Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
FASEB J ; 32(3): 1602-1612, 2018 03.
Article in English | MEDLINE | ID: mdl-29183962

ABSTRACT

The intestine is involved in whole-body lipid and cholesterol homeostasis and secretes lipoproteins containing apolipoprotein (Apo)B48 and discrete ApoA-I into the mesenteric lymph. The lymphatic system has been proposed to have a significant role in the reverse cholesterol transport pathway associated with HDL-ApoA-I. In conditions of insulin resistance (IR), there is intestinal overproduction of chylomicrons containing ApoB48; however, there is limited data on the intestinal synthesis and secretion of HDL-ApoA-I. microRNA (miR)-223 has been shown to regulate peripheral HDL metabolism and may impact intestinal-derived HDL. Niacin (nicotinic acid; vitamin B3) is known to regulate lipid metabolism, but the role of niacin in modulating intestinal lipid and lipoprotein (ApoB48 and ApoA-I) metabolism is unknown. The aim of this study was to determine the secretion of intestinal lymphatic HDL-ApoA-I and the effect of dietary intervention with niacin on these pathways in a rodent model of IR. HDL was isolated from intestinal mesenteric lymph by density ultracentrifugation, and subsequent HDL miR analysis was developed in collaboration with Exiqon Services. Insulin-resistant rodents were fed chow or chow with niacin (1% w/w) for 6 wk. Intestinal lymph HDL-ApoA-I and miR-223 expression were lower by at least 45 and 60%, respectively, and lymph HDL was associated with 85% higher triglyceride (TG) content in IR compared to non-IR control group. Niacin was found to increase secretion of lymph HDL and miR-223 by at least 50-60% and to deplete the TGs associated with HDL compared with the nontreated IR group. Niacin significantly increased peroxisome proliferator-activating nuclear receptor α and carnitine palmitoyltransferase I α mRNA and annulled Tnf-α mRNA expression in intestinal (jejunal) explants. Altered intestinal lymphatic HDL-ApoA-I and miR-223 metabolism in IR and modulation by niacin may provide insight into the intestinal-mediated regulation of the reverse cholesterol transport pathway.-Mangat, R., Borthwick, F., Haase, T., Jacome, M., Nelson, R., Kontush, A., Vine, D. F., Proctor, S. D. Intestinal lymphatic HDL miR-223 and ApoA-I are reduced during insulin resistance and restored with niacin.


Subject(s)
Apolipoprotein A-I/biosynthesis , Gene Expression Regulation/drug effects , Insulin Resistance/ethnology , Intestinal Mucosa/metabolism , Lipoproteins, HDL/biosynthesis , Lymph Nodes/metabolism , MicroRNAs/biosynthesis , Niacin/pharmacology , Animals , Apolipoprotein A-I/genetics , Lipoproteins, HDL/genetics , Male , Mesentery/metabolism , Mice , Mice, Transgenic , MicroRNAs/genetics
2.
Front Nutr ; 3: 44, 2016.
Article in English | MEDLINE | ID: mdl-27777929

ABSTRACT

Obesity and its metabolic complications have emerged as the epidemic of the new millennia. The use of obese rodent models continues to be a productive component of efforts to understand the concomitant metabolic complications of this disease. In 1978, the JCR:LA-cp rat model was developed with an autosomal recessive corpulent (cp) trait resulting from a premature stop codon in the extracellular domain of the leptin receptor. Rats that are heterozygous for the cp trait are lean-prone, while those that are homozygous (cp/cp) spontaneously display the pathophysiology of obesity as well as a metabolic syndrome (MetS)-like phenotype. Over the years, there have been formidable scientific contributions that have originated from this rat model, much of which has been reviewed extensively up to 2008. The premise of these earlier studies focused on characterizing the pathophysiology of MetS-like phenotype that was spontaneously apparent in this model. The purpose of this review is to highlight areas of recent advancement made possible by this model including; emerging appreciation of the "thrifty gene" hypothesis in the context of obesity, the concept of how chronic inflammation may drive obesogenesis, the impact of acute forms of inflammation to the brain and periphery during chronic obesity, the role of dysfunctional insulin metabolism on lipid metabolism and vascular damage, and the mechanistic basis for altered vascular function as well as novel parallels between the human condition and the female JCR:LA-cp rat as a model for polycystic ovary disease (PCOS).

