Reviews/Commentaries/ADA Statements REVIEW ARTICLE

Lipids and Glucose in What is the cause and effect?

1 GUENTHER BODEN, MD viving times of famine would be increased 2 MARKKU LAAKSO, MD if they could maximize energy storage (as fat) during times of surplus food availabil- ity. The stored fat could then be used dur- ing periods of starvation. istorically, type 2 diabetes was con- If this had happened, our understanding Adipocytes take up and store FFAs. sidered to revolve around a glu- of the pathogenesis of type 2 diabetes may The two main types of are H cose-insulin axis. The foundations have developed along a different route subcutaneous and visceral adipose tissue. for this thinking were probably laid down and led us more quickly to our current About 80% of body fat is located in the by two momentous discoveries in diabe- awareness that , or more accu- subcutaneous adipose tissue, and ϳ10% tes research. According to popular leg- rately, the products of excess adipose tis- is located in visceral adipose tissue (7). end, Oskar Minkowski noticed that urine sue, precede the perturbations of glucose The remainder is in various other loca- from his pancreatectomized dogs at- metabolism. tions, such as perirenal and peritoneal ad- tracted an inordinate number of flies. He It is now apparent that elevation of ipose tissue (7). is then alleged to have tasted the urine plasma free fatty acids (FFAs) plays a piv- The body uses its fat reserves during and noted its sweetness. From this ob- otal role in the development of type 2 di- periods of low energy intake, when FFAs servation came the supposition that the abetes by causing insulin resistance. Type are being released for other tissues to be pancreas produced a substance that con- 2 diabetes develops because pancreatic used as fuel. However, if plasma FFA lev- trolled sugar concentration, and diabetes ␤-cells eventually fail to produce enough els are elevated for more than a few hours, occurred in the absence of this substance. insulin to compensate for the ongoing in- they will cause insulin resistance (8). In The second landmark was the discovery sulin resistance. There is a tight associa- certain conditions, the FFA-induced in- by Frederick Banting that insulin was the tion between type 2 diabetes and sulin resistance has the beneficial effect of active element from the pancreas. As a dyslipidemia. The latter is characterized preserving carbohydrate for use by vital consequence of these two discoveries, the by raised small, dense LDL levels, ele- tissues, such as the central nervous sys- concept that the glucose-insulin relation- vated levels of triglycerides, and low lev- tem. This is the case during starvation and ship was the central element of fuel me- els of HDL. Individually, the latter two during the second half of pregnancy, tabolism gained a firm hold. Diabetes has factors increase the risk of cardiovascular when the insulin resistance of the mother since been considered to be a disorder disease, and the combination of the two is preserves glucose for the growing fetus. primarily associated with abnormal glu- a risk factor for cardiovascular heart dis- cose metabolism. This notion is given cre- ease that is at least as strong as a high level PATHOLOGICAL dence by the considerable amount of data of LDL cholesterol (2). Some (3,4) but not CONSEQUENCES OF FAT indicating that the chronic elevation of all (5) studies have demonstrated that ACCUMULATION plasma glucose causes many of the micro- increased levels of small, dense LDL are In contrast to its beneficial effects during vascular complications of diabetes. associated with an elevated risk of cardio- periods of starvation and gestation, dur- In 1992, McGarry (1) asked what vascular disease. ing prolonged periods of energy excess, would have happened if Minkowski had the FFA-induced insulin resistance be- lacked a sense of taste but had a good EVOLUTIONARY comes counterproductive because there is nose. If that had been the case, he may RATIONALE FOR FAT no need for preservation of carbohydrate have smelled acetone, and this may have ACCUMULATION for use by vital tissues. Under these con- led him to the conclusion that removal of According to the thrifty gene hypothesis ditions, glucose levels remain normal the pancreas affects fatty acid metabolism. (6), the probability of an individual sur- only as long as the basal and postprandial ●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●●● secretion of insulin by the pancreas is From the 1Division of Endocrinology/Diabetes/Metabolism and the General Clinical Research Center, Tem- 2 sufficient to compensate for the insulin ple University Hospital, Philadelphia, Pennsylvania; and the Department of Medicine, University of Kuopio, resistance. Kuopio, Finland. Address correspondence and reprint requests to G. Boden, MD, Temple University Hospital, 3401 N. Broad St., Philadelphia, PA 19140. E-mail: [email protected]. INDUCTION OF INSULIN Received for publication 27 May 2004 and accepted in revised form 10 June 2004. G.B. and M.L. have received honoraria from AstraZeneca Pharmaceuticals. RESISTANCE BY FFAS Abbreviations: DAG, diacylglycerol; FFA, free fatty acid; IL, interleukin; IRS, insulin receptor substrate; The mechanisms by which elevated FFA NF, nuclear factor; NHANES III, Third National Health and Nutrition Examination Survey; PKC, protein levels result in insulin resistance have kinase C; PPAR, peroxisome proliferator–activated receptor; TNF, tumor necrosis factor; TZD, thiazo- been determined in skeletal muscle, lidinedione. A table elsewhere in this issue shows conventional and Syste`me International (SI) units and conversion where most insulin-stimulated glucose factors for many substances. uptake occurs. Formerly, it was believed © 2004 by the American Diabetes Association. that FFA production from overloaded fat

