Role of the Adipocyte, Free Fatty Acids, and Ectopic Fat in Pathogenesis of Type 2 Diabetes Mellitus: Peroxisomal Proliferator-Activated Receptor Agonists Provide a Rational Therapeutic Approach

Harold Bays, Lawrence Mandarino and Ralph A. DeFronzo

Diabetes Division, University of Texas Health Science Center (L.M., R.A.D.), San Antonio, Texas 78229; and Louisville Metabolic and Atherosclerosis Research Center (H.B.), Louisville, Kentucky 40213

Address all correspondence and requests for reprints to: Ralph A. DeFronzo, M.D., Diabetes Division, University of Texas Health Science Center, 7703 Floyd Curl Drive, San Antonio, Texas 78229. E-mail: albarado{at}uthscsa.edu.


    Introduction
 Top
 Introduction
 I. Pathogenesis of T2DM
 II. Role of the...
 III. The Fat Cell...
 IV. Fat Topography: Ectopic...
 V. "Dysfunctional" Fat vs....
 References
 
Type 2 diabetes mellitus (T2DM) is characterized by insulin resistance in liver and muscle and impaired insulin secretion. Considerable evidence also implicates deranged adipocyte metabolism and altered fat topography in the pathogenesis of glucose intolerance in T2DM. 1) Fat cells are resistant to insulin’s antilipolytic effect, leading to day-long elevated plasma FFA levels. Chronically increased plasma FFA stimulates gluconeogenesis, induces hepatic/muscle insulin resistance, and impairs insulin secretion in genetically predisposed individuals. These FFA-induced disturbances are referred to as lipotoxicity. 2) Dysfunctional fat cells produce excessive amounts of insulin resistance-inducing, inflammatory, and atherosclerotic-provoking cytokines and fail to secrete normal amounts of insulin-sensitizing adipocytokines. 3) Enlarged fat cells are insulin resistant and have diminished capacity to store fat. When adipocyte storage capacity is exceeded, lipid "overflows" into muscle, liver, and perhaps ß-cells, causing muscle/hepatic insulin resistance and impaired insulin secretion. Thiazolidinediones enhance adipocyte insulin sensitivity, inhibit lipolysis, reduce plasma FFA, and favorably influence the production of adipocytokines. Thiazolidinediones also redistribute fat within the body (decreased visceral and hepatic fat; increased sc fat) and decrease intracellular concentrations of triglyceride metabolites in muscle, liver, and ß-cells, contributing to improvements in muscle/hepatic insulin sensitivity and pancreatic function in type 2 diabetics.


    I. Pathogenesis of T2DM
 Top
 Introduction
 I. Pathogenesis of T2DM
 II. Role of the...
 III. The Fat Cell...
 IV. Fat Topography: Ectopic...
 V. "Dysfunctional" Fat vs....
 References
 
The pathophysiology of glucose intolerance in T2DM is complex and involves both genetic and acquired factors (1, 2). Among the acquired factors, weight gain (1, 2, 3) and physical inactivity (1, 2, 4) are of paramount importance. In diabetic individuals with overt fasting hyperglycemia, the triumvirate (1, 2) of 1) impaired insulin secretion by the pancreatic ß-cells, 2) muscle insulin resistance, and 3) hepatic insulin resistance all play central roles in the development and progression of glucose intolerance.

A. Insulin secretion

Before the onset of postprandial and fasting hyperglycemia, individuals genetically predisposed to develop T2DM are known to be resistant to the action of insulin (5, 6, 7, 8). Nonetheless, glucose homeostasis remains normal because of a marked increase in insulin secretion by pancreatic ß-cells (1, 2, 5, 6, 7, 8, 9, 10, 11). The increase in the fasting plasma insulin concentration is sufficient to offset the hepatic insulin resistance, and the basal rate of hepatic glucose production (HGP) remains normal, whereas the hyperinsulinemic response to carbohydrate ingestion is sufficient to cause normal suppression of basal HGP and to overcome the defect in muscle glucose uptake (1, 2, 5, 6, 7, 8, 12, 13). With the onset of impaired glucose tolerance (IGT), there is a worsening of the insulin resistance in muscle and liver (1, 2, 6, 13, 14) and a further increase in the total insulin response to an oral glucose load (1, 2, 9, 12). However, the timing of insulin release in IGT is disrupted, with a decline in early phase insulin secretion, followed by an excessive late phase insulin response (1, 6, 9, 12, 13, 14, 15, 16, 17). The progression from IGT to overt T2DM is associated with little or no further deterioration in insulin resistance (1, 2, 11, 18). Rather, the ability of the pancreas to maintain its high insulin secretory rate declines (1, 2, 9, 11, 13, 15, 16, 18, 19), leading to a worsening of glucose intolerance and eventual development of overt T2DM. The inverted U-shaped curve of insulin secretion as individuals progress from normal glucose tolerance to IGT to T2DM has been referred to as Starling’s curve of the pancreas (1). Initially, the defect in glucose homeostasis is evident by an excessive rise in the postprandial glucose excursion (IGT and mild diabetes), and this is followed by a rise in the fasting plasma glucose concentration.

B. Muscle glucose uptake

Muscle tissue in T2DM individuals is moderately to severely resistant to the action of insulin (1, 2, 20, 21). When insulin is infused systemically or locally into the leg or forearm, the stimulatory effect of hyperinsulinemia on muscle glucose uptake is markedly reduced (20). This defect in muscle glucose uptake in concert with the impaired ability of insulin to suppress the elevated basal rate of hepatic glucose production (20, 21, 22) is responsible for the excessive rise in the plasma glucose concentration after glucose ingestion, i.e. postprandial hyperglycemia (23). In nondiabetic individuals, approximately one third of the glucose that is taken up by muscle in response to a physiological insulin stimulus is oxidized, and the other two thirds is converted to glycogen (1, 2, 21, 24). In type 2 diabetics, the ability of physiological and pharmacological increases in the plasma insulin concentration to augment muscle glucose oxidation and glycogen synthesis is severely impaired (1, 2, 21, 24).

C. HGP

In T2DM individuals with established fasting hyperglycemia, the basal rate of hepatic glucose production is increased despite fasting plasma insulin concentrations that are 2- to 4-fold elevated (1, 2, 21, 22), indicating the presence of hepatic resistance to the action of insulin. The hepatic insulin resistance is further evident by the impaired ability of hyperinsulinemia to suppress basal HGP (1, 2, 21). The elevated basal rate of HGP is closely correlated with the increase in fasting plasma glucose concentration in T2DM patients (1, 22) and primarily is accounted for by an increase in gluconeogenesis (25).


    II. Role of the Adipocyte and FFA in the Pathogenesis of T2DM: The Dysharmonious Quartet
 Top
 Introduction
 I. Pathogenesis of T2DM
 II. Role of the...
 III. The Fat Cell...
 IV. Fat Topography: Ectopic...
 V. "Dysfunctional" Fat vs....
 References
 
A. Overview

The majority (>80%) of patients with T2DM in the U.S. are overweight (26), and both lean and especially obese type 2 diabetics are characterized by day-long elevations in the plasma free fatty acid (FFA) concentration which fail to suppress normally following ingestion of a mixed meal or oral glucose load (27) or in response to insulin (21, 28). Similar abnormalities in FFA metabolism have been documented in individuals with IGT and nondiabetic, insulin resistant obese individuals (29, 30). FFA are stored as triglycerides in adipocytes and serve as a source of energy during fasting conditions. Insulin is a potent inhibitor of lipolysis (21) and restrains the release of FFA from the adipocyte by inhibiting the enzyme hormone-sensitive lipase. In type 2 diabetics, the ability of insulin to inhibit lipolysis (as reflected by suppression of radioactive palmitate turnover) and to reduce the plasma FFA concentration is markedly impaired (21). It is now recognized that chronically elevated plasma FFA concentrations cause insulin resistance in muscle and liver (1, 2, 28, 30, 31) and impair insulin secretion (32, 33). Thus, elevated plasma FFA levels can cause/aggravate the three major pathogenic disturbances that are responsible for impaired glucose homeostasis in T2DM individuals, and the time has arrived for the "triumvirate" (muscle, liver, and ß-cell) to be joined by the "fourth musketeer" (29) to form the "dysharmonious quartet" (Fig. 1Go). In addition to FFA that circulate in plasma in increased amounts, T2DM patients have increased stores of triglycerides in muscle (34, 35) and liver (36, 37), which correlate closely with the presence of insulin resistance in these tissues. The triglycerides in liver and muscle are in a state of constant turnover, and the metabolites [i.e. fatty acyl coenzymes A (CoAs), ceramides, diacyglycerol, etc.] of intracellular triglyceride lipolysis impair insulin action in both liver and muscle (2, 38). This sequence of events has been referred to as lipotoxicity (2, 32, 39). Evidence also has accumulated to implicate lipotoxicity as an important cause of ß-cell dysfunction (32, 40).



View larger version (22K):
[in this window]
[in a new window]
 
FIG. 1. Pathogenesis of T2DM: the "dysharmonious quartet."

