Hostname: page-component-848d4c4894-xfwgj Total loading time: 0 Render date: 2024-06-27T04:28:42.094Z Has data issue: false hasContentIssue false

Mechanisms of the components of the metabolic syndrome that predispose to diabetes and atherosclerotic CVD

Published online by Cambridge University Press:  28 February 2007

Robert H. Eckel*
Affiliation:
Division of Endocrinology, Metabolism and Diabetes, University of Colorado at Denver & Health Sciences Center, PO Box 6511, MS8106, Aurora, Colorado 80045, USA
*
Corresponding author: Professor Robert H. Eckel, fax +1 303 724 3924, Robert.Eckel@UCHSC.edu
Rights & Permissions [Opens in a new window]

Abstract

The metabolic syndrome represents a summation of obesity-driven risk factors for atherosclerotic CVD and type 2 diabetes. Definitions of the syndrome vary but in general agree closely in identifying subjects. The relationships between the metabolic syndrome and atherosclerotic CVD and diabetes also vary, with relative risks of approximately 1·5–3·0 and approximately 3·0–5·0 respectively. Insulin resistance appears to explain much of the pathophysiology of the syndrome. Both increased fatty acid flux and an excess of circulating pro-inflammatory cytokines are likely mediators. With increased waist circumference, increases in fatty acid delivery to the liver result in higher rates of hepatic glucose production and increases in the secretion of apoB-containing lipoproteins. Concomitant changes in HDL ensue, including a replacement of the cholesterol content with TAG, an accelerated clearance from the plasma and thus a reduced number of HDL particles. Typically also present are increases in small dense LDL. Hypertension in part relates to the insulin resistance, but may involve other mechanisms. Impaired fasting glucose often relates to defects in insulin secretion in addition to insulin resistance, and probably more than any other component of the syndrome predicts the increased incidence of type 2 diabetes. Although not included in the diagnostic criteria, increases in pro-inflammatory cytokines and pro-thrombotic factors, in addition to decreases in plasma adiponectin, may also contribute to the increased incidence of atherosclerotic CVD and diabetes. In general, the greater the number of metabolic syndrome components, the greater the risk for these outcomes. The cytokines and pro-thrombotic factors also appear to contribute.

Type
Plenary Lecture
Copyright
Copyright © The Author 2007

Abbreviations:
CRP

C-reactive protein

LPL

lipoprotein lipase

The metabolic syndrome: definition

The metabolic syndrome is a recently defined set of criteria that has been prompted by the worldwide obesity epidemic. Although multiple definitions have been offered, to make the approach to the metabolic syndrome easier, perhaps the criteria proposed by the National Cholesterol Education Program Adult Treatment Program III Committee (Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults, 2001) and/or those of the International Diabetes Federation (Alberti et al. Reference Alberti, Zimmet and Shaw2005) should be emphasized (Table 1). The sensitivity for the presence of the syndrome may be greater with the International Diabetes Federation criteria because an expanded waist circumference must be present. A case in point is the higher prevalence of the syndrome in the National Health and Nutrition Examination Survey III population using the International Diabetes Federation v. National Cholesterol Education Program Adult Treatment Program III Committee definitions (Ford, Reference Ford2005).

Table 1. National Cholesterol Education Program Adult Treatment Program III Committee (Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults, 2001; NCEP:ATPIII 2001) and International Diabetes Federation (Alberti et al. Reference Alberti, Zimmet and Shaw2005; IDF) criteria for the metabolic syndrome

M, males; F, females.

* In this analysis, the following thresholds for waist circumference (cm) were used: M: white ≥94, African-American ≥94, Mexican-American ≥90; F: white ≥80, African-American ≥80, Mexican-American ≥80. For participants whose designation was ‘other race, including multiracial’ the thresholds (cm) that were once based on Europid cut-off points (M≥94, F≥80) and South Asian cut-off points (M≥90, F≥80) were used. For participants who were considered ‘other Hispanic’ the IDF thresholds for ethnic South and Central Americans were used.

Recent controversy has surrounded the diagnosis. In the latter half of 2005 a joint statement by the American Diabetes Association and the European Association for the Study of Diabetes (Kahn et al. Reference Kahn, Buse, Ferrannini and Stern2005) has raised a number of concerns about the scientific merit of the metabolic syndrome. Many of these points are valid and need further consideration, but responses to follow justify the continued importance of the syndrome to scientists and practitioners (Grundy et al. Reference Grundy, Cleeman, Daniels, Donato, Eckel and Franklin2005; Grundy, Reference Grundy2006). With time, consolidation of the definition, mechanisms, implications and therapeutics will ensue.

Metabolic syndrome: risk for atherosclerosis and type 2 diabetes mellitus

The metabolic syndrome confers risk for atherosclerotic CVD and type 2 diabetes. It is important to note that the selection criteria, end points and the interval for outcome evaluation used in the many trials have varied. Thus, broad sweeping conclusions related to the impact of the metabolic syndrome on any outcome need to be carefully considered.

Atherosclerotic CVD

The risk for new-onset CHD in patients with the metabolic syndrome in the absence of diabetes has varied substantially from population to population but averages between about 1·5-fold and 3-fold higher (Eckel et al. Reference Eckel, Grundy and Zimmet2005). However, the population-attributable risk for the development of CHD over a follow-up period of 8 years in middle-aged men and women with the metabolic syndrome in the Framingham Offspring Study was found to be 34% and 16% respectively (Rutter et al. Reference Rutter, Meigs, Sullivan, D'Agostino and Wilson2005). Use of the Framingham risk scoring system for patients without diabetes but with the metabolic syndrome enhances the predictability of CHD. Elevations in C-reactive protein (CRP) also influence this risk, and more so in patients with the metabolic syndrome and type 2 diabetes (Rutter et al. Reference Rutter, Meigs, Sullivan, D'Agostino and Wilson2005).

Although the metabolic syndrome is a substantial predictor of CHD and atherosclerotic CVD in general (which includes CHD and stroke), there is a paucity of information about the specific risk of stroke and peripheral vascular disease. In the absence of diabetes and previous atherosclerotic CVD the relative risk of ischaemic stroke in Finnish men with the metabolic syndrome was found to be approximately 2·5 (Kurl et al. Reference Kurl, Laukkanen, Niskanen, Laaksonen, Sivenius, Nyyssonen and Salonen2006). In the Framingham Offspring Study both the metabolic syndrome and diabetes were found to predict ischaemic stroke, with the population-attributable risk being greater for patients with the metabolic syndrome than for patients with diabetes alone (19% v. 7%), particularly in women (27% v. 5%; Najarian et al. Reference Najarian, Sullivan, Kannel, Wilson, D'Agostino and Wolf2006).

Type 2 diabetes

Overall, the risk for type 2 diabetes in patients with the metabolic syndrome is 3-fold–5-fold higher (Eckel et al. Reference Eckel, Grundy and Zimmet2005). In an 8-year follow-up of middle-aged men and women in the Framingham Offspring Study the population-attributable risk for developing type 2 diabetes was found to be 62% and 47% respectively (Wilson et al. Reference Wilson, D'Agostino, Parise, Sullivan and Meigs2005). Clearly, patients with impaired fasting glucose or impaired glucose tolerance have a high risk for type 2 diabetes; a risk that may be independent of other components of the metabolic syndrome. However, in the Insulin Resistance Atherosclerosis Study (Hanley et al. Reference Hanley, Karter, Williams, Festa, D'Agostino, Wagenknecht and Haffner2005) the definitions of the International Diabetes Federation (Alberti et al. Reference Alberti, Zimmet and Shaw2005) and the National Cholesterol Education Program Adult Treatment Program III Committee (Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults, 2001) were shown to predict type 2 diabetes equally well despite not requiring the use of oral glucose tolerance testing or measures of insulin resistance or microalbuminuria. The risk for new-onset type 2 diabetes may be as strong for the metabolic syndrome as for impaired glucose tolerance and may not require the presence of impaired fasting glucose as a predictor.

Mechanisms for CHD and type 2 diabetes mellitus underlying the metabolic syndrome

Insulin resistance

The most accepted and unifying hypothesis to describe the pathophysiology of the metabolic syndrome is insulin resistance (Eckel et al. Reference Eckel, Grundy and Zimmet2005). Insulin resistance has been most often defined using a glucocentric view, i.e. when a defect in insulin action results in fasting hyperinsulinaemia to maintain euglycaemia. Yet, even before fasting hyperinsulinaemia develops, postprandial hyperinsulinaemia exists. A series of biomarkers are available to assess the presence of insulin resistance, including fasting insulin, the homeostatic model assessment, pro-insulin and TAG:HDL-cholesterol, yet a number of limitations exist for each one. Although the modified intravenous tolerance test of Bergman (Boston et al. Reference Boston, Stefanovski, Moate, Sumner, Watanabe and Bergman2003) has proved useful in research settings, its practical application is lacking. The euglycaemic clamp continues to be the gold standard.

A major contributor to the development of insulin resistance is circulating fatty acids. Albumin-bound NEFA are derived predominantly from TAG stores in adipose tissue. Historically, the cAMP-dependent enzyme hormone-sensitive lipase was considered to be the lipolytic enzyme; however, recent evidence from murine models indicates that adipose TAG lipase may be more important (Haemmerle et al. Reference Haemmerle, Lass, Zimmermann, Gorkiewicz, Meyer and Rozman2006). Lipoprotein lipase (LPL) also provides fatty acids but through the lipolysis of circulating TAG-rich lipoproteins (Eckel, Reference Eckel1989). Insulin is important to both anti-lipolysis and the stimulation of LPL in adipose tissue. Importantly, the most sensitive pathway of insulin action is the inhibition of lipolysis (Jensen et al. Reference Jensen, Caruso, Heiling and Miles1989). Paracrine regulation of lipolysis in adipose tissue may also be an important contributor to the circulating NEFA burden. One such paracrine affecter is the cytokines. With increasing adipose tissue mass, infiltration of monocyte-derived macrophages occurs and inflammatory cytokines are released. This local effect promotes additional impairment in the anti-lipolytic effect of insulin, creating more lipolysis (Suganami et al. Reference Suganami, Nishida and Ogawa2005).

In muscle excessive fatty acids decrease insulin sensitivity (Fig. 1(a)). Increased fatty acid availability results in the accumulation of intramyocellular fatty acyl-CoA, which then unfavourably modifies a number of downstream pathways including: insulin signalling; insulin-(in)dependent glucose transport and phosphorylation; insulin-stimulated glycogen synthesis; insulin-stimulated oxidative phosphorylation (ATP synthesis); accumulation of TAG; expression of PPARγ coactivator-1 and PPARγ coactivator-1-controlled genes involved in mitochondrial biogenesis and oxidative phosphorylation and potentially the initiation of inflammatory processes by activation of protein kinase C and NF-κB; expression of matrix metalloproteinases (for review, see Roden, Reference Roden2005). This effect of fatty acids on muscle insulin sensitivity is more attributable to SFA than unsaturated fatty acids (Storlien et al. Reference Storlien, Higgins, Thomas, Brown, Wang, Huang and Else2000; Lee et al. Reference Lee, Pinnamaneni, Eo, Cho, Pyo, Kim, Sinclair, Febbraio and Watt2006).