DIABETES CARE, VOLUME 27, NUMBER 9, SEPTEMBER 2004 2253 Lipids, glucose, and type 2 diabetes

Figure 1—Potential mechanism of FFA on insulin resistance and atherogenesis in hu- man muscle. The key initiating event is an increase in plasma FFA followed by in- creased uptake of FFA into muscle. This leads to intramyocellular accumulation of fatty acyl-CoA and DAG and activation of PKC (the ␤ II and ␦ isoforms). It is assumed that activation of PKC interrupts insulin sig- naling by serine phosphorylation of IRS-1, resulting in a decrease in tyrosine phosphor- ylation of IRS-1. At the same time, activation of PKC also leads to production of inflam- matory and proatherogenic proteins through activation of the I␬B-␣/NF-␬B pathway. The broken lines indicate that ac- tivation of PKC by reactive oxygen species (ROS) and activation of the I␬B-␣/NF-␬B pathway by ROS has not been demonstrated in human muscle but only in bovine aortic smooth muscle and endothelial cells (ref. 84). PI, phosphatidylinositol. cells disrupted glucose homeostasis via However, it is probably not the accumu- can cause insulin resistance is by increas- the Randle glucose–fatty acid cycle. First lation of fat in muscle cells that causes ing oxidative stress (26). Reactive oxygen described by Randle et al. (9) in 1963, the insulin resistance but rather the accumu- species can activate PKC and the NF-␬B hypothesis was that glucose uptake is re- lation of other metabolites, including di- pathway (Fig. 1) and thereby contribute duced when tissue energy needs are being acylglycerol (DAG), that occur at the same to insulin resistance (17,27). met by FFA oxidation. The oxidation of time (17). FFAs also affect the functioning of in- FFAs was thought to result in decreased DAG, which is an intermediate of tri- sulin in the liver and thus contribute to glucose oxidation and an increase in in- glyceride metabolism, is a potent activa- hepatic overproduction of glucose and to tracellular citrate levels, which would tor of protein kinase C (PKC) (18). In elevated circulating blood glucose levels decrease glycolysis and glucose uptake. In healthy volunteers, along with the rise in (28). The main role of insulin in the liver is vivo and in vitro studies have only par- intramyocellular levels of DAG, there was control of glucose production. The mecha- tially confirmed Randle’s hypothesis (8, a concomitant increase in PKC activity nism by which insulin acutely (within 1–2 10–12). For instance, insulin-stimulated (17). PKC is an enzyme that can phos- h) suppresses hepatic glucose production is glucose uptake has been found to proceed phorylate serine and threonine residues by inhibiting glycogenolysis (29). FFAs normally for several hours after maximal on both the insulin receptor (19,20) and produce insulin resistance in the liver by inhibition of carbohydrate oxidation by insulin receptor substrate (IRS)-1 (20, inhibiting the acute insulin suppression of fatty acids (8,11–13). 21). The latter two molecules are impor- glycogenolysis (30). Insulin also promotes It is now thought that FFAs induce tant for insulin signaling. Serine phos- hepatic uptake of FFAs and production of insulin resistance in human muscle at the phorylation of IRS-1 can lead to its de- intracellular triglycerides. Thus, insulin re- level of insulin-stimulated glucose trans- struction and to insulin resistance (22). sistance in the liver may contribute to ele- port or phosphorylation by impairing the A change in intracellular DAG levels vated plasma FFA levels. insulin-signaling pathway (Fig. 1) is also accompanied by activation of the An increase in visceral fat could also (13,14). In a study of nondiabetic men nuclear factor (NF)-␬B pathway (Fig. 1) cause insulin resistance by mechanisms and women, insulin resistance developed (17). NF-␬B has been linked to fatty acid– that do not directly involve FFAs (Fig. 1). 