3.
Food Funct ; 7(9): 3943-52, 2016 Sep 14.
Article in English | MEDLINE | ID: mdl-27538786

ABSTRACT

Hawthorn is a widely used herbal alternative medicine for the treatment of various cardiovascular diseases. However, the attributed health benefits, purported to be due to the presence of phenolic compounds, may depend on both the specific species and plant part. Studies to date investigating effects of hawthorn on heart disease(s) have used well-described European and/or Asian species, while little is known regarding the bioactivity of species native to North America. Six weeks of supplementation of both fireberry hawthorn berry (native Crataegus chrysocarpa) and English hawthorn leaf (C. monogyna, naturalized in North America) in the JCR:LA-cp rat, resulted in a significant reduction in heart weight, fasting LDL-C and improved heart function (p < 0.05). Fasting triglyceride and myocardial fibrosis were also reduced, but only by the berry extract. We demonstrate that both of the Canadian-sourced hawthorn extracts (introduced leaf and native berry) have cardioprotective benefits, likely via increased availability of nitric oxide.


Subject(s)
Cardiotonic Agents/therapeutic use , Crataegus/chemistry , Dietary Supplements , Dyslipidemias/prevention & control , Heart Diseases/prevention & control , Hypolipidemic Agents/therapeutic use , Plant Extracts/therapeutic use , Animals , Canada , Cholesterol, LDL/blood , Crataegus/growth & development , Dyslipidemias/blood , Dyslipidemias/pathology , Dyslipidemias/physiopathology , Fibrosis , Fruit/chemistry , Fruit/growth & development , Heart/physiopathology , Heart Diseases/blood , Heart Diseases/pathology , Heart Diseases/physiopathology , Introduced Species , Male , Myocardium/pathology , Organ Size , Plant Leaves/chemistry , Plant Leaves/growth & development , Random Allocation , Rats, Mutant Strains
4.
Lipids ; 51(7): 821-31, 2016 07.
Article in English | MEDLINE | ID: mdl-27072368

ABSTRACT

The main dietary sources of trans fatty acids are partially hydrogenated vegetable oils (PHVO), and products derived from polyunsaturated fatty acid biohydrogenation (PUFA-BHP) in ruminants. Trans fatty acid intake has historically been associated with negative effects on health, generating an anti-trans fat campaign to reduce their consumption. The profiles and effects on health of PHVO and PUFA-BHP can, however, be quite different. Dairy products naturally enriched with vaccenic and rumenic acids have many purported health benefits, but the putative benefits of beef fat naturally enriched with PUFA-BHP have not been investigated. The objective of the present experiment was to determine the effects of beef peri-renal fat (PRF) with differing enrichments of PUFA-BHP on lipid and insulin metabolism in a rodent model of dyslipidemia and insulin resistance (JCR:LA-cp rat). The results showed that 6 weeks of diet supplementation with beef PRF naturally enriched due to flaxseed (FS-PRF) or sunflower-seed (SS-PRF) feeding to cattle significantly improved plasma fasting insulin levels and insulin sensitivity, postprandial insulin levels (only in the FS-PRF) without altering dyslipidemia. Moreover, FS-PRF but not SS-PRF attenuated adipose tissue accumulation. Therefore, enhancing levels of PUFA-BHP in beef PRF with FS feeding may be a useful approach to maximize the health-conferring value of beef-derived fats.


Subject(s)
Dietary Fats, Unsaturated/administration & dosage , Dyslipidemias/diet therapy , Fatty Acids, Unsaturated/administration & dosage , Fatty Acids, Unsaturated/metabolism , Red Meat/analysis , Trans Fatty Acids/analysis , Animal Feed , Animals , Cattle , Dairy Products/analysis , Dietary Supplements , Disease Models, Animal , Hydrogenation , Insulin Resistance , Linseed Oil/administration & dosage , Lipids/blood , Male , Plant Oils/administration & dosage , Rats , Sunflower Oil
5.
J Endocrinol ; 226(3): 193-206, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26187902