Fig. 1. Pathophysiology of the metabolic syndrome (insulin resistance and NEFA), (a) NEFA are released in abundance from an expanded adipose tissue mass. In the liver NEFA result in an increased production of glucose and TAG and secretion of VLDL. Associated lipid and lipoprotein abnormalities include reductions in HDL-cholesterol and an increased density of LDL. NEFA also reduce insulin sensitivity in muscle by inhibiting insulin-mediated glucose uptake. Associated defects include a reduction in glucose partitioning to glycogen and increased lipid accumulation in TAG. Increases in circulating glucose, and to some extent NEFA, increase pancreatic insulin secretion, resulting in hyperinsulinaemia. Hyperinsulinaemia may result in enhanced sodium reabsorption and increased sympathetic nervous system (SNS) activity and contribute to the hypertension, as might increased circulating levels of NEFA. (b) Superimposed and contributory to the insulin resistance produced by excessive NEFA is the paracrine and endocrine effect of the pro-inflammatory state. Produced by a variety of cells in adipose tissue, including adipocytes and monocyte-derived macrophages, the enhanced secretion of IL-6 and TNFα, among others, results in more insulin resistance and lipolysis of adipose tissue TAG stores to circulating NEFA. IL-6 and other cytokines also are increased in the circulation and may enhance hepatic glucose production, the production of VLDL by the liver and insulin resistance in muscle. Cytokines and NEFA also increase the production of fibrinogen and plasminogen activator inhibitor-1 (PAI-1) by the liver that complements the overproduction of PAI-1 by adipose tissue. This action results in a pro-thrombotic state. Higher circulating levels of cytokines also stimulate the hepatic production of C-reactive protein (CRP). Reductions in the production of the anti-inflammatory and insulin-sensitizing cytokine adiponectin are also associated with the metabolic syndrome and may contribute to the pathophysiology of the syndrome. , Increased; , reduced; , effects relatively reduced. (From Eckel et al. Reference Eckel, Grundy and Zimmet2005, with permission from Elsevier.)

In human subjects the infusion of a lipid emulsion with the simultaneous administration of intravenous heparin to release lipases and then lipoprotein-TAG fatty acids into the plasma compartment results in insulin resistance (Boden et al. Reference Boden, Cheung, Stein, Kresge and Mozzoli2002), which leads to defects in insulin action not only in skeletal muscle but also in the liver (Boden, Reference Boden2003). In the liver of high-fat-fed rats insulin resistance can be attributed to a defect in insulin-stimulated insulin receptor substrate-1 and insulin receptor substrate-2 tyrosine phosphorylation. These changes have been associated with activation of protein kinase C-ε and c-Jun N-terminal kinase-1 (Samuel et al. Reference Samuel, Liu, Qu, Elder, Bilz, Befroy, Romanelli and Shulman2004). Yet, while circulating fatty acids increase hepatic glucose production and diminish the inhibition of glucose production by insulin (Boden & Shulman, Reference Boden and Shulman2002), lipogenesis as a result of the stimulatory effects of fatty acids and insulin on sterol response element-binding protein-1c continues (Shimomura et al. Reference Shimomura, Bashmakov, Ikemoto, Horton, Brown and Goldstein1999). Very recent evidence has demonstrated that the pharmacological inhibition of the lipid biosynthetic enzymes acetyl-CoA carboxylase-1 and -2 increases fat oxidation and reduces hepatic steatosis and insulin resistance (Savage et al. Reference Savage, Choi, Samuel, Liu, Zhang and Wang2006).

More detailed studies in human subjects have examined the cellular and molecular basis of insulin resistance in more detail (Petersen et al. Reference Petersen, Befroy, Dufour, Dziura, Ariyan, Rothman, DiPietro, Cline and Shulman2003, Reference Petersen, Dufour, Befroy, Garcia and Shulman2004; Barbato et al. Reference Barbato, Cappuccio, Folkerd, Strazzullo, Sampson, Cook and Alberti2004), and in insulin-resistant subjects with obesity and/or type 2 diabetes and in the elderly a defect in mitochondrial oxidative phosphorylation has been identified. More recently, a common variant in mitochondrial DNA at bp 16189 (T→C transition) has been associated with the metabolic syndrome in Chinese adults (Weng et al. Reference Weng, Liou, Lin, Wei, Lee and Eng2005). Although the mitochondrial ribosomal RNA and transfer RNA are encoded by mitochondrial DNA in human subjects, all proteins involved in mitochondrial translation are encoded by nuclear genes. Moreover, increasingly nuclear genes associated with the metabolic syndrome are being recognized (Matsunaga & Muramatsu, Reference Matsunaga and Muramatsu2005).

Murine models of the metabolic syndrome are becoming more evident. In mice made deficient in the endoplasmic reticulum X-box-binding protein-1 hyperactivation of c-Jun N-terminal kinase increases serine phosphorylation of insulin receptor substrate-1 and insulin resistance (Ozcan et al. Reference Ozcan, Cao, Yilmaz, Lee, Iwakoshi, Ozdelen, Tuncman, Gorgun, Glimcher and Hotamisligil2004). Among other models addressing the mechanism of the metabolic syndrome in rodents are mice lacking the fatty acid-binding proteins aP2 and mal1. These mice exhibit a striking phenotype with strong protection from diet-induced obesity, insulin resistance, type 2 diabetes and fatty liver disease (Maeda et al. Reference Maeda, Cao, Kono, Gorgun, Furuhashi and Uysal2005).

A new report (Shuldiner & McLenithan, Reference Shuldiner and McLenithan2004) indicates that PPARγ coactivator-1α and -1β mRNA levels decrease with age in individuals with a genetic variant in PPARγ coactivator-1α, and that these decreases correlate with alterations in whole-body glucose and fatty acid oxidation. These data provide insights into how aging modifies genetic susceptibility to alterations in oxidative phosphorylation and type 2 diabetes.

Oxidative stress has also been viewed as a cellular mechanism for insulin resistance. One enzyme that initiates base excision repair of ring-fragmented purines and some pyrimidines is neil1. In the absence of oxidative stress neil1-knock-out (neil1−/−) and heterozygotic (neil1+/−) mice develop severe obesity, dyslipidaemia and hepatic steatosis with more modest hyperinsulinaemia (Vartanian et al. Reference Vartanian, Lowell, Minko, Wood, Ceci, George, Ballinger, Corless, McCullough and Lloyd2006).

Thus, with time more basic mechanisms of insulin resistance are being discovered. Presumably, these biochemical changes in insulin-mediated signalling pathways result in decreases in insulin-mediated glucose transport and metabolism in the metabolic syndrome as well.

Obesity and increased waist circumference

Although the first description of the metabolic syndrome occurred in the early 20th century (Kylin, Reference Kylin1923), the worldwide obesity epidemic has been the most important driving force for the much more recent recognition of the syndrome. Despite the importance of obesity in the model, it is relevant to remember that patients who are normal weight may also be insulin resistant (Ruderman et al. Reference Ruderman, Chisholm, Pi-Sunyer and Schneider1998).

For the International Diabetes Federation (Alberti et al. Reference Alberti, Zimmet and Shaw2005) definition of the metabolic syndrome the waist circumference as defined must be increased (Table 1). It is important to distinguish between a large waist circumference that is a result of increases in subcutaneous adipose tissue from that in which the visceral depot is expanded. This distinction can be made using computerized tomography or MRI (Lee et al. Reference Lee, Janssen and Ross2004). With increases in intra-abdominal or visceral adipose tissue a higher rate of flux of adipose tissue-derived NEFA to the liver would be expected, whereas increases in abdominal subcutaneous fat would release lipolysis products into the systemic circulation and avoid more direct effects on hepatic metabolism, i.e. glucose production, lipid synthesis, lipoprotein secretion and the secretion of pro-thrombotic proteins such as fibrinogen and plasminogen activator inhibitor-1 (Aubert et al. Reference Aubert, Frere, Aillaud, Morange, Juhan-Vague and Alessi2003).

Despite these potential differences in mechanisms related to excessive abdominal adipose tissue distribution, the clinical diagnosis of the metabolic syndrome does not distinguish between increases in subcutaneous and visceral fat. Yet, perhaps by a mechanism related to NEFA flux and metabolism, the predominance of visceral v. subcutaneous adipose tissue with increasing waist circumference in Asians and Asian Indians (Bajaj & Banerji, Reference Bajaj and Banerji2004) renders the relative prevalence of the syndrome higher than that in African-American men in whom subcutaneous fat predominates (Tanaka et al. Reference Tanaka, Horimai and Katsukawa2003). However, there is evidence that the elevated postprandial NEFA release in upper-body obese women originates from the non-splanchnic upper body fat, and not from the visceral depot (Guo et al. Reference Guo, Hensrud, Johnson and Jensen1999). These results suggest that visceral fat may be a marker for, but not the source of, excess postprandial fatty acids in obesity.

The amount of epicardial fat assessed by echocardiography could be a simple and practical tool for cardiovascular risk stratification in clinical practice and research (Iacobellis et al. Reference Iacobellis, Corradi and Sharma2005). Epicardial fat is a metabolically-active organ that generates various bioactive molecules, some of which might markedly affect cardiac function. This small ‘visceral fat’ depot is now recognized as a rich source of NEFA and a number of bioactive molecules, such as inflammatory cytokines and adiponectin. Presently, validation and more clinical significance are needed.

Murine examples of the impact of body fat distribution on insulin resistance and the metabolic syndrome include mice with over-expression of 11-hydroxysteroid dehydrogenase 1 that have elevated intra-adipose corticosterone levels but normal circulating corticosterone levels (Masuzaki et al. Reference Masuzaki, Paterson, Shinyama, Morton, Mullins, Seckl and Flier2001). These mice exhibit central obesity, hypertension, impaired glucose tolerance and hypertriacylglycerolaemia. Conversely, mice that fail to express 11-hydroxysteroid dehydrogenase 1 are resistant to the development of metabolic syndrome (Kotelevtsev et al. Reference Kotelevtsev, Holmes, Burchell, Houston, Schmoll, Jamieson, Best, Brown, Edwards, Seckl and Mullins1997).

More recent studies have shown that the ectopic expression of very low levels of uncoupling protein 1 in epididymal fat reverses both insulin and leptin resistance. Uncoupling protein 1 expression in epididymal fat improves glucose tolerance and decreases food intake in both diet-induced and genetically-obese mouse models (Yamada et al. Reference Yamada, Katagiri, Ishigaki, Ogihara, Imai and Uno2006). Moreover, in wild-type mice local-nerve dissection in the epididymis or pharmacological afferent blockade blunts the decrease in food intake, suggesting that afferent-nerve signals from intra-abdominal adipose tissue regulate food intake.

