You are on page 1of 10

European Journal of Clinical Investigation (2002) 32 (Suppl.

3), 14 23

Free fatty acids in obesity and type 2 diabetes: dening their role in the development of insulin resistance and -cell dysfunction
Blackwell Science, Ltd

G. Boden and G. I. Shulman*


Division of Endocrinology/Diabetes/Metabolism and the General Clinical Research Center, Temple University Hospital, Philadelphia PA, USA, *Howard Hughes Medical Institute, Departments of Internal Medicine and Cellular & Molecular Physiology and the General Clinical Research Center, Yale University School of Medicine, New Haven, CT, USA

Abstract

Plasma free fatty acids (FFA) play important physiological roles in skeletal muscle, heart, liver and pancreas. However, chronically elevated plasma FFA appear to have pathophysiological consequences. Elevated FFA concentrations are linked with the onset of peripheral and hepatic insulin resistance and, while the precise action in the liver remains unclear, a model to explain the role of raised FFA in the development of skeletal muscle insulin resistance has recently been put forward. Over 30 years ago, Randle proposed that FFA compete with glucose as the major energy substrate in cardiac muscle, leading to decreased glucose oxidation when FFA are elevated. Recent data indicate that high plasma FFA also have a signicant role in contributing to insulin resistance. Elevated FFA and intracellular lipid appear to inhibit insulin signalling, leading to a reduction in insulinstimulated muscle glucose transport that may be mediated by a decrease in GLUT-4 translocation. The resulting suppression of muscle glucose transport leads to reduced muscle glycogen synthesis and glycolysis. In the liver, elevated FFA may contribute to hyperglycaemia by antagonizing the effects of insulin on endogenous glucose production. FFA also affect insulin secretion, although the nature of this relationship remains a subject for debate. Finally, evidence is discussed that FFA represent a crucial link between insulin resistance and -cell dysfunction and, as such, a reduction in elevated plasma FFA should be an important therapeutic target in obesity and type 2 diabetes. Keywords insulin secretion, magnetic resonance spectroscopy, nonesteried fatty acids, noninsulin-dependent diabetes mellitus Eur J Clin Invest 2002; 32 (Suppl. 3): 1423

Introduction
Plasma free fatty acid (FFA) concentrations are commonly elevated in obese individuals [1], most likely due to increased FFA release associated with an expansion in fat mass [2,3]. Elevated plasma FFA are common in type 2 diabetes [4] and early changes may be predictive for the transition of patients from impaired glucose tolerance (IGT) to type 2 diabetes [5,6]. Furthermore, studies indicate that
Correspondence to: Dr Guenther Boden, Division of Endocrinology/Diabetes/Metabolism and the General Clinical Research Center, Temple University Hospital, Philadelphia PA 19140, USA. Tel: 215 707 8984; Fax: 2157071560; E-mail: bodengh@tuhs.temple.edu and Dr Gerald I. Shulman, Boyer Center for Molecular Medicine 254, 295 Congress Avenue, New Haven CT 06510, USA. Tel: +1 2037855447, Fax: +1 203737 4059; E-mail: gerald.shulman@yale.edu 2002 Blackwell Science Ltd

elevated circulating FFA may directly contribute to the underlying pathophysiology of type 2 diabetes, in particular, the development of insulin resistance both in the periphery and the liver [7,8]. High plasma FFA concentrations are associated with a number of cardiovascular risk factors linked to insulin resistance including hypertension, dyslipidaemia, hyperuricaemia and abnormal brinolysis [9,10]. Moreover, evidence from in vitro studies supports the suggestion that chronically elevated plasma FFA may contribute to -cell dysfunction in type 2 diabetes [1113]. Despite the link between elevated plasma FFA concentrations and the development of several pathological conditions, the signicant physiological role of FFA tends to be overlooked. FFA constitute an important energy source in most body tissues, representing the primary oxidative fuel for liver, resting skeletal muscle, renal cortex and myocardium [14]. When demand for fuel rises, adipose tissue lipolysis is stimulated, thus increasing systemic FFA availability

Free fatty acids in obesity and T2D

15

and preserving glucose for cerebral requirements. The demand for FFA is particularly high during starvation or exercise; for example, lipid oxidation can account for more than 70% of total body energy expenditure following an overnight fast [15]. Another situation where FFA have an important physiological role is during pregnancy. In the early stages, additional maternal fat is laid down while during late pregnancy, lipolytic hormones stimulate fat breakdown, leading to elevations in plasma FFA concentrations. This induces peripheral insulin resistance and causes a switch in fuel metabolism from carbohydrate to fat oxidation, thus maximizing the availability of glucose for the developing foetus [16]. In addition, FFA have other important physiological functions, for instance, FFA enhance both basal and glucose-stimulated insulin secretion and are essential for stimulussecretion coupling in the pancreatic -cell [17,18]. Different types of FFA inuence glucosestimulated insulin secretion to varying extents, for example longer chain FFA have a greater stimulatory effect on secretion compared with short-chain FFA [19]. Here, we review the inuence of FFA on insulin sensitivity and -cell function in both physiological and pathophysiological circumstances and discuss whether the effect of FFA on these two variables may be linked.

Figure 1 The glucosefatty acid cycle: relationship between glucose and free fatty acid metabolism (Reprinted with permission from Elsevier Science; The Lancet, 1963;1:785 89 [33]).

Role of FFA in the development of insulin resistance


Over 80% of people with type 2 diabetes are obese, while virtually all are insulin resistant [7]. The association between obesity and insulin resistance is likened to a cause and effect relationship since both human and animal studies indicate that weight loss/gain correlates closely with increasing/ decreasing insulin sensitivity, respectively [20 23]. Several factors are proposed to link obesity and insulin resistance and promising candidates include FFA, tumour necrosis factor- (TNF-) and leptin. This review will focus on the potential role of FFA while TNF- and leptin are reviewed elsewhere [2426]. Adipose tissue has been proposed to be a site of insulin resistance [27]. As insulin resistance develops, there is a decrease in insulin-mediated suppression of lipolysis leading to increased circulating FFA and ultimately insulin resistance in skeletal muscle and liver [7,8]. A strong correlation is observed between increased plasma FFA, intramyocellular lipid accumulation and insulin resistance [2831]. A number of hypotheses have been put forward to explain the role of FFA and intracellular lipid in the pathogenesis of type 2 diabetes, but, until recently, a more detailed biochemical explanation for FFA-induced insulin resistance has remained elusive.