2–4 h after an acute elevation in plasma induced impairment of insulin action in Adipose tissue is a source of inflammatory FFA concentration and took a similar rodents (23,24). NF-␬B is also increas- mediators, such as tumor necrosis factor amount of time to disappear after plasma ingly recognized as playing a crucial role (TNF)-␣ (31) and interleukin (IL)-6 (32) FFA levels had returned to normal (8,15). in the pathogenesis of coronary artery dis- and peptides that include resistin (33), This delay indicates that FFAs have an in- ease (25). Thus, the activation of NF-␬B leptin (34), and adiponectin (35). So far, direct effect, a contention that is sup- may also help to explain the increased however, the physiological relevance of ported by the observation that acute prevalence of vascular disease in obese these adipokines for the development of increases in plasma FFAs increased tri- patientswithtype2diabetes.Consequent- insulin resistance in humans has not been glyceride content in muscle cells of hu- ly, lowering FFA concentrations may pre- established. man volunteers (16). This rise in vent activation of the NF-␬B pathway and intramyocellular triglyceride concentra- have benefits beyond increasing insulin ADIPONECTIN AND tion occurred several hours after the ele- sensitivity and improving regulation of INSULIN RESISTANCE vation of FFAs and coincided with the glucose levels. Of all the adipokines mentioned, adi- development of insulin resistance (16). Another mechanism by which FFAs ponectin is most likely to affect insulin

2254 DIABETES CARE, VOLUME 27, NUMBER 9, SEPTEMBER 2004 Boden and Laakso sensitivity. Adiponectin is produced ex- basis of this new definition, the actual nicotinic acid analogs are drugs that lower clusively in adipocytes. It stimulates fatty proportion of overweight people with im- plasma FFA levels. Their usefulness is acid oxidation, decreases plasma triglyc- paired glucose metabolism will be higher limited, however, because the initial low- erides, and improves glucose metabolism than the estimates from published sur- ering of plasma FFA levels by nicotinic by increasing insulin sensitivity (36). Adi- veys (including NHANES III) that used acid is invariably followed by a sharp FFA ponectin levels are negatively correlated the older definition. rebound (51) that increases insulin resis- with the development of insulin resis- Recently, it has become clearer why tance, at least temporarily. There has tance in rhesus monkeys (37). The plasma many obese, insulin-resistant people will therefore been considerable interest in levels of adiponectin in Caucasians and never develop diabetes. In obese people drugs that activate the peroxisome prolif- Pima Indians are negatively correlated with normal pancreatic ␤-cells, FFAs are erator–activated receptors (PPARs), with percent body fat and plasma insulin potent insulin secretagogues and can which are nuclear transcription factors levels and are positively correlated with compensate for the insulin resistance that that regulate the expression of numerous insulin-mediated glucose uptake (35,38). they produce. Acute elevations of plasma genes involved in lipid and carbohydrate In addition, plasma levels of adiponectin FFAs have long been known to stimulate metabolism, inflammation, and vascular are lower in individuals with type 2 dia- insulin secretion (42). More importantly, tone. betes than in age- and body mass– prolonged elevations of plasma FFAs matched individuals without diabetes (2–4 days) have been shown to potentiate PPAR-␥ (35). Thus, the impaired release of adipo- glucose-stimulated insulin secretion in Thiazolidinediones (TZDs), in contrast to nectin that occurs in obesity may contrib- healthy volunteers (43–45). Moreover, nicotinic acid analogs, lower plasma FFA ute to insulin resistance and development when chronic elevated plasma FFA levels levels without a rebound phenomenon of type 2 diabetes. were lowered in obese diabetic and non- (52–54). The binding affinity between The mechanism leading to decreased diabetic subjects, insulin secretion rates TZDs and PPAR-␥ correlates well with adiponectin levels in obesity (39) is not decreased by 30–50% (46), indicating their insulin-sensitizing activity. There- clear. Adiponectin is inhibited by insulin that the elevated plasma FFAs had sup- fore, it is generally accepted that TZDs and TNF-␣. Therefore, hyperinsulinemia ported 30–50% of basal insulin secretion. exert their action through PPAR-␥ caused by obesity-induced insulin resis- In contrast, in first-degree relatives of (55,56). PPAR-␥ is expressed at highest tance, together with enhanced TNF-␣ patients with type 2 diabetes (i.e., in indi- concentrations in adipose tissue and at expression, may contribute to reduced viduals who are genetically predisposed much lower concentrations in liver and adiponectin secretion. It has also been to develop diabetes), FFAs are unable to muscle (57,58), suggesting that the pri- suggested that visceral adipose tissue may fully compensate with adequately in- mary action of TZDs is on adipose tissue. produce an as yet unidentified substance creased insulin secretion for the insulin In support, it has been shown that TZDs that destabilizes adiponectin mRNA (40). resistance that they produce (45,47). This play an important role in adipocyte devel- defect in FFA-stimulated insulin secre- opment (59) and that they lower plasma FFAs AND INSULIN tion can also be demonstrated in patients levels of FFAs (Fig. 2) (52–54). In addi- SECRETION with impaired glucose tolerance (47,48) tion, TZDs have been postulated to im- Insulin resistance in the liver results in and in patients with overt type 2 diabetes prove insulin sensitivity by redistributing overproduction of glucose, whereas insu- (46). These