ABSTRACT

Polycystic ovary syndrome (PCOS) is one of the most common endocrine-metabolic disorders in women of reproductive age characterized by ovulatory dysfunction, hyperandrogenism and cardiometabolic risk. The overweight-obese PCOS phenotype appears to have exacerbated reproductive dysfunction and cardiometabolic risk. In overweight-obese adult women with PCOS, exercise and energy restricted diets have shown limited and inconsistent effects on both cardiometabolic indices and reproductive outcomes. We hypothesized that an early lifestyle intervention involving exercise and dietary energy restriction to prevent or reduce the propensity for adiposity would modulate reproductive indices and cardiometabolic risk in an obese PCOS-prone rodent model. Weanling obese PCOS-prone and Lean-Control JCR:LA-cp rodents were given a chow diet ad libitum or an energy-restricted diet combined with or without voluntary exercise (4  h/day) for 8 weeks. Dietary energy restriction and exercise lowered total body weight gain and body fat mass by 30% compared to free-fed sedentary or exercising obese PCOS-prone animals (P<0.01). Energy restriction induced an increase in exercise intensity compared to free-feeding plus exercise conditions. Energy restriction and exercise decreased fasting plasma triglycerides and apoB48 concentrations in obese PCOS-prone animals compared to free-fed and exercise or sedentary groups. The energy restriction and exercise combination in obese PCOS-prone animals significantly increased plasma sex-hormone binding globulin, hypothalamic cocaine-and amphetamine-regulated transcript (CART) and Kisspeptin mRNA expression to levels of the Lean-Control group, and this was further associated with improvements in estrous cyclicity. The combination of exercise and dietary energy restriction when initiated in early life exerts beneficial effects on cardiometabolic and reproductive indices in an obese PCOS-prone rodent model, and this may be associated with normalization of the hypothalamic neuropeptides, Kisspeptin and CART.


Subject(s)
Caloric Restriction , Diet , Obesity/metabolism , Physical Conditioning, Animal/physiology , Polycystic Ovary Syndrome/metabolism , Adipose Tissue/metabolism , Animals , Body Weight/physiology , Disease Models, Animal , Female , Insulin Resistance/physiology , Life Style , Obesity/prevention & control , Rats
6.
FASEB J ; 28(9): 4088-99, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24903275

ABSTRACT

Inadequate cell proliferation is considered a major causative factor for impaired coronary collateral growth (CCG). Proangiogenic growth factors (GFs) stimulate cell proliferation, but their administration does not promote CCG in patients. These GFs are increased in patients with metabolic syndrome and in animal models, where CCG is impaired. Here, we investigated whether excessive cell proliferation underlies impaired CCG in metabolic syndrome. Normal [Sprague-Dawley (SD)] and metabolic syndrome [James C. Russell (JCR)] rats underwent repetitive ischemia (RI; transient, repetitive coronary artery occlusion and myocardial ischemia). We have shown that CCG was maximal at d 9 of RI in SD rats but did not occur in JCR rats. The increase in cell proliferation (PCNA, Ki-67, cyclin A, phospho- cdc2, p21Waf, p27Kip) was transient (∼4-fold, d 3 RI) in SD rats but greater and sustained in JCR rats (∼8- to 6-fold, d 3-9 RI). In JCR rats, this was associated with increased and sustained miR-21 expression and accumulation of proliferating synthetic vascular smooth muscle cells in the lumen of small arterioles, which failed to undergo outward expansion. Administration of anti-miR-21 blocked RI-induced cell proliferation and significantly improved CCG in JCR rats (∼60%). miR-21-dependent excessive cell proliferation in the later stages of collateral remodeling correlates with impaired CCG in metabolic syndrome.


Subject(s)
Cell Proliferation/genetics , Collateral Circulation/physiology , Coronary Artery Disease/prevention & control , Coronary Circulation/physiology , Metabolic Syndrome/physiopathology , MicroRNAs/metabolism , Muscle, Smooth, Vascular/cytology , Myocardial Ischemia/prevention & control , Animals , Apoptosis , Blotting, Western , Cells, Cultured , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Disease Models, Animal , Immunoenzyme Techniques , Male , MicroRNAs/genetics , Muscle, Smooth, Vascular/metabolism , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Neovascularization, Pathologic/prevention & control , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
7.
J Nutr Biochem ; 25(7): 692-701, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24775093