Insulin resistance in skeletal muscle Skeletal muscle is a major contributor to insulin resistance in type 2 diabetes. In a study investigating glucose

metabolism in individuals with type 2 diabetes, insulinstimulated total body glucose uptake decreased by 30 40% compared with nondiabetic controls under hyperinsulinaemic conditions [32]. Measurement of peripheral and splanchnic glucose uptake and hepatic glucose output in the same individuals indicated that nearly 90% of this decrease was accounted for by the peripheral tissues (predominantly skeletal muscle) [32]. In 1963, Randle et al. proposed a connection between muscle insulin resistance and elevated FFA concentrations [33]. They demonstrated that FFA compete with glucose as an energy substrate in muscle and adipose tissue, describing the relationship between glucose and FFA metabolism in terms of a glucosefatty acid cycle (Fig. 1). Studying rat cardiac muscle in vitro, they observed that the rate of fat oxidation increased relative to carbohydrate oxidation in response to elevated FFA concentrations [33]. This was associated with a decrease in insulin-stimulated muscle glucose uptake and utilization. Almost 30 years later, Boden et al. observed similar effects in healthy human skeletal muscle [34]. Under euglycaemichyperinsulinaemic clamp conditions, they demonstrated a decrease in carbohydrate oxidation associated with increased fat oxidation following 1 h of lipid infusion [34]. In the same healthy individuals, they observed a signicant inhibition of insulin-stimulated glucose uptake following 34 h of lipid infusion [34]. This observation was conrmed in subsequent studies in healthy volunteers [35,36] and in individuals with type 2 diabetes [37]. In both of these groups, there was a consistent and signicant reduction in insulin-stimulated glucose uptake of 40 55% following lipid infusion compared with controls. Studies in diabetic rats demonstrate that anti-lipolytic agents (nicotinic acid and phenylisopropyladenosine) exert potent blood glucose-lowering effects by decreasing plasma FFA concentrations and thus improving insulin sensitivity [38]. To investigate the effect of anti-lipolytic agents in

2002 Blackwell Science Ltd, European Journal of Clinical Investigation, 32 (Suppl. 3), 1423

16

G. Boden and G. I. Shulman

humans, a long-acting nicotinic acid analogue, Acipimox, was used to lower chronically elevated FFA in obese individuals, with or without type 2 diabetes [39]. After overnight administration of Acipimox, fasting plasma FFA decreased by 6070% in all groups. This was accompanied by a twofold increase in insulin-stimulated glucose uptake in the obese, nondiabetic subgroup, sufcient to normalize glucose uptake to the levels seen in lean, nondiabetic individuals. Insulin-stimulated glucose uptake also improved in obese, diabetic subjects following Acipimox administration, although the compensation was not complete, indicating that FFA was only partially responsible for inducing the insulin resistance observed in type 2 diabetes. Importantly, the improvements in insulin sensitivity detected after overnight Acipimox administration were accompanied by signicant improvements in glucose tolerance [39], consistent with earlier ndings in the diabetic rat model. Hence, there is a gathering body of evidence linking elevated FFA and the development of muscle insulin resistance. Further clarication of the cellular mechanism for skeletal muscle insulin resistance is needed to determine the precise role of FFA. Key ndings to date are outlined below. Reduced glycogen synthesis plays a dominant role in inducing muscle insulin resistance In 1990, Shulman et al. used 13C nuclear magnetic resonance (NMR) spectroscopy to measure the rate of muscle glycogen synthesis in situ [40]. Under hyperglycaemic hyperinsulinaemic clamp conditions, they observed a 60% reduction in subjects with type 2 diabetes relative to nondiabetic, healthy individuals [40]. Muscle glycogen synthesis was estimated to account for 70% of whole-body glucose metabolism and 90% of nonoxidative glucose metabolism [40]. These results indicated that decreased muscle glycogen synthesis plays a central role in the insulin resistance seen in type 2 diabetes, although the precise rate-controlling step was not established. Decreased glucose transport leads to reduced muscle glycogen synthesis Candidate steps in the muscle glycogen synthesis pathway that may be responsible for this decrease include glycogen synthase, hexokinase and glucose transporter enzymes (Fig. 2). If glycogen synthase activity is suppressed in type 2 diabetes, then the rate of glycogen synthesis would be expected to decrease and glucose-6-phosphate would accumulate in the muscle of diabetic individuals in response to insulin. In fact, reductions in both muscle glycogen synthesis and glucose-6-phosphate concentrations have been observed in individuals with type 2 diabetes [40,41], consistent with either hexokinase activity or glucose transport being the rate limiting step in this pathway. In order to differentiate between decreased hexokinase activity and glucose transport as potential rate-limiting steps, the intracellular concentrations of muscle glycogen, glucose-6-phosphate and glucose were measured in patients

Figure 2 Potential rate-controlling steps responsible for reduced insulin-stimulated muscle glycogen synthesis in patients with type 2 diabetes mellitus; adapted with permission from Shulman G.I. J Clin Invest 2000;106:1716 [8].

with type 2 diabetes and in healthy, nondiabetic subjects using 13C and 31P NMR spectroscopy [42]. Under hyperglycaemic, hyperinsulinaemic conditions, the muscle glycogen synthesis rate and glucose-6-phosphate concentrations were found to be lower in diabetic individuals relative to control subjects, consistent with previous results. Since intracellular glucose represents an intermediate step between glucose transport and hexokinase activity, a substantial accumulation of intracellular glucose would be expected in subjects with type 2 diabetes if hexokinase activity was the rate-controlling step. In fact, the small change in intracellular glucose concentration detected in the diabetic patients was proportional to the observed change in glucose-6-phosphate, indicating that the reduction in muscle glycogen synthesis observed in type 2 diabetes primarily results from decreased insulin-stimulated glucose transport activity [42]. Identifying the role of FFA revisiting Randles hypothesis How are FFA involved in the development of muscle insulin resistance? Several modications have been made since Randle et al. rst proposed that elevated FFA induce insulin resistance in muscle solely by increasing the oxidation of fat relative to carbohydrate [33]. Boden et al. investigated potential mechanisms by which elevated FFA inhibit insulinstimulated glucose uptake both in healthy individuals [35] and in patients with type 2 diabetes (Fig. 3) [37]. In agreement with the previous ndings, they observed an inhibition of carbohydrate oxidation in healthy volunteers after an hour of lipid infusion under euglycaemic, hyperinsulinaemic conditions [35]. This effect alone is unlikely to be responsible for the signicant reduction in insulinstimulated glucose uptake that is observed some 2 3 h later [35]. In a subsequent study of type 2 diabetes patients, they reported that the decrease in insulin-stimulated glucose uptake occurring after 3 4 h lipid infusion is accompanied by a proportional inhibition of both glycogen synthesis and glycolysis [37]. Hence the major inhibitory effect of elevated FFA may occur at an early stage in glucose utilization, i.e. at the level of glucose transport/phosphorylation, since a primary effect on either glycogen synthesis or glycolysis would lead to disproportionate actions on these two

2002 Blackwell Science Ltd, European Journal of Clinical Investigation, 32 (Suppl. 3), 14 23

Free fatty acids in obesity and T2D

17

Figure 3 Inhibitory effects of FFAs on glucose utilization. The inhibition of carbohydrate oxidation (1) was the earliest demonstrable effect. It developed during the initial 2 h of lipid infusion but did not inhibit insulin-stimulated glucose uptake or glycolysis. The inhibition of glucose transport /phosphorylation (2) developed after 3 4 h, whereas the inhibition of glycogen synthesis (3) developed after 4 6 h of high plasma levels of free fatty acids; adapted with permission from Boden G. Endocrine Practice 2001;7:44 51 [93].