observations suggest that the fat from visceral to subcutaneous adipose lin resistance in skeletal muscle produces obese individuals who develop type 2 di- tissue (60,61) and by increasing blood underutilization of glucose. Because FFAs abetes have a genetic predisposition to levels of adiponectin (62,63). can induce insulin resistance in both liver pancreatic ␤-cell failure. This hypothesis The available TZDs (pioglitazone and and muscle, all overweight or obese peo- is supported by the fact that these individ- rosiglitazone) have a generally good safety ple, who are likely to have elevated uals have a defect in both FFA and glu- and tolerability profile. They have been plasma FFA levels, might be expected to cose-stimulated insulin secretion (49) associated with increased development of have elevated glucose levels. This, how- and by in vitro studies showing that basal edema, which can exacerbate or lead to ever, is not the case because only approx- insulin secretion from normal Wistar rat congestive heart failure (64,65) in high- imately half of overweight individuals islets increased when cultured for 7 days risk patients with preexisting vascular have abnormal glucose levels; in the Third in 2 mmol/l FFAs but decreased in islets disease (66,67). The highest incidences of National Health and Nutrition Examina- from Zucker fatty rats who are predis- edema and congestive heart failure were tion Survey (NHANES III), 23% of over- posed to develop diabetes (50). seen when TZDs were used in combina- weight or obese (BMI Ն25 kg/m2) tion with insulin (65,68,69). Alongside individuals had impaired fasting glucose PEROXISOME improvements in glycemia and in some (fasting glucose concentration of 110– PROLIFERATOR–ACTIVATED lipid parameters, e.g., HDL, clinical trials 125 mg/dl) or impaired glucose tolerance RECEPTORS: A TARGET FOR have also reported dose-dependent (2-h glucose concentration of 140–199 THERAPEUTIC weight gain compared with placebo and mg/dl), and 23% had diabetes (41). INTERVENTION small decreases in hemoglobin and he- NHANES III was conducted using a lower Prolonged exposure to elevated FFA lev- matocrit after treatment with TZDs (70,71). limit of fasting glucose of 110 mg/dl to els is central to the development of insulin define impaired fasting glucose, but the resistance and type 2 diabetes, making PPAR-␣ American Diabetes Association has since modulation of these levels an attractive FFAs are natural ligands for PPAR-␣, defined the cutoff as 100 mg/dl. On the therapeutic strategy. Nicotinic acid and which is preferentially expressed in tis-

DIABETES CARE, VOLUME 27, NUMBER 9, SEPTEMBER 2004 2255 Lipids, glucose, and type 2 diabetes

Figure 2—Potential contributions of PPAR-␣ and PPAR-␥ to improvements in insulin sensitivity. sues where fatty acids are oxidized such as sitivity. Specific PPAR-␣ agonism can proach may offer an attractive option for the liver, muscle, kidney, and heart. Acti- lower lipid levels in rats and improve in- therapeutic intervention. Ongoing stud- vation of PPAR-␣ stimulates the expres- sulin sensitivity (76,77), whereas studies ies are examining the efficacy and safety of sion of genes involved in fatty acid and of fibrates in humans have either reported these new agents. lipoprotein oxidation in the liver and improved (78,79) or unimproved (80,81) muscle (72,73). PPAR-␣ activators, such insulin sensitivity. SUMMARY as fibrates, decrease plasma FFA and tri- From an initial perception that a disorder glyceride concentrations by stimulating Dual PPAR-␣/␥ activation of glucose metabolism was the primary several metabolic pathways (Fig. 2). Fi- PPAR-␥ exerts its beneficial effect by low- event in the pathogenesis of type 2 diabe- brates increase fatty acid uptake and oxi- ering plasma FFA levels, increasing tes, there is now a growing appreciation dation by increasing the expression of plasma adiponectin levels, and redistrib- that chronic elevation of FFA levels is an lipoprotein lipase in the liver and by de- uting fat from visceral to subcutaneous early event that contributes to the devel- creasing apolipoprotein C-III concentra- depots, and PPAR-␣ activation lowers opment of this disease. FFAs induce insu- tions. Apolipoprotein C-III is a protein plasma FFA levels through increased fat lin resistance, which increases with FFA that inhibits triglyceride hydrolysis by li- oxidation. Therefore, dual PPAR-␣/␥ ago- levels, and this can be a beneficial adap- poprotein lipase, and its downregulation nism may have advantages over selective tive response during starvation and preg- by fibrates results in reduced triglyceride PPAR subtype activation (82,83). Specif- nancy. However, insulin resistance can and VLDL production by the liver (74). ically, dual PPAR-␣/␥ agonism may lower become counterproductive when there is Simultaneously, these agents enhance in- plasma FFAs more than either PPAR-␣ or an excess of energy intake associated with travascular triglyceride metabolism (75). PPAR-␥ alone (Fig. 2). Given the central physical inactivity. The extra fuel is stored The lowering of FFAs through increased role of FFAs in the development of insulin in visceral and subcutaneous fat depots. oxidation may well improve insulin sen- resistance and type 2 diabetes, this ap- As fat accumulates, there is an ongoing