In the setting of partial or complete lipoatrophy insulin resistance and the metabolic syndrome typically co-exist (Garg & Misra, Reference Garg and Misra2004). Evidence from these less-common disorders does support a genetic basis of the syndrome, including single-gene defects in PPAR-λ, lamin A/C, 1-acylglycerol-3-phosphate, O-acyltransferase, seipin (Hegele, Reference Hegele2003), the β-2 adrenergic receptor (Dallongeville et al. Reference Dallongeville, Helbecque, Cottel, Amouyel and Meirhaeghe2003) and adiponectin (Fumeron et al. Reference Fumeron, Aubert, Siddiq, Betoulle, Pean and Hadjadj2004).

Dyslipidaemia

With increased NEFA flux to the liver, increased production of apoB-containing TAG-rich VLDL occurs (Lewis et al. Reference Lewis, Uffelman, Szeto, Weller and Steiner1995). The effect of insulin on this process is somewhat complex. In the setting of insulin resistance the increased flux of NEFA to the liver increases hepatic TAG synthesis; however, under physiological conditions insulin inhibits rather than increases the secretion of VLDL into the systemic circulation (Lewis & Steiner, Reference Lewis and Steiner1996). This inhibitory effect in part is an effect of insulin on the degradation of apoB (Taghibiglou et al. Reference Taghibiglou, Rashid-Kolvear, Van Iderstine, Le Tien, Fantus, Lewis and Adeli2002). Yet, insulin is also lipogenic, increasing the transcription and enzyme activity of many genes that relate to TAG biosynthesis (Foufelle & Ferre, Reference Foufelle and Ferre2002). Not completely addressed is whether or not this pathway remains operational in the setting of systemic insulin resistance.

In addition, insulin resistance may also reduce the level of LPL in peripheral tissues, i.e. in adipose tissue more than in muscle (Eckel et al. Reference Eckel, Yost and Jensen1995). This alteration in LPL, however, appears to contribute less to the hypertriacylglycerolaemia. However, mutations in the LPL gene affect LPL expression and are associated with both dyslipidaemia and insulin resistance (Holzl et al. Reference Holzl, Iglseder, Sandhofer, Malaimare, Lang, Paulweber and Sandhofer2002; Ma et al. Reference Ma, Thomas, Ng, Critchley, Chan and Tomlinson2003; Goodarzi et al. Reference Goodarzi, Guo, Taylor, Quinones, Saad, Yang, Hsueh and Rotter2004). Recent evidence even suggests that levels of LPL mass in pre-heparin plasma may be a quantitative indicator of whole-body insulin resistance (Miyashita & Shirai, Reference Miyashita and Shirai2005).

The other major lipoprotein disturbance in the metabolic syndrome is a reduction in HDL-cholesterol. This reduction is a consequence of changes in HDL composition and metabolism. In the presence of hypertriacylglycerolaemia a decrease in the cholesterol content of HDL is a consequence of decreases in the cholesteryl ester content of the lipoprotein core, with variable increases in TAG making the particle small and dense, a function in part of cholesteryl ester transfer protein (Murakami et al. Reference Murakami, Michelagnoli, Longhi, Gianfranceschi, Pazzucconi, Calabresi, Sirtori and Franceschini1995). Although HDL typically possess many features that appear to contribute to the association between increased HDL-cholesterol and protection from atherosclerosis, HDL may be modified to become pro-inflammatory (Ansell et al. Reference Ansell, Watson, Fogelman, Navab and Fonarow2005). The ability of HDL to inhibit or paradoxically to enhance vascular inflammation, lipid oxidation, plaque growth and thrombosis reflects changes in specific enzyme and protein components, e.g. increases in serum amyloid A, an acute-phase reactant and pro-inflammatory molecule (Kontush & Chapman, Reference Kontush and Chapman2006). This change in lipoprotein composition also results in an increased clearance of HDL from the circulation (Brinton et al. Reference Brinton, Eisenberg and Breslow1991; Chan et al. Reference Chan, Barrett and Watts2006). Thus, reductions in plasma levels of apoA-1 (a reflection of HDL particle number) are seen.

The relationship between these changes in HDL and insulin resistance is probably indirect, occurring in concert with the changes in TAG-rich lipoprotein metabolism. Although small dense HDL particles are typically better lipid acceptors, the formation of HDL that are small and dense in the setting of the dyslipidaemia of the metabolic syndrome may attenuate anti-atherogenic properties (Kontush & Chapman, Reference Kontush and Chapman2006). Thus, low circulating levels of HDL-cholesterol might, therefore, be associated with small HDL particles of abnormal structure and composition with defective functionality.

In addition to HDL, LDL are also modified in composition in a similar way. In fact, with fasting serum TAG of >2·0 mm almost all patients have a predominance of small dense LDL (de Graaf et al. Reference de Graaf, Hendriks, Demacker and Stalenhoef1993; Manzato et al. Reference Manzato, Zambon, Zambon, Cortella, Sartore and Crepaldi1993). This change in LDL composition is attributable to relative depletion of non-esterified cholesterol, esterified cholesterol and phospholipid with either no change or an increase in LDL-TAG (Halle et al. Reference Halle, Berg, Baumstark, Konig, Huonker and Keul1999; Kwiterovich, Reference Kwiterovich2002). Small dense LDL may be more atherogenic because they are more toxic to the endothelium, more able to transit through the endothelial basement membrane, more likely to adhere to arterial wall glycosaminoglycans, have increased susceptibility to oxidation and/or are more selectively bound to scavenger receptors on monocyte-derived macrophages (Packard, Reference Packard1996; Krauss, Reference Krauss1995). However, the direct relationship between small dense LDL and the natural history of atherosclerotic CVD is not entirely accepted (Lada & Rudel, Reference Lada and Rudel2004; Jungner et al. Reference Jungner, Sniderman, Furberg, Aastveit, Holme and Walldius2006). In some studies this alteration in LDL composition is an independent risk factor for CVD (Zambon et al. Reference Zambon, Hokanson, Brown and Brunzell1999). Recently, the effects of gemfibrozil on NMR-measured LDL and HDL particle subclasses, not reflected by conventional lipoprotein-cholesterol measures, has helped to explain the beneficial effect of the fibrate in patients with low HDL-cholesterol and CHD. However, most often the association between particle composition and atherosclerotic CVD events is not independent, but related to the concomitant changes in other lipoproteins and other risk factors (Sacks & Campos, Reference Sacks and Campos2003). It is possible that when circulating levels of LDL-cholesterol are lower, measurements of LDL particle number may ultimately prove to be more useful.

Glucose intolerance

The defects in insulin action in glucose metabolism include deficiencies in the ability of the hormone to suppress glucose production by the liver and kidney and mediate glucose uptake and metabolism in insulin-sensitive tissues, i.e. muscle and adipose tissue. The relationship between impaired fasting glucose or glucose tolerance and insulin resistance is well supported by human studies (Abdul-Ghani et al. Reference Abdul-Ghani, Tripathy and DeFronzo2006). To compensate for defects in insulin action insulin secretion and/or clearance must be modified to sustain euglycaemia. On failure of this compensation defects in insulin secretion rather than insulin clearance predominate.

Insulin resistance in pancreatic islet β-cells implies that the signals that generate glucose-dependent insulin secretion have been adversely modified, and fatty acids are prime candidates. Although NEFA can stimulate insulin secretion, increasing and prolonged exposure to excessive levels results in decreases in insulin secretion (Lee et al. Reference Lee, Hirose, Ohneda, Johnson, McGarry and Unger1994). The mechanism for this alteration has been attributed to lipotoxicity through a number of potential different mechanisms (Shimabukuro et al. Reference Shimabukuro, Zhou, Levi and Unger1998; Yaney & Corkey, Reference Yaney and Corkey2003; Boucher et al. Reference Boucher, Lu, Burgess, Telemaque-Potts, Jensen, Mulder, Wang, Unger, Sherry and Newgard2004; Joseph et al. Reference Joseph, Koshkin, Saleh, Sivitz, Zhang, Lowell, Chan and Wheeler2004).

Insulin also feeds back on its own secretion. The importance of this feedback relates to experiments in rodents in which the insulin receptor is tissue-specifically deleted. When the insulin receptor is deleted in skeletal muscle hyperglycaemia does not result (Bruning et al. Reference Bruning, Michael, Winnay, Hayashi, Horsch, Accili, Goodyear and Kahn1998); however, the β-cell-specific knock-out of the insulin receptor produces progressive glucose intolerance and diabetes (Kulkarni et al. Reference Kulkarni, Bruning, Winnay, Postic, Magnuson and Kahn1999). In human subjects with genetic predispositions to develop diabetes the presumed stress of the insulin-resistant environment on β-cell function produces glucose intolerance and ultimately the higher risk of diabetes.

Hypertension

The relationship between insulin resistance and hypertension is well established (Ferrannini et al. Reference Ferrannini, Buzzigoli, Bonadonna, Giorico, Oleggini, Graziadei, Pedrinelli, Brandi and Bevilacqua1987), and relates to several different mechanisms. Insulin is a vasodilator when given intravenously to normal-weight subjects (Steinberg et al. Reference Steinberg, Brechtel, Johnson, Fineberg and Baron1994), with secondary effects on Na re-absorption in the kidney (DeFronzo et al. Reference DeFronzo, Cooke, Andres, Faloona and Davis1975). Of interest, Na re-absorption is increased in Caucasians but not in Africans or Asians with the metabolic syndrome (Barbato et al. Reference Barbato, Cappuccio, Folkerd, Strazzullo, Sampson, Cook and Alberti2004). In insulin-resistant subjects the vasodilatory effect of insulin may be lost (Tooke & Hannemann, Reference Tooke and Hannemann2000), but the renal effect on Na re-absorption preserved (Kuroda et al. Reference Kuroda, Uzu, Fujii, Nishimura, Nakamura, Inenaga and Kimura1999). Insulin also increases the activity of the sympathetic nervous system (Anderson et al. Reference Anderson, Hoffman, Balon, Sinkey and Mark1991), an effect that may also be preserved in the setting of the insulin resistance (Egan, Reference Egan2003). In addition, fatty acids themselves can produce vasoconstriction (Tripathy et al. Reference Tripathy, Mohanty, Dhindsa, Syed, Ghanim, Aljada and Dandona2003).

The spontaneous-hypertensive rat is a widely-studied model of hypertension that exhibits metabolic abnormalities that share features with the human metabolic syndrome. Genetic-linkage studies have revealed a defective CD36 gene encoding a membrane fatty acid transporter and hyperinsulinaemia in the spontaneous-hypertensive rat. However, there is no unifying mechanism that can explain these phenotypes as a consequence of a defective CD36 gene (Tanaka et al. Reference Tanaka, Sohmiya, Kono, Terasaki, Horie, Ohkaru, Muramatsu, Takai, Miyazaki and Kitaura2006). Of interest, the hearts of CD36-defective spontaneous-hypertensive rats exhibit uncoupling of glucose oxidation from its uptake and enhanced protein O-linked N-acetylglucosaminylation, suggesting increased glucose shunt through the hexose monophosphate pathway. Thiamine repletion in the CD36-defective spontaneous-hypertensive rat results in correction of the uncoupling of glucose oxidation to glucose uptake, reduced protein O-linked N-acetylglucosaminylation and the expression of mRNA involved in the hexose monophosphate shunt, renin–angiotensin system and adipokines in epididymal adipose tissue, and reduced hypertension and hyperinsulinaemia.