glucose transport /phosphorylation rather than at glycogen synthesis. It should be noted, however, that studies from another laboratory indicate that elevated FFA signicantly increase glucose-6-phosphate concentrations in healthy subjects [35]. One possible reason for this inconsistency is that the observed increase is an artefact caused by glycogen hydrolysis in the muscle biopsy sample that can be avoided by performing 31pNMR measurements in situ [36]. To differentiate between an action of FFA on phosphorylation or glucose transport activity, intramuscular glucose concentrations were measured using a novel 13C NMR technique as described in Cline et al. [42]. As before, if hexokinase activity is the rate-limiting step, a signicant accumulation of intracellular glucose is expected in response to high FFA concentrations. In fact, intracellular glucose concentrations decreased signicantly in healthy volunteers following a 5-h infusion of lipid, supporting the hypothesis that elevated FFA induce insulin resistance principally at the level of glucose transport [43]. Elevated FFA inhibit glucose transport by inhibiting insulin signalling How do elevated plasma FFA inhibit glucose transport? One possibility is a direct effect on the GLUT-4 glucose transporter, for example, FFA-induced changes in GLUT4 synthesis or vesicle trafcking, budding, or fusion [44]. Alternatively, elevated FFA may indirectly affect GLUT-4 activity by modifying upstream insulin signalling events. To examine the latter possibility, Dresner et al. [43] investigated the impact of raising FFA concentrations on phosphatidylinositol 3-kinase (PI 3-kinase), an important component of the insulin signalling cascade involved in regulating GLUT-4 translocation [45,46]. They found that the increase in PI 3-kinase activity observed in response to insulin stimulation in control subjects was virtually abolished in individuals given lipid infusions [43]. It is not clear from this study whether elevated plasma FFA inhibit insulin-stimulated PI 3-kinase activity directly or by modifying other components of the insulin signalling pathway. For a comprehensive review of the insulin signalling cascade, see White [47]. Subsequent experiments demonstrated that high FFA concentrations may affect several upstream proteins in the pathway, including insulin receptor substrate 1 (IRS-1) and protein kinase C theta (PKC; Fig. 4) [48]. In rats infused with lipid under euglycaemic hyperinsulinaemic conditions, a 50% decrease in PI 3kinase activity was observed compared with control rats. In these animals, insulin-stimulated IRS-1 tyrosine phosphorylation decreased, and membrane-bound PKC concentrations increased, relative to controls [48]. These results suggested that increased circulating FFA may induce insulin resistance in skeletal muscle by activating PKC, conrming previous observations where alterations in PKC and PKC were linked to the development of muscle insulin resistance in high-fat-fed rats [49]. It is proposed that the resulting decrease in IRS-1 tyrosine phosphorylation suppresses PI 3-kinase activity and decreases GLUT-4 translocation, culminating in a reduction of glucose transport [8].

pathways. There may also be a secondary abnormality in glycogen synthesis caused by elevated FFA concentrations, since a reduction in muscle glycogen synthase activity was observed following 4 6 h lipid infusion that was not apparent after 3 4 h [35]. From these data, as well as from measurements of intramuscular glucose-6-phosphate concentrations, these workers suggested that two different defects contribute to the impairment of glycogen synthesis depending on the FFA concentration. At FFA concentrations of 075 m, they found increased intramuscular glucose-6-phosphate concentrations suggesting a FFA-induced inhibition of glycogen synthase activity, whereas at FFA concentrations of 05 m, they were not able to nd any difference in intramuscular glucose-6-phosphate. From these data, the authors concluded that, at this lower FFA concentration, decreased muscle glucose transport/phosphorylation activity accounted for the reduced rates of muscle glucose uptake. Roden et al. conrmed that an increase in plasma FFA (2 m) caused a profound inhibition of insulin-mediated glucose uptake in skeletal muscle following 4 6 h of lipid infusion [36]. Since glucose-6-phosphate lies between glycogen synthase and hexokinase in the glycogen synthetic pathway (Fig. 2), accumulation of glucose-6-phosphate in response to lipid infusion would indicate an effect at the level of glycogen synthesis. In fact, under euglycaemic hyperinsulinaemic clamp conditions, they reported a signicant decrease in intramuscular glucose-6-phosphate concentrations in healthy individuals as measured by 31P NMR spectroscopy in response to lipid infusion associated with a 50% reduction in insulin-stimulated muscle glycogen synthesis as measured by 13C NMR spectroscopy [36]. These results denitively demonstrate that elevated plasma FFA inhibit insulin-stimulated glucose uptake at the level of

2002 Blackwell Science Ltd, European Journal of Clinical Investigation, 32 (Suppl. 3), 1423

18

G. Boden and G. I. Shulman

contrast, liver-LPL-overexpressing mice had a twofold increase in liver triglyceride content and were insulin resistant due to the impaired ability of insulin to suppress endogenous glucose production, which was associated with defects in insulin activation of IRS-2-associated PI 3-kinase activity. These defects in insulin action and signalling were associated with increases in intracellular fat metabolites (diacylglycerol, fatty acyl coenzyme A). These ndings suggest a direct and causative relationship between intracellular fat metabolites and insulin resistance mediated via alterations in the insulin signalling pathway.

Insulin resistance in liver


Figure 4 Proposed alternative mechanism for fatty acid-induced insulin resistance in human skeletal muscle; adapted with permission from Shulman G.I. J Clin Invest 2000;106:1716 [8].

If this proposal is true, then it is conceivable that increased intracellular lipid accumulation may inhibit insulin signalling. This is supported by evidence from A-ZIP/ F-1 transgenic mice. A-ZIP/F-1 is a protein that inhibits the function of transcription factors critical for adipose tissue development. Overexpression of this protein in mice causes abnormal fat deposition or lipodystrophy [50,51]. These lipodystrophic, or fatless, mice are severely insulin resistant and have a reduced response to insulin-stimulated IRS-1 and PI 3-kinase activity [51]. In addition, there is a twofold increase in intracellular lipid content in both muscle and liver. When normal adipose tissue is transplanted into these mice, insulin signalling and intracellular lipid content are simultaneously restored to normal levels, supporting the proposal that intracellular lipid accumulation may lead to abnormal insulin signalling and insulin resistance [51]. The introduction of normal adipose tissue into lipodystrophic mice may be analogous to the mechanism by which the thiazolidinediones (troglitazone, rosiglitazone and pioglitazone) improve insulin sensitivity [8]. For example, rosiglitazone activates peroxisome proliferator-activated receptor gamma (PPAR) in adipose tissue, leading to increased adipocyte differentiation, redistribution of fat from muscle and liver to subcutaneous adipose tissue and improved insulin sensitivity [52,53]. Specically, these changes may be mediated by alterations in muscle PKC and PKC activity, as evidenced by data from rosiglitazonetreated high-fat-fed rats [54]. Research is currently underway to investigate further how fat redistribution mediated by the thiazolidinediones affects the insulin signalling pathway and thus improves insulin sensitivity. Further evidence for the role of intracellular lipid in mediating insulin resistance has been obtained from transgenic mice with muscle-specic and liver-specic overexpression of lipoprotein lipase (LPL) [55]. Muscle-LPL-overexpressing mice had a threefold increase in muscle triglyceride content and were insulin resistant due to decreases in insulinstimulated glucose transport in skeletal muscle and insulin activation of IRS-1 associated PI 3-kinase activity. In

