo B-Lp-containing very low-density lipoprotein (VLDL), an LDL progenitor. Sortilin also regulates the trafficking of the subtilase PCSK9, which when secreted binds the LDL receptor (LDLR), resulting in its endocytosis and destruction at the lysosome. We show that the site 2 binding compound (cpd984) has multiple effects in hepatocytes, including (1) enhanced Apo-Lp secretion, (2) increased cellular PCSK9 retention, and (3) augmented levels of LDLR at the plasma membrane. We postulate that cpd984 enhances apo B-Lp secretion in part through binding the lipid phosphatidylinositol 3,4,5-trisphosphate (PIP3), which is present at higher levels on circulating VLDL form fed rats relative to after fasting. We attribute the enhanced VLDL secretion to its increased binding affinity for sortilin site 1 induced by cpd984 binding site 2. This hinders PCSK9 binding and secretion, which would subsequently prevent its binding to LDLR leading to its degradation. This suggests that site 2 is an allosteric regulator of site 1 binding. This effect is not limited to VLDL, as cpd984 augments binding of the neuropeptide neurotensin (NT) to sortilin site 1. Molecular dynamics simulations demonstrate that the C-terminus of NT (Ct-NT) stably binds site 1 through an electrostatic interaction. This was bolstered by the ability of Ct-NT to disrupt lower-affinity interactions between sortilin and the site 1 ligand PIP3. Together, these data show that binding cargo at sortilin site 1 is allosterically regulated through site 2 binding, with important ramifications for cellular lipid homeostasis involving proteins such as PCSK9 and LDLR.


Subject(s)
Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/metabolism , Hepatocytes/metabolism , Lipoproteins, VLDL/metabolism , Proprotein Convertase 9/metabolism , Receptors, LDL/metabolism , Allosteric Regulation , Animals , Binding Sites , Humans , Molecular Dynamics Simulation , Protein Conformation , Protein Transport , Rats , Rats, Sprague-Dawley
2.
Curr Opin Lipidol ; 28(4): 383-384, 2017 08.
Article in English | MEDLINE | ID: mdl-28700380
3.
Biochem Biophys Res Commun ; 479(3): 551-556, 2016 Oct 21.
Article in English | MEDLINE | ID: mdl-27666481

ABSTRACT

Sortilin is a multi-ligand sorting receptor that interacts with B100-containing VLDL and LDL as well as other ligands including neurotensin (NT). The current study investigates the hypothesis that phosphatidylinositol (3,4,5)-trisphosphate (PIP3) generated downstream of insulin action can directly bind to sortilin. NT binds to sortilin at a well characterized site via its carboxy terminus (C-term). Using a crystal structure of human sortilin (hsortilin), PIP3 is predicted to bind at this C-term site. Binding of PIP3 to hsortilin is demonstrated using surface plasmon resonance (SPR) flowing PIP3 nanodiscs over immobilized hsortilin. Studies were performed using SPR where dibutanoyl PIP3 is shown to compete with NT for sortilin binding. Rat VLDL and LDL were evaluated for PIP3 content immunologically using monoclonal antibodies directed against PIP3. Rat plasma VLDL contained three times more immunoreactive PIP3 than LDL per µg of protein. Because VLDL contains additional ligands that bind sortilin, to distinguish specific PIP3 binding, we used PIP3 liposomes. Liposome floatation assays were used to demonstrate PIP3 liposome binding to sortilin. Using SPR and immobilized hsortilin, the C-term NT tetrapeptide (P-Y-I-L) is shown to bind to hsortilin. A compound (cpd984) was identified with strong theoretical binding to the site on sortilin involved in NT N-terminal binding. When cpd984 is co-incubated with the tetrapeptide, the affinity of binding to sortilin is increased. Similarly, the affinity of PIP3 liposome binding increased in the presence of cpd984. Overall, results demonstrate that sortilin is a PIP3 binding protein with binding likely to occur at the C-term NT binding site. The presence of multiple ligands on B100-containing lipoproteins, VLDL and LDL, raises the interesting possibility for increased interaction with sortilin based on the presence of PIP3.