ABSTRACT

Trans11-18:1 (vaccenic acid, VA) is one of the most predominant naturally occurring trans fats in our food chain and has recently been shown to exert hypolipidemic effects in animal models. In this study, we reveal new mechanism(s) by which VA can alter body fat distribution, energy utilization and dysfunctional lipid metabolism in an animal model of obesity displaying features of the metabolic syndrome (MetS). Obese JCR:LA-cp rats were assigned to a control diet that included dairy-derived fat or the control diet supplemented with 1% VA. VA reduced total body fat (-6%), stimulated adipose tissue redistribution [reduced mesenteric fat (-17%) while increasing inguinal fat mass (29%)] and decreased adipocyte size (-44%) versus control rats. VA supplementation also increased metabolic rate (7%) concomitantly with an increased preference for whole-body glucose utilization for oxidation and increased insulin sensitivity [lower HOMA-IR (-59%)]. Further, VA decreased nonalcoholic fatty liver disease activity scores (-34%) and reduced hepatic (-27%) and intestinal (-39%) triglyceride secretion relative to control diet, while exerting differential transcriptional regulation of SREBP1 and FAS amongst other key genes in the liver and the intestine. Adding VA to dairy fat alleviates features of MetS potentially by remodeling adipose tissue and attenuating ectopic lipid accumulation in a rat model of obesity and MetS. Increasing VA content in the diet (naturally or by fortification) may be a useful approach to maximize the health value of dairy-derived fats.


Subject(s)
Dietary Fats/pharmacology , Metabolic Syndrome/metabolism , Non-alcoholic Fatty Liver Disease/physiopathology , Oleic Acids/pharmacology , Adipocytes/drug effects , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Dairy Products , Disease Models, Animal , Disease Progression , Fatty Acids/pharmacology , Insulin/metabolism , Insulin Resistance , Liver/drug effects , Liver/metabolism , Male , Metabolic Syndrome/prevention & control , Non-alcoholic Fatty Liver Disease/prevention & control , Obesity/metabolism , Rats
8.
Atherosclerosis ; 232(1): 141-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24401228

ABSTRACT

OBJECTIVE: Statins are widely used for the treatment of hyperlipidemia to reduce cardiovascular disease (CVD) risk. Intriguingly, recent reports suggest that whilst statins are effective in reducing hepatic cholesterol synthesis, they in turn may up-regulate intestinal cholesterol absorption. The direct effects and/or mechanisms of this phenomenon remain largely unknown. The aim of this study was to investigate the potential for statins to increase intestinal lipid absorption and/or secretion in a rodent model of the metabolic syndrome (MetS). METHODS AND RESULTS: Mets JCR:LA-cp rats received a 1% cholesterol diet containing Simvastatin (0.01% w/w), for 8 weeks. Fasting and postprandial plasma biochemical profile was assessed using enzymatic assays and a modified apoB48 (chylomicron; CM) western blotting protocol. Statin treatment reduced fasting plasma TG (-49%), cholesterol (-24%) and postprandial plasma apoB48 (-58%). The intestinal secretion of lipids into mesenteric lymph was assessed using lymph fistulae procedures. Interestingly, MetS rats treated with statin secreted greater cholesterol (1.9-fold) and TG (1.5-fold) per apoB48 particle, into mesenteric lymph. This was shown to be as a result of simvastatin-induced increase in intestinal cholesterol absorption (31.5%). Experiments using in vivo inhibition of lipoprotein lipase (LPL; poloxamer-407) demonstrated statin treatment reduced hepatic cholesterol secretion (-49%), but significantly increased hepatic (73%) TG secretion in MetS rats. Statin treatment also increased the expression of genes involved in lipid synthesis (Hmgcr, Srebp1, Fas, Acc; 33-67%) and reduced those involved in efflux (Abca1, Abcg8; -36 to 73%) in enterocytes and liver of MetS rats versus untreated control. CONCLUSIONS: In a rodent model of MetS, statin treatment adversely up-regulates intestinal lipid secretion as a result of increased intestinal cholesterol absorption, and increases the intestinal expression of genes involved in lipid synthesis; effects which may confound clinical benefits to remnant dyslipidemia.