 
B. FFA and insulin secretion

After the ingestion of a mixed meal or infusion of lipid, the plasma FFA concentration rises, and FFA are transported into the ß-cell via fatty acid-binding protein 2 (32, 40, 41, 42). Within the cytosol, fatty acids are converted to their fatty acyl CoA derivative, which, in turn, augments insulin secretion via a variety of mechanisms (32, 40, 41, 42): 1) increased formation of phosphatidic acid and diacylglycerol, which directly and indirectly (through activation of protein kinase C) enhance exocytosis of insulin stored within secretory granules; 2) stimulation of endoplasmic reticulum Ca2+-adenosine triphosphatase, leading to an increase in intracellular calcium concentration and augmentation of insulin secretion; and 3) closure of the K+-ATP channel with resultant depolarization of the ß-cell membrane, which causes an increase in intracellular calcium and stimulation of exocytosis of insulin-containing granules. After ingestion of a mixed meal, the resultant hyperglycemia leads to an increase in malonyl CoA concentration within the ß-cell. Malonyl CoA inhibits carnitine palmitoyl transferase-1, thus impairing the transport of fatty acyl CoAs into the mitochondria where they are oxidized in the Krebs cycle. The resulting increase in cytosolic fatty acyl CoAs (i.e. consequent to malonyl CoA-induced inhibition of carnitine palmitoyl transferase-1) works in tandem with hyperglycemia to augment insulin secretion. Consistent with these in vitro observations, short-term (2- to 6-h) elevation of the plasma FFA concentration in rodents and man has been shown to enhance insulin secretion (32, 43, 44, 45), whereas an acute decrease in the plasma FFA concentration inhibits glucose-stimulated insulin secretion (32, 43).

In contrast to the acute effect of elevated plasma FFA to enhance insulin secretion, longer-term (>=48 h) exposure results in an impaired ß-cell response to glucose both in vitro and in vivo in animals (46, 47, 48, 49, 50, 51) and humans (33, 52, 53, 54). The inhibitory effect of chronically elevated plasma FFA appears to be more prominent in individuals with a genetic predisposition to develop T2DM (33), but has also been observed in healthy carriers of the Pro12Ala polymorphism of the peroxisome proliferator-activated receptor-{gamma}2 (PPAR{gamma}) gene (55). Conversely, a reduction in the plasma FFA concentration in type 2 diabetics has been shown to improve insulin secretion (33, 56, 57). Several groups have shown that chronic (6- to 7-wk) overfeeding of a high fat diet impairs insulin secretion in dogs (58, 59). Elevated plasma FFA levels also have been shown to be a risk marker for the long-term development of impaired glucose tolerance and T2DM (50) in Caucasians (60) and Pima Indians (61). Unger (62) has coined the term lipotoxicity to describe the deleterious effect of chronic FFA elevation on insulin secretion by the pancreatic ß-cells. In the Zucker diabetic fatty rat, chronically increased plasma FFA levels initially lead to a physiological impairment in insulin secretion. With time, however, ß-cell apoptosis ensues and ß-cell mass is reduced by more than 50% (50). Within the ß-cell, elevated fatty acyl CoAs increase the formation of ceramide. Ceramide, in turn, augments nitric oxide formation, which is cidal to the ß-cell (40). Obviously, similar studies cannot be performed in man in vivo, but, at least in the Zucker diabetic fatty rat model, the biochemical/molecular basis of FFA-induced ß-cell toxicity has been established. Incubation of human islets with FFA also has been shown to cause ß-cell apoptosis (51). Regardless of the mechanism(s) involved, chronic physiological (478–666 µmol/liter) (33) elevation of the plasma FFA concentration in nondiabetic humans has been shown to impair insulin secretion (33, 52, 53, 54, 55).

C. FFA and muscle glucose metabolism

Four decades ago, Randle and colleagues (63) proposed that increased FFA oxidation restrains glucose oxidation in muscle by altering the redox potential of the cell and by inhibiting key glycolytic enzymes. Excessive FFA oxidation 1) leads to the intracellular accumulation of acetyl CoA, a potent inhibitor of pyruvate dehydrogenase, 2) increases the NADH/NAD ratio, causing a slowing of the Krebs cycle, and 3) results in the accumulation of citrate, a powerful inhibitor of phosphofructokinase. Inhibition of phosphofructokinase, in turn, causes product inhibition of the early steps involved in glucose metabolism, leading to the accumulation of glucose-6-phosphate (G-6-P), which inhibits hexokinase II. The block in glucose phosphorylation results in a build-up of intracellular free glucose, which restrains glucose transport into the cell via the GLUT4 transporter. The resultant decrease in glucose transport impairs glycogen synthesis, although a direct inhibitory effect of fatty acyl CoAs on glycogen synthase also has been demonstrated (64). This sequence of events, via which accelerated plasma FFA oxidation inhibits muscle glucose transport, glucose oxidation, and glycogen synthesis, is referred to as the Randle cycle (63). It should be noted that the same scenario would ensue if the FFA were derived from triglycerides stored in muscle (34, 35) or from plasma (21, 28, 29).

Felber and co-workers (30, 65, 66) were among the first to demonstrate that in diabetic (and obese nondiabetic) humans, basal plasma FFA levels and lipid oxidation are increased and fail to suppress normally after glucose ingestion. The elevated basal rate of lipid oxidation was strongly correlated with a decreased basal rate of glucose oxidation as well as with the reduced rate of nonoxidative glucose disposal, which primarily reflects glycogen synthesis (24), after ingestion of a glucose load. Further validation of the Randle cycle in man has come from studies employing the euglycemic insulin clamp (67). When Intralipid is infused concomitantly with insulin, either to prevent the decline in plasma FFA concentration or to increase the plasma FFA concentration above basal levels, both glucose oxidation and nonoxidative glucose disposal are inhibited in a dose-dependent fashion (31, 67). Using magnetic resonance imaging, Roden et al. (68) have shown that the FFA-induced inhibition of nonoxidative glucose disposal reflects impaired glycogen synthesis.

D. Randle cycle revisited: biochemical/molecular basis of FFA-induced insulin resistance

The Randle cycle was formulated based upon experiments performed in rat diaphragm and heart muscle (63). Recent studies performed in human skeletal muscle implicate mechanisms, in addition to those proposed by Randle, in the FFA-induced insulin resistance (Fig. 2Go). Several investigators (68, 69, 70) failed to observe a rise in muscle G-6-P, an increase in muscle citrate levels, or an inhibition of phosphofructokinase when insulin-stimulated glucose metabolism was inhibited by a lipid infusion to elevate the plasma FFA concentration. Thus, although increased FFA/lipid oxidation and decreased glucose oxidation are closely coupled, as originally demonstrated by Randle, mechanisms other than product (i.e. elevated G-6-P) inhibition of the early steps of glucose metabolism must be invoked to explain the defects in glucose transport, glucose phosphorylation, and glycogen synthesis.



View larger version (31K):
[in this window]
[in a new window]
 
FIG. 2. Randle cycle revisited; see text for a detailed discussion. The negative sign within the enclosed circle indicates a site of inhibition by intracellular metabolites of triglyceride metabolism.

 
Studies in humans and animals have shown a strong negative correlation between insulin-stimulated glucose metabolism and increased intramuscular lipid pools, including triglycerides (71, 72, 73),1 diacylglycerol (DAG) (77, 78), and long chain fatty acyl CoAs (FA-CoA) (79). An acute elevation in plasma FFA concentration leads to an increase in muscle fatty acyl CoA and DAG concentrations. Both long chain fatty acyl CoAs and DAG activate protein kinase C{theta} (77), which increases serine phosphorylation with subsequent inhibition of insulin receptor substrate-1 (IRS-1) tyrosine phosphorylation (80, 81, 82) (Fig. 2Go). Consistent with this observation, both acute (4 h) and chronic (5 d) elevation in the plasma FFA concentration inhibits insulin-stimulated tyrosine phosphorylation of IRS-1, the association of the p85 subunit of PI-3 kinase with IRS-1, and activation of phosphoinositol 3-kinase in human skeletal muscle (83, 84, 85). A direct effect of long chain FA-CoAs on glucose transport (86), glucose phosphorylation (87), and glycogen synthase (64) also has been demonstrated in muscle (Fig. 2Go). Lastly, increased muscle ceramide levels (secondary to increased long chain FA-CoAs) interfere with glucose transport and inhibit glycogen synthase in muscle via activation of protein kinase B (88, 89) (Fig. 2Go).

E. FFA and hepatic glucose metabolism

The liver plays a central role in the regulation of glucose metabolism (1, 2). After ingestion of a carbohydrate meal, the combination of hyperinsulinemia and hyperglycemia suppresses basal hepatic glucose production (23). If glucose were to enter the systemic circulation simultaneously from both the gastrointestinal tract and the liver, marked hyperglycemia would ensue. In addition, the liver takes up approximately one third of the glucose in the ingested carbohydrate meal (23, 90). Collectively, suppression of hepatic glucose production and augmentation of hepatic glucose uptake account for the maintenance of about half of the rise in plasma glucose concentration after carbohydrate ingestion (1, 2, 23, 90).