It is commonly accepted that endogenous glucose production (mainly contributed by the liver) is increased in type 2 diabetes and a close correlation has been observed between fasting plasma glucose concentrations and the rate of hepatic glucose production [56,57]. While endogenous glucose production is reported to be 25% higher in individuals with poorly controlled type 2 diabetes relative to healthy controls [58,59], there has been much controversy as to whether changes in gluconeogenesis or glycogenolysis are responsible for this increase. In order to address this question, Magnusson et al. used 13C NMR spectroscopy to assess directly the rates of net hepatic glycogenolysis and gluconeogenesis in control and poorly controlled type 2 diabetic subjects. Using this approach, they found that the increased rates of glucose production in the diabetic subjects could be entirely accounted for by a 60% increase in the rate of gluconeogenesis [58]. These data are supported by recent evidence using other approaches to assess gluconeogenesis in severely hyperglycaemic type 2 diabetes patients [60,61]. Although little is known concerning the mechanism of insulin resistance in the liver, a role for elevated plasma FFA has been postulated. While raised FFA concentrations promote gluconeogenesis in rat liver [62], the effect in humans has only been examined recently. Nicotinic acid was administered to healthy individuals from 16 to 20 h of a 24-h fast to lower plasma FFA [63]. Nicotinic acid discontinuation at 20 h resulted in a rebound in plasma FFA concentrations above basal fasting levels. Changes in gluconeogenesis mirrored the changes in plasma FFA, while glycogenolysis was inversely correlated with FFA concentrations. Overall, these changes in plasma FFA concentrations had little effect on endogenous glucose production as a balance was maintained between changes in gluconeogenesis and glycogenolysis due to hepatic autoregulation [63]. A similar approach was used in subjects with type 2 diabetes [64]. As before, reducing plasma FFA inhibited, while increasing FFA stimulated, gluconeogenesis but the impact on glycogenolysis differed to that in healthy subjects. When plasma FFA concentration decreased with nicotinic acid administration, glycogenolysis also decreased, causing a signicant overall reduction in endogenous glucose production. These observations support the idea that autoregulation of endogenous glucose production in

2002 Blackwell Science Ltd, European Journal of Clinical Investigation, 32 (Suppl. 3), 14 23

Free fatty acids in obesity and T2D

19

response to decreasing gluconeogenesis is abnormal in subjects with diabetes. Hence pharmacological agents that effectively lower FFA in people with type 2 diabetes are likely to have a signicant effect in reducing fasting plasma glucose [64]. In addition to stimulating gluconeogenesis, elevated FFA also affect insulin secretion [65] and hepatic insulin clearance [66], both of which affect endogenous glucose production. Further experiments are required to investigate the role of elevated FFA in suppressing insulinstimulated inhibition of endogenous glucose production.

Therapeutic approaches to lowering FFA/insulin resistance As outlined above, elevated plasma FFA contribute signicantly to skeletal muscle insulin resistance. While data concerning the involvement of FFA in hepatic insulin resistance appear contradictory, there is a suggestion that high circulating FFA may also stimulate endogenous glucose production. Therefore, it appears logical that lowering FFA should be a therapeutic target in obesity and type 2 diabetes. Weight loss leads to decreased circulating FFA and improved insulin sensitivity in individuals with type 2 diabetes [2123,67]. However, patient compliance to diet and exercise therapy is often poor and weight loss may not be maintained in the longer term, so additional therapeutic options are required [68]. Several pharmacological agents, including the thiazolidinediones, reduce elevated FFA and/or insulin resistance in people with type 2 diabetes [69]. For example, insulinstimulated glucose disposal improved by 40 50% following troglitazone therapy in subjects with type 2 diabetes [59,70], while a similar increase was demonstrated in individuals treated with pioglitazone [71]. Recent data indicate that the same is true for rosiglitazone [72]. It is proposed that the thiazolidinediones reduce insulin resistance by enhancing insulin-stimulated glucose transport, leading to increased rates of muscle glycogen synthesis and glucose oxidation [73]. Two main effects of these drugs on glucose transport are proposed: rstly, a direct increase in the expression of glucose transporters in skeletal muscle [73] and secondly, a reduction in circulating FFA concentrations leading to enhanced insulin signalling and increased GLUT-4 translocation [8]. In addition to their primary action in adipose tissue leading to important effects in skeletal muscle, the thiazolidinediones also improve insulin sensitivity in the liver. For example, high doses of troglitazone (600 mg) induced a small but signicant decrease in endogenous glucose production in subjects with type 2 diabetes, possibly as a result of decreased systemic FFA [59]. Recently, rosiglitazone treatment was shown to stimulate fat redistribution from the liver to subcutaneous tissue [74,75], indicating a potential mechanism by which the thiazolidinediones may improve hepatic insulin sensitivity [8]. These agents may also be of use in the treatment of nonalcoholic steatohepatitis (NASH) or fatty liver, a condition closely associated with abdominal obesity and insulin resistance. NASH is common in individuals with type 2 diabetes and is characterized

by hyperinsulinaemia and an increased supply of FFA to the liver [76]. In contrast to the thiazolidinediones, metformin appears to act primarily in the liver. The major effect of metformin is a reduction in endogenous glucose production [70,77]. Following some debate as to whether decreased endogenous glucose production results from a direct effect of metformin on gluconeogenesis [77] or glycogenolysis [78], recent studies by Hundal et al. using 13C NMR to measure directly the rates of net hepatic glycogenolysis and using 2H2O to measure directly the rates of gluconeogenesis found that metformin lowers the rates of glucose production exclusively by inhibiting gluconeogenesis [61]. In addition, metformin therapy appears to cause a small increase in peripheral glucose disposal [70]. Since chronic hyperglycaemia may contribute to peripheral insulin resistance [79], the effect of metformin on glucose disposal may be a secondary effect in response to reduced glucose toxicity [70].