2256 DIABETES CARE, VOLUME 27, NUMBER 9, SEPTEMBER 2004 Boden and Laakso increase in the levels of plasma FFAs, tein particles with the incidence of coro- healthy subjects. Diabetes 50:1612–1617, which causes insulin resistance. In addi- nary artery disease in men and women. 2001 tion, the deficit of another product of ad- JAMA 276:875–881, 1996 17. Itani SI, Ruderman NB, Schmieder F, ipose tissue (e.g., adiponectin) may 4. Lamarche B, Tchernof A, Moorjani S, Boden G: Lipid-induced insulin resis- contribute to increased insulin resistance. Cantin B, Dagenais GR, Lupien PJ, Des- tance in human muscle is associated with pres JP: Small, dense low-density lipopro- changes in diacylglycerol, protein kinase To counter insulin resistance and prevent tein particles as a predictor of the risk of C, and I␬B-␣. Diabetes 51:2005–2011, hyperglycemia, insulin levels increase. In ischemic heart disease in men: prospec- 2002 individuals with a genetic predisposition tive results from the Quebec Cardiovascu- 18. Bronfman M, Morales MN, Orellana A: for diabetes, however, the pancreas can- lar Study. Circulation 95:69–75, 1997 Diacylglycerol activation of protein kinase not compensate for the increased secre- 5. Mykkanen L, Kuusisto J, Haffner SM, C is modulated by long-chain acyl-CoA. tory demands placed on it, resulting in Laakso M, Austin MA: LDL size and risk of Biochem Biophys Res Commun 152:987– type 2 diabetes. coronary heart disease in elderly men and 992, 1988 The pivotal role of FFAs in the devel- women. Arterioscler Thromb Vasc Biol 19: 19. Itani SI, Zhou Q, Pories WJ, MacDonald opment of insulin resistance and type 2 2742–2748, 1999 KG, Dohm GL: Involvement of protein ki- diabetes suggests that the optimal thera- 6. Hales CN, Barker DJ: Type 2 (non-insu- nase C in human skeletal muscle insulin lin-dependent) diabetes mellitus: the resistance and obesity. Diabetes 49:1353– peutic intervention should decrease thrifty phenotype hypothesis. Diabetolo- 1358, 2000 plasma FFA levels. The PPAR family is gia 35:595–601, 1992 20. Yu C, Chen Y, Cline GW, Zhang D, Zong intimately involved in lipid metabolism. 7. Arner P: Free fatty acids: do they play a H, Wang Y, Bergeron R, Kim JK, Cush- Two subtypes of these receptors are the central role in type 2 diabetes? Diabetes man SW, Cooney GJ, Atcheson B, White site of action of synthetic PPAR agonists: Obes Metab 3 (Suppl. 1):11–19, 2001 MF, Kraegen EW, Shulman GI: Mecha- PPAR-␣ and PPAR-␥. The former in- 8. Boden G, Jadali F, White J, Liang Y, Moz- nism by which fatty acids inhibit insulin creases fatty acid oxidation, whereas the zoli M, Chen X, Coleman E, Smith C: activation of insulin receptor substrate-1 latter results in the redistribution of fat Effects of fat on insulin-stimulated carbo- (IRS-1)-associated phosphatidylinositol from visceral to subcutaneous body fat hydrate metabolism in normal men. J Clin 3-kinase activity in muscle. J Biol Chem and an increase in adiponectin. The out- Invest 88:960–966, 1991 277:50230–50236, 2002 ␥ 9. Randle PJ, Garland PB, Hales CN, News- 21. De Fea K, Roth RA: Protein kinase C mod- come of activation of PPAR- is a lowering holme EA: The glucose fatty-acid cycle: its ulation of insulin receptor substrate-1 of plasma FFA concentrations and im- role in insulin sensitivity and the meta- tyrosine phosphorylation requires serine proved insulin sensitivity. The effects of bolic disturbances of diabetes mellitus. 612. Biochemistry 36:12939–12947, 1997 PPAR-␣ on lipid metabolism may also Lancet 1:785–789, 1963 22. Ravichandran LV, Esposito DL, Chen J, bring about improvements in insulin sen- 10. Rennie MJ, Holloszy JO: Inhibition of Quon MJ: Protein kinase C-zeta phos- sitivity. The currently available PPAR ago- glucose uptake and glycogenolysis by phorylates insulin receptor substrate-1 nists selectively activate either PPAR-␣ availability of oleate in well-oxygenated and impairs its ability to activate phos- (i.e., fibrates) or PPAR-␥ (i.e., TZDs) and perfused skeletal muscle. Biochem J 168: phatidylinositol 3-kinase in response to have been shown to improve lipid metab- 161–170, 1977 insulin. J Biol Chem 276:3543–3549, 2001 olism. It may be that dual agonism with 11. Boden G, Chen X, Ruiz J, White JV, Ros- 23. Kim JK, Kim YJ, Fillmore JJ, Chen Y, ␣ ␥ setti L: Mechanisms of fatty acid-induced Moore I, Lee J, Yuan M, Li ZW, Karin M, PPAR- / agonists will provide addi- inhibition of glucose uptake. J Clin Invest Perret P, Shoelson SE, Shulman GI: Pre- tional benefits above and beyond those 93:2438–2446, 1994 vention of fat-induced insulin resistance achieved with sole activation of either 12. Roden M, Price TB, Perseghin G, Petersen by salicylate. J Clin Invest 108:437–446, PPAR-␣ or PPAR-␥. KF, Rothman DL, Cline GW, Shulman GI: 2001 Mechanism of free fatty acid-induced in- 24. Yuan M, Konstantopoulos N, Lee J, sulin resistance in humans. J Clin Invest Hansen L, Li ZW, Karin M, Shoelson SE: Acknowledgments— This work was sup- 97:2859–2865, 1996 Reversal of obesity- and diet-induced in- ported by National Institutes of Health Grants 13. Boden G, Chen X: Effects of fat on glucose sulin resistance with salicylates or tar- R01-AG15353, R01-DK58895, R01- uptake and utilization in patients with geted disruption of Ikkbeta. Science 293: HL0733267, and R01-DK066003 and a Men- non-insulin-dependent diabetes. J Clin In- 1673–1677, 2001 tor-Based Training Grant from the American vest 96:1261–1268, 1995 25. Ross R: Atherosclerosis: an inflammatory Diabetes Association (all to G.B.). 