The regulation of HGP is controlled by a number of factors, of which insulin (inhibits HGP) (90, 91) and glucagon and FFA (stimulate HGP) (92) are the most important. In vitro studies have demonstrated that plasma FFA are potent stimulators of HGP and do so by stimulating pyruvate carboxylase and phosphoenolpyruvate carboxykinase, the rate-limiting enzymes for gluconeogenesis (93, 94, 95), and by increasing the activity of glucose-6-phosphatase, the enzyme that ultimately controls the release of glucose by the liver (96). In normal subjects, an increase in plasma FFA concentration stimulates gluconeogenesis (97, 98), whereas a decrease in the plasma FFA concentration reduces gluconeogenesis (97, 99). Bergman, Cherrington, and colleagues (92, 100) have shown that a significant component of the suppressive effect of insulin on HGP is mediated via inhibition of lipolysis and reduction of the plasma FFA concentration. Moreover, FFA infusion in normal humans under conditions that simulate the diabetic state (101) and in obese insulin-resistant subjects (102) enhances HGP due to stimulation of gluconeogenesis (97).

In type 2 diabetics the fasting plasma FFA concentration and lipid oxidation rate are increased and are strongly correlated with both the elevated fasting plasma glucose concentration and the basal rate of HGP (1, 2, 21, 29, 30, 102, 103). Moreover, reduction of the plasma FFA concentration with nicotinic acid reduces HGP and gluconeogenesis in T2DM subjects (104). In the insulin-resistant, normal glucose-tolerant offspring of two diabetic parents, suppression of HGP by insulin is reduced and basal, and insulin-suppressed rates of plasma FFA and lipid oxidation are impaired (5, 12). In other insulin-resistant states, including obesity and IGT, insulin-suppressed HGP also is reduced and correlates with both the elevated basal FFA levels and the increased lipid oxidation rate (18, 105, 106). In a recent study (106), normal glucose-tolerant, insulin-resistant men demonstrated impaired suppression of HGP and plasma FFA levels by insulin and increased hepatic fat content, with strong correlations among the three independent variables. The relationship among elevated plasma FFA concentration, FFA oxidation, and HGP in T2DM (and obesity) can be explained as follows. 1) Increased plasma FFA, by mass action, augments FFA uptake by hepatocytes, leading to accelerated lipid oxidation and accumulation of acetyl CoA. Increased acetyl CoA stimulates pyruvate carboxylase and phosphoenolpyruvate carboxykinase, the rate-limiting enzymes in gluconeogenesis (94, 95), as well as glucose-6-phosphatase, the rate-controlling enzyme for glucose release from the hepatocyte (96). 2) Increased FFA oxidation provides a source of energy (in the form of ATP) and reduced nucleotides (NADH) to drive gluconeogenesis. 3) Elevated plasma FFA induce hepatic insulin resistance by inhibiting the insulin signal transduction system (80, 81, 82, 83, 84, 85, 107, 108). In T2DM patients, these deleterious effects of elevated plasma FFA concentration occur in concert with increased plasma glucagon levels (109, 110), increased hepatic sensitivity to glucagon (110), and increased hepatic uptake of circulating gluconeogenic precursors (111).


    III. The Fat Cell as an Endocrine Organ
 Top
 Introduction
 I. Pathogenesis of T2DM
 II. Role of the...
 III. The Fat Cell...
 IV. Fat Topography: Ectopic...
 V. "Dysfunctional" Fat vs....
 References
 
The preceding discussion emphasized the adipocyte as a storage depot for FFA. In insulin-resistant states, such as T2DM (and obesity), stored FFA are released in excessive amounts and travel to the muscle/liver and pancreas where they induce insulin resistance and impair insulin secretion, respectively. It now is recognized that the adipocyte is a metabolic factory that produces a wide variety of adipocytokines (112). In type 2 diabetics there is a reduction in the production of some factors that normally are synthesized by the adipocyte, i.e. adiponectin, whereas there is accelerated release of other adipocytokines, i.e. resistin, angiotensinogen, plasminogen activator inhibitor-1, TNF{alpha}, ILs, leptin, and others. Thus, the fat cell has become "dysfunctional."

A. Adiponectin

Adiponectin (Acrp 30 or AdipoQ) possesses two unique characteristics: 1) it is expressed only in adipocytes, from where it is secreted into the circulation; and 2) it is the only known adipocyte-secreted factor that increases tissue sensitivity to insulin (113, 114, 115). Plasma adiponectin levels are markedly decreased in T2DM (and obese) subjects (116, 117, 118, 119, 120, 121), and the magnitude of the reduction is strongly correlated with the severity of insulin resistance in peripheral tissues (muscle) (116, 117, 118, 119) and liver (120, 121). Recent studies have shown that adiponectin is a potent antiinflammatory agent that inhibits a number of steps involved in the development of atherosclerosis in animals and humans (reviewed in Refs. 122 and 123). In cultured adipocytes, insulin increases adiponectin expression (124). As adipocytes from type 2 diabetics are resistant to insulin (125), one could postulate that the decline in plasma adiponectin concentration in type 2 diabetics results from diminished insulin sensitivity. Alternatively, decreased adiponectin expression could reflect the dysfunctional fat cell syndrome.

B. Resistin

Resistin is a novel protein, discovered in preadipocytes that were undergoing differentiation into mature adipocytes (126). When injected into normal animals, resistin produces insulin resistance. Conversely, neutralization of resistin with antiresistin antibodies leads to a decline in the plasma glucose concentration and enhanced insulin-stimulated glucose uptake in cultured cells (127, 128). Recent studies have demonstrated that in human T2DM (and obesity), circulating resistin levels and resistin expression in fat cells are increased (129, 130, 131, 132, 133). In humans with T2DM we and others (Bajaj, M., S. Suraamornkul, L. Hardies, and R. A. DeFronzo, submitted for publication) have shown that plasma resistin levels are elevated and correlate closely with hepatic, but not muscle, insulin resistance (134). Consistent with this observation, infusion of resistin into normal animals causes severe hepatic insulin resistance, but has no effect on muscle insulin sensitivity (135). Resistin also is a member of the FIZZ family of proteins and is a potent inflammatory agent (136). Insulin inhibits resistin expression in adipocytes (137, 138, 139). As T2DM individuals have elevated basal plasma resistin levels (133, 134) despite fasting plasma insulin concentrations that are increased 2- to 4-fold (1, 2), this suggests that the inhibitory effect of insulin on fat cell synthesis and/or secretion of resistin may be resistant to the action of insulin.

C. Angiotensinogen

Although the liver is usually thought to be the primary source of angiotensin in the circulation, fat cells from rodents (140) and humans (141) express the message for angiotensin in abundance and represent an important source of angiotensinogen in the body. Although little is known about angiotensinogen expression in fat cells in human T2DM and obesity, angiotensinogen expression in adipocytes and plasma levels of the hormone have been shown to be increased in rodent models of obesity (142). Angiotensin II is a potent vasoconstrictor and has been implicated in the development of atherosclerosis in muscle by inhibiting the insulin signaling cascade (143, 144). Therefore, it is interesting to speculate that the strong associations among insulin resistance, hypertension, and accelerated atherosclerosis in both T2DM and obesity are a manifestation of the dysfunctional fat cell syndrome.

D. Plasminogen activator inhibitor-1 (PAI-1)

PAI-1 is a serine protease that inhibits tissue plasminogen activator, which cleaves plasminogen to plasmin, thus activating the fibrinolytic cascade. Elevated PAI-1 levels upset the balance between the thrombotic and fibrinolytic systems, favoring the formation of microthrombi on blood vessels and accelerating the atherosclerotic process. Both T2DM and obese subjects have elevated PAI-1 levels (145), which are correlated with the development of coronary artery disease and myocardial infarction (146). Adipose tissue is a major source of PAI-1, and PAI-1 expression and secretion by adipocytes are markedly increased in diabetic obese nondiabetic individuals (147, 148). Moreover, insulin, which is characteristically increased in insulin-resistant states, is a potent stimulus for PAI-1 secretion by fat cells (147, 149). Thus, the dysfunctional fat cell through enhancing PAI-1 secretion may provide an important link between accelerated cardiovascular disease and insulin resistance/obesity/T2DM.

E. TNF{alpha}

TNF{alpha} is an adipocyte-derived cytokine that plays a central role in the insulin resistance of sepsis and cancer (150, 151). When infused into rodents, it produces severe insulin resistance (152), stimulates lipolysis, and activates the inflammatory MAPK isoforms c-Jun N-terminal kinase and p38 MAPK (153). TNF{alpha} impairs insulin signaling by inhibiting the function of IRS-1 through serine phosphorylation (154, 155). Neutralization of circulating TNF{alpha} enhances insulin sensitivity in insulin-resistant rodent models of diabetes and obesity (153). The role of TNF{alpha} in the insulin resistance of human T2DM and obesity is less clear. Circulating TNF{alpha} levels are increased in obese nondiabetic (156) and T2DM individuals (157, 158), but the correlation between insulin resistance and plasma TNF{alpha} levels is weak in both obese (159) and diabetic (157) individuals. It should be emphasized, however, that TNF{alpha} exerts local paracrine effects, and the induction of insulin resistance may be more related to the local tissue TNF{alpha} concentration than to the levels in plasma (160). TNF{alpha} also is a potent inflammatory cytokine that has been implicated in the development of atherosclerosis in nonhuman models (160).