Impact of FFA on insulin secretion


It is commonly accepted that changes in plasma FFA concentrations inuence insulin secretion. While it is well documented that acute FFA administration leads to stimulation of glucose-stimulated insulin secretion [80 84], the impact of chronic FFA exposure remains controversial. A growing body of evidence supports the hypothesis that chronically elevated FFA have a lipotoxic effect on the pancreas [11,12,85]. For example, prolonged lipid infusion has been shown to have a biphasic inuence on insulin secretion from the perfused pancreas of normal rats [13]. Following an initial stimulation of glucose-induced insulin secretion in response to lipid after 3 h, a marked decrease in secretion was observed following 48 h of lipid infusion. The observed inhibitory effect of lipid only affected glucose-induced secretion, was coupled to decreased glucose oxidation and appeared to be dependent on fatty acid oxidation [13]. The importance of FFA was later conrmed in isolated rat islets, where long-term exposure to FFA substantially inhibited glucose-stimulated insulin secretion by decreasing glucose oxidation [86]. Further support for the lipotoxicity hypothesis comes from experiments investigating the effect of troglitazone in islets from Zucker Diabetic Fatty (ZDF) rats [87]. These rats are hyperglycaemic with signicant abnormalities in insulin secretion that have been attributed to a 50-fold increase in islet triglyceride content relative to nondiabetic controls [88]. The inhibitory effect of triglyceride was conrmed by the improvement in insulin secretion following troglitazone incubation, which simultaneously reduced islet triglyceride content [87]. These data support the hypothesis that increased circulating FFA and islet lipid accumulation are responsible for the suppression of insulin secretion seen in ZDF rats. One possible mechanism for the proposed lipotoxicity is that surplus unoxidized FFA (characterized by elevated islet triglyceride content) increase the formation of nitric oxide, which induces -cell apoptosis [85].

2002 Blackwell Science Ltd, European Journal of Clinical Investigation, 32 (Suppl. 3), 1423

20

G. Boden and G. I. Shulman

In contrast, data in humans indicate an increase in glucose-stimulated insulin secretion following prolonged FFA administration [65]. In healthy volunteers, plasma glucose was clamped at a level that would stimulate insulin secretion and lipid was infused continuously for 48 h. Contrary to in vitro observations in rats [13], lipid administration increased the mean insulin secretion rate by almost 50% over the duration of the study compared with low FFA controls [65]. However, when these data were adjusted for insulin resistance [89], the enhanced insulin secretion rate was found to compensate precisely for the FFA-induced insulin resistance. Nevertheless, it may be necessary to study the effect of FFA over a more prolonged period. Since it is not feasible in humans to perform lipid infusions for periods longer than 48 h, other methods are needed that more closely relate to human obesity. One approach involved using nicotinic acid or Acipimox to lower elevated FFA concentrations in obese subjects with and without diabetes [17,39]. In these subjects, signicant decreases in insulin secretion (30 50%) were observed, indicating that basal plasma FFA support between 30 and 50% of basal insulin levels. These data suggest that physiological increases in plasma FFA concentrations in humans potentiate glucosestimulated insulin secretion and are unlikely to be lipotoxic to cells.

reects a genetic predisposition to -cell dysfunction. Despite their elevated plasma FFA concentrations, these 20% are unable to compensate for insulin resistance efciently, resulting in a decit in insulin secretion relative to insulin resistance and, ultimately, the onset of hyperglycaemia. The remaining 80%, while insulin resistant, can compensate by stimulating the pancreas via FFA to secrete more insulin and so do not progress to diabetes. Inevitably, there is a slight overcompensation and this leads to the hyperinsulinaemia that is characteristic in these individuals. If this theory is correct, then the insulin secretory response to FFA is likely to be reduced in diabetic individuals. However, preliminary experiments designed to test this hypothesis indicate a normal insulin secretory response in individuals with type 2 diabetes, either following lipid infusion or during the FFA rebound that occurs after discontinuation of nicotinic acid administration [94]. Interestingly, a twofold increase in ketone bodies was also observed in the diabetic subjects [94]. Since ketone bodies also act as insulin secretagogues [95 97], these data suggest that individuals with type 2 diabetes may increase plasma ketone bodies in order to compensate for the abnormal insulin secretory response to FFA. Validation of the relationship between FFA, ketone bodies and insulin resistance is required in order to explore this hypothesis further.

Do FFA provide the link between insulin resistance and -cell dysfunction?
It is now well established that high plasma FFA concentrations have a major inuence on the underlying pathophysiology of type 2 diabetes, primarily by inducing insulin resistance in the periphery. Recent evidence indicates that insulin resistance is also present in the cell and may contribute to the abnormalities in insulin secretion observed in type 2 diabetes [90,91]. Since high plasma FFA and increased intracellular lipid inhibit insulin signalling in muscle [48,49,51], it is possible that elevations in FFA may also contribute to -cell insulin resistance, representing a secondary pathogenetic mechanism. According to the lipotoxicity hypothesis, chronically elevated FFA may exert a direct toxic action on the cells of the pancreas by increasing the rate of nitric oxide formation [11,12,85]. In addition, evidence from transgenic mice expressing human islet amyloid polypeptide (IAPP) indicates that increased dietary fat consumption may play a role in the development of -cell dysfunction by inducing islet amyloid deposition [92]. However, despite growing support from in vitro studies, there is no substantial in vivo evidence to support the existence of lipotoxicity in humans as yet. The concept that chronically elevated FFA concentrations are solely responsible for both insulin resistance and -cell dysfunction is problematic. If this were true, since obesity is commonly associated with increased plasma FFA concentrations, all obese individuals would ultimately have type 2 diabetes. In fact, only approximately 20% of obese people go on to develop the condition [93] and this probably

Conclusion
Recent ndings support the idea of a causal relationship between elevated FFA and peripheral insulin resistance. Under specic circumstances, a rise in FFA concentration has important physiological consequences, for example, during pregnancy, elevated FFA induce insulin resistance and so valuable glucose is conserved for the developing foetus. The divide between physiology and pathophysiology appears to be narrow and high plasma FFA are intimately associated with obesity and type 2 diabetes. In obese individuals, plasma FFA concentrations are chronically elevated and, besides producing insulin resistance in skeletal muscle, may have additional actions in the liver and pancreas that may contribute to the development of type 2 diabetes. Although the exact relationship between elevated FFA, insulin resistance and -cell dysfunction requires further investigation, there is mounting evidence that elevated FFA should represent an important therapeutic target in obesity and type 2 diabetes. Agents such as the thiazolidinediones that provide long-term reductions in plasma FFA concentrations may be of considerable benet in the management of these and related conditions.

Acknowledgement
Dr G. Bodens work was supported by US Public Health Service Grants RO1AG 07988, RO1OK 588895 and MO1RR00349 (GCRC Branch of NCRR).