Subjects with CD36 mutations have also been identified (Yamashita et al. Reference Yamashita, Hirano, Kuwasako, Janabi, Toyama, Ishigami and Sakai2006). In addition to mild hypertension and dyslipidaemia, whole-body insulin-mediated body glucose uptake is reduced in CD36-deficient patients, indicating the presence of insulin resistance. The frequency of CD36 deficiency is also higher in patients with CHD than in control subjects.

Yet, despite the plethora of potential mechanisms by which insulin resistance might cause hypertension, with the current level of understanding insulin resistance appears to contribute only moderately to the increased prevalence of hypertension in the metabolic syndrome (Hanley et al. Reference Hanley, Karter, Festa, D'Agostino, Wagenknecht, Savage, Tracy, Saad and Haffner2002).

Other manifestations

Insulin resistance is accompanied by many other alterations that are not included in the diagnostic criteria for the metabolic syndrome (Table 2). Increases in apoB and apoC-III, uric acid, pro-thrombotic factors (fibrinogen, plasminogen activator inhibitor-1), serum viscosity, asymmetric dimethylarginine, homocysteine, leucocyte count, pro-inflammatory cytokines, the presence of microalbuminuria, non-alcoholic fatty liver disease and/or non-alcoholic steatohepatitis, polycystic ovarian disease, obstructive sleep apnea and cholesterol gallstones and a decrease in adiponectin are all associated with insulin resistance.

Table 2. Other alterations that accompany insulin resistance that are not included in the diagnostic criteria for the metabolic syndrome

↑, Increased; ↓, decreased.

Cigarette smoking (Eliasson et al. Reference Eliasson, Attvall, Taskinen and Smith1994) and a sedentary lifestyle (Lakka et al. Reference Lakka, Laaksonen, Lakka, Mannikko, Niskanen, Rauramaa and Salonen2003) can also produce many of the major criteria of the syndrome and beyond. Increases in apoB, apoC-III (Onat et al. Reference Onat, Hergenc, Sansoy, Fobker, Ceyhan, Toprak and Assmann2003) and non-alcoholic steatohepatitis (Medina et al. Reference Medina, Fernandez-Salazar, Garcia-Buey and Moreno-Otero2004) are tied to the effects of fatty acids on VLDL production by the liver, and in the case of apoB and apoC-III provide evidence of an increased number of pro-atherogenic particles in the circulation. Hyperuricaemia reflects defects in insulin action on the renal tubular re-absorption of uric acid (Facchini et al. Reference Facchini, Chen, Hollenbeck and Reaven1991), whereas the increase in asymmetric dimethylarginine, an endogenous inhibitor of NO synthase, relates to endothelial dysfunction (Egan, Reference Egan2003). An extended form of endothelial pathophysiology in insulin-resistant states could be microalbuminuria (Rowley et al. Reference Rowley, O'Dea and Best2003). Insulin resistance is also associated with gall bladder disease (Nervi et al. Reference Nervi, Miquel, Alvarez, Ferreccio, Garcia-Zattera, Gonzalez, Perez-Ayuso, Rigotti and Villarroel2006). Bile is typically more lithogenic when it contains a higher cholesterol:bile acids and phospholipids. The primary reason for lithogenic bile in obese patients is an increase in total body cholesterol synthesis, possibly secondary to the overload of tissues with fatty acids that are precursors for cholesterol synthesis (Grundy, Reference Grundy2004).

Pro-inflammatory cytokines

The association between the metabolic syndrome and inflammation is well documented (Sutherland et al. Reference Sutherland, McKinnley and Eckel2004). The increases in pro-inflammatory cytokines including IL-6, IL-18, resistin, TNFα and CRP (Fernandez-Real & Ricart, Reference Fernandez-Real and Ricart2003; Fig. 1(b)) reflect overproduction by the expanded adipose tissue mass (Trayhurn & Wood, Reference Trayhurn and Wood2004). Recent evidence (Weisberg et al. Reference Weisberg, McCann, Desai, Rosenbaum, Leibel and Ferrante2003; Xu et al. Reference Xu, Barnes, Yang, Tan, Yang, Chou, Sole, Nichols, Ross, Tartaglia and Chen2003) suggests that in adipose tissue monocyte-derived macrophages reside and may be at least in part the source of the generation of pro-inflammatory cytokines locally and in the plasma. Insulin resistance in the liver, muscle and adipose tissue is not only associated with the abundance of pro-inflammatory cytokines (and relative deficiency of the anti-inflammatory cytokine adiponectin), but a direct result of this burden (Neuschwander-Tetri & Caldwell, Reference Neuschwander-Tetri and Caldwell2003). It remains unclear, however, how much of the insulin resistance related to the adipose tissue content of macrophages is paracrine v. endocrine.

As a general index of inflammation, CRP levels vary with ethnicity, level of fitness, and gender (Chambers et al. Reference Chambers, Eda, Bassett, Karim, Thompson, Gallimore, Pepys and Kooner2001; LaMonte et al. Reference LaMonte, Durstine, Yanowitz, Lim, DuBose, Davis and Ainsworth2002; Patel et al. Reference Patel, Srinivasan, Xu, Li, Chen and Berenson2006). For instance, CRP concentrations are higher in healthy Indian Asians than in European whites and relate to greater central obesity and insulin resistance in Indian Asians (Chambers et al. Reference Chambers, Eda, Bassett, Karim, Thompson, Gallimore, Pepys and Kooner2001). In the Bogalusa Heart Study plasma CRP levels were found to show a significant race (black>Caucasian; P=0·01) and gender (female>male; P=0·0001) difference (Patel et al. Reference Patel, Srinivasan, Xu, Li, Chen and Berenson2006). At present, it remains unclear whether these differences in CRP when adjusted for other covariates will relate to different rates of development of diabetes and/or atherosclerotic CVD. Of interest, a state of low-grade systemic inflammation evidenced by higher levels of CRP has been found in subjects with a normal BMI who show subclinical insulin resistance but no other metabolic abnormalities (Bo et al. Reference Bo, Gambino, Uberti, Mangiameli, Colosso, Repetti, Gentile, Cassader and Pagano2005).

Within the setting of the metabolic syndrome increases in inflammatory markers do affect CHD mortality (Coppola et al. Reference Coppola, Corrado, Muratori, Tantillo, Vitale, Lo and Novo2006; Langenberg et al. Reference Langenberg, Bergstrom, Scheidt-Nave, Pfeilschifter and Barrett-Connor2006; Linnemann et al. Reference Linnemann, Voigt, Nobel and Janka2006). Increases in CRP also appear to impact on the incidence of peripheral vascular disease in patients with diabetes and the metabolic syndrome (Vu et al. Reference Vu, Vu, Pio, Malik, Franklin, Chen and Wong2005). In addition, elevated CRP concentrations are a predictor of diabetes in the general Japanese population, an effect independent of obesity and insulin resistance (Doi et al. Reference Doi, Kiyohara, Kubo, Ninomiya, Wakugawa, Yonemoto, Iwase and Iida2005). A similar observation has been reported in west of Scotland men (Sattar et al. Reference Sattar, Scherbakova, Ford, O'Reilly, Stanley, Forrest, Macfarlane, Packard, Cobbe and Shepherd2004). When the risks for atherosclerotic CVD and diabetes are considered sufficient data appear to have been accumulated to modify the definition of the metabolic syndrome to include a measure of inflammation (Haffner, Reference Haffner2006).

Visfatin (also known as pre-B-cell colony-enhancing factor) is a cytokine that is highly expressed in visceral fat and its blood levels correlate with obesity (Stephens & Vidal-Puig, Reference Stephens and Vidal-Puig2006). Circulating visfatin concentrations are increased by hyperglycaemia. This effect is suppressed by exogenous hyperinsulinaemia or somatostatin infusion. Glucose signalling for visfatin release in adipocytes involves the phosphatidylinositol 3-kinase/Akt pathway (Haider et al. Reference Haider, Schaller, Kapiotis, Maier, Luger and Wolzt2006). Additional insight into mechanisms and the relationship between this cytokine and atherosclerotic CVD and diabetes is needed.

A large amount of information indicates that inflammation may be involved in the initiation as well as the development of hypertension. Evidence from animal models as well as patients have indicated that hypertension may exert pro-inflammatory actions through the increased expression of several mediators, including leucocyte adhesion molecules, chemokines, growth factors, heat-shock proteins, endothelin-1, and angiotensin. This association between inflammation and hypertension relates to previous documentation that endothelial dysfunction and increased serum levels of CRP are present in patients with hypertension (Li et al. Reference Li, Fang and Hui2005).

Elevated levels of the protease inhibitor plasminogen activator inhibitor-1 are well known molecular markers of the metabolic syndrome. However, it has been proposed that plasminogen activator inhibitor-1 could also act as an aetiological factor in the development of metabolic syndrome and its sequelae. This effect could be a result of plasminogen activator inhibitor-1 inhibition of the activity of members of the proprotein convertase class of serine proteases (Griffiths & Grainger, Reference Griffiths and Grainger2006).

Adiponectin

Adiponectin is an anti-inflammatory cytokine that is produced exclusively by adipocytes. Adiponectin has been shown to both enhance insulin sensitivity and inhibit many steps in the inflammatory process (Nawrocki & Scherer, Reference Nawrocki and Scherer2004). In the liver adiponectin inhibits both the expression of hepatic gluconeogenic enzymes and the rate of endogenous glucose production (Combs et al. Reference Combs, Berg, Obici, Scherer and Rossetti2001). In muscle adiponectin increases glucose transport and enhances fatty acid oxidation, effects that result in part from the activation of AMP kinase (Xu et al. Reference Xu, Barnes, Yang, Tan, Yang, Chou, Sole, Nichols, Ross, Tartaglia and Chen2003). Adiponectin and the two receptors of adiponectin adipoR1 and adipoR2 are all associated with body composition, insulin sensitivity and metabolic variables (Bluher et al. Reference Bluher, Bullen, Lee, Kralisch, Fasshauer, Kloting, Niebauer, Schon, Williams and Mantzoros2006).

Recent evidence suggests that the high-molecular-weight form of adiponectin has better predictive power in relation to insulin resistance and the metabolic syndrome than plasma total adiponectin level (Hara et al. Reference Hara, Horikoshi, Yamauchi, Yago, Miyazaki, Ebinuma, Imai, Nagai and Kadowaki2006; Lara-Castro et al. Reference Lara-Castro, Luo, Wallace, Klein and Garvey2006). Moreover, adiponectin relates strongly to levels of HDL-cholesterol. The relationship appears to be a consequence of a strong negative correlation between adiponectin and the fractional clearance rate of apoA-1 (Verges et al. Reference Verges, Petit, Duvillard, Dautin, Florentin, Galland and Gambert2006). Interestingly, there are reports that link low levels of adiponectin to myocardial infarction (Pischon et al. Reference Pischon, Girman, Hotamisligil, Rifai, Hu and Rimm2004) and to the progression of subclinical CHD (Maahs et al. Reference Maahs, Ogden, Kinney, Wadwa, Snell-Gergeon, Dabelea, Hokanson, Ehrlich, Eckel and Rewers2005).