F. IL-6

IL-6, an inflammatory cytokine, is highly expressed in adipocytes (161) and plays an important role in inflammation and the regulation of T cell and ß-cell function (162). In rodent models of diabetes, IL-6 has been implicated in the development of muscle insulin resistance (163) and ß-cell apoptosis (40). In humans with T2DM, IL-6 levels are increased and correlate with the severity of glucose intolerance (164) and with the severity of inflammation, as indicated by the highly sensitive C-reactive protein (CRP) serum concentration (165). The correlation between IL-6 and the CRP reflects both a direct effect of IL-6 on CRP secretion and the inflammatory-provoking actions of IL-6 that secondarily augment CRP secretion.

G. Leptin

Leptin is produced exclusively by adipocytes and secreted into the bloodstream (166). It is an appetite suppressant that exerts its effects by interacting with a type 2 cytokinase receptor in the hypothalamus, where it interacts with neuropeptide Y, melanocyte-stimulating hormone, and the melanocortin-4 receptor (167, 168). Leptin-deficient rodents (167) and humans (169) develop hyperphagia and marked obesity, which are reversed by administration of the cytokine. When administered to rodents, some (170, 171, 172), but not all (173), studies have shown an insulin-sensitizing effect in muscle and adipocytes. Although large scale clinical studies in humans have failed to identify an effect of leptin on glucose metabolism in obese and T2DM subjects (174), mutations in the leptin gene in man are associated with severe obesity, glucose intolerance, and insulin resistance, which are reversed by leptin replacement therapy (175, 176).

An interesting aspect of leptin function that has received little attention is its potential role in inflammation. Leptin acts directly on macrophages to augment their phagocytic activity and increase their production of inflammatory cytokines (177). Leptin also is required for macrophages to manifest full immunocompetency (178, 179). Thus, leptin could play an important role in the inflammation associated with T2DM and atherosclerosis (180).

H. Adipsin and acylation-stimulating protein (ASP)

The proteins adipsin and ASP are highly expressed in adipocytes and are integral proximal components of the alternate complement pathway (181, 182). Because adipocytes lack the distal components of the complement pathway, cell lysis does not occur. Rather, ASP increases lipid deposition in adipocytes by enhancing glucose uptake and deposition into triglycerides (182). Consistent with this observation, both ASP (markedly) and adipsin (moderately) levels have been shown to be increased in human obesity and T2DM (183).

I. Inflammation, insulin resistance, and the adipocyte: a hypothesis

Considerable evidence links chronic inflammation, insulin resistance, T2DM, and atherosclerosis (180, 184). Based upon the preceding discussion, it is reasonable to hypothesize that dysfunctional fat cells provide a link that connects these seemingly diverse metabolic and cardiovascular disorders.

Acute phase proteins (CRP, sialic acid, fibrinogen, complement factors, PAI-1, and serum amyloid A) are increased in T2DM subjects and individuals with coronary artery disease (180, 184). Highly sensitive CRP is strongly predictive of the inflammatory atherosclerotic process and future myocardial infarction (185). Many acute phase proteins are stimulated by factors such as TNF{alpha}, ILs, and leptin, and they are released from multiple sites, including not only white blood cells, but also adipocytes (see preceding discussion). These fat cell-derived proteins are increased in insulin-resistant conditions, i.e. T2DM, obesity, atherosclerosis, and the metabolic syndrome (186, 187), and play important roles in the immune response. Thus, leptin enhances macrophage function and is required for a normal immune response (188). TNF{alpha} and IL-6, which also are highly expressed in fat cells, have clear roles in immune modulation (151, 160, 162, 189). The presence in adipose tissue of proteins (adipsin and ASP) in the proximal portion of the alternative complement pathway raises questions about their possible roles in the inflammatory process. Resistin, initially characterized as an insulin resistance factor, is a member of a family of resistin-like molecules that belong to the FIZZ (found in inflammatory zone) protein family (resistin is FIZZ3) (136, 190). Similarly, the adipokines adiponectin (191) and PAI-1 (147) have been implicated in the inflammatory process.

The mechanisms via which chronic inflammation produces insulin resistance in such diverse tissues as liver, skeletal muscle, fat, and vascular smooth muscle cells have yet to be elucidated. One possibility might be related to activation of serine/threonine phosphorylation cascades that lead, on the one hand, to activation of nuclear factor-{kappa}B and inflammation-related transcriptional regulation and, on the other hand, to serine phosphorylation of elements of the insulin receptor signaling system, especially IRS-1 (192). Evidence supporting this schema arises from the observation that high dose salicylates can improve insulin sensitivity through a mechanism that involves inhibition of I{kappa}ß kinase (IKK-ß) (193). Consistent with this hypothesis, heterozygous IKK-ß knockout mice have increased insulin sensitivity, and salicylate administration improves insulin sensitivity in insulin-resistant mice (194). IKK-ß induces insulin resistance by causing serine phosphorylation of IRS-1 and subsequent inhibition of the insulin signaling cascade. Salicylates also inhibit the activity of RSK2 (p90 ribosomal S6 kinase) (195), and IRS-1 contains several consensus serine/threonine phosphorylation sites for RSK2. Our laboratory has shown that RSK2 activity is increased 5-fold in skeletal muscle biopsies from patients with T2DM (196). Based upon the preceding discussion, some investigators have proposed an adipocentric view of insulin signaling and intracellular trafficking (197).


    IV. Fat Topography: Ectopic Fat, Insulin Resistance, and ß-Cell Failure
 Top
 Introduction
 I. Pathogenesis of T2DM
 II. Role of the...
 III. The Fat Cell...
 IV. Fat Topography: Ectopic...
 V. "Dysfunctional" Fat vs....
 References
 
A. Overview

The association between T2DM and obesity is well established. Cross-sectional (198, 199) and prospective (200, 201) studies have documented that the incidence of diabetes rises steeply with increasing body weight. The diabetogenic effect of obesity is related to three factors: attained body mass index, duration of obesity, and recent increase in body weight (202). Epidemiologic data (203, 204) as well as direct measurement with the euglycemic insulin clamp technique (205, 206, 207) have established obesity as an insulin-resistant state, and both obesity and insulin resistance are risk factors for the development of T2DM (1, 2, 208, 209). Like T2DM (21), the insulin resistance of obesity involves muscle (205, 206, 210), liver (203, 206, 211), and adipocytes (105, 212, 213).

In addition to total fat content, the pattern of fat distribution is also an important predictor of the body’s sensitivity to insulin. Individuals with preferential upper body fat accumulation (android) are more insulin resistant, hyperinsulinemic, and dyslipidemia than people with a preponderance of lower body fat (gynecoid) (214, 215). Using magnetic resonance imaging and computed tomography, increased visceral fat has been shown to be specifically related to the presence of insulin resistance (215, 216, 217, 218, 219). This association has been attributed to the enhanced lipolytic activity of visceral fat cells, with increased delivery of FFA into the portal (causing hepatic insulin resistance) and systemic (causing muscle insulin resistance) circulations (220, 221). However, there also is evidence to indicate that visceral fat cells produce excessive amounts of insulin-resistant, inflammatory-provoking adipocytokines (i.e. resistin, TNF{alpha}, IL-6, PAI-1, etc.) and undersecrete insulin-sensitizing adipocytokines (i.e. adiponectin) (222, 223, 224, 225).

B. Ectopic fat deposition: an emerging paradigm

Recent studies have demonstrated that organ-specific deposition of fat is a strong predictor of insulin resistance in muscle and liver. Increased intramyocellular triglyceride content, assessed by muscle biopsy (226) or magnetic resonance/computed tomography (34, 35, 227, 228, 229, 230), correlates closely with muscle insulin resistance and is a better predictor of impaired insulin action than visceral adiposity (231). Conversely, a decrease in intramyocellular lipid content brought about by weight loss is a strong correlate of the improved muscle insulin sensitivity (232, 233, 234).

Fat accumulation within the liver also is tightly correlated with hepatic insulin resistance. In type 2 diabetics, increased hepatic fat content, quantitated by magnetic resonance spectroscopy, is closely related to the basal hepatic insulin resistance index (HGP x fasting plasma insulin) and impaired suppression of HGP by insulin (37, 120, 121). Consistent with this observation, insulin requirements in T2DM patients are closely related to the hepatic fat content (235), and in nondiabetic individuals, hepatic fat stores are strongly correlated with hepatic sensitivity to insulin (36).