2002 Blackwell Science Ltd, European Journal of Clinical Investigation, 32 (Suppl. 3), 14 23

Free fatty acids in obesity and T2D

21

References
22 1 Gordon ES. Non-esteried fatty acids in blood of obese and lean subjects. Am J Clin Nutr 1960;8:7407. 2 Jensen MD, Haymond MW, Rizza RA, Cryer PE, Miles JM. Inuence of body fat distribution on free fatty acid metabolism in obesity. J Clin Invest 1989;83:116873. 3 Bjrntorp P, Bergman H, Varnauskas E. Plasma free fatty acid turnover rate in obesity. Acta Med Scand 1969;185:3516. 4 Reaven GM, Hollenbeck CB, Jeng CY, Wu MS, Chen YD. Measurement of plasma glucose, free fatty acid, lactate, and insulin for 24 h in patients with NIDDM. Diabetes 1988;37:1020 4. 5 Charles MA, Eschwege E, Thibult N, Claude JR, Warnet JM, Rosselin GE et al. The role of non-esteried fatty acids in the deterioration of glucose tolerance in Caucasian subjects: results of the Paris Prospective Study. Diabetologia 1997;40:11016. 6 Paolisso G, Tataranni PA, Foley JE, Bogardus C, Howard BV, Ravussin E. A high concentration of fasting plasma nonesteried fatty acids is a risk factor for the development of NIDDM. Diabetologia 1995;38:121317. 7 Boden G. Role of fatty acids in the pathogenesis of insulin resistance and NIDDM. Diabetes 1997;46:310. 8 Shulman GI. Cellular mechanisms of insulin resistance. J Clin Invest 2000;106:1716. 9 Reaven GM. Banting lecture 1988. Role of insulin resistance in human disease. Diabetes 1988;37:1595607. 10 Fagot-Campagna A, Balkau B, Simon D, Warnet JM, Claude JR, Ducimetiere P et al. High free fatty acid concentration: an independent risk factor for hypertension in the Paris Prospective Study. Int J Epidemiol 1998;27:808 13. 11 Unger RH. Lipotoxicity in the pathogenesis of obesitydependent NIDDM. Genetic and clinical implications. Diabetes 1995;44:863 70. 12 McGarry JD, Dobbins RL. Fatty acids, lipotoxicity and insulin secretion. Diabetologia 1999;42:12838. 13 Sako Y, Grill VE. A 48-hour lipid infusion in the rat timedependently inhibits glucose-induced insulin secretion and B cell oxidation through a process likely coupled to fatty acid oxidation. Endocrinology 1990;127:15809. 14 Coppack SW, Jensen MD, Miles JM. In vivo regulation of lipolysis in humans. J Lipid Res 1994;35:17793. 15 Felber JP, Magnenat G, Casthelaz M, Geser CA, Muller-Hess R, de Kalbermatten N et al. Carbohydrate and lipid oxidation in normal and diabetic subjects. Diabetes 1977;26:6939. 16 Boden G. Fuel metabolism in pregnancy and in gestational diabetes mellitus. Obstet Gynecol Clin North Am 1996;23:1 10. 17 Boden G, Chen X, Iqbal N. Acute lowering of plasma fatty acids lowers basal insulin secretion in diabetic and nondiabetic subjects. Diabetes 1998;47:1609 12. 18 Dobbins RL, Chester MW, Daniels MB, McGarry JD, Stein DT. Circulating fatty acids are essential for efcient glucose-stimulated insulin secretion after prolonged fasting in humans. Diabetes 1998;47:1613 18. 19 Stein DT, Stevenson BE, Chester MW, Basit M, Daniels MB, Turley SD et al. The insulinotropic potency of fatty acids is inuenced profoundly by their chain length and degree of saturation. J Clin Invest 1997;100:398403. 20 Sims EA, Danforth EJ, Horton ES, Bray GA, Glennon JA, Salans LB. Endocrine and metabolic effects of experimental obesity in man. Recent Prog Horm Res 1973;29:45796. 21 Beck-Nielsen H, Pedersen O, Lindskov HO. Normalization of the insulin sensitivity and the cellular insulin binding during

23

24

25

26 27 28

29

30

31

32

33

34

35

36

37

38

39

treatment of obese diabetics for one year. Acta Endocrinol (Copenh) 1979;90:103 12. Freidenberg GR, Reichart D, Olefsky JM, Henry RR. Reversibility of defective adipocyte insulin receptor kinase activity in non-insulin-dependent diabetes mellitus. Effect of weight loss. J Clin Invest 1988;82:1398 406. Bak JF, Mller N, Schmitz O, Saaek A, Pedersen O. In vivo insulin action and muscle glycogen synthase activity in type 2 (non-insulin-dependent) diabetes mellitus: effects of diet treatment. Diabetologia 1992;35:777 84. Hotamisligil GS, Spiegelman BM. Tumor necrosis factor : a key component of the obesity-diabetes link. Diabetes 1994;43:12718. Greenberg AS, McDaniel ML. Identifying the links between obesity, insulin resistance and -cell function: potential role of adipocyte-derived cytokines in the pathogenesis of type 2 diabetes. Eur J Clin Invest 2002;32 (Suppl. 3):24 34. Girard J. Is leptin the link between obesity and insulin resistance? Diabetes Metab 1997;23:16 24. Bergman RN, Mittelman SD. Central role of the adipocyte in insulin resistance. J Basic Clin Physiol Pharmacol 1998;9:20521. Perseghin G, Ghosh S, Gerow K, Shulman GI. Metabolic defects in lean nondiabetic offspring of NIDDM parents: a cross-sectional study. Diabetes 1997;46:10019. Krssak M, Falk PK, Dresner A, DiPietro L, Vogel SM, Rothman DL et al. Intramyocellular lipid concentrations are correlated with insulin sensitivity in humans: a 1H NMR spectroscopy study. Diabetologia 1999;42:113 16. Perseghin G, Scifo P, De Cobelli F, Pagliato E, Battezzati A, Arcelloni C et al. Intramyocellular triglyceride content is a determinant of in vivo insulin resistance in humans: a 1H-13C nuclear magnetic resonance spectroscopy assessment in offspring of type 2 diabetic parents. Diabetes 1999;48:1600 6. Boden G, Lebed B, Schatz M, Homko C, Lemieux S. Effects of acute changes of plasma free fatty acids on intramyocellular fat content and insulin resistance in healthy subjects. Diabetes 2001;50:1612 17. DeFronzo RA, Gunnarsson R, Bjorkman O, Olsson M, Wahren J. Effects of insulin on peripheral and splanchnic glucose metabolism in noninsulin-dependent (type II) diabetes mellitus. J Clin Invest 1985;76:149 55. Randle PJ, Garland PB, Hales CN, Newsholme EA. The glucose fatty-acid cycle: its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus. Lancet 1963;1:785 9. Boden G, Jadali F, White J, Liang Y, Mozzoli M, Chen X et al. Effects of fat on insulin-stimulated carbohydrate metabolism in normal men. J Clin Invest 1991;88:960 6. Boden G, Chen X, Ruiz J, White JV, Rossetti L. Mechanisms of fatty acid-induced inhibition of glucose uptake. J Clin Invest 1994;93:2438 46. Roden M, Price TB, Perseghin G, Petersen KF, Rothman DL, Cline GW et al. Mechanism of free fatty acid-induced insulin resistance in humans. J Clin Invest 1996;97:2859 65. Boden G, Chen X. Effects of fat on glucose uptake and utilization in patients with non-insulin-dependent diabetes. J Clin Invest 1995;96:12618. Reaven GM, Chang H, Ho H, Jeng CY, Hoffman BB. Lowering of plasma glucose in diabetic rats by antilipolytic agents. Am J Physiol 1988;254:E23 E30. Santomauro AT, Boden G, Silva ME, Rocha DM, Santos RF, Ursich MJ et al. Overnight lowering of free fatty acids with Acipimox improves insulin resistance and glucose tolerance in obese diabetic and nondiabetic subjects. Diabetes 1999;48:1836 41.