In mice decreased circulating levels of adiponectin appear to be important in producing metabolic changes consistent with the metabolic syndrome (van Puijenbroek et al. Reference van Puijenbroek, Du Buf-Vereijken, Spooren and van Doormaal1996; Yamauchi et al. Reference Yamauchi, Hara, Kubota, Terauchi, Tobe, Froguel, Nagai and Kadowaki2003; Ohashi et al. Reference Ohashi, Kihara, Ouchi, Kumada, Fujita and Hiuge2006). Reductions in adiponectin are also apparent in human subjects with the metabolic syndrome (Bergman et al. Reference Bergman, Murphy, Burke, Zhao, Valesky and Liu2004; Matsuzawa et al. Reference Matsuzawa, Funahashi, Kihara and Shimomura2004). Additional properties of adiponectin related to the metabolic syndrome have recently emerged. Adiponectin-knock-out mice develop hypertension when maintained on a high-salt diet, but in the absence of insulin resistance (Ohashi et al. Reference Ohashi, Kihara, Ouchi, Kumada, Fujita and Hiuge2006). The hypertension of adiponectin-knock-out mice is associated with reduced mRNA levels of endothelial NO synthase and PGI2 synthase in aorta and low levels of endothelial NO synthase metabolites and PGI2 synthase in plasma. Moreover, adiponectin therapy lowers the elevated blood pressure and corrects the altered mRNA to normal. In addition, adiponectin-induced cyclooxygenase-2-dependent synthesis of PGE2 in cardiac cells and cyclooxygenase-2 inhibition reverses the inhibitory effects of adiponectin on TNFα production and infarct size (Shibata et al. Reference Shibata, Sato, Pimentel, Takemura, Kihara, Ohashi, Funahashi, Ouchi and Walsh2005). Overall, adiponectin is thought to be cardio-protective (Ouchi et al. Reference Ouchi, Shibata and Walsh2006). Presently, the relative contribution of the deficiency in adiponectin v. the overabundance of the pro-inflammatory cytokines remains unclear.

Beyond insulin resistance

An alternative concept suggested by Unger (Reference Unger2003) to explain the metabolic syndrome is leptin resistance. In general, conditions in which leptin deficiency or resistance are present are associated with TAG accumulation in non-adipose organs, e.g. liver, muscle and the islets. This pathophysiology could relate to the absence of down-regulation of sterol response element-binding protein-1c by leptin (Kakuma et al. Reference Kakuma, Lee, Higa, Wang, Pan, Shimomura and Unger2000) and/or the inability of leptin to activate AMP kinase in muscle (Minokoshi & Kahn, Reference Minokoshi and Kahn2003). Leptin also appears to decrease insulin secretion (Cases et al. Reference Cases, Gabriely, Ma, Yang, Michaeli, Fleischer, Rossetti and Barzilai2001; Sung et al. Reference Sung, Lee, Jung, Kim, Cheon, Yang and Rhee2005), a result secondary to suppressor of cytokine signalling-3-mediated transcriptional inhibition of pre-proinsulin gene expression (Laubner et al. Reference Laubner, Kieffer, Lam, Niu, Jakob and Seufert2005). Leptin resistance could relate to the hyperinsulinaemia that develops in the setting of the metabolic syndrome before defects in insulin secretion lead to the development of diabetes (Seufert, Reference Seufert2004).

In addition to linking adiposity and central nervous circuits to reduce appetite and enhance energy expenditure, leptin has been shown to increase overall sympathetic nerve activity, facilitate glucose utilization and improve insulin sensitivity. In addition, leptin is capable of regulating cardiac and vascular contractility through a local NO-dependent mechanism (Ren, Reference Ren2004). The recent discovery that leptin and insulin share many of the same signalling pathways (Xu et al. Reference Xu, Kaelin, Takeda, Akira, Schwartz and Barsh2005) suggests that both peptides could share responsibility for the metabolic syndrome.

Summary

Nearly all the criteria included in the metabolic syndrome relate to or are a cause of insulin resistance and confer increased risk for atherosclerotic CVD and diabetes. Moreover, most of the components that relate to the syndrome but are not included in the definition, e.g. pro-inflammatory cytokines, also relate to insulin resistance and atherosclerotic CVD and diabetes risk. With time, the syndrome will hopefully be clarified in definition and mechanisms so that the relationship with atherosclerotic CVD and diabetes will result in targeting either the syndrome or the individual components to reduce risk. At present, an emphasis on lifestyle is most appropriate to address the syndrome; in addition, steps should be taken to modify risk factors.