Inherited forms of lipodystrophy in humans, although rare, provide additional insight into the importance of ectopic fat deposition in the pathogenesis of insulin resistance and glucose intolerance. Human (236, 237) and animal (238) forms of lipodystrophy are characterized by selective loss of sc and visceral fat and are associated with a number of metabolic abnormalities, such as dyslipidemia, hyperglycemia, and insulin resistance. Insufficient adipose tissue mass leads to excessive storage of ingested fat in muscle and liver and the development of severe insulin resistance in these organs. Replacement therapy with leptin mobilizes fat out of the liver and muscle, leading to dramatic improvements in hepatic and muscle sensitivity to insulin and improved glycemic control (239, 240). Additional support for the ectopic fat paradigm comes from transgenic animal models with defects in adipogenesis (241, 242, 243, 244, 245). These lipodystrophic animals manifest marked fatty infiltration of the liver and muscle, hepatic and muscle insulin resistance, and IGT. Surgical transplantation of adipose tissue into these lipotrophic animals results in mobilization of fat out of the liver and muscle, improved insulin sensitivity in these organs, and correction of the glucose intolerance (246). Conversely, surgical removal of adipose tissue in normal glucose tolerant hamsters results in fat accumulation in liver and muscle, insulin resistance, and glucose intolerance (247).

The sequence of events by which the inability to store fat in adipose tissue leads to ectopic accumulation of triglycerides in muscle and liver, hepatic and muscle insulin resistance, glucose intolerance, and overt diabetes has been referred to as the overflow hypothesis (248). We believe that the overflow hypothesis has received sufficient scientific validation so that it no longer should be considered hypothetical. The overflow hypothesis also explains the well documented observation that enlarged fat cells correlate better with insulin resistance than any other measure of adiposity (249, 250, 251, 252) and that enlarged fat cells are a strong, independent predictor of the development of type 2 diabetes (253). Once the capacity of the fat cell to store triglycerides is exceeded, fat overflows to other tissues (muscle and liver), where the intracellular metabolites of triglyceride metabolism interfere with insulin signaling, glucose transport/phosphorylation, and glycogen synthesis in muscle and augment hepatic gluconeogenesis/total glucose production.

On a more speculative note, there are experimental data in rodent models of diabetes to support the overflow hypothesis of ß-cell dysfunction. In the genetically obese Zucker fatty rat, overt diabetes develops at about 10–12 wk of life, and this is associated with a marked increase in islet triglyceride content and FA-CoA levels (40, 46, 49, 50). The rise in ß-cell FA-CoA concentration leads to increased ceramide formation, stimulation of inducible nitric oxide synthase, and elevated nitric oxide levels, which, in concert with the release of inflammatory cytokines (TNF{alpha}, IL-6, and others), lead to ß-cell dysfunction and apoptosis (40, 46, 49, 50). At present, it remains unknown whether a similar sequence of events occurs in human ß-cells. However, chronic physiological and pharmacological elevation of the plasma FFA concentration, which would be expected to increase FFA transport into the ß-cell by mass action, has been shown to impair insulin secretion (32, 33, 52, 53, 54), whereas chronic reduction of the plasma FFA concentration with acipimox in genetically predisposed individuals augments insulin secretion (254).


    V. "Dysfunctional" Fat vs. "Healthy" Fat
 Top
 Introduction
 I. Pathogenesis of T2DM
 II. Role of the...
 III. The Fat Cell...
 IV. Fat Topography: Ectopic...
 V. "Dysfunctional" Fat vs....
 References
 
Most type 2 diabetics 1) possess too much fat; 2) have an abnormal distribution of fat with excessive fat deposition in muscle, liver, and visceral adipocytes; and 3) have large, insulin-resistant fat cells whose capacity to store triglycerides is compromised. These dysfunctional fat cells produce excessive amounts of adipocytokines that cause insulin resistance, inflammation, hypercoagulability, dyslipidemia, and possibly hypertension. They also underproduce adipocytokines (adiponectin) that play an essential role in maintaining normal insulin sensitivity. When viewed in this context, it is clear that the glucose intolerance of T2DM results at least in part secondary to a disordered fat cell metabolism. Because the derangements in adipocyte metabolism are present in normal glucose-tolerant, insulin-resistant, genetically predisposed individuals (5, 7, 12, 73), in people with IGT (11, 13, 29, 30, 31, 36, 72, 103, 187), and in insulin-resistant, normal glucose-tolerant obese individuals (30, 34, 36, 65, 66, 105), some authorities have considered disordered lipid/FFA metabolism to represent the primary disturbance in the pathogenesis of T2DM (255).

An important question is whether dysfunctional fat cells can be converted to healthy adipocytes, and whether the abnormal pattern of fat distribution can be reversed, leading to enhanced muscle/hepatic sensitivity to insulin and improved ß-cell function. Thiazolidinediones represent a novel class of antidiabetic agents that improve glycemic control (256, 257, 258, 259, 260), enhance hepatic and muscle insulin sensitivity (261, 262, 263, 264, 265, 266, 267, 268), and improve ß-cell function in both human and animal models of T2DM (40, 269, 270, 271, 272). A consistent finding in these studies is the association between weight gain and reduction in hemoglobin A1c (HbA1c) with troglitazone (273, 274, 275, 276), rosiglitazone (263, 277, 278), and pioglitazone (261, 262, 279). The greater the weight gain, the better the improvement in glycemic control. This observation is paradoxical, because weight gain brought about by overeating is associated with insulin resistance and deterioration in glycemic control (3, 280, 281). It also is noteworthy that obese individuals respond better than lean subjects to thiazolidinediones, and that women, who have a higher percentage of body fat, respond better than men (279, 282).

The glucose-lowering efficacy of the thiazolidinediones is related to their ability to bind to PPAR{gamma} (160, 266, 283). Similar beneficial effects on glucose homeostasis can be expected from nonthiazolidinedione PPAR{gamma} agonists (283, 284). PPAR{gamma} is one of a group of three nuclear receptor isoforms (the other two are PPAR{alpha} and PPAR{delta}) that are encoded by different genes (284, 285, 286). PPARs are ligand-regulated nuclear transcription factors that regulate gene expression by binding to specific peroxisome proliferator response elements within the promoter region (160, 258, 283, 284, 285). PPAR{gamma} exists as two isoforms, {gamma}1 and {gamma}2. PPAR{gamma}1 is primarily and highly expressed in adipose tissue, whereas PPAR{gamma}2 is expressed in a wide variety of tissues (160, 283, 284, 285, 287, 288, 289). Of note is the low expression of both PPAR{gamma}1 and PPAR{gamma}2 in skeletal muscle (283, 288, 289), making this tissue an unlikely target for any direct action of the thiazolidinediones. PPARs associate with the retinoic acid receptor, and the PPAR/retinoic acid X receptor complex binds with cofactors to initiate gene transcription (160, 266, 283, 284, 285). The naturally occurring ligands for the PPAR{gamma} are fatty acids and eicosanoids, which are active at micromolar concentrations (283, 284, 290, 291). Thiazolidinediones are synthetic ligands that are potent PPAR{gamma} agonists (160, 266, 283, 284, 285).

PPAR{gamma} is a critical transcription factor in the differentiation of preadipocytes into adipocytes (292, 293, 294, 295). Three families of transcription factors play central roles in the differentiation and maturation of preadipocytes into adipocytes: 1) PPAR{gamma}; 2) CCAAT/enhancer-binding proteins (C/EBPs) {alpha}, ß, and {delta}; 3) a sterol response element-binding protein (296, 297, 298, 299). PPAR{gamma} is the master regulator of adipogenesis, being both essential and sufficient for adipocyte differentiation (300). C/EBPß, C/EBP{delta}, and sterol response element-binding protein-1c act early in adipogenesis to induce PPAR{gamma} expression, whereas C/EBP{alpha} interacts with PPAR{gamma} to facilitate the expression of multiple genes in the mature fat cell, including the insulin-sensitive glucose transport system (301, 302) and other key genes involved in lipid homeostasis (including phosphoenolpyruvate carboxykinase, acyl-CoA synthetase, lipoprotein lipase, and adipocyte P2) (303, 304, 305, 306). Each of these genes contains a PPAR response element in its regulatory region. PPAR{gamma} also up-regulates the expression of fatty acid transporter proteins (FATP-1 and D036) (307, 308) and c-Cbl-associated protein (309). The net result of these changes is to enhance glucose (providing the carbon skeleton) and FFA transport into the adipocyte and to stimulate triglyceride synthesis. Consistent with these molecular actions, all thiazolidinediones cause a marked reduction in plasma FFA concentration and inhibit lipolysis in T2DM patients (261, 262, 263, 264, 265, 266, 268). Thiazolidinediones also inhibit the expression of the leptin gene in adipocytes (160, 228), and the resultant decline in the plasma leptin concentration (157) may increase appetite and contribute to weight gain (167, 169).