Subject(s)
Adaptor Proteins, Vesicular Transport/chemistry , Lipoproteins, VLDL/chemistry , Neurotensin/chemistry , Phosphatidylinositol Phosphates/chemistry , Animals , Binding Sites , Computer Simulation , Humans , Lipoproteins, VLDL/blood , Liposomes/chemistry , Phosphatidylinositols/chemistry , Protein Binding , Protein Domains , Rats , Rats, Sprague-Dawley , Surface Plasmon Resonance
4.
Biochem Biophys Res Commun ; 478(2): 546-52, 2016 09 16.
Article in English | MEDLINE | ID: mdl-27495870

ABSTRACT

Studies examining the relationship between cellular sortilin and VLDL-B100 secretion demonstrate inconsistent results. Current studies explore the possibility that discrepancies may be related to insulin sensitivity. McArdle RH7777 cells (McA cells) cultured under serum enriched conditions lose sensitivity to insulin. Following incubation in serum-free DMEM containing 1% BSA, McA cells become insulin responsive and demonstrate reduced apo B secretion. Current studies indicate that insulin sensitive McA cells express lower cellular sortilin that corresponds with reduction in VLDL-B100 secretion without changes in mRNA of either sortilin or apo B. When sortilin expression is further reduced by siRNA knockdown (KD), there are additional decreases in VLDL-B100 secretion. A crystal structure of human sortilin (hsortilin) identifies two binding sites on the luminal domain for the N- and C-termini of neurotensin (NT). A small organic compound (cpd984) was identified that has strong theoretical binding to the N-terminal site. Both cpd984 and NT bind hsortilin by surface plasmon resonance. In incubations with insulin sensitive McA cells, cpd984 was shown to enhance VLDL-B100 secretion at each level of sortilin KD suggesting cpd984 acted through sortilin in mediating its effect. Current results support a role for sortilin to facilitate VLDL-B100 secretion which is limited to insulin sensitive McA cells. Inconsistent reports of the relationship between VLDL-B100 secretion and sortilin in previous studies may relate to differing functions of sortilin in VLDL-B100 secretion depending upon insulin sensitivity.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Apolipoprotein B-100/metabolism , Insulin Resistance , Insulin/metabolism , Lipoproteins, VLDL/metabolism , Adaptor Proteins, Vesicular Transport/chemistry , Adaptor Proteins, Vesicular Transport/genetics , Animals , Binding Sites , Cell Line , Gene Knockdown Techniques , Humans , Molecular Docking Simulation , Rats, Sprague-Dawley
5.
Biochem Biophys Res Commun ; 469(2): 326-31, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26616056

ABSTRACT

Previous studies in rat hepatocytes demonstrated that insulin-dependent apolipoprotein (apo) B degradation (IDAD) is lost when cells are maintained for 3 d under enriched culture conditions. Loss of IDAD correlates with increased expression of protein tyrosine phosphatase 1B (PTP1B) known to be associated with resistance to insulin signaling in the liver. McArdle RH7777 hepatoma (McA) cells cultured in serum containing medium are resistant to IDAD; demonstrate a 30% increase in apo B secretion, and express increased levels of PTP1B protein and mRNA. In addition, insulin-stimulated Class I phosphatidylinositide 3-kinase (PI3K) activity of anti-pY immunoprecipitates is severely blunted. IDAD resistance in McA cells correlates with diminished translocation of insulin-stimulated pY-IRS1 to intracellular membranes. Incubation of McA cells with RK682, a protein tyrosine phosphatase inhibitor, is sufficient to restore IDAD in resistant McA cells. Overall, results further support the importance of Class I PI3K activity in IDAD, and suggest that loss of this activity is sufficient to cause resistance. Although other factors are involved in downstream events including sortilin binding to apo B, autophagy, and lysosomal degradation, loss of signal generation and reduced localization of Class I PI3K to intracellular membranes plays a significant role in IDAD resistance.