References

Abdul-Ghani, MA, Tripathy, D & DeFronzo, RA (2006) Contributions of beta-cell dysfunction and insulin resistance to the pathogenesis of impaired glucose tolerance and impaired fasting glucose. Diabetes Care 29, 11301139.Google Scholar
Alberti, KG, Zimmet, P & Shaw, J (2005) The metabolic syndrome – a new worldwide definition. Lancet 366, 10591062.Google Scholar
Anderson, EA, Hoffman, RP, Balon, TW, Sinkey, CA & Mark, AL (1991) Hyperinsulinemia produces both sympathetic neural activation and vasodilation in normal humans. Journal of Clinical Investigation 87, 22462252.Google Scholar
Ansell, BJ, Watson, KE, Fogelman, AM, Navab, M & Fonarow, GC (2005) High-density lipoprotein function recent advances. Journal of the American College of Cardiology 46, 17921798.Google Scholar
Aubert, H, Frere, C, Aillaud, MF, Morange, PE, Juhan-Vague, I & Alessi, MC (2003) Weak and non-independent association between plasma TAFI antigen levels and the insulin resistance syndrome. Journal of Thrombosis and Haemostasis 1, 791797.CrossRefGoogle ScholarPubMed
Bajaj, M & Banerji, MA (2004) Type 2 diabetes in South Asians: a pathophysiologic focus on the Asian-Indian epidemic. Current Diabetes Reports 4, 213218.Google Scholar
Barbato, A, Cappuccio, FP, Folkerd, EJ, Strazzullo, P, Sampson, B, Cook, DG & Alberti, KG (2004) Metabolic syndrome and renal sodium handling in three ethnic groups living in England. Diabetologia 47, 4046.CrossRefGoogle ScholarPubMed
Bergman, AJ, Murphy, G, Burke, J, Zhao, JJ, Valesky, R, Liu, L et al. (2004) Simvastatin does not have a clinically significant pharmacokinetic interaction with fenofibrate in humans. Journal of Clinical Pharmacology 44, 10541062.Google Scholar
Bluher, M, Bullen, JW Jr, Lee, JH, Kralisch, S, Fasshauer, M, Kloting, N, Niebauer, J, Schon, MR, Williams, CJ & Mantzoros, CS (2006) Circulating adiponectin and expression of adiponectin receptors in human skeletal muscle: associations with metabolic parameters and insulin resistance and regulation by physical training. Journal of Clinical Endocrinology and Metabolism 91, 23102316.CrossRefGoogle ScholarPubMed
Bo, S, Gambino, R, Uberti, B, Mangiameli, MP, Colosso, G, Repetti, E, Gentile, L, Cassader, M & Pagano, GF (2005) Does C-reactive protein identify a subclinical metabolic disease in healthy subjects? European Journal of Clinical Investigation 35, 265270.Google Scholar
Boden, G (2003) Effects of free fatty acids (FFA) on glucose metabolism: significance for insulin resistance and type 2 diabetes. Experimental and Clinical Endocrinology and Diabetes 111, 121124.Google Scholar
Boden, G, Cheung, P, Stein, TP, Kresge, K & Mozzoli, M (2002) FFA cause hepatic insulin resistance by inhibiting insulin suppression of glycogenolysis. American Journal of Physiology 283, E12E19.Google ScholarPubMed
Boden, G & Shulman, GI (2002) Free fatty acids in obesity and type 2 diabetes: defining their role in the development of insulin resistance and beta-cell dysfunction. European Journal of Clinical Investigation 32, Suppl. 3, 1423.Google Scholar
Boston, RC, Stefanovski, D, Moate, PJ, Sumner, AE, Watanabe, RM & Bergman, RN (2003) MINMOD Millennium: a computer program to calculate glucose effectiveness and insulin sensitivity from the frequently sampled intravenous glucose tolerance test. Diabetes Technology and Therapeutics 5, 10031015.CrossRefGoogle ScholarPubMed
Boucher, A, Lu, D, Burgess, SC, Telemaque-Potts, S, Jensen, MV, Mulder, H, Wang, MY, Unger, RH, Sherry, AD & Newgard, CB (2004) Biochemical mechanism of lipid-induced impairment of glucose-stimulated insulin secretion and reversal with a malate analogue. Journal of Biological Chemistry 279, 2726327271.CrossRefGoogle ScholarPubMed
Brinton, EA, Eisenberg, S & Breslow, JL (1991) Increased apo A-I and apo A-II fractional catabolic rate in patients with low high density lipoprotein-cholesterol levels with or without hypertriglyceridemia. Journal of Clinical Investigation 87, 536544.Google Scholar
Bruning, JC, Michael, MD, Winnay, JN, Hayashi, T, Horsch, D, Accili, D, Goodyear, LJ & Kahn, CR (1998) A muscle-specific insulin receptor knockout exhibits features of the metabolic syndrome of NIDDM without altering glucose tolerance. Molecular Cell 2, 559569.Google Scholar
Cases, JA, Gabriely, I, Ma, XH, Yang, XM, Michaeli, T, Fleischer, N, Rossetti, L & Barzilai, N (2001) Physiological increase in plasma leptin markedly inhibits insulin secretion in vivo. Diabetes 50, 348352.CrossRefGoogle ScholarPubMed
Chambers, JC, Eda, S, Bassett, P, Karim, Y, Thompson, SG, Gallimore, JR, Pepys, MB & Kooner, JS (2001) C-reactive protein, insulin resistance, central obesity, and coronary heart disease risk in Indian Asians from the United Kingdom compared with European whites. Circulation 104, 145150.CrossRefGoogle ScholarPubMed
Chan, DC, Barrett, PH & Watts, GF (2006) Recent studies of lipoprotein kinetics in the metabolic syndrome and related disorders. Current Opinion in Lipidology 17, 2836.Google Scholar
Combs, TP, Berg, AH, Obici, S, Scherer, PE & Rossetti, L (2001) Endogenous glucose production is inhibited by the adipose-derived protein Acrp30. Journal of Clinical Investigation 108, 18751881.CrossRefGoogle ScholarPubMed
Coppola, G, Corrado, E, Muratori, I, Tantillo, R, Vitale, G, Lo, CL & Novo, S (2006) Increased levels of C-reactive protein and fibrinogen influence the risk of vascular events in patients with NIDDM. International Journal of Cardiology 106, 1620.CrossRefGoogle ScholarPubMed
Dallongeville, J, Helbecque, N, Cottel, D, Amouyel, P & Meirhaeghe, A (2003) The Gly16→Arg16 and Gln27→Glu27 polymorphisms of beta2-adrenergic receptor are associated with metabolic syndrome in men. Journal of Clinical Endocrinology and Metabolism 88, 48624866.Google Scholar
DeFronzo, RA, Cooke, CR, Andres, R, Faloona, GR & Davis, PJ (1975) The effect of insulin on renal handling of sodium, potassium, calcium, and phosphate in man. Journal of Clinical Investigation 55, 845855.CrossRefGoogle ScholarPubMed
de Graaf, J, Hendriks, JC, Demacker, PN & Stalenhoef, AF (1993) Identification of multiple dense LDL subfractions with enhanced susceptibility to in vitro oxidation among hypertriglyceridemic subjects. Normalization after clofibrate treatment. Arteriosclerosis and Thrombosis 13, 712719.Google Scholar
Doi, Y, Kiyohara, Y, Kubo, M, Ninomiya, T, Wakugawa, Y, Yonemoto, K, Iwase, M & Iida, M (2005) Elevated C-reactive protein is a predictor of the development of diabetes in a general Japanese population: the Hisayama Study. Diabetes Care 28, 24972500.CrossRefGoogle Scholar
Eckel, RH (1989) Lipoprotein lipase. A multifunctional enzyme relevant to common metabolic diseases. New England Journal of Medicine 320, 10601068.Google Scholar
Eckel, RH, Grundy, SM & Zimmet, PZ (2005) The metabolic syndrome. Lancet 365, 14151428.CrossRefGoogle ScholarPubMed
Eckel, RH, Yost, TJ & Jensen, DR (1995) Alterations in lipoprotein lipase in insulin resistance. International Journal of Obesity and related Metabolic Disorders 19, Suppl. 1, S16S21.Google Scholar
Egan, BM (2003) Insulin resistance and the sympathetic nervous system. Current Hypertension Reports 5, 247254.Google Scholar
Eliasson, B, Attvall, S, Taskinen, MR & Smith, U (1994) The insulin resistance syndrome in smokers is related to smoking habits. Arteriosclerosis and Thrombosis 14, 19461950.Google Scholar
Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (2001) The Third Report of The National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III). Executive summary. Journal of the American Medical Association 285, 24862497.CrossRefGoogle Scholar
Facchini, F, Chen, YD, Hollenbeck, CB & Reaven, GM (1991) Relationship between resistance to insulin-mediated glucose uptake, urinary uric acid clearance, and plasma uric acid concentration. Journal of the American Medical Association 266, 30083011.Google Scholar
Fernandez-Real, JM & Ricart, W (2003) Insulin resistance and chronic cardiovascular inflammatory syndrome. Endocrine Reviews 24, 278301.Google Scholar
Ferrannini, E, Buzzigoli, G, Bonadonna, R, Giorico, MA, Oleggini, M, Graziadei, L, Pedrinelli, R, Brandi, L & Bevilacqua, S (1987) Insulin resistance in essential hypertension. New England Journal of Medicine 317, 350357.Google Scholar
Ford, ES (2005) Prevalence of the metabolic syndrome defined by the International Diabetes Federation among adults in the U.S. Diabetes Care 28, 27452749.CrossRefGoogle ScholarPubMed
Foufelle, F & Ferre, P (2002) New perspectives in the regulation of hepatic glycolytic and lipogenic genes by insulin and glucose: a role for the transcription factor sterol regulatory element binding protein-1c. Biochemical Journal 366, 377391.CrossRefGoogle ScholarPubMed
Fumeron, F, Aubert, R, Siddiq, A, Betoulle, D, Pean, F, Hadjadj, S et al. (2004) Adiponectin gene polymorphisms and adiponectin levels are independently associated with the development of hyperglycemia during a 3-year period: the epidemiologic data on the insulin resistance syndrome prospective study. Diabetes 53, 11501157.Google Scholar
Garg, A & Misra, A (2004) Lipodystrophies: rare disorders causing metabolic syndrome. Endocrinology and Metabolism Clinics of North America 33, 305331.Google Scholar
Goodarzi, MO, Guo, X, Taylor, KD, Quinones, MJ, Saad, MF, Yang, H, Hsueh, WA & Rotter, JI (2004) Lipoprotein lipase is a gene for insulin resistance in Mexican Americans. Diabetes 53, 214220.Google Scholar
Griffiths, SL & Grainger, DJ (2006) Proposal of a novel diabetogenic mechanism involving the serpin PAI-1. Bioessays 28, 629641.Google Scholar
Grundy, SM (2004) Cholesterol gallstones: a fellow traveler with metabolic syndrome? American Journal of Clinical Nutrition 80, 12.CrossRefGoogle ScholarPubMed
Grundy, SM (2006) Does a diagnosis of metabolic syndrome have value in clinical practice? American Journal of Clinical Nutrition 83, 12481251.CrossRefGoogle ScholarPubMed
Grundy, SM, Cleeman, JI, Daniels, SR, Donato, KA, Eckel, RH, Franklin, BA et al. (2005) Diagnosis and management of the metabolic syndrome: an American Heart Association/National Heart, Lung, and Blood Institute Scientific Statement. Circulation 112, 27352752.CrossRefGoogle ScholarPubMed
Guo, Z, Hensrud, DD, Johnson, CM & Jensen, MD (1999) Regional postprandial fatty acid metabolism in different obesity phenotypes. Diabetes 48, 15861592.Google Scholar
Haemmerle, G, Lass, A, Zimmermann, R, Gorkiewicz, G, Meyer, C, Rozman, J et al. (2006) Defective lipolysis and altered energy metabolism in mice lacking adipose triglyceride lipase. Science 312, 734737.Google Scholar
Haffner, SM (2006) The metabolic syndrome: inflammation, diabetes mellitus, and cardiovascular disease. American Journal of Cardiology 97, 3A11A.Google Scholar
Haider, DG, Schaller, G, Kapiotis, S, Maier, C, Luger, A & Wolzt, M (2006) The release of the adipocytokine visfatin is regulated by glucose and insulin. Diabetologia 49, 19091914.CrossRefGoogle ScholarPubMed
Halle, M, Berg, A, Baumstark, MW, Konig, D, Huonker, M & Keul, J (1999) Influence of mild to moderately elevated triglycerides on low density lipoprotein subfraction concentration and composition in healthy men with low high density lipoprotein cholesterol levels. Atherosclerosis 143, 185192.Google Scholar