In rodents, thiazolidinediones increase the number of small fat cells by stimulating adipogenesis in sc fat depots (295, 310, 311, 312). Although PPAR{gamma} receptors are present in visceral adipocytes and preadipocytes in rodents, they are not activated to divide. Thiazolidinediones also cause apoptosis of large fat cells in both visceral and sc regions in rodents (295, 311, 312). These observations are consistent with ex vivo studies in which thiazolidinediones (rosiglitazone and pioglitazone) were shown to markedly enhance the differentiation of human sc preadipocytes into mature small adipocytes (310). Moreover, prior transfection of these human preadipocytes with a mutant PPAR{gamma} blocked this differentiation process (313). Two studies have examined sc abdominal fat cell size in adipose depots after treatment with thiazolidinediones in humans. One study reported a trend toward smaller fat cells (314), whereas the other study reported a slight increase in fat cell size (315). However, the number of diabetics treated with thiazolidinediones in both studies was small, and even if thiazolidinediones caused sc preadipocytes to differentiate into small fat cells, they would eventually accumulate fat and increase in size. Studies using magnetic resonance imaging consistently have shown that troglitazone, pioglitazone, and rosiglitazone cause a shift in fat topography, characterized by a decrease in intraabdominal fat and an increase in sc fat (261, 262, 263, 268, 275, 316, 317) (Fig. 3Go). Whether the increase in sc fat mass observed in humans is due to increased fat cell number (adipogenesis), increased fat cell size (induction of genes involved in lipogenesis and inhibition of lipoprotein lipase), or some combination of the two remains to be determined. As visceral fat is strongly related to increased gluconeogenesis and HGP in T2DM individuals (211), it is not surprising that thiazolidinedione therapy improves hepatic insulin sensitivity and reduces hepatic gluconeogenesis and total hepatic glucose output (261, 262, 264, 265). It is noteworthy that surgical removal of visceral fat in rodent models of type 2 diabetes also improves hepatic insulin sensitivity, reduces HGP and gluconeogenesis, and improves glucose tolerance (277, 318). Although the improvement in hepatic insulin sensitivity has been attributed to decreased portal FFA delivery to the liver from the lipolytically more active visceral fat cells (220, 319, 320, 321, 322, 323), one also could postulate that a reduction in insulin-resistant adipocytokines (such as resistin or TNF{alpha}) or an increase in insulin-sensitizing adipocytokines (such as adiponectin) is responsible for the improvement in hepatic insulin sensitivity (116, 118, 119). In fat, evidence for this later scenario has been provided in both man (120, 121, 134) and animals (133).



View larger version (26K):
[in this window]
[in a new window]
 
FIG. 3. Effect of PPAR{gamma} agonists on fat distribution. [Adapted from Treatment of Type 2 Diabetes Mellitus: A Sound Approach Based upon its Pathophysiology, a CME program sponsored by the Academy of Medicine of New Jersey (Lawrenceville, NJ).]

 
The increase in FFA uptake by sc fat cells (whether due to hyperplasia or hypertrophy) also explains the consistent decrease in plasma FFA concentration observed with all thiazolidinediones and the strong correlation between improved muscle insulin sensitivity and reduction in plasma FFA concentration/FFA turnover (261, 262, 263, 265, 268). As the plasma FFA concentration declines, one also would predict a mobilization of fat out of the muscle and liver (Fig. 3Go), with improved insulin sensitivity in these organs. Indeed, both pioglitazone (120, 121) and rosiglitazone (268) been shown to reduce hepatic fat content in association with an improvement in basal hepatic insulin sensitivity and enhanced insulin-mediated suppression of HGP in T2DM subjects. Pioglitazone therapy also enhances splanchnic glucose uptake of an oral glucose load, although the increased splanchnic glucose uptake correlates more closely with the reduction in HbA1c than the decrease in hepatic fat content (120). In the one published study that examined intramyocellular fat content after rosiglitazone therapy, no change was observed (268). However, it is the decrease in intracellular concentration of metabolites of muscle triglycerides (FA-CoAs, DAG, ceramides, etc.) that best predicts the improvement in muscle sensitivity to insulin (71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 86, 87, 88, 89), and these metabolites were not measured in this study (268). Recently, we have shown that the reduction in plasma FFA concentration is associated with improved insulin sensitivity in muscle and enhanced insulin receptor and IRS-1 tyrosine phosphorylation, increased association of IRS-1 with phosphoinositol 3-kinase, and increased phosphoinositol 3-kinase activity (324).

The sequence of events described above explains why weight gain (261, 262, 263, 273, 279) and reduced plasma FFA concentrations (261, 262, 263, 265, 268) are strongly correlated with the reduction in HbA1c in T2DM patients treated with thiazolidinediones. In the absence of an isocaloric diet or caloric restriction, weight gain is a predictable consequence of thiazolidinedione therapy. When viewed in this context, weight gain, along with the reduction in plasma FFA concentration, indicates that the thiazolidinedione is working and exerting beneficial effects to improve tissue sensitivity to insulin and glucose homeostasis. Further, the decline in plasma FFA is indicative of a "healthier" fat cell, which now responds well to insulin stimulation of glucose uptake and suppression of lipolysis (263, 324). Other indications that the thiazolidinediones have converted the dysfunctional fat cell in type 2 diabetics into a healthier fat cell include 1) inhibition of resistin, TNF{alpha}, and PAI-1 gene expression in adipocytes and reduction in their circulating levels (127, 128, 160, 197, 228, 325, 326), and 2) stimulation of adiponectin gene expression in adipocytes and increase in the plasma adiponectin concentration (121, 326, 327).

In rodent models of diabetes, thiazolidinediones improve ß-cell function in concert with a reduction in islet fat content (32, 40, 50) and preserve islet histology and ß-cell mass (328, 329). In diabetic rats, thiazolidinediones prevent the loss of ß-cell mass by maintaining ß-cell proliferation and inhibiting ß-cell apoptosis (330). In cultured human islets, FFA down-regulate insulin mRNA expression, inhibit glucose-stimulated insulin secretion, and promote ß-cell death. All of these deleterious effects of FFA are prevented when islets are incubated with rosiglitazone (331, 332). In humans, thiazolidinedione therapy has been shown to improve the insulinogenic index (incremental insulin response/incremental glucose response during the oral glucose tolerance test) in a dose-dependent fashion (269). In the Troglitazone in Prevention of Diabetes (TRIPOD) study, treatment of women with a prior history of gestational diabetes mellitus with troglitazone markedly reduced the conversion rate to T2DM over a 30-month period (333). Although troglitazone consistently improved insulin sensitivity (measured with the frequently sampled iv glucose tolerance test) in gestational diabetes mellitus responders, improved ß-cell function was the best predictor of response (333). The protective effect of troglitazone against the development of T2DM and the drug’s beneficial effect on ß-cell function persisted for an additional 8 months after cessation of troglitazone (333). An improved ß-cell response to glucose in subjects with IGT also has been reported by other investigators (334). One could speculate that a mobilization of fat out of ß-cells into sc fat depots explains the long-lasting (8 months) beneficial effects of troglitazone on ß-cell function as well as on tissue sensitivity to insulin. Moreover, one would predict that as long as the triglyceride remained within sc fat cells, the improvements in ß-cell function and tissue insulin sensitivity would persist.


    Footnotes
 
Abbreviations: ASP, Acylation-stimulating protein; C/EBP, CCAAT/enhancer-binding protein; CoA, coenzyme A; CRP, C-reactive protein; DAG, diacylglycerol; FA, fatty acyl; FFA, free fatty acid; G-6-P, glucose-6-phosphate; HbA1c, hemoglobin A1c; HGP, hepatic glucose production; IGT, impaired glucose tolerance; IKK-ß, I{kappa}B kinase; IRS-1, insulin receptor substrate-1; PAI-1, plasminogen activator inhibitor-1; PPAR, peroxisomal proliferator-activated receptor; RSK2, p90 ribosomal S6 kinase; T2DM, type 2 diabetes mellitus.

Received April 24, 2003.

Accepted November 3, 2003.


    References
 Top
 Introduction
 I. Pathogenesis of T2DM
 II. Role of the...
 III. The Fat Cell...
 IV. Fat Topography: Ectopic...
 V. "Dysfunctional" Fat vs....
 References
 