14. Dresner A, Laurent D, Marcucci M, Griffin disease. N Engl J Med 340:115–126, 1999 ME, Dufour S, Cline GW, Slezak LA, 26. Ceriello A: Oxidative stress and glycemic Andersen DK, Hundal RS, Rothman DL, regulation. Metabolism 49:27–29, 2000 References Petersen KF, Shulman GI: Effects of free 27. Griffin ME, Marcucci MJ, Cline GW, Bell 1. McGarry JD: What if Minkowski had been fatty acids on glucose transport and IRS- K, Barucci N, Lee D, Goodyear LJ, Krae- ageusic? An alternative angle on diabetes. 1-associated phosphatidylinositol 3-ki- gen EW, White MF, Shulman GI: Free Science 258:766–770, 1992 nase activity. J Clin Invest 103:253–259, fatty acid-induced insulin resistance is as- 2. Jeppesen J, Hein HO, Suadicani P, Gyn- 1999 sociated with activation of protein kinase telberg F: Relation of high TG-low HDL 15. Homko CJ, Cheung P, Boden G: Effects of C ␪ and alterations in the insulin signaling cholesterol and LDL cholesterol to the in- free fatty acids on glucose uptake and uti- cascade. Diabetes 48:1270–1274, 1999 cidence of ischemic heart disease: an lization in healthy women. Diabetes 52: 28. Ferrannini E, Barrett EJ, Bevilacqua S, De- 8-year follow-up in the Copenhagen Male 487–491, 2003 Fronzo RA: Effect of fatty acids on glucose Study. Arterioscler Thromb Vasc Biol 17: 16. Boden G, Lebed B, Schatz M, Homko C, production and utilization in man. J Clin 1114–1120, 1997 Lemieux S: Effects of acute changes of Invest 72:1737–1747, 1983 3. Gardner CD, Fortmann SP, Krauss RM: plasma free fatty acids on intramyocellu- 29. Gastaldelli A, Toschi E, Pettiti M, Fras- Association of small low-density lipopro- lar fat content and insulin resistance in cerra S, Quinones-Galvan A, Sironi AM,

DIABETES CARE, VOLUME 27, NUMBER 9, SEPTEMBER 2004 2257 Lipids, glucose, and type 2 diabetes

Natali A, Ferrannini E: Effect of physio- chard SM: Secretion of adiponectin and Invest 103:365–372, 1999 logical hyperinsulinemia on gluconeo- regulation of apM1 gene expression in hu- 52. Ghazzi MN, Perez JE, Antonucci TK, genesis in nondiabetic subjects and in man visceral adipose tissue. Biochem Bio- Driscoll JH, Huang SM, Faja BW, Whit- type 2 diabetic patients. Diabetes 50:1807– phys Res Commun 288:1102–1107, 2001 comb RW: Cardiac and glycemic benefits 1812, 2001 41. Benjamin SM, Valdez R, Geiss LS, Rolka of troglitazone treatment in NIDDM: the 30. Boden G, Cheung P, Stein TP, Kresge K, DB, Narayan KM: Estimated number of Troglitazone Study Group. Diabetes 46: Mozzoli M: FFA cause hepatic insulin re- adults with in the US in 2000: 433–439, 1997 sistance by inhibiting insulin suppression opportunities for prevention. Diabetes 53. Maggs DG, Buchanan TA, Burant CF, of glycogenolysis. Am J Physiol Endocrinol Care 26:645–649, 2003 Cline G, Gumbiner B, Hsueh WA, Inzuc- Metab 283:12–19, 2002 42. Crespin SR, Greenough WB III, Steinberg chi S, Kelley D, Nolan J, Olefsky JM, 31. Peraldi P, Spiegelman B: TNF-alpha and D: Stimulation of insulin secretion by Polonsky KS, Silver D, Valiquett TR, Shul- insulin resistance: summary and future long-chain free fatty acids: a direct pan- man GI: Metabolic effects of troglitazone prospects. Mol Cell Biochem 182:169–175, creatic effect. J Clin Invest 52:1979–1984, monotherapy in type 2 diabetes mellitus: 1998 1973 a randomized, double-blind, placebo- 32. Pradhan AD, Manson JE, Rifai N, Buring 43. Boden G, Chen X, Rosner J, Barton M: controlled trial. Ann Intern Med 128:176– JE, Ridker PM: C-reactive protein, inter- Effects of a 48-h fat infusion on insulin 185, 1998 leukin 6, and risk of developing type 2 secretion and glucose utilization. Diabetes 54. Mayerson AB, Hundal RS, Dufour S, diabetes mellitus. JAMA 286:327–334, 44:1239–1242, 1995 Lebon V, Befroy D, Cline GW, Enocksson 2001 44. Jensen CB, Storgaard H, Holst JJ, Dela F, S, Inzucchi SE, Shulman GI, Petersen KF: 33. Steppan CM, Bailey ST, Bhat S, Brown EJ, Madsbad S, Vaag AA: Insulin secretion The effects of rosiglitazone on insulin sen- Banerjee RR, Wright CM, Patel HR, and cellular glucose metabolism after pro- sitivity, lipolysis, and hepatic and skeletal Ahima RS, Lazar MA: The hormone resis- longed low-grade intralipid infusion in muscle triglyceride content in patients tin links obesity to diabetes. Nature 409: young men. J Clin Endocrinol Metab 88: with type 2 diabetes. Diabetes 51:797– 307–312, 2001 2775–2783, 2003 802, 2002 34. Kahn BB, Flier JS: Obesity and insulin re- 45. Kashyap S, Belfort R, Gastaldelli A, 55. Lehmann JM, Moore LB, Smith-Oliver sistance. J Clin Invest 106:473–481, 2000 Pratipanawatr T, Berria R, Pratipanawatr TA, Wilkison WO, Willson TM, Kliewer 35. Weyer C, Funahashi T, Tanaka S, Hotta W, Bajaj M, Mandarino L, DeFronzo R, SA: An antidiabetic