Subject(s)
Apolipoproteins B/metabolism , Hepatocytes/metabolism , Insulin Resistance/physiology , Insulin/metabolism , Microsomes, Liver/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Animals , Cell Line , Enzyme Activation , Protein Transport/physiology , Rats , Serum/metabolism , Stress, Physiological/physiology
6.
Hypertension ; 66(4): 881-91, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26324502

ABSTRACT

Sex difference is a risk factor for abdominal aortic aneurysm (AAA) formation yet the reason for male predominance remains unclear. Androgen and the androgen receptor (AR) influence the male sex difference, indicating that AR signaling may affect AAA development. Using angiotensin II­induced AAA in apolipoprotein E null mouse models (82.4% AAA incidence), we found that mice lacking AR failed to develop AAA and aorta had dramatically reduced macrophages infiltration and intact elastic fibers. These findings suggested that AR expression in endothelial cells, macrophages, or smooth muscle cells might play a role in AAA development. Selective knockout of AR in each of these cell types further demonstrated that mice lacking AR in macrophages (20% AAA incidence) or smooth muscle cells (12.5% AAA incidence) but not in endothelial cells (71.4% AAA incidence) had suppressed AAA development. Mechanism dissection showed that AR functioned through modulation of interleukin-1α (IL-1α) and transforming growth factor-ß1 signals and by targeting AR with the AR degradation enhancer ASC-J9 led to significant suppression of AAA development. These results demonstrate the underlying mechanism by which AR influences AAA development is through IL-1α and transforming growth factor-ß1, and provides a potential new therapy to suppress/prevent AAA by targeting AR with ASC-J9.


Subject(s)
Aortic Aneurysm, Abdominal/genetics , Gene Expression Regulation , Inflammation/genetics , Interleukin-1alpha/genetics , RNA/genetics , Receptors, Androgen/genetics , Transforming Growth Factor beta1/genetics , Animals , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Cells, Cultured , Disease Models, Animal , Female , Inflammation/metabolism , Inflammation/pathology , Interleukin-1alpha/biosynthesis , Macrophages/metabolism , Male , Mice , Mice, Knockout , Polymerase Chain Reaction , Receptors, Androgen/biosynthesis , Signal Transduction , Transforming Growth Factor beta1/biosynthesis
8.
Diabetes ; 63(10): 3180-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25249645

ABSTRACT

Prostate cancer (PCa) is one of the most frequently diagnosed malignancies in men. Androgen-deprivation therapy (ADT) is the first-line treatment and fundamental management for men with advanced PCa to suppress functions of androgen/androgen receptor (AR) signaling. ADT is effective at improving cancer symptoms and prolonging survival. However, epidemiological and clinical studies support the notion that testosterone deficiency in men leads to the development of metabolic syndrome that increases cardiovascular disease risk. The underlying mechanisms by which androgen/AR signaling regulates metabolic homeostasis in men are complex, and in this review, we discuss molecular mechanisms mediated by AR signaling that link ADT to metabolic syndrome. Results derived from various AR knockout mouse models reveal tissue-specific AR signaling that is involved in regulation of metabolism. These data suggest that steps be taken early to manage metabolic complications associated with PCa patients receiving ADT, which could be accomplished using tissue-selective modulation of AR signaling and by treatment with insulin-sensitizing agents.


Subject(s)
Androgens/metabolism , Insulin Resistance/physiology , Metabolic Syndrome/etiology , Obesity/etiology , Receptors, Androgen/metabolism , Animals , Humans , Male , Metabolic Syndrome/metabolism , Mice , Obesity/metabolism
9.
Hypertension ; 63(6): 1345-53, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24688120