  1. DeFronzo RA 1988 Lilly Lecture. The triumvirate: ß cell, muscle, liver. A collusion responsible for NIDDM. Diabetes 37:667–687[Medline]
  2. DeFronzo RA 1997 Pathogenesis of type 2 diabetes: metabolic and molecular implications for identifying diabetes genes. Diabetes Rev 4:177–269
  3. Kelley DE 1995 Effects of weight loss on glucose homeostasis in NIDDM. Diabetes Rev 3:366–377
  4. Schneider SH, Morgado A 1995 Effects of fitness and physical training on carbohydrate metabolism and associated cardiovascular risk factors in patients with diabetes. Diabetes Rev 3:378–407
  5. Gulli G, Ferrannini E, Stern M, Haffner S, DeFronzo RA 1992 The metabolic profile of NIDDM is fully established in glucose-tolerant offspring of two Mexican-American NIDDM parents. Diabetes 41:1575–1586[Abstract]
  6. Tripathy D, Carlsson M, Almgren P, Isomaa B, Taskinen M-R, Tuomi T, Groop LC 2000 Insulin secretion and insulin sensitivity in relation to glucose tolerance. Lessons from the Botnia Study. Diabetes 49:975–980[Abstract]
  7. Pratipanawatr W, Pratipanawatr T, Cusi K, Berria R, Adams JM, Jenkinson CP, Maezono K, DeFronzo RA, Mandarino LJ 2001 Skeletal muscle insulin resistance in normoglycemic subjects with a strong family history of type 2 diabetes is associated with decreased insulin-stimulated insulin receptor substrate-1 tyrosine phosphorylation. Diabetes 50:2572–2578[Abstract/Free Full Text]
  8. Jackson S, Bagstaff SM, Lynn S, Yeaman SJ, Turnbull DM, Walker M 2000 Decreased insulin responsiveness of glucose uptake in cultured human skeletal muscle cells from insulin-resistant nondiabetic relatives of type 2 diabetic families. Diabetes 49:1169–1177[Abstract]
  9. Yalow RS, Berson SA 1960 Immunoassay of endogenous plasma insulin in man. J Clin Invest 39:1157–1175
  10. Diamond MP, Jacobs R, Connolly-Howard M, DeFronzo RA 1993 Glucose metabolism during the menstrual cycle: assessment by the euglycemic, hyperinsulinemic clamp technique. J Reprod Med 38:417–421[Medline]
  11. Reaven GM, Hollenbeck CB, Chen YDI 1989 Relationship between glucose tolerance, insulin secretion, and insulin action in non-obese individuals with varying degrees of glucose tolerance. Diabetologia 32:52–55[Medline]
  12. Vauhkonen I, Niskanen L, Vanninen E, Kainulainen S, Uusitupa M, Laakso M 1997 Defects in insulin secretion and insulin action in non-insulin-dependent diabetes mellitus are inherited. Metabolic studies on offspring of diabetic probands. J Clin Invest 100:86–96
  13. Weyer C, Bogardus C, Pratley RE 1999 Metabolic characteristics of individuals with impaired fasting glucose and/or impaired glucose tolerance. Diabetes 48:2197–2203[Abstract]
  14. Mirrakou A, Kelly D, Mokan M, Veneman T, Pangburn T, Reilly J, Gerich J 1992 Role of reduced suppression of glucose production and diminished early insulin release in impaired glucose tolerance. N Engl J Med 326:22–29[Abstract]
  15. Polonsky KS, Sturis J, Bell GI 1996 Non-insulin-dependent diabetes mellitus: a genetically programmed failure of the ß cell to compensate for insulin resistance. N Engl J Med 334:777–783[Free Full Text]
  16. Kahn SE 2001 The importance of ß-cell failure in the development and progression of type 2 diabetes. J Clin Endocrinol Metab 86:4047–4058[Free Full Text]
  17. Jensen CC, Cnop M, Hull RL, Fujimoto WY, Kahn SE, and the American Diabetes Association GENNID Study Group 2002 ß-Cell function is a major contributor to oral glucose tolerance in high-risk relatives of four ethnic groups in the U.S. Diabetes 51:2170–2178[Abstract/Free Full Text]
  18. Bogardus C, Lillioja S, Howard BV, Reaven G, Mott D 1984 Relationships between insulin secretion, insulin action, and fasting plasma glucose concentration in nondiabetic and noninsulin-dependent diabetic subjects. J Clin Invest 74:1238–1246[Medline]
  19. Saad MF, Knowler WC, Pettitt DJ, Nelson RG, Mott DM, Bennett PH 1989 Sequential changes in serum insulin concentration during development of non-insulin-dependent diabetes. Lancet 1:1356–1359[Medline]
  20. DeFronzo RA, Gunnarsson R, Bjorkman O, Olsson M, Wahren J 1985 Effects of insulin on peripheral and splanchnic glucose metabolism in non-insulin-dependent (type II) diabetes mellitus. J Clin Invest 76:149–155[Medline]
  21. Groop LC, Bonadonna RC, Del Prato S, Ratheiser K, Zyck K, DeFronzo RA 1989 Effect of insulin on oxidative and non-oxidative pathways of glucose and FFA metabolism in NIDDM. Evidence for multiple sites of insulin resistance. J Clin Invest 84:205–213[Medline]
  22. DeFronzo RA, Ferrannini E, Simonson DC 1989 Fasting hyperglycemia in non-insulin dependent diabetes mellitus: contributions of excessive hepatic glucose production and impaired tissue glucose uptake. Metabolism 38:387–395[Medline]
  23. Ferrannini E, Simonson DC, Katz LD, Reichard G, Bevilacqua S, Barrett EJ, Olsson M, DeFronzo RA 1988 The disposal of an oral glucose load in patients with non-insulin dependent diabetes. Metabolism 37:79–85[Medline]
  24. Shulman GI, Rothman DL, Jue T, Stein P, DeFronzo RA, Shulman RG 1990 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 322:223–228[Abstract]
  25. Magnusson I, Rothman D, Katz L, Shulman R, Shulman G 1992 Increased rate of gluconeogenesis in type II diabetes: a 13C nuclear magnetic resonance study. J Clin Invest 90:1323–1327[Medline]
  26. Mokdad AH, Ford ES, Bowman BA, Nelson DE, Engelgau MM, Vinicor F, Marks JS 2000 Diabetes trends in the United States, 1990–1998. Diabetes Care 23:1278–1283[Abstract]
  27. Reaven GM, Hollenbeck C, Jeng C-Y, Wu MS, Chen Y-DI 1988 Measurement of plasma glucose, free fatty acid, lactate, and insulin for 24 hours in patients with NIDDM. Diabetes 37:1020–1024[Abstract]
  28. Boden G, Shulman GI 2002 Free fatty acids in obesity and type 2 diabetes: defining their role in the development of insulin resistance and ß cell dysfunction. Eur J Clin Invest 32(Suppl 3):14–23
  29. Reaven GM 1995 The fourth musketeer—from Alexandre Dumas to Claude Bernard. Diabetologia 38:3–13[CrossRef][Medline]
  30. Golay A, Felber JP, Jequier E, DeFronzo RA, Ferrannini E 1988 Metabolic basis of obesity and noninsulin-dependent diabetes mellitus. Diabetes Metab Rev 4:727–747[Medline]
  31. Boden G 1997 Role of fatty acids in the pathogenesis of insulin resistance and NIDDM. Diabetes 46:3–10[Abstract]
  32. McGarry JD 2002 Banting Lecture 2001: dysregulation of fatty acid metabolism in the etiology of type 2 diabetes. Diabetes 51:7–18[Free Full Text]
  33. Kashyap S, Belfort R, Pratipanawatr T, Berria R, Pratipanawatr W, Bajaj M, Mandarino L, DeFronzo R, Cusi K 2002 Chronic elevation in plasma free fatty acids impairs insulin secretion in non-diabetic offspring with a strong family history of T2DM. Diabetes 51(Suppl 2):A12
  34. Goodpaster BH, Thaete FL, Kelley BE 2000 Thigh adipose tissue distribution is associated with insulin resistance in obesity and in type 2 diabetes mellitus. Am J Clin Nutr 71:885–892[Abstract/Free Full Text]
  35. Greco AV, Mingrone G, Giancaterini A, Manco M, Morroni M, Cinti S, Granzotto M, Vettor R, Camastra S, Ferrannini E 2002 Insulin resistance in morbid obesity. Reversal with intramyocellular fat depletion. Diabetes 51:144–151[Abstract/Free Full Text]
  36. Seppala-Lindroos A, Vehkavaara S, Hakkinen A-M, Goto T, Westerbacka J, Sovijarvi A, Halavaara J, Yki-Jarvinen H 2002 Fat accumulation in the liver is associated with defects in insulin suppression of glucose production and serum free fatty acids independent of obesity in normal men. J Clin Endocrinol Metab 87:3023–3028[Abstract/Free Full Text]
  37. Miyazaki Y, Hardies LJ, Wajcberg E, Glass L, Triplitt C, Bajaj M, Cersosimo E, Mandarion LJ, DeFronzo RA 2002 Effect of pioglitazone on liver fat content, abdominal fat distribution, and insulin sensitivity in patients with type 2 diabetes mellitus. Diabetes 51(Suppl 2):A69
  38. Prentki M, Corkey BE 1996 Are the B-cell signaling molecules malonyl-CoA and cytosolic long-chain acyl-CoA implicated in multiple tissue defects of obesity and NIDDM? Diabetes 45:273–283[Abstract]
  39. Unger RH 1996 Lipotoxicity in the pathogenesis of obesity-dependent NIDDM: genetic and clinical implications. Diabetes 45:273–283[Abstract]
  40. Shimabukuro M, Zhou YT, Levi M, Unger RH 1998 Fatty acid induced ß cell apoptosis. Proc Natl Acad Sci USA 95:2498–2502[Abstract/Free Full Text]