thiazolidinedione is a K, Matsuzawa Y, Pratley RE, Tataranni Cusi K: A sustained increase in plasma high affinity ligand for peroxisome prolif- PA: Hypoadiponectinemia in obesity and free fatty acids impairs insulin secretion in erator-activated receptor gamma (PPAR type 2 diabetes: close association with in- nondiabetic subjects genetically predis- gamma). J Biol Chem 270:12953–12956, sulin resistance and hyperinsulinemia. posed to develop type 2 diabetes. Diabetes 1995 J Clin Endocrinol Metab 86:1930–1935, 52:2461–2474, 2003 56. Willson TM, Cobb JE, Cowan DJ, Wiethe 2001 46. Boden G, Chen X, Iqbal N: Acute lower- RW, Correa ID, Prakash SR, Beck KD, 36. Yamauchi T, Kamon J, Waki H, Terauchi ing of plasma fatty acids lowers basal Moore LB, Kliewer SA, Lehmann JM: The Y, Kubota N, Hara K, Mori Y, Ide T, Mu- insulin secretion in diabetic and nondia- structure-activity relationship between rakami K, Tsuboyama-Kasaoka N, Ezaki betic subjects. Diabetes 47:1609–1612, peroxisome proliferator-activated recep- O, Akanuma Y, Gavrilova O, Vinson 1998 tor gamma agonism and the antihypergly- C, Reitman ML, Kagechika H, Shudo 47. Storgaard H, Jensen CB, Vaag AA, Volund cemic activity of thiazolidinediones. J Med K, Yoda M, Nakano Y, Tobe K, Nagai A, Madsbad S: Insulin secretion after Chem 39:665–668, 1996 R, Kimura S, Tomita M, Froguel P, short- and long-term low-grade free fatty 57. Tontonoz P, Hu E, Graves RA, Budavari Kadowaki T: The fat-derived hormone acid infusion in men with increased risk AI, Spiegelman BM: mPPAR gamma 2: tis- adiponectin reverses insulin resistance as- of developing type 2 diabetes. Metabolism sue-specific regulator of an adipocyte en- sociated with both lipoatrophy and obe- 52:885–894, 2003 hancer. Genes Dev 8:1224–1234, 1994 sity. Nat Med 7:941–946, 2001 48. Stefan N, Stumvoll M, Bogardus C, 58. Kruszynska YT, Mukherjee R, Jow L, 37. Hotta K, Funahashi T, Bodkin NL, Ort- Tataranni PA: Elevated plasma nonesteri- Dana S, Paterniti JR, Olefsky JM: Skeletal meyer HK, Arita Y, Hansen BC, Matsu- fied fatty acids are associated with deteri- muscle peroxisome proliferator-activated zawa Y: Circulating concentrations of the oration of acute insulin response in IGT receptor-gamma expression in obesity adipocyte protein adiponectin are de- but not NGT. Am J Physiol Endocrinol and non-insulin-dependent diabetes mel- creased in parallel with reduced insulin Metab 284:E1156–E1161, 2003 litus. J Clin Invest 101:543–548, 1998 sensitivity during the progression to type 49. Pimenta W, Korytkowski M, Mitrakou A, 59. Tontonoz P, Hu E, Spiegelman BM: Stim- 2 diabetes in rhesus monkeys. Diabetes Jenssen T, Yki-Jarvinen H, Evron W, Dai- ulation of adipogenesis in fibroblasts 50:1126–1133, 2001 ley G, Gerich J: Pancreatic beta-cell dys- by PPAR gamma 2, a lipid-activated 38. Goldfine AB, Kahn RC: Apidonectin: link- function as the primary genetic lesion in transcription factor. Cell 79:1147–1156, ing the fat cell to insulin sensitivity. Lancet NIDDM: evidence from studies in normal 1994 362:1431–1432, 2003 glucose-tolerant individuals with a first- 60. Adams M, Montague CT, Prins JB, Holder 39. Arita Y, Kihara S, Ouchi N, Takahashi M, degree NIDDM relative. JAMA 273:1855– JC, Smith SA, Sanders L, Digby JE, Sewter Maeda K, Miyagawa J, Hotta K, Shimo- 1861, 1995 CP, Lazar MA, Chatterjee VK, O’Rahilly mura I, Nakamura T, Miyaoka K, Kuri- 50. Hirose H, Lee YH, Inman LR, Nagasawa Y, S: Activators of peroxisome proliferator- yama H, Nishida M, Yamashita S, Okubo Johnson JH, Unger RH: Defective fatty ac- activated receptor gamma have depot- K, Matsubara K, Muraguchi M, Ohmoto id-mediated beta-cell compensation in specific effects on human preadipocyte Y, Funahashi T, Matsuzawa Y: Paradoxi- Zucker diabetic fatty rats: pathogenic im- differentiation. J Clin Invest 100:3149– cal decrease of an adipose-specific pro- plications for obesity-dependent diabetes. 3153, 1997 tein, adiponectin, in obesity. Biochem J Biol Chem 271:5633–5637, 1996 61. de Souza CJ, Eckhardt M, Gagen K, Dong Biophys Res Commun 257:79–83, 1999 51. Chen X, Iqbal N, Boden G: The effects of M, Chen W, Laurent D, Burkey BF: Effects 40. Halleux CM, Takahashi M, Delporte ML, free fatty acids on gluconeogenesis and of pioglitazone on adipose tissue remod- Detry R, Funahashi T, Matsuzawa Y, Bri- glycogenolysis in normal subjects. J Clin eling within the setting of obesity and in-

2258 DIABETES CARE, VOLUME 27, NUMBER 9, SEPTEMBER 2004 Boden and Laakso