Hanley, AJ, Karter, AJ, Festa, A, D'Agostino, R Jr, Wagenknecht, LE, Savage, P, Tracy, RP, Saad, MF & Haffner, S (2002) Factor analysis of metabolic syndrome using directly measured insulin sensitivity: The Insulin Resistance Atherosclerosis Study. Diabetes 51, 26422647.Google Scholar
Hanley, AJ, Karter, AJ, Williams, K, Festa, A, D'Agostino, RB Jr, Wagenknecht, LE & Haffner, SM (2005) Prediction of type 2 diabetes mellitus with alternative definitions of the metabolic syndrome: the Insulin Resistance Atherosclerosis Study 1. Circulation 112, 37133721.Google Scholar
Hara, K, Horikoshi, M, Yamauchi, T, Yago, H, Miyazaki, O, Ebinuma, H, Imai, Y, Nagai, R & Kadowaki, T (2006) Measurement of the high-molecular weight form of adiponectin in plasma is useful for the prediction of insulin resistance and metabolic syndrome. Diabetes Care 29, 13571362.Google Scholar
Hegele, RA (2003) Monogenic forms of insulin resistance: apertures that expose the common metabolic syndrome. Trends in Endocrinology and Metabolism 14, 371377.Google Scholar
Holzl, B, Iglseder, B, Sandhofer, A, Malaimare, L, Lang, J, Paulweber, B & Sandhofer, F (2002) Insulin sensitivity is impaired in heterozygous carriers of lipoprotein lipase deficiency. Diabetologia 45, 378384.Google Scholar
Iacobellis, G, Corradi, D & Sharma, AM (2005) Epicardial adipose tissue: anatomic, biomolecular and clinical relationships with the heart. Nature Clinical Practice Cardiovascular Medicine 2, 536543.Google Scholar
Jensen, MD, Caruso, M, Heiling, V & Miles, JM (1989) Insulin regulation of lipolysis in nondiabetic and IDDM subjects. Diabetes 38, 15951601.Google Scholar
Joseph, JW, Koshkin, V, Saleh, MC, Sivitz, WI, Zhang, CY, Lowell, BB, Chan, CB & Wheeler, MB (2004) Free fatty acid induced beta-cell defects are dependent on uncoupling protein 2 expression. Journal of Biological Chemistry 279, 5104951056.Google Scholar
Jungner, I, Sniderman, AD, Furberg, C, Aastveit, AH, Holme, I & Walldius, G (2006) Does low-density lipoprotein size add to atherogenic particle number in predicting the risk of fatal myocardial infarction? American Journal of Cardiology 97, 943946.Google Scholar
Kahn, R, Buse, J, Ferrannini, E & Stern, M (2005) The metabolic syndrome: time for a critical appraisal. Joint statement from the American Diabetes Association and the European Association for the Study of Diabetes. Diabetologia 48, 16841699.Google Scholar
Kakuma, T, Lee, Y, Higa, M, Wang, Z, Pan, W, Shimomura, I & Unger, RH (2000) Leptin, troglitazone, and the expression of sterol regulatory element binding proteins in liver and pancreatic islets. Proceedings of the National Academy of Sciences USA 97, 85368541.Google Scholar
Kontush, A & Chapman, MJ (2006) Antiatherogenic small, dense HDL – guardian angel of the arterial wall? Nature Clinical Practice Cardiovascular Medicine 3, 144153.Google Scholar
Kotelevtsev, Y, Holmes, MC, Burchell, A, Houston, PM, Schmoll, D, Jamieson, P, Best, R, Brown, R, Edwards, CR, Seckl, JR & Mullins, JJ (1997) 11beta-hydroxysteroid dehydrogenase type 1 knockout mice show attenuated glucocorticoid-inducible responses and resist hyperglycemia on obesity or stress. Proceedings of the National Academy of Sciences USA 94, 1492414929.Google Scholar
Krauss, RM (1995) Dense low density lipoproteins and coronary artery disease. American Journal of Cardiology 75, 53B57B.Google Scholar
Kulkarni, RN, Bruning, JC, Winnay, JN, Postic, C, Magnuson, MA & Kahn, CR (1999) Tissue-specific knockout of the insulin receptor in pancreatic beta cells creates an insulin secretory defect similar to that in type 2 diabetes. Cell 96, 329339.Google Scholar
Kurl, S, Laukkanen, JA, Niskanen, L, Laaksonen, D, Sivenius, J, Nyyssonen, K & Salonen, JT (2006) Metabolic syndrome and the risk of stroke in middle-aged men. Stroke 37, 806811.CrossRefGoogle ScholarPubMed
Kuroda, S, Uzu, T, Fujii, T, Nishimura, M, Nakamura, S, Inenaga, T & Kimura, G (1999) Role of insulin resistance in the genesis of sodium sensitivity in essential hypertension. Journal of Human Hypertension 13, 257262.Google Scholar
Kwiterovich, PO Jr (2002) Clinical relevance of the biochemical, metabolic, and genetic factors that influence low-density lipoprotein heterogeneity. American Journal of Cardiology 90, 30i47i.Google Scholar
Kylin, E (1923) Studien über das Hypertonie-Hyperglykamie-Hyperurikamiesyndrome (Studies on hypertension-hyperglycaemia-hyperuricaemia syndrome). Zentralblatt für innere Medizin 44, 105127.Google Scholar
Lada, AT & Rudel, LL (2004) Associations of low density lipoprotein particle composition with atherogenicity. Current Opinion in Lipidology 15, 1924.CrossRefGoogle ScholarPubMed
Lakka, TA, Laaksonen, DE, Lakka, HM, Mannikko, N, Niskanen, LK, Rauramaa, R & Salonen, JT (2003) Sedentary lifestyle, poor cardiorespiratory fitness, and the metabolic syndrome. Medicine and Science in Sports and Exercise 35, 12791286.Google Scholar
LaMonte, MJ, Durstine, JL, Yanowitz, FG, Lim, T, DuBose, KD, Davis, P & Ainsworth, BE (2002) Cardiorespiratory fitness and C-reactive protein among a tri-ethnic sample of women. Circulation 106, 403406.Google Scholar
Langenberg, C, Bergstrom, J, Scheidt-Nave, C, Pfeilschifter, J & Barrett-Connor, E (2006) Cardiovascular death and the metabolic syndrome: role of adiposity-signaling hormones and inflammatory markers. Diabetes Care 29, 13631369.CrossRefGoogle ScholarPubMed
Lara-Castro, C, Luo, N, Wallace, P, Klein, RL & Garvey, WT (2006) Adiponectin multimeric complexes and the metabolic syndrome trait cluster. Diabetes 55, 249259.Google Scholar
Laubner, K, Kieffer, TJ, Lam, NT, Niu, X, Jakob, F & Seufert, J (2005) Inhibition of preproinsulin gene expression by leptin induction of suppressor of cytokine signaling 3 in pancreatic {beta}-cells. Diabetes 54, 34103417.Google Scholar
Lee, JS, Pinnamaneni, SK, Eo, SJ, Cho, IH, Pyo, JH, Kim, CK, Sinclair, AJ, Febbraio, MA & Watt, MJ (2006) Saturated, but not n-6 polyunsaturated, fatty acids induce insulin resistance: role of intramuscular accumulation of lipid metabolites. Journal of Applied Physiology 100, 14671474.Google Scholar
Lee, S, Janssen, I & Ross, R (2004) Interindividual variation in abdominal subcutaneous and visceral adipose tissue: influence of measurement site. Journal of Applied Physiology 97, 948954.Google Scholar
Lee, Y, Hirose, H, Ohneda, M, Johnson, JH, McGarry, JD & Unger, RH (1994) Beta-cell lipotoxicity in the pathogenesis of non-insulin-dependent diabetes mellitus of obese rats: impairment in adipocyte-beta-cell relationships. Proceedings of the National Academy of Sciences USA 91, 1087810882.CrossRefGoogle ScholarPubMed
Lewis, GF & Steiner, G (1996) Acute effects of insulin in the control of VLDL production in humans. Implications for the insulin-resistant state. Diabetes Care 19, 390393.Google Scholar
Lewis, GF, Uffelman, KD, Szeto, LW, Weller, B & Steiner, G (1995) Interaction between free fatty acids and insulin in the acute control of very low density lipoprotein production in humans. Journal of Clinical Investigation 95, 158166.Google Scholar
Li, JJ, Fang, CH & Hui, RT (2005) Is hypertension an inflammatory disease? Medical Hypotheses 64, 236240.Google Scholar
Linnemann, B, Voigt, W, Nobel, W & Janka, HU (2006) C-reactive protein is a strong independent predictor of death in type 2 diabetes: association with multiple facets of the metabolic syndrome. Experimental and Clinical Endocrinology and Diabetes 114, 127134.Google Scholar
Ma, YQ, Thomas, GN, Ng, MC, Critchley, JA, Chan, JC & Tomlinson, B (2003) The lipoprotein lipase gene HindIII polymorphism is associated with lipid levels in early-onset type 2 diabetic patients. Metabolism 52, 338343.Google Scholar
Maahs, DM, Ogden, LG, Kinney, GL, Wadwa, RP, Snell-Gergeon, J, Dabelea, D, Hokanson, JE, Ehrlich, J, Eckel, RH & Rewers, M (2005) Low plasma adiponectin levels predict progression of coronary artery calcification. Circulation 111, 747753.Google Scholar
Maeda, K, Cao, H, Kono, K, Gorgun, CZ, Furuhashi, M, Uysal, KT et al. (2005) Adipocyte/macrophage fatty acid binding proteins control integrated metabolic responses in obesity and diabetes. Cell Metabolism 1, 107119.Google Scholar
Manzato, E, Zambon, S, Zambon, A, Cortella, A, Sartore, G & Crepaldi, G (1993) Levels and physicochemical properties of lipoprotein subclasses in moderate hypertriglyceridemia. Clinica Chimica Acta 219, 5765.Google Scholar
Masuzaki, H, Paterson, J, Shinyama, H, Morton, NM, Mullins, JJ, Seckl, JR & Flier, JS (2001) A transgenic model of visceral obesity and the metabolic syndrome. Science 294, 21662170.Google Scholar
Matsunaga, T & Muramatsu, MA (2005) Knowledge-based computational search for genes associated with the metabolic syndrome. Bioinformatics 21, 31463154.Google Scholar
Matsuzawa, Y, Funahashi, T, Kihara, S & Shimomura, I (2004) Adiponectin and metabolic syndrome. Arteriosclerosis, Thrombosis, and Vascular Biology 24, 2933.CrossRefGoogle ScholarPubMed
Medina, J, Fernandez-Salazar, LI, Garcia-Buey, L & Moreno-Otero, R (2004) Approach to the pathogenesis and treatment of nonalcoholic steatohepatitis. Diabetes Care 27, 20572066.CrossRefGoogle Scholar
Minokoshi, Y & Kahn, BB (2003) Role of AMP-activated protein kinase in leptin-induced fatty acid oxidation in muscle. Biochemical Society Transactions 31, 196201.Google Scholar
Miyashita, Y & Shirai, K (2005) Clinical determination of the severity of metabolic syndrome: preheparin lipoprotein lipase mass as a new marker of metabolic syndrome. Current Medicinal Chemistry Cardiovascular and Hematological Agents 3, 377381.Google Scholar
Murakami, T, Michelagnoli, S, Longhi, R, Gianfranceschi, G, Pazzucconi, F, Calabresi, L, Sirtori, CR & Franceschini, G (1995) Triglycerides are major determinants of cholesterol esterification/transfer and HDL remodeling in human plasma. Arteriosclerosis and Thrombosis 15, 18191828.Google Scholar
Najarian, RM, Sullivan, LM, Kannel, WB, Wilson, PW, D'Agostino, RB & Wolf, PA (2006) Metabolic syndrome compared with type 2 diabetes mellitus as a risk factor for stroke: the Framingham Offspring Study. Archives of Internal Medicine 166, 106111.Google Scholar
Nawrocki, AR & Scherer, PE (2004) The delicate balance between fat and muscle: adipokines in metabolic disease and musculoskeletal inflammation. Current Opinion in Pharmacology 4, 281289.Google Scholar
Nervi, F, Miquel, JF, Alvarez, M, Ferreccio, C, Garcia-Zattera, MJ, Gonzalez, R, Perez-Ayuso, RM, Rigotti, A & Villarroel, L (2006) Gallbladder disease is associated with insulin resistance in a high risk Hispanic population. Journal of Hepatology 45, 299305.Google Scholar
Neuschwander-Tetri, BA & Caldwell, SH (2003) Nonalcoholic steatohepatitis: summary of an AASLD Single Topic Conference. Hepatology 37, 12021219.Google Scholar
Ohashi, K, Kihara, S, Ouchi, N, Kumada, M, Fujita, K, Hiuge, A et al. (2006) Adiponectin replenishment ameliorates obesity-related hypertension. Hypertension 47, 11081116.Google Scholar
Onat, A, Hergenc, G, Sansoy, V, Fobker, M, Ceyhan, K, Toprak, S & Assmann, G (2003) Apolipoprotein C-III, a strong discriminant of coronary risk in men and a determinant of the metabolic syndrome in both genders. Atherosclerosis 168, 8189.Google Scholar