  41. Matschinsky FM 1996 Banting Lecture 1995: a lesson in metabolic regulation inspired by the glucokinase glucose sensor paradigm. Diabetes 45:223–241[Abstract]
  42. Newgard CB, McGarry JD 1995 Metabolic coupling factors in pancreatic ß cell signal transduction. Annu Rev Biochem 64:689–719[CrossRef][Medline]
  43. McGarry JD, Dobbins RL 1999 Fatty acids, lipotoxicity and insulin secretion. Diabetologia 42:128–138[CrossRef][Medline]
  44. Warnotte C, Gilon P, Nenquin M, Henquin JC 1994 Mechanisms of the stimulation of insulin release by saturated fatty acids: a study of palmitate effects of mouse B-cells. Diabetes 43:703–711[Abstract]
  45. Stein DT, Esser V, Stevenson BE, Lane KE, Whiteside JH, Daniels MB, Chen S, McGarry JD 1996 Essentially of circulating fatty acids for glucose-stimulated insulin secretion in the fasted rat. J Clin Invest 97:2728–2735[Abstract/Free Full Text]
  46. Zhou UP, Grill VE 1994 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 93:870–876[Medline]
  47. Sako Y, Grill VE 1990 A 48-h lipid infusion in the rat time-dependently inhibits glucose-induced insulin secretion and ß cell oxidation through a process likely coupled to fatty acid oxidation. Endocrinology 127:1580–1589[Abstract]
  48. Mason TM, Goh T, Tchipashvili V, Sandhu H, Gupta N, Lewis GF, Giacca A 1999 Prolonged elevation of plasma free fatty acids desensitizes the insulin secretory response to glucose in vivo in rats. Diabetes 48:524–530[Abstract]
  49. Zhou YP, Grill V 1995 Long term exposure to fatty acids and ketones inhibits b-cell function in human pancreatic islets of Langerhans. J Clin Endocrinol Metab 80:1584–1590[Abstract]
  50. Unger RH 1997 How obesity causes diabetes in Zucker diabetic fatty rats. Trends Endocrinol Metab 7:276–282
  51. Lupi R, Dotta F, Marselli L, Del Guerra S, Masini M, Santangelo C, Patane G, Boggi U, Piro S, Anello M, Bergamini E, Mosca F, Di Mario U, Del Prato S, Marchetti P 2002 Prolonged exposure to free fatty acids has cytostatic and pro-apoptotic effects on human pancreatic islets. Evidence that ß cell death is caspase mediated, partially dependent on ceramide pathway, and Bcl-2 regulated. Diabetes 51:1437–1442[Abstract/Free Full Text]
  52. Carpentier A, Mittelman SD, Lamarche B, Bergman RN, Giacca A, Lewis GF 1999 Acute enhancement of insulin secretion by FFA in humans is lost with prolonged FFA elevation. Am J Physiol 276:E1055–E1066
  53. Paolisso G, Gambardella A, Amato L, Tortoriello R, D’Amore A, Varricchio M, D’Onofrio F 1995 Opposite effects of short- and long-term fatty acid infusion on insulin secretion in healthy subjects. Diabetologia 38:1295–1299[CrossRef][Medline]
  54. Carpentier A, Mittelman SD, Bergman RN, Giacca A, Lewis GF 2000 Prolonged elevation of plasma free fatty acids impair pancreatic B-cell function in obese nondiabetic humans but not in individuals with type 2 diabetes. Diabetes 49:399–408[Abstract]
  55. Stefan N, Fristche A, Haring H, Stumvoll M 2001 Effect of experimental elevation of free fatty acids on insulin secretion and insulin sensitivity in healthy carriers of the Pro12Ala polymorphism of the peroxisome proliferators-activated receptor-{gamma}2 gene. Diabetes 50:1143–1148[Abstract/Free Full Text]
  56. Paolisso G, Tagliamonte MR, Rizzo MR, Gualdiero P, Saccomanno F, Gambardella A, Giugliano D, D’Onofrio F, Howard BV 1998 Lowering fatty acids potentiates acute insulin response in first degree relatives of people with type II diabetes. Diabetologia 41:1127–1132[CrossRef][Medline]
  57. Qvigstad E, Mostad IL, Bjerve KS, Grill VE 2003 Acute lowering of circulating fatty acids improves insulin secretion in a subset of type 2 diabetes subjects. Am J Physiol 284:E129–E137
  58. Kaiyala KJ, Prigeon RL, Kahn SE, Woods SC, Porte Jr D, Schwartz MW 1999 Reduced ß-cell function contributes to impaired glucose tolerance in dogs made obese by high-fat feeding. Am J Physiol 277:E659–E667
  59. Bergman RN, Ader M 2000 Free fatty acids and pathogenesis of type 2 diabetes mellitus. TEM 11:351–356[Medline]
  60. Charles MA, Eschwege E, Thibult N, Claude JR, Warnet JM, Rosselin GE, Girard J, Balkau B 1997 The role of non-esterified fatty acids in the deterioration of glucose tolerance in Caucasian subjects: results of the Paris Prospective Study. Diabetologia 40:1101–1106[CrossRef][Medline]
  61. Knowler WC, Pettitt DJ, Saad MF, Bennett PH 1990 Diabetes mellitus in the Pima Indians: incidence, risk factors and pathogenesis. Diab Metab Rev 6:1–27[Medline]
  62. Unger RH 2002 Lipotoxic diseases. Annu Rev Med 53:319–336[CrossRef][Medline]
  63. Randle PJ, Garland PB, Hales CN, Newsholme EA 1963 The glucose fatty acid cycle: its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus. Lancet 1:785–789.[Medline]
  64. Wititsuwannakul D, Kim K 1977 Mechanism of palmityl coenzyme A inhibition of liver glycogen synthase. J Biol Chem 252:7812–7817[Abstract]
  65. Felber JP, Meyer HU, Curchod B, Iselin HU, Rousselle J, Maeder E, Pahud P, Jequier E 1981 Glucose storage and oxidation in different degrees of human obesity measured by continuous indirect calorimetry. Diabetologia 20:39–44[Medline]
  66. Golay A, Felber JP, Meyer HU, Curchod B, Maeder E, Jequier E 1984 Study on lipid metabolism in obesity diabetes. Metabolism 33:111–116[Medline]
  67. Thiebaud D, DeFronzo RA, Jacot E, Golay A, Acheson K, Maeder E, Jequier E, Felber JP 1982 Effect of long chain triglyceride infusion on glucose metabolism in man. Metabolism 21:1128–1136
  68. Roden M, Krssak M, Stingl H, Gruber S, Hofer A, Furnsinn C, Moser E, Waldhausl W 1999 Rapid impairment of skeletal muscle glucose transport/phosphorylation by free fatty acids in humans. Diabetes 48:358–364[Abstract/Free Full Text]
  69. Boden G 1999 Free fatty acids, insulin resistance, and type 2 diabetes mellitus. Proc Assoc Am Physicians 111:241–248[Medline]
  70. Boden G, Chen X, Ruiz J, White JV, Rossetti L 1994 Mechanisms of fatty acid-induced inhibition of glucose uptake. J Clin Invest 93:2438–2446[Medline]
  71. Pan DA, Lillioja S, Kriketos AD, Milner MR, Baur LA, Bogardus C, Jenkins AB, Storlien LH 1997 Skeletal muscle triglyceride levels are inversely related to insulin action. Diabetes 46:983–988[Abstract]
  72. Krssak M, Falk Petersen K, Dresner A, DiPietro L, Vogel SM, Rothman DL, Roden M, Shulman GI 1999 Intramyocellular lipid concentrations are correlated with insulin sensitivity in humans: a 1HNMR spectroscopy study. Diabetologia 42:113–116[CrossRef][Medline]
  73. Jacobs S, Machann J, Rett K, Brechtel K, Volk A, Renn W, Maerker E, Matthaei S, Schick F, Claussen CD, Hearing H-U 1999 Association of increased intramyocellular lipid content with insulin resistance in lean non-diabetic offspring of type 2 diabetic subjects. Diabetes 48:1113–1119[Abstract]
  74. Martin WH 1996 Effects of acute and chronic exercise on fat metabolism. Exercise Sport Sci Rev 24:203–231[Medline]
  75. Abernethy PJ, Thaver R, Taylor AW 1990 Acute and chronic responses of skeletal muscle to endurance and sprint exercise. A review. Sports Med 10:365–389[Medline]
  76. Kelley DE, He J, Menshikova EV, Ritov VB 2002 Dysfunction of mitochondria in human skeletal muscle in type 2 diabetes. Diabetes 51:2944–2950[Abstract/Free Full Text]
  77. Kelley DE, Mandarino LJ 2000 Fuel selection in human skeletal muscle in insulin resistance. Diabetes 49:677–683[Abstract]
  78. Itani SI, Ruderman NB, Schmieder F, Boden G 2002 Lipid-induced insulin resistance in human muscle is associated with changes in diacylglycerol, protein kinase C, and I{kappa}B-{alpha}. Diabetes 51:2005–2011[Abstract/Free Full Text]
  79. Ellis BA, Poynten A, Lowy AJ, Furler SM, Chisholm DJ, Kraegen EW, Cooney GJ 2000 Long-chain acyl-CoA esters as indicators of lipid metabolism and insulin sensitivity in rat and human muscle. Am J Physiol 279:E554–E560
  80. De Fea K, Roth RA 1997 Protein kinase C modulation of insulin receptor substrate-1 tyrosine phosphorylation requires serine 612. Biochemistry 36:12939–12947[CrossRef][Medline]
  81. Ravichandran LV, Esposito DL, Chen J, Quon MJ 2001 PKC-{zeta} phosphorylates IRS-1 and impairs its ability to activate P-3-kinase in response to insulin. J Biol Chem 276:3543–3549[Abstract/Free Full Text]
  82. Faergeman NJ, Knudsen J 1997 Role of long-chain fatty acyl-CoA esters in the regulation of metabolism and in cell signaling. Biochem J 323:1–12[Medline]
  83. Kruszynska YT, Worrall DS, Ofrecio J, Frias JP, Macaraeg G, Olefsky JM 2002 