sulin resistance. Diabetes 50:1863–1871, ment of patients with type 2 diabetes: a study. Diabetes Care 11:495–499, 1988 2001 6-month randomized placebo-controlled 79. Mussoni L, Mannucci L, Sirtori C, Pazzuc- 62. Maeda N, Takahashi M, Funahashi T, dose-response study: the Pioglitazone 001 coni F, Bonfardeci G, Cimminiello C, Kihara S, Nishizawa H, Kishida K, Naga- Study Group. Diabetes Care 23:1605– Notarbartolo A, Scafidi V, Bittolo BG, retani H, Matsuda M, Komuro R, Ouchi 1611, 2000 Alessandrini P, Nenci G, Parise P, Co- N, Kuriyama H, Hotta K, Nakamura T, 71. Phillips LS, Grunberger G, Miller E, Pat- lombo L, Piliego T, Tremoli E: Effects of Shimomura I, Matsuzawa Y: PPAR-␥ li- wardhan R, Rappaport EB, Salzman A: gemfibrozil on insulin sensitivity and on gands increase expression and plasma Once- and twice-daily dosing with rosigli- haemostatic variables in hypertriglyceri- concentrations of adiponectin, an adi- tazone improves glycemic control in pa- demic patients. Atherosclerosis 148:397– pose-derived protein. Diabetes 50:2094– tients with type 2 diabetes. Diabetes Care 406, 2000 2099, 2001 24:308–315, 2001 80. Sane T, Knudsen P, Vuorinen-Markkola 63. Boden G, Cheung P, Mozzoli M, Fried SK: 72. Gulick T, Cresci S, Caira T, Moore DD, H, Yki-Jarvinen H, Taskinen MR: De- Effect of thiazolidinediones on glucose Kelly DP: The peroxisome proliferator-ac- creasing triglyceride by gemfibrozil ther- and fatty acid metabolism in patients with tivated receptor regulates mitochondrial apy does not affect the glucoregulatory or type 2 diabetes. Metabolism 52:753–759, fatty acid oxidative enzyme gene expres- antilipolytic effect of insulin in nondia- 2003 sion. Proc Natl Acad SciUSA91:11012– betic subjects with mild hypertriglyceri- 64. Nesto RW, Bell D, Bonow RO, Fonseca V, 11016, 1994 demia. Metabolism 44:589–596, 1995 Grundy SM, Horton ES, Le Winter M, 73. Muoio DM, Way JM, Tanner CJ, Winegar 81. Asplund-Carlson A: Effects of gemfibrozil Porte D, Semenkovich CF, Smith S, DA, Kliewer SA, Houmard JA, Kraus WE, therapy on glucose tolerance, insulin sen- Young LH, Kahn R: Thiazolidinedione Dohm GL: Peroxisome proliferator-acti- sitivity and plasma plasminogen activator use, fluid retention, and congestive heart vated receptor-␣ regulates fatty acid utili- inhibitor activity in hypertriglyceridae- failure: a consensus statement from the zation in primary human skeletal muscle mia. J Cardiovasc Risk 3:385–390, 1996 American Heart Association and Ameri- cells. Diabetes 51:901–909, 2002 82. Ljung B, Bamberg K, Dahllof B, Kjellstedt can Diabetes Association: October 7, 74. Staels B, Dallongeville J, Auwerx J, A, Oakes ND, Ostling J, Svensson L, Cam- 2003. Circulation 108:2941–2948, 2003 Schoonjans K, Leitersdorf E, Fruchart JC: ejo G: AZ 242, a novel PPARalpha/gamma 65. Raskin P, Rendell M, Riddle MC, Dole JF, Mechanism of action of fibrates on lipid agonist with beneficial effects on insulin Freed MI, Rosenstock J: A randomized and lipoprotein metabolism. Circulation resistance and carbohydrate and lipid me- trial of rosiglitazone therapy in patients 98:2088–2093, 1998 tabolism in ob/ob mice and obese Zucker with inadequately controlled insulin- 75. Minnich A, Tian N, Byan L, Bilder G: A rats. J Lipid Res 43:1855–1863, 2002 treated type 2 diabetes. Diabetes Care 24: potent PPARalpha agonist stimulates mi- 83. Etgen GJ, Oldham BA, Johnson WT, 1226–1232, 2001 tochondrial fatty acid beta-oxidation in Broderick CL, Montrose CR, Brozinick JT, 66. Cheng AY, Fantus IG: Thiazolidinedione- liver and skeletal muscle. Am J Physiol En- Misener EA, Bean JS, Bensch WR, Brooks induced congestive heart failure. Ann Phar- docrinol Metab 280:E270–E279, 2001 DA, Shuker AJ, Rito CJ, McCarthy JR, macother 38:817–820, 2004 76. Ye JM, Doyle PJ, Iglesias MA, Watson DG, Ardecky RJ, Tyhonas JS, Dana SL, Bila- 67. Kermani A, Garg A: Thiazolidinedione- Cooney GJ, Kraegen EW: Peroxisome kovics JM, Paterniti JR Jr, Ogilvie KM, Liu associated congestive heart failure and proliferator-activated receptor (PPAR)-␣ S, Kauffman RF: A tailored therapy for the pulmonary edema. Mayo Clin Proc 78: activation lowers muscle lipids and im- : the dual peroxisome 1088–1091, 2003 proves insulin sensitivity in high fat-fed proliferator-activated receptor-␣/␥ ago- 68. Rubin C, Egan J, Schneider R: Combina- rats: comparison with PPAR-␥ activation. nist LY465608 ameliorates insulin resis- tion therapy with pioglitazone and insulin Diabetes 50:411–417, 2001 tance and diabetic hyperglycemia while in patients with type 2 diabetes (Ab- 77. Guerre-Millo M, Gervois P, Raspe E, Mad- improving cardiovascular risk factors in stract). Diabetes 48 (Suppl. 1):A110, 1999 sen L, Poulain P, Derudas B, Herbert JM, preclinical models. Diabetes 51:1083– 69. Rosenstock J, Einhorn D, Hershon K, Winegar DA, Willson TM, Fruchart JC, 1087, 2002 Glazer NB, Yu S: Efficacy and safety of Berge RK, Staels B: Peroxisome prolif- 84. Inoguchi T, Li P, Umeda F, Yu HY, Kaki- pioglitazone in type 2 diabetes: a random- erator-activated receptor alpha activators moto M, Imamura M, Aoki T, Etoh T, ised, placebo-controlled study in patients improve insulin sensitivity and reduce ad- Hashimoto T, Naruse M, Sano H, Utsumi receiving stable insulin therapy. Int J Clin iposity. J Biol Chem 275:16638–16642, H, Nawata H: High glucose level and free Pract 56:251–257, 2002 2000 fatty acid stimulate reactive oxygen spe- 70. Aronoff S, Rosenblatt S, Braithwaite S, 78. Kobayashi M, Shigeta Y, Hirata Y, Omori cies production through protein kinase Egan JW, Mathisen AL, Schneider RL: Y, Sakamoto N, Nambu S, Baba S: Im- C–dependent activation of NAD(P)H ox- Pioglitazone hydrochloride monotherapy provement of glucose tolerance in NIDDM idase in cultured vascular cells. Diabetes improves glycemic control in the treat- by clofibrate: randomized double-blind 49:1939–1945, 2000

DIABETES CARE, VOLUME 27, NUMBER 9, SEPTEMBER 2004 2259