2002 Blackwell Science Ltd, European Journal of Clinical Investigation, 32 (Suppl. 3), 1423

22

G. Boden and G. I. Shulman

40 Shulman GI, Rothman DL, Jue T, Stein P, DeFronzo RA, Shulman RG. Quantitation of muscle glycogen synthesis in normal subjects and subjects with non-insulin-dependent diabetes by 13C nuclear magnetic resonance spectroscopy. N Engl J Med 1990;322:223 8. 41 Rothman DL, Shulman RG, Shulman GI. 31P nuclear magnetic resonance measurements of muscle glucose-6phosphate. Evidence for reduced insulin-dependent muscle glucose transport or phosphorylation activity in non-insulin-dependent diabetes mellitus. J Clin Invest 1992;89:1069 75. 42 Cline GW, Petersen KF, Krssak M, Shen J, Hundal RS, Trajanoski Z et al. Impaired glucose transport as a cause of decreased insulin-stimulated muscle glycogen synthesis in type 2 diabetes. N Engl J Med 1999;341:2406. 43 Dresner A, Laurent D, Marcucci M, Grifn ME, Dufour S, Cline GW et al. Effects of free fatty acids on glucose transport and IRS-1-associated phosphatidylinositol 3-kinase activity. J Clin Invest 1999;103:253 9. 44 Kahn BB. Lilly lecture 1995. Glucose transport: pivotal step in insulin action. Diabetes 1996;45:164454. 45 Okada T, Kawano Y, Sakakibara T, Hazeki O, Ui M. Essential role of phosphatidylinositol 3-kinase in insulin-induced glucose transport and antilipolysis in rat adipocytes. Studies with a selective inhibitor wortmannin. J Biol Chem 1994;269:3568 73. 46 Clarke JF, Young PW, Yonezawa K, Kasuga M, Holman GD. Inhibition of the translocation of GLUT1 and GLUT4 in 3T3L1 cells by the phosphatidylinositol 3-kinase inhibitor, wortmannin. Biochem J 1994;300:6315. 47 White MF. The insulin signalling system and the IRS proteins. Diabetologia 1997;40 (Suppl. 2):S217. 48 Grifn ME, Marcucci MJ, Cline GW, Bell K, Barucci N, Lee D et al. Free fatty acid-induced insulin resistance is associated with activation of protein kinase C theta and alterations in the insulin signaling cascade. Diabetes 1999;48:12704. 49 Schmitz-Peiffer C, Browne CL, Oakes ND, Watkinson A, Chisholm DJ, Kraegen EW et al. Alterations in the expression and cellular localization of protein kinase C isozymes epsilon and theta are associated with insulin resistance in skeletal muscle of the high-fat-fed rat. Diabetes 1997;46:169 78. 50 Gavrilova O, Marcus-Samuels B, Graham D, Kim JK, Shulman GI, Castle AL et al. Surgical implantation of adipose tissue reverses diabetes in lipoatrophic mice. J Clin Invest 2000;105:271 8. 51 Kim JK, Gavrilova O, Chen Y, Reitman ML, Shulman GI. Mechanism of insulin resistance in A-ZIP/ F-1 fatless mice. J Biol Chem 2000;275:8456 60. 52 Oakes ND, Kennedy CJ, Jenkins AB, Laybutt DR, Chisholm DJ, Kraegen EW. A new antidiabetic agent, BRL 49653, reduces lipid availability and improves insulin action and glucoregulation in the rat. Diabetes 1994;43:120310. 53 Oakes ND, Camilleri S, Furler SM, Chisholm DJ, Kraegen EW. The insulin sensitizer, BRL 49653, reduces systemic fatty acid supply and utilization and tissue lipid availability in the rat. Metabolism 1997;46:935 42. 54 Schmitz-Peiffer C, Oakes ND, Browne CL, Kraegen EW, Biden TJ. Reversal of chronic alterations of skeletal muscle protein kinase C from fat-fed rats by BRL-49653. Am J Physiol 1997;273:E915 E921. 55 Kim JK, Fillmore JJ, Chen Y, Yu C, Moore IK, Pypaert M et al. Tissue-specic overexpression of lipoprotein lipase causes tissue-specic insulin resistance. Proc Natl Acad Sci USA 2001;98:7522 7.

56 DeFronzo RA, Simonson D, Ferrannini E. Hepatic and peripheral insulin resistance: a common feature of type 2 (noninsulin-dependent) and type 1 (insulin-dependent) diabetes mellitus. Diabetologia 1982;23:31319. 57 Jeng CY, Sheu WH, Fuh MM, Chen YD, Reaven GM. Relationship between hepatic glucose production and fasting plasma glucose concentration in patients with NIDDM. Diabetes 1994;43:14404. 58 Magnusson I, Rothman DL, Katz LD, Shulman RG, Shulman GI. Increased rate of gluconeogenesis in type II diabetes mellitus. A 13C nuclear magnetic resonance study. J Clin Invest 1992;90:1323 7. 59 Maggs DG, Buchanan TA, Burant CF, Cline G, Gumbiner B, Hsueh WA et al. Metabolic effects of troglitazone monotherapy in type 2 diabetes mellitus. A randomized, double-blind, placebo-controlled trial. Ann Intern Med 1998;128:176 85. 60 Boden G, Chen X, Stein TP. Gluconeogenesis in moderately and severely hyperglycemic patients with type 2 diabetes mellitus. Am J Physiol Endocrinol Metab 2001;280:E23 E30. 61 Hundal RS, Krssak M, Dufour S, Laurent D, Lebon V, Chandramouli V et al. Mechanism by which metformin reduces glucose production in type 2 diabetes. Diabetes 2000;49:2063 9. 62 Williamson JR, Kreisberg RA, Felts PW. Mechanism for the stimulation of gluconeogenesis by fatty acids in perfused rat liver. Proc Natl Acad Sci USA 1966;56:24754. 63 Chen X, Iqbal N, Boden G. The effects of free fatty acids on gluconeogenesis and glycogenolysis in normal subjects. J Clin Invest 1999;103:365 72. 64 Boden G, Chen X, Capulong E, Mozzoli M. Effects of free fatty acids on gluconeogenesis and autoregulation of glucose production in type 2 diabetes. Diabetes 2001;50:810 16. 65 Boden G, Chen X, Rosner J, Barton M. Effects of a 48-h fat infusion on insulin secretion and glucose utilization. Diabetes 1995;44:1239 42. 66 Wiesenthal SR, Sandhu H, McCall RH, Tchipashvili V, Yoshii H, Polonsky K et al. Free fatty acids impair hepatic insulin extraction in vivo. Diabetes 1999;48:766 74. 67 Henry RR, Wallace P, Olefsky JM. Effects of weight loss on mechanisms of hyperglycemia in obese non-insulin-dependent diabetes mellitus. Diabetes 1986;35:990 8. 68 Mann JI. Can dietary intervention produce long-term reduction in insulin resistance? Br J Nutr 2000;83 (Suppl. 1):S169 S172. 69 Lebovitz HE, Banerji MA. Insulin resistance and its treatment by thiazolidinediones. Recent Prog Horm Res 2001;56:265 94. 70 Yu JG, Kruszynoka YT, Mulford MI, Olefsky JM. A comparison of troglitazone and metformin on insulin requirements in euglycemic intensively treated type 2 diabetes patients. Diabetes 1999;48:2414 21. 71 Yamasaki Y, Kawamori R, Wasada T, Sato A, Omori Y, Eguchi H et al. Pioglitazone (AD-4833) ameliorates insulin resistance in patients with NIDDM. AD-4833 Glucose Clamp Study Group, Japan Tohoku. J Exp Med 1997;183:173 83. 72 Carey DG, Cowin GJ, Galloway GJ, Dodrell D, Richards J, Jones NP. Rosiglitazone increases insulin sensitivity and reduces factors associated with insulin resistance in type 2 diabetics. Diabetes Res Clin Pract 2000;50 (Suppl. 1):S64. 73 Petersen KF, Krssak M, Inzucchi S, Cline GW, Dufour S, Shulman GI. Mechanism of troglitazone action in type 2 diabetes. Diabetes 2000;49:82731. 74 Carey DG, Galloway D, Dodrell D, Richards J, Jones NP, Zhou B. Rosiglitazone reduces hepatic fat and increases subcutaneous but not intra-abdominal fat depots. Diabetologia 2000;43 (Suppl. 1):A68.