ABSTRACT

The male sex has a higher risk to develop coronary artery diseases, including atherosclerosis. The androgen receptor (AR) is expressed in several atherosclerosis-associated cell types, including monocytes/macrophages, endothelial cells (ECs), and smooth muscle cells (SMCs), but its pathophysiological role in each cell type during the development of atherosclerotic lesions remains unclear. Using the Cre-loxP system, we selectively knocked out AR in these 3 cell types and the resultant AR knockout (ARKO) mice, monocyte/macrophage ARKO, EC-ARKO, and SMC-ARKO, were then crossed with the low-density lipoprotein receptor (LDLR) deficient (LDLR(-/-)) mice to develop monocyte/macrophage ARKO-LDLR(-/-), EC-ARKO-LDLR(-/-), and SMC-ARKO-LDLR(-/-) mice for the study of atherosclerosis. The results showed that the monocyte/macrophage ARKO-LDLR(-/-) mice had reduced atherosclerosis compared with the wild-type-LDLR(-/-) control mice. However, no significant difference was detected in EC-ARKO-LDLR(-/-) and SMC-ARKO-LDLR(-/-) mice compared with wild-type-LDLR(-/-) mice, suggesting that the AR in monocytes/macrophages, and not in ECs and SMCs, plays a major role to promote atherosclerosis. Molecular mechanism dissection suggested that AR in monocytes/macrophages upregulated the tumor necrosis factor-α, integrin ß2, and lectin-type oxidized LDL receptor 1 molecules that are involved in 3 major inflammation-related processes in atherosclerosis, including monocytes/macrophages migration and adhesion to human umbilical vein ECs, and subsequent foam cell formation. Targeting AR via the AR degradation enhancer, ASC-J9, in wild-type-LDLR(-/-) mice showed similar effects as seen in monocyte/macrophage ARKO-LDLR(-/-) mice with little influence on lipid profile. In conclusion, the AR in monocytes/macrophages plays key roles in atherosclerosis and targeting AR with ASC-J9 may represent a new potential therapeutic approach to battle atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Macrophages/metabolism , Monocytes/metabolism , Receptors, Androgen/deficiency , Animals , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Blotting, Western , CD18 Antigens/metabolism , Cell Adhesion/genetics , Cell Line, Tumor , Cell Movement/genetics , Cells, Cultured , Curcumin/analogs & derivatives , Curcumin/pharmacology , Diet, High-Fat , Foam Cells/cytology , Human Umbilical Vein Endothelial Cells/cytology , Humans , Macrophages/drug effects , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Monocytes/cytology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Receptors, Androgen/genetics , Receptors, LDL/deficiency , Receptors, LDL/genetics , Tumor Necrosis Factor-alpha/metabolism
11.
PLoS One ; 9(1): e85848, 2014.
Article in English | MEDLINE | ID: mdl-24454937

ABSTRACT

Steatosis, oxidative stress, and apoptosis underlie the development of nonalcoholic steatohepatitis (NASH). Protein kinase C delta (PKCδ) has been implicated in fatty liver disease and is activated in the methionine and choline-deficient (MCD) diet model of NASH, yet its pathophysiological importance towards steatohepatitis progression is uncertain. We therefore addressed the role of PKCδ in the development of steatosis, inflammation, oxidative stress, apoptosis, and fibrosis in an animal model of NASH. We fed PKCδ(-/-) mice and wildtype littermates a control or MCD diet. PKCδ(-/-) primary hepatocytes were used to evaluate the direct effects of fatty acids on hepatocyte lipid metabolism gene expression. A reduction in hepatic steatosis and triglyceride levels were observed between wildtype and PKCδ(-/-) mice fed the MCD diet. The hepatic expression of key regulators of ß-oxidation and plasma triglyceride metabolism was significantly reduced in PKCδ(-/-) mice and changes in serum triglyceride were blocked in PKCδ(-/-) mice. MCD diet-induced hepatic oxidative stress and hepatocyte apoptosis were reduced in PKCδ(-/-) mice. MCD diet-induced NADPH oxidase activity and p47(phox) membrane translocation were blunted and blocked, respectively, in PKCδ(-/-) mice. Expression of pro-apoptotic genes and caspase 3 and 9 cleavage in the liver of MCD diet fed PKCδ(-/-) mice were blunted and blocked, respectively. Surprisingly, no differences in MCD diet-induced fibrosis or pro-fibrotic gene expression were observed in 8 week MCD diet fed PKCδ(-/-) mice. Our results suggest that PKCδ plays a role in key pathological features of fatty liver disease but not ultimately in fibrosis in the MCD diet model of NASH.


Subject(s)
Apoptosis , Fatty Liver/enzymology , Lipid Metabolism , Oxidative Stress , Protein Kinase C-delta/physiology , Animals , Biomarkers/metabolism , Cells, Cultured , Choline Deficiency/enzymology , Diet , Endoplasmic Reticulum Stress , Enzyme Activation , Female , Gene Expression , Hepatocytes/physiology , Liver/enzymology , Liver/pathology , Liver Cirrhosis/enzymology , Male , Methionine/deficiency , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease , Primary Cell Culture
13.
Biochem Biophys Res Commun ; 435(4): 616-20, 2013 Jun 14.
Article in English | MEDLINE | ID: mdl-23685141

ABSTRACT

Insulin acutely stimulates the degradation of apolipoprotein B (apo B) which decreases very low density lipoprotein (VLDL) secretion by liver. Insulin-dependent apo B degradation (IDAD) occurs following phosphatidylinositide 3-kinase (PI3K) activation and involves lysosomal degradation. Insulin suppression of apo B secretion is blocked by over-expression of phosphatase and tensin homologue (PTEN) in McArdle RH7777 (McA) cells suggesting the importance of Class I PI3K generated PI (3,4,5) triphosphate (PIP3) in IDAD. Classical autophagy inhibitors including 3-methyladenine, L-asparagine and bafilomycin A1 also blocked the ability of insulin to suppress apo B secretion by rat hepatocytes (RH) suggesting that IDAD occurs through an autophagy-related mechanism. IDAD is also blocked following over-expression in McA cells of a dominant negative kinase-defective Vps34, a class III PI3K that generates PI 3-monophosphate required for autophagy. Vps34 inhibition of IDAD occurs without altering insulin-dependent S473 phosphorylation of Akt indicating PI3K/PIP3/Akt signaling is intact. Cellular p62/SQSTM1, an inverse indicator of autophagy, is increased with insulin treatment consistent with the known ability of insulin to inhibit autophagy, and therefore the role of insulin in utilizing components of autophagy for apo B degradation is unexpected. Thapsigargan, an inducer of endoplasmic reticulum (ER) stress, and a recently demonstrated autophagy inhibitor, blocked apo B secretion which contrasted with other autophagy inhibitors and mutant Vps34 results which were permissive with respect to apo B secretion. Pulse chase studies indicated that intact B100 and B48 proteins were retained in cells treated with thapsigargan consistent with their accumulation in autophagosomal vacuoles. Differences between IDAD and ER stress-coupled autophagy mediated by thapsgargin suggest that IDAD involves an unique form of autophagy. Insulin action resulting in hepatic apo B degradation is novel and important in understanding regulation of hepatic VLDL metabolism.


Subject(s)
Apolipoproteins B/metabolism , Autophagy/physiology , Hepatocytes/metabolism , Insulin/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Cell Line , Humans , Rats
14.
Diabetes ; 62(2): 411-23, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23139353

ABSTRACT

Clinical investigations highlight the increased incidence of metabolic syndrome in prostate cancer (PCa) patients receiving androgen deprivation therapy (ADT). Studies using global androgen receptor (AR) knockout mice demonstrate that AR deficiency results in the development of insulin resistance in males. However, mechanisms by which AR in individual organs coordinately regulates insulin sensitivity remain unexplored. Here we tested the hypothesis that functional AR in the brain contributes to whole-body insulin sensitivity regulation and to the metabolic abnormalities developed in AR-deficient male mice. The mouse model selectively lacking AR in the central nervous system and AR-expressing GT1-7 neuronal cells were established and used to delineate molecular mechanisms in insulin signaling modulated by AR. Neuronal AR deficiency leads to reduced insulin sensitivity in middle-aged mice. Neuronal AR regulates hypothalamic insulin signaling by repressing nuclear factor-κB (NF-κB)-mediated induction of protein-tyrosine phosphatase 1B (PTP1B). Hypothalamic insulin resistance leads to hepatic insulin resistance, lipid accumulation, and visceral obesity. The functional deficiency of AR in the hypothalamus leads to male mice being more susceptible to the effects of high-fat diet consumption on PTP1B expression and NF-κB activation. These findings suggest that in men with PCa undergoing ADT, reduction of AR function in the brain may contribute to insulin resistance and visceral obesity. Pharmacotherapies targeting neuronal AR and NF-κB may be developed to combat the metabolic syndrome in men receiving ADT and in elderly men with age-associated hypogonadism.


Subject(s)
Gene Expression Regulation/physiology , Hypothalamus/metabolism , Insulin Resistance/physiology , NF-kappa B/metabolism , Neurons/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/biosynthesis , Receptors, Androgen/metabolism , Animals , Brain/metabolism , Cell Line , Diet, High-Fat , Insulin/blood , Insulin/metabolism , Insulin Resistance/genetics , Leptin/blood , Leptin/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity, Abdominal/genetics , Obesity, Abdominal/metabolism , Receptors, Androgen/genetics , Signal Transduction/genetics , Signal Transduction/physiology
15.
Biochem Biophys Res Commun ; 430(1): 66-71, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23159624

ABSTRACT

Insulin suppresses secretion of very low density lipoprotein (VLDL) apolipoprotein (apo) B in primary rodent hepatocytes (RH) by favoring the degradation of B100, the larger form of apo B, through post-endoplasmic reticulum proteolysis. Sortilin 1 (sort1), a multi-ligand sorting receptor, has been proposed as a mediator of lysosomal B100 degradation by directing B100 in pre-VLDL to lysosomes rather than allowing maturation to VLDL and secretion. The purpose of our studies was to investigate the role of sort1 in insulin-dependent degradation of apo B. Using liver derived McArdle RH7777 (McA) cells, we demonstrate that insulin suppresses VLDL B100 secretion via a phosphatidylinositide 3-kinase (PI3K) dependent process that is inhibitable by wortmannin in a fashion similar to RH. Using McA cells and in situ cross-linking, we demonstrate that insulin acutely (30min) stimulates the interaction of B100 with sort1. The insulin-induced interaction of sort1-B100 is markedly enhanced when lysosomal degradation is inhibited by Bafilomycin A1 (BafA1), an inhibitor of lysosomal acidification. As BafA1 also prevents insulin suppressive effects on apo B secretion, our results suggest that sort1-B100 interaction stimulated by insulin transiently accumulates with BafA1 and favors B100 secretion by default.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Apolipoproteins B/antagonists & inhibitors , Hepatocytes/drug effects , Insulin/pharmacology , Lysosomes/drug effects , Animals , Apolipoproteins B/metabolism , Cell Line , Hepatocytes/metabolism , Lipoproteins, VLDL/antagonists & inhibitors , Lipoproteins, VLDL/metabolism , Lysosomes/metabolism , Macrolides/pharmacology , Rats
16.
Int J Inflam ; 2012: 678190, 2012.
Article in English | MEDLINE | ID: mdl-22991685

ABSTRACT

Although the exact mechanism through which NADPH oxidases (Nox's) generate reactive oxygen species (ROS) is still not completely understood, it is widely considered that ROS accumulation is the cause of oxidative stress in endothelial cells. Increasing pieces of evidence strongly indicate the role for ROS in endothelial inflammation and dysfunction and subsequent development of atherosclerotic plaques, which are causes of various pathological cardiac events. An overview for a causative relationship between ROS and endothelial inflammation will be provided in this review. Particularly, a crucial role for specific protein SUMOylation in endothelial inflammation will be presented. Given that SUMOylation of specific proteins leads to increased endothelial inflammation, targeting specific SUMOylated proteins may be an elegant, effective strategy to control inflammation. In addition, the involvement of ROS production in increasing the risk of recurrent coronary events in a sub-group of non-diabetic, post-infarction patients with elevated levels of HDL-cholesterol will be presented with the emphasis that elevated HDL-cholesterol under certain inflammatory conditions can lead to increased incidence of cardiovascular events.

17.
Arterioscler Thromb Vasc Biol ; 32(9): 2104-12, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22796579

ABSTRACT

Insulin plays a central role in regulating energy metabolism, including hepatic transport of very low-density lipoprotein (VLDL)-associated triglyceride. Hepatic hypersecretion of VLDL and consequent hypertriglyceridemia leads to lower circulating high-density lipoprotein levels and generation of small dense low-density lipoproteins characteristic of the dyslipidemia commonly observed in metabolic syndrome and type 2 diabetes mellitus. Physiological fluctuations of insulin modulate VLDL secretion, and insulin inhibition of VLDL secretion upon feeding may be the first pathway to become resistant in obesity that leads to VLDL hypersecretion. This review summarizes the role of insulin-related signaling pathways that determine hepatic VLDL production. Disruption in signaling pathways that reduce generation of the second messenger phosphatidylinositide (3,4,5) triphosphate downstream of activated phosphatidylinositide 3-kinase underlies the development of VLDL hypersecretion. As insulin resistance progresses, a number of pathways are altered that further augment VLDL hypersecretion, including hepatic inflammatory pathways. Insulin plays a complex role in regulating glucose metabolism, and it is not surprising that the role of insulin in VLDL and lipid metabolism will prove equally complex.