Subject(s)
Anticholesteremic Agents/therapeutic use , Lipids/blood , Metabolic Syndrome/blood , Simvastatin/therapeutic use , Up-Regulation , Animals , Anticholesteremic Agents/blood , Cardiovascular Diseases/prevention & control , Cholesterol/chemistry , Cholesterol/pharmacokinetics , Disease Models, Animal , Dyslipidemias/drug therapy , Food Deprivation , Gene Expression Regulation , Hydrolysis , Intestines/drug effects , Lipoprotein Lipase/blood , Male , Metabolic Syndrome/drug therapy , Postprandial Period , Rats , Simvastatin/blood , Triglycerides/blood
9.
Br J Nutr ; 110(1): 11-9, 2013 Jul 14.
Article in English | MEDLINE | ID: mdl-23151363

ABSTRACT

We have previously shown nutritional intervention with fish oil (n-3 PUFA) to reduce numerous complications associated with the metabolic syndrome (MetS) in the JCR:LA-corpulent (cp) rat. In the present study, we sought to explore the potential role of fish oil to prevent glomerulosclerosis in JCR:LA-cp rats via renal eicosanoid metabolism and lipidomic analysis. Male lean and MetS JCR:LA-cp rats were fed a lipid-balanced diet supplemented with fish oil (5 or 10 % of total fat). After 16 weeks of feeding, albuminuria was significantly reduced in MetS rats supplemented with 5 or 10 % fish oil ( - 53 and - 70 %, respectively, compared with the untreated MetS rats). The 5 % fish oil diet resulted in markedly lower glomerulosclerosis ( - 43 %) in MetS rats and to a lesser extent in those supplemented with 10 % fish oil. Interestingly, untreated MetS rats had higher levels of 11- and 12-hydroxyeicosatetraenoic acids (HETE) v. lean rats. Dietary fish oil reduced these levels, as well as other (5-, 9- and 15-) HETE. Whilst genotype did not alter prostanoid levels, fish oil reduced endogenous renal levels of 6-keto PGF1α (PGI2 metabolite), thromboxane B2 (TxB2), PGF2α and PGD2 by approximately 60 % in rats fed 10 % fish oil, and TxB2 ( - 50 %) and PGF2α ( - 41 %) in rats fed 5 % fish oil. In conclusion, dietary fish oil prevented glomerular damage in MetS rats and mitigated the elevation in renal HETE levels. These results suggest a potential role for dietary fish oil to improve dysfunctional renal eicosanoid metabolism associated with kidney damage during conditions of the MetS.


Subject(s)
Dietary Fats/therapeutic use , Fish Oils/therapeutic use , Hydroxyeicosatetraenoic Acids/blood , Kidney Diseases/prevention & control , Kidney Glomerulus/drug effects , Metabolic Syndrome/complications , 6-Ketoprostaglandin F1 alpha/metabolism , Albuminuria/prevention & control , Animals , Dietary Fats/metabolism , Dietary Fats/pharmacology , Dietary Supplements , Dinoprost/metabolism , Disease Models, Animal , Fish Oils/metabolism , Fish Oils/pharmacology , Genotype , Kidney Diseases/etiology , Kidney Diseases/metabolism , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Male , Metabolic Syndrome/metabolism , Prostaglandin D2/metabolism , Prostaglandins/metabolism , Rats , Rats, Inbred Strains , Thromboxane B2/metabolism
10.
Atherosclerosis ; 222(2): 402-8, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22483015

ABSTRACT

OBJECTIVE: Low plasma high-density lipoprotein cholesterol (HDL-C) concentration is associated with the metabolic syndrome (MetS) and increased prevalence of cardiovascular disease (CVD). Animal and human studies report infusion of apolipoprotein A-1 (apoA-1) can reduce endothelial dysfunction, and/or induce regression of atherosclerosis. However, the direct mechanisms underlying the vascular benefits of either apoA-1 or HDL-C remain unclear. In this study, we assessed the ability of reconstituted HDL (rHDL) to improve vascular complications of MetS, including left ventricular (LV)-hypertrophy, arterial cholesterol deposition and myocardial lesion development. METHODS AND RESULTS: Obese insulin resistant (IR) JCR:LA-cp rats were infused with rHDL (0.4 mg/kg) over 3 days before assessing cardiac function (Echocardiography) at days 7 and 50 post-infusion, as well as haematoxylin and eosin staining of myocardial lesions at day 50. Acute ex vivo arterial cholesterol deposition was assessed with acute infusion of rHDL ex-vivo. Infusion of rHDL partially corrected abnormal diastolic compliance (18%; *p<0.05) and improved parameters of cardiac function in IR rats. Further, acute rHDL infusion in carotid vessels reduced remnant lipoprotein associated-cholesterol deposition (30-86%; **p<0.01) ex vivo in IR and male Wistar rats and reduced (41%; *p<0.05) the frequency of early-stage myocardial lesions in IR rats. CONCLUSION: Short-term infusion of rHDL may beneficially reduce chronic vascular sequelae of MetS, including temporary improvement in LV-dysfunction, acute reduction of acute arterial cholesterol deposition and the development of early-stage myocardial lesions in the JCR:LA-cp rat.