2002 Blackwell Science Ltd, European Journal of Clinical Investigation, 32 (Suppl. 3), 14 23

Free fatty acids in obesity and T2D

23

75 Mayerson AB, Hendal RS, Dufour S, Lebon V, Befroy D, Cline GW et al. The effects of rosiglatazone on insulin sensitivity, lipolysis and tepanic and skeletal muscle triglyceride content in patients with type 2 diabetes. Diabetes 2002;51:797 802. 76 Luyckx FH, Lefebvre PJ, Scheen AJ. Non-alcoholic steatohepatitis: association with obesity and insulin resistance, and inuence of weight loss. Diabetes Metab 2000;26:98 106. 77 Stumvoll M, Nurjhan N, Perriello G, Dailey G, Gerich JE. Metabolic effects of metformin in non-insulin-dependent diabetes mellitus. N Engl J Med 1995;333:5504. 78 Cusi K, Consoli A, DeFronzo RA. Metabolic effects of metformin on glucose and lactate metabolism in noninsulindependent diabetes mellitus. J Clin Endocrinol Metab 1996;81:4059 67. 79 Rossetti L, Giaccari A, DeFronzo RA. Glucose toxicity. Diabetes Care 1990;13:610 30. 80 Felber JP, Vannotti A. Effects of fat infusions on glucose tolerance and insulin plasma levels. Med Exp 1964;10:153 6. 81 Malaisse WJ, Malaisse-Lagae F. Stimulation of insulin secretion by noncarbohydrate metabolites. J Lab Clin Med 1968;72:438 48. 82 Pelkonen R, Miettinen TA, Taskinen MR, Nikkila EA. Effect of acute elevation of plasma glycerol, triglyceride and FFA levels on glucose utilization and plasma insulin. Diabetes 1968;17:76 82. 83 Crespin SR, Greenough WB, Steinberg D. Stimulation of insulin secretion by long-chain free fatty acids. A direct pancreatic effect. J Clin Invest 1973;52:197984. 84 Goberna R, Tamarit JJ, Osorio J, Fussganger R, Tamarit J, Pfeiffer EF. Action of B-hydroxy butyrate, acetoacetate and palmitate on the insulin release in the perfused isolated rat pancreas. Horm Metab Res 1974;6:25660. 85 Unger RH, Zhou YT. Lipotoxicity of beta-cells in obesity and in other causes of fatty acid spillover. Diabetes 2001;50 (Suppl. 1):S118 S121.

86 Zhou YP, Grill VE. Long-term exposure of rat pancreatic islets to fatty acids inhibits glucose-induced insulin secretion and biosynthesis through a glucose fatty acid cycle. J Clin Invest 1994;93:870 6. 87 Shimabukuro M, Zhou YT, Lee Y, Unger RH. Troglitazone lowers islet fat and restores beta cell function of Zucker diabetic fatty rats. J Biol Chem 1998;273:354750. 88 Lee Y, Hirose H, Zhou YT, Esser V, McGarry JD, Unger RH. Increased lipogenic capacity of the islets of obese rats: a role in the pathogenesis of NIDDM. Diabetes 1997;46:408 13. 89 Bergman RN, Ader M. Free fatty acids and pathogenesis of type 2 diabetes mellitus. Trends Endocrinol Metab 2000;11:351 6. 90 Withers DJ, Gutierrez JS, Towery H, Burks DJ, Ren JM, Previs S et al. Disruption of IRS-2 causes type 2 diabetes in mice. Nature 1998;391:900 4. 91 Kulkarni RN, Bruning JC, Winnay JN, Postic C, Magnuson MA, Kahn CR. Tissue-specic knockout of the insulin receptor in pancreatic beta cells creates an insulin secretory defect similar to that in type 2 diabetes. Cell 1999;96:329 39. 92 Kahn SE, Andrikopoulos S, Verchere CB. Islet amyloid: a longrecognized but underappreciated pathological feature of type 2 diabetes. Diabetes 1999;48:24153. 93 Boden G. Free fatty acids the link between obesity and insulin resistance. Endocr Pract 2001;7:44 51. 94 Boden G, Chen X. Effects of fatty acids and ketone bodies on basal insulin secretion in type 2 diabetes. Diabetes 1999;48:577 83. 95 Madison LL, Mebane D, Unger RH, Lochner A. The hypoglycemic action of ketones II. Evidence for a stimulatory feedback of ketones on the pancreatic beta cells. J Clin Invest 1964;43:408 11. 96 Jenkins DJ, Hunter WM, Goff DV. Ketone bodies and evidence for increased insulin secretion. Nature 1970;227:384 5. 97 Balasse EO, Ooms HA, Lambilliotte JP. Evidence for a stimulatory effect of ketone bodies on insulin secretion in man. Horm Metab Res 1970;2:3712.

2002 Blackwell Science Ltd, European Journal of Clinical Investigation, 32 (Suppl. 3), 1423

You might also like