Subject(s)
Hypertriglyceridemia/etiology , Insulin Resistance , Insulin/metabolism , Lipoproteins, VLDL/biosynthesis , Liver/metabolism , Animals , Apolipoproteins B/genetics , Apolipoproteins B/metabolism , Blood Glucose/metabolism , Cytokines/metabolism , Energy Metabolism , Humans , Hypertriglyceridemia/genetics , Hypertriglyceridemia/immunology , Hypertriglyceridemia/metabolism , Hypertriglyceridemia/physiopathology , Inflammation Mediators/metabolism , Liver/immunology , Liver/physiopathology , Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol Phosphates/metabolism , Signal Transduction , Up-Regulation
19.
Diabetes ; 60(5): 1493-503, 2011 May.
Article in English | MEDLINE | ID: mdl-21478464

ABSTRACT

OBJECTIVE: TR4 is a nuclear receptor without clear pathophysiological roles. We investigated the roles of hepatic TR4 in the regulation of lipogenesis and insulin sensitivity in vivo and in vitro. RESEARCH DESIGN AND METHODS: TR4 activity and phosphorylation assays were carried out using hepatocytes and various TR4 wild-type and mutant constructs. Liver tissues from TR4 knockout, C57BL/6, and db/db mice were examined to investigate TR4 target gene stearoyl-CoA desaturase (SCD) 1 regulation. RESULTS: TR4 transactivation is inhibited via phosphorylation by metformin-induced AMP-activated protein kinase (AMPK) at the amino acid serine 351, which results in the suppression of SCD1 gene expression. Additional mechanistic dissection finds TR4-transactivated SCD1 promoter activity via direct binding to the TR4-responsive element located at -243 to -255 on the promoter region. The pathophysiological consequences of the metformin→AMPK→TR4→SCD1 pathway are examined via TR4 knockout mice and primary hepatocytes with either knockdown or overexpression of TR4. The results show that the suppression of SCD1 via loss of TR4 resulted in reduced fat mass and increased insulin sensitivity with increased ß-oxidation and decreased lipogenic gene expression. CONCLUSIONS: The pathway from metformin→AMPK→TR4→SCD1→insulin sensitivity suggests that TR4 may function as an important modulator to control lipid metabolism, which sheds light on the use of small molecules to modulate TR4 activity as a new alternative approach to battle the metabolic syndrome.


Subject(s)
Liver/metabolism , Metformin/pharmacology , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Stearoyl-CoA Desaturase/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Glucose Tolerance Test , Immunoprecipitation , Insulin/pharmacology , Liver/drug effects , Liver/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/drug effects , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Stearoyl-CoA Desaturase/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism
20.
Biochem Biophys Res Commun ; 406(2): 252-6, 2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21316344

ABSTRACT

Secretion of apolipoprotein (apo) B-containing lipoproteins by the liver depends mainly upon apo B availability and microsomal triglyceride transfer protein (MTP) activity and is subject to insulin regulation. Hepatic MTP mRNA expression is negatively regulated by insulin which correlates with inhibition of apo B secretion suggesting that insulin might suppress apo B secretion through an MTP-dependent mechanism. To investigate this possibility, we examined the acute effect of insulin on hepatic MTP expression and activity levels in vivo utilizing apobec-1(-/-) mice. Insulin did not significantly alter hepatic MTP mRNA levels or lipid transfer activity 2h following injection, but suppressed expression of genes important in gluconeogenesis. To study the specific role of MTP, we expressed human MTP (hMTP) in primary rat hepatocytes using adenoviral gene transfer. Increased expression of hMTP resulted in a 47.6±17.9% increase in total apo B secreted. Incubation of hepatocytes with insulin suppressed apo B secretion by 50.1±10.8% in cells over-expressing hMTP and by 53.0±12.4% in control transfected hepatocytes. Results indicate that even under conditions of increased hepatic apo B secretion mediated by MTP, responsiveness of hepatocytes to insulin to suppress apo B secretion is maintained.


Subject(s)
Apolipoproteins B/metabolism , Carrier Proteins/metabolism , Insulin/physiology , Lipoproteins, VLDL/metabolism , Liver/metabolism , Adenoviridae , Animals , Carrier Proteins/genetics , Cells, Cultured , Humans , Insulin/pharmacology , Liver/drug effects , Male , Mice , Mice, Mutant Strains , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...