Subject(s)
Apolipoprotein A-I/administration & dosage , Carotid Arteries/drug effects , Carotid Artery Diseases/drug therapy , Cholesterol/metabolism , Insulin Resistance , Lipoproteins, HDL/administration & dosage , Metabolic Syndrome/drug therapy , Myocardium/pathology , Ventricular Dysfunction, Left/drug therapy , Ventricular Function, Left/drug effects , Animals , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Diseases/etiology , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/pathology , Disease Models, Animal , Echocardiography, Doppler , Humans , Infusions, Parenteral , Male , Metabolic Syndrome/etiology , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Myocardium/metabolism , Obesity/complications , Rats , Rats, Wistar , Time Factors , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology
11.
J Am Heart Assoc ; 1(5): e003434, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23316299

ABSTRACT

BACKGROUND: Literature supports the "response-to-retention" hypothesis-that during insulin resistance, impaired metabolism of remnant lipoproteins can contribute to accelerated cardiovascular disease progression. We used the JCR:LA-cp rat model of metabolic syndrome (MetS) to determine the extent of arterial accumulation of intestinal-derived remnants ex vivo and potential mechanisms that contribute to exacerbated cholesterol deposition in insulin resistance. METHODS AND RESULTS: Arteries from control and MetS (insulin-resistant) JCR:LA-cp rats were perfused ex vivo with Cy5-labeled remnant lipoproteins, and their arterial retention was quantified by confocal microscopy. Arterial proteoglycans were isolated from control and MetS rats at 6, 12, and 32 weeks of age. There was a significant increase in the arterial retention of remnants and in associated cholesterol accumulation in MetS rats as compared to control rats. Mechanistic studies reveal that increased cholesterol deposition is a result of greater arterial biglycan content; longer glycosaminoglycans and increased production of cholesterol-rich intestinal-derived remnants, as compared to controls. Additionally, perfusion of vessels treated with ezetimibe, alone or in combination with simvastatin, with remnants isolated from the respective treatment group reduced ex vivo arterial retention of remnant-derived cholesterol ex vivo as compared to untreated controls. CONCLUSIONS: Increased progression of atherosclerotic cardiovascular disease in MetS and type 2 diabetes mellitus might be explained in part by an increase in the arterial retention of cholesterol-rich remnants. Furthermore, ezetimibe alone or in combination treatment with simvastatin could be beneficial in ameliorating atherosclerotic cardiovascular disease in insulin resistance and MetS.


Subject(s)
Anticholesteremic Agents/therapeutic use , Arteries/metabolism , Atherosclerosis/physiopathology , Azetidines/therapeutic use , Biglycan/metabolism , Cholesterol/metabolism , Insulin Resistance/physiology , Lipoproteins/metabolism , Metabolic Syndrome/physiopathology , Animals , Atherosclerosis/drug therapy , Ezetimibe , Male , Metabolic Syndrome/drug therapy , Rats , Simvastatin/therapeutic use
12.
Br J Nutr ; 105(11): 1572-82, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21276281

ABSTRACT

There is increasing interest in the potential chronic beneficial effects of dietary n-3 PUFA on the metabolic syndrome (MetS) and associated cardiovascular complications. We have recently established that increased dietary n-3 PUFA has a profound acute benefit on fasting lipids and the postprandial pro-inflammatory response in the JCR:LA-cp rat, a model of the MetS. However, it is unclear to what extent chronic dietary n-3 PUFA intervention can modulate the progression of end-stage metabolic and vascular complications. The present study aimed to determine the chronic effects of dietary n-3 PUFA supplementation on fasting and non-fasting dyslipidaemia, insulin resistance and vascular complications in the JCR:LA-cp rodent model. JCR:LA-cp rats were fed an isoenergetic lipid-balanced diet supplemented with 5 % n-3 PUFA (w/w) of the total fat (fish oil-derived EPA/DHA) for 16 weeks. Fasting and non-fasting (postprandial) plasma lipid profile was assessed. Hepatic and adipose tissue was probed for the expression of lipogenic proteins (acyl-CoA carboxylase (ACC), fatty acid synthase (FAS) and sterol regulatory element-binding protein-1 (SREBP-1)), while the activity of Jun N-terminal kinase (JNK) was assessed via Western blot to target phosphorylated JNK protein in primary enterocytes. The frequency of myocardial lesions was assessed by haematoxylin and eosin staining. Increased dietary n-3 PUFA improved both the fasting and postprandial lipid profiles (TAG, cholesterol and apoB48) in the JCR:LA-cp rat, potentially via the down-regulation of the hepatic or adipose tissue expression of lipogenic enzymes (ACC, FAS and SREBP-1). Rats fed the 5 % n-3 PUFA diet had lower (58·2 %; P < 0·01) enterocytic phosphorylated JNK protein and secreted less cholesterol (30 %; P < 0·05) into mesenteric lymph compared with the control. The chronic metabolic benefits of dietary n-3 PUFA may underlie the potential to reduce vascular complications during the MetS, including the observed reduction in the frequency (approximately 80 %) of late-stage 3 myocardial lesions.


Subject(s)
Cardiovascular Diseases/prevention & control , Diet , Dyslipidemias/diet therapy , Fatty Acids, Omega-3/administration & dosage , Metabolic Syndrome/diet therapy , Animals , Body Weight/drug effects , Disease Models, Animal , Drug Administration Schedule , Dyslipidemias/blood , Eating/drug effects , Enterocytes/metabolism , Fatty Acids, Omega-3/pharmacology , Gene Expression Regulation/drug effects , Jejunum/cytology , Lipids/blood , Lymph/chemistry , Male , Myocardium/pathology , Obesity/genetics , Postprandial Period , Random Allocation , Rats
13.
Clin Sci (Lond) ; 119(7): 265-72, 2010 Jun 22.
Article in English | MEDLINE | ID: mdl-20491656

ABSTRACT

Dysregulated macrophage cholesterol homoeostasis lies at the heart of early and developing atheroma, and removal of excess cholesterol from macrophage foam cells, by efficient transport mechanisms, is central to stabilization and regression of atherosclerotic lesions. The present study demonstrates that transient overexpression of STARD3 {START [StAR (steroidogenic acute regulatory protein)-related lipid transfer] domain 3; also known as MLN64 (metastatic lymph node 64)}, an endosomal cholesterol transporter and member of the 'START' family of lipid trafficking proteins, induces significant increases in macrophage ABCA1 (ATP-binding cassette transporter A1) mRNA and protein, enhances [(3)H]cholesterol efflux to apo (apolipoprotein) AI, and reduces biosynthesis of cholesterol, cholesteryl ester, fatty acids, triacylglycerol and phospholipids from [(14)C]acetate, compared with controls. Notably, overexpression of STARD3 prevents increases in cholesterol esterification in response to acetylated LDL (low-density lipoprotein), blocking cholesteryl ester deposition. Thus enhanced endosomal trafficking via STARD3 induces an anti-atherogenic macrophage lipid phenotype, positing a potentially therapeutic strategy.


Subject(s)
Atherosclerosis/metabolism , Carrier Proteins/biosynthesis , Lipid Metabolism/physiology , Macrophages/metabolism , Membrane Proteins/biosynthesis , Monocytes/metabolism , Aorta/metabolism , Carrier Proteins/genetics , Cells, Cultured , Foam Cells/metabolism , Gene Expression Regulation/physiology , Humans , Membrane Proteins/genetics , Phenotype , Polymerase Chain Reaction/methods , RNA, Messenger/genetics
14.
Cardiovasc Res ; 86(3): 526-34, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20083572

ABSTRACT

AIMS: In this study, we investigated the impact of enhancing cholesterol delivery to mitochondrial sterol 27-hydroxylase, via steroidogenic acute regulatory protein (StAR), on the expression of genes involved in macrophage cholesterol homeostasis and efflux of cholesterol to apolipoprotein (apo) AI. METHODS AND RESULTS: Stably transfected, murine (RAW 264.7) macrophages were used to investigate the role of StAR in cholesterol homeostasis. Cellular responses were analysed using quantitative PCR, immunoblotting, and an LXRE reporter plasmid; [3H]cholesterol efflux was measured in the presence or absence of apoAI. Macrophage overexpression of mitochondrial cholesterol trafficking protein, StAR, activates and induces expression of liver X receptors (LXRs), and significantly alters expression of genes involved in cholesterol homeostasis, decreasing Fdps, Hmgcr, Mvk, Ldlr, and Scap, and markedly increasing Abca1 mRNA and protein. Overexpression of StAR, but not mutated 'loss-of-function' (R181L) StAR, enhanced efflux of [3H]cholesterol to apoAI, and this effect was maintained in macrophages pretreated with LDL or acetylated LDL. The effect of StAR overexpression on apoAI-dependent [3H]cholesterol efflux was mimicked by non-sterol agonist, T901317, and 27-hydroxycholesterol, and blocked by LXR inhibitor, geranylgeranyl pyrophosphate, sterol 27-hydroxylase inhibitor, GW273297x, and probucol, inhibitor of ATP binding cassette transporter A1 (ABCA1). Importantly, all observed effects of StAR overexpression were dependent upon cyclic AMP (cAMP analogue, dibutyryl cAMP), which is required for the full activity of the StAR protein to be manifested. CONCLUSION: Macrophage overexpression of StAR significantly enhances LXR-dependent apoAI- and ABCA1-dependent cholesterol efflux, by which disposal of excess arterial cholesterol deposits and atheroma regression can be achieved.


Subject(s)
Apolipoprotein A-I/metabolism , Cholesterol/metabolism , Macrophages/metabolism , Phosphoproteins/metabolism , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Biological Transport , Cell Line , Cholestanetriol 26-Monooxygenase/antagonists & inhibitors , Cholestanetriol 26-Monooxygenase/metabolism , Cyclic CMP/analogs & derivatives , Cyclic CMP/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Genes, Reporter , Homeostasis , Hydrocarbons, Fluorinated/pharmacology , Hydroxycholesterols/metabolism , Immunoblotting , Lipid Metabolism/genetics , Lipoproteins, LDL/metabolism , Liver X Receptors , Macrophages/drug effects , Mice , Mutation , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/metabolism , Phosphoproteins/genetics , Polyisoprenyl Phosphates/pharmacology , Probucol/pharmacology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sulfonamides/pharmacology , Transfection , Triglycerides/metabolism , Up-Regulation
15.
FEBS Lett ; 583(7): 1147-53, 2009 Apr 02.
Article in English | MEDLINE | ID: mdl-19272380

ABSTRACT

The STARD1 subfamily of 'START' lipid trafficking proteins can reduce macrophage lipid content and inflammatory status (STARD1; StAR), and traffic cholesterol from endosomes (STARD3/MLN64). During macrophage differentiation, STARD1 mRNA and protein increase with sterol content, while the reverse is true for STARD3. Sterol depletion (methyl beta-cyclodextrin) enhances STARD3, and represses STARD1 expression. Agonists of Liver X receptors, peroxisome proliferator activated receptor-gamma and retinoic acid X receptors increase STARD1 expression, while hypocholesterolaemic agent, LY295427, reveals both STARD1 and STARD3 as putative SREBP-target genes. Pathophysiological 'foam cell' formation, induced by acetylated or oxidized LDL, significantly reduced both STARD1 and STARD3 gene expression. Differential regulation of STARD1 and D3 reflects their distinct roles in macrophage cholesterol metabolism, and may inform anti-atherogenic strategies.


Subject(s)
Carrier Proteins/metabolism , Cholesterol/metabolism , Foam Cells/metabolism , Gene Expression Regulation/physiology , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Biological Transport/drug effects , Biological Transport/physiology , Carrier Proteins/genetics , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Cholestanols/pharmacology , Cholesterol/genetics , DNA-Binding Proteins/agonists , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Foam Cells/cytology , Gene Expression Regulation/drug effects , Humans , Lipoproteins, LDL/genetics , Lipoproteins, LDL/metabolism , Liver X Receptors , Membrane Proteins/genetics , Orphan Nuclear Receptors , PPAR gamma/agonists , PPAR gamma/genetics , PPAR gamma/metabolism , Phosphoproteins/genetics , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Retinoid X Receptors/agonists , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/metabolism , beta-Cyclodextrins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...