Ouchi, N, Shibata, R & Walsh, K (2006) Cardioprotection by adiponectin. Trends in Cardiovascular Medicine 16, 141146.Google Scholar
Ozcan, U, Cao, Q, Yilmaz, E, Lee, AH, Iwakoshi, NN, Ozdelen, E, Tuncman, G, Gorgun, C, Glimcher, LH & Hotamisligil, GS (2004) Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science 306, 457461.CrossRefGoogle ScholarPubMed
Packard, CJ (1996) LDL subfractions and atherogenicity: an hypothesis from the University of Glasgow. Current Medical Research and Opinion 13, 379390.CrossRefGoogle ScholarPubMed
Patel, DA, Srinivasan, SR, Xu, JH, Li, S, Chen, W & Berenson, GS (2006) Distribution and metabolic syndrome correlates of plasma C-reactive protein in biracial (black-white) younger adults: the Bogalusa Heart Study. Metabolism 55, 699705.CrossRefGoogle ScholarPubMed
Petersen, KF, Befroy, D, Dufour, S, Dziura, J, Ariyan, C, Rothman, DL, DiPietro, L, Cline, GW & Shulman, GI (2003) Mitochondrial dysfunction in the elderly: possible role in insulin resistance. Science 300, 11401142.Google Scholar
Petersen, KF, Dufour, S, Befroy, D, Garcia, R & Shulman, GI (2004) Impaired mitochondrial activity in the insulin-resistant offspring of patients with type 2 diabetes. New England Journal of Medicine 350, 664671.Google Scholar
Pischon, T, Girman, CJ, Hotamisligil, GS, Rifai, N, Hu, FB & Rimm, EB (2004) Plasma adiponectin levels and risk of myocardial infarction in men. Journal of the American Medical Association 291, 17301737.Google Scholar
Ren, J (2004) Leptin and hyperleptinemia – from friend to foe for cardiovascular function. Journal of Endocrinology 181, 110.Google Scholar
Roden, M (2005) Muscle triglycerides and mitochondrial function: possible mechanisms for the development of type 2 diabetes. International Journal of Obesity 29, Suppl. 2, S111S115.Google Scholar
Rowley, K, O'Dea, K & Best, JD (2003) Association of albuminuria and the metabolic syndrome. Current Diabetes Reports 3, 8086.Google Scholar
Ruderman, N, Chisholm, D, Pi-Sunyer, X & Schneider, S (1998) The metabolically obese, normal-weight individual revisited. Diabetes 47, 699713.Google Scholar
Rutter, MK, Meigs, JB, Sullivan, LM, D'Agostino, RB Sr & Wilson, PW (2005) Insulin resistance, the metabolic syndrome, and incident cardiovascular events in the Framingham Offspring Study. Diabetes 54, 32523257.Google Scholar
Sacks, FM & Campos, H (2003) Clinical review 163: Cardiovascular endocrinology: Low-density lipoprotein size and cardiovascular disease: a reappraisal. Journal of Clinical Endocrinology and Metabolism 88, 45254532.Google Scholar
Samuel, VT, Liu, ZX, Qu, X, Elder, BD, Bilz, S, Befroy, D, Romanelli, AJ & Shulman, GI (2004) Mechanism of hepatic insulin resistance in non-alcoholic fatty liver disease. Journal of Biological Chemistry 279, 3234532353.Google Scholar
Sattar, N, Scherbakova, O, Ford, I, O'Reilly, DS, Stanley, A, Forrest, E, Macfarlane, PW, Packard, CJ, Cobbe, SM & Shepherd, J (2004) Elevated alanine aminotransferase predicts new-onset type 2 diabetes independently of classical risk factors, metabolic syndrome, and C-reactive protein in the West of Scotland Coronary Prevention Study. Diabetes 53, 28552860.Google Scholar
Savage, DB, Choi, CS, Samuel, VT, Liu, ZX, Zhang, D, Wang, A et al. (2006) Reversal of diet-induced hepatic steatosis and hepatic insulin resistance by antisense oligonucleotide inhibitors of acetyl-CoA carboxylases 1 and 2. Journal of Clinical Investigation 116, 817824.Google Scholar
Seufert, J (2004) Leptin effects on pancreatic beta-cell gene expression and function. Diabetes 53, Suppl. 1, S152S158.Google Scholar
Shibata, R, Sato, K, Pimentel, DR, Takemura, Y, Kihara, S, Ohashi, K, Funahashi, T, Ouchi, N & Walsh, K (2005) Adiponectin protects against myocardial ischemia-reperfusion injury through AMPK- and COX-2-dependent mechanisms. Nature Medicine 11, 10961103.Google Scholar
Shimabukuro, M, Zhou, YT, Levi, M & Unger, RH (1998) Fatty acid-induced beta cell apoptosis: a link between obesity and diabetes. Proceedings of the National Academy of Sciences USA 95, 24982502.Google Scholar
Shimomura, I, Bashmakov, Y, Ikemoto, S, Horton, JD, Brown, MS & Goldstein, JL (1999) Insulin selectively increases SREBP-1c mRNA in the livers of rats with streptozotocin-induced diabetes. Proceedings of the National Academy of Sciences USA 96, 1365613661.Google Scholar
Shuldiner, AR & McLenithan, JC (2004) Genes and pathophysiology of type 2 diabetes: more than just the Randle cycle all over again. Journal of Clinical Investigation 114, 14141417.Google Scholar
Steinberg, HO, Brechtel, G, Johnson, A, Fineberg, N & Baron, AD (1994) Insulin-mediated skeletal muscle vasodilation is nitric oxide dependent. A novel action of insulin to increase nitric oxide release. Journal of Clinical Investigation 94, 11721179.Google Scholar
Stephens, JM & Vidal-Puig, AJ (2006) An update on visfatin/pre-B cell colony-enhancing factor, an ubiquitously expressed, illusive cytokine that is regulated in obesity. Current Opinion in Lipidology 17, 128131.Google Scholar
Storlien, LH, Higgins, JA, Thomas, TC, Brown, MA, Wang, HQ, Huang, XF & Else, PL (2000) Diet composition and insulin action in animal models. British Journal of Nutrition 83, Suppl. 1, S85S90.Google Scholar
Suganami, T, Nishida, J & Ogawa, Y (2005) A paracrine loop between adipocytes and macrophages aggravates inflammatory changes: role of free fatty acids and tumor necrosis factor alpha. Arteriosclerosis, Thrombosis, and Vascular Biology 25, 20622068.Google Scholar
Sung, YY, Lee, YS, Jung, WH, Kim, HY, Cheon, HG, Yang, SD & Rhee, SD (2005) Glucose intolerance in young TallyHo mice is induced by leptin-mediated inhibition of insulin secretion. Biochemical and Biophysical Research Communications 338, 17791787.Google Scholar
Sutherland, J, McKinnley, B & Eckel, RH (2004) The metabolic syndrome and inflammation. Metabolic Syndrome and Related Disorders 2, 82104.Google Scholar
Taghibiglou, C, Rashid-Kolvear, F, Van Iderstine, SC, Le Tien, H, Fantus, IG, Lewis, GF & Adeli, K (2002) Hepatic very low density lipoprotein-ApoB overproduction is associated with attenuated hepatic insulin signaling and overexpression of protein-tyrosine phosphatase 1B in a fructose-fed hamster model of insulin resistance. Journal of Biological Chemistry 277, 793803.Google Scholar
Tanaka, S, Horimai, C & Katsukawa, F (2003) Ethnic differences in abdominal visceral fat accumulation between Japanese, African-Americans, and Caucasians: a meta-analysis. Acta Diabetologia 40, Suppl. 1, S302S304.Google Scholar
Tanaka, T, Sohmiya, K, Kono, T, Terasaki, F, Horie, R, Ohkaru, Y, Muramatsu, M, Takai, S, Miyazaki, M & Kitaura, Y (2006) Thiamine attenuates the hypertension and metabolic abnormalities in CD36-defective SHR: Uncoupling of glucose oxidation from cellular entry accompanied with enhanced protein O-GlcNAcylation in CD36 deficiency. Molecular and Cellular Biochemistry (Epublication ahead of print version).Google Scholar
Tooke, JE & Hannemann, MM (2000) Adverse endothelial function and the insulin resistance syndrome. Journal of Internal Medicine 247, 425431.Google Scholar
Trayhurn, P & Wood, IS (2004) Adipokines: inflammation and the pleiotropic role of white adipose tissue. British Journal of Nutrition 92, 347355.Google Scholar
Tripathy, D, Mohanty, P, Dhindsa, S, Syed, T, Ghanim, H, Aljada, A & Dandona, P (2003) Elevation of free fatty acids induces inflammation and impairs vascular reactivity in healthy subjects. Diabetes 52, 28822887.Google Scholar
Unger, RH (2003) Lipid overload and overflow: metabolic trauma and the metabolic syndrome. Trends in Endocrinology and Metabolism 14, 398403.Google Scholar
van Puijenbroek, EP, Du Buf-Vereijken, PW, Spooren, PF & van Doormaal, JJ (1996) Possible increased risk of rhabdomyolysis during concomitant use of simvastatin and gemfibrozil. Journal of Internal Medicine 240, 403404.Google Scholar
Vartanian, V, Lowell, B, Minko, IG, Wood, TG, Ceci, JD, George, S, Ballinger, SW, Corless, CL, McCullough, AK & Lloyd, RS (2006) The metabolic syndrome resulting from a knockout of the NEIL1 DNA glycosylase. Proceedings of the National Academy of Sciences USA 103, 18641869.Google Scholar
Verges, B, Petit, JM, Duvillard, L, Dautin, G, Florentin, E, Galland, F & Gambert, P (2006) Adiponectin is an important determinant of apoA-I catabolism. Arteriosclerosis, Thrombosis, and Vascular Biology 26, 13641369.Google Scholar
Vu, JD, Vu, JB, Pio, JR, Malik, S, Franklin, SS, Chen, RS & Wong, ND (2005) Impact of C-reactive protein on the likelihood of peripheral arterial disease in United States adults with the metabolic syndrome, diabetes mellitus, and preexisting cardiovascular disease. American Journal of Cardiology 96, 655658.Google Scholar
Weisberg, SP, McCann, D, Desai, M, Rosenbaum, M, Leibel, RL & Ferrante, AW Jr (2003) Obesity is associated with macrophage accumulation in adipose tissue. Journal of Clinical Investigation 112, 17961808.CrossRefGoogle ScholarPubMed
Weng, SW, Liou, CW, Lin, TK, Wei, YH, Lee, CF, Eng, HL et al. (2005) Association of mitochondrial deoxyribonucleic acid 16 189 variant (T→C transition) with metabolic syndrome in Chinese adults. Journal of Clinical Endocrinology and Metabolism 90, 50375040.Google Scholar
Wilson, PW, D'Agostino, RB, Parise, H, Sullivan, L & Meigs, JB (2005) Metabolic syndrome as a precursor of cardiovascular disease and type 2 diabetes mellitus. Circulation 112, 30663072.Google Scholar
Xu, AW, Kaelin, CB, Takeda, K, Akira, S, Schwartz, MW & Barsh, GS (2005) PI3K integrates the action of insulin and leptin on hypothalamic neurons. Journal of Clinical Investigation 115, 951958.Google Scholar
Xu, H, Barnes, GT, Yang, Q, Tan, G, Yang, D, Chou, CJ, Sole, J, Nichols, A, Ross, JS, Tartaglia, LA & Chen, H (2003) Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. Journal of Clinical Investigation 112, 18211830.Google Scholar
Yamada, T, Katagiri, H, Ishigaki, Y, Ogihara, T, Imai, J, Uno, K et al. (2006) Signals from intra-abdominal fat modulate insulin and leptin sensitivity through different mechanisms: neuronal involvement in food-intake regulation. Cell Metabolism 3, 223229.Google Scholar
Yamashita, S, Hirano, KI, Kuwasako, T, Janabi, M, Toyama, Y, Ishigami, M & Sakai, N (2006) Physiological and pathological roles of a multi-ligand receptor CD36 in atherogenesis; insights from CD36-deficient patients. Molecular and Cellular Biochemistry (Epublication ahead of print version).Google Scholar
Yamauchi, T, Hara, K, Kubota, N, Terauchi, Y, Tobe, K, Froguel, P, Nagai, R & Kadowaki, T (2003) Dual roles of adiponectin/Acrp30 in vivo as an anti-diabetic and anti-atherogenic adipokine. Current Drug Targets Immune, Endocrine and Metabolic Disorders 3, 243254.Google Scholar
Yaney, GC & Corkey, BE (2003) Fatty acid metabolism and insulin secretion in pancreatic beta cells. Diabetologia 46, 12971312.Google Scholar
Zambon, A, Hokanson, JE, Brown, BG & Brunzell, JD (1999) Evidence for a new pathophysiological mechanism for coronary artery disease regression: hepatic lipase-mediated changes in LDL density. Circulation 99, 19591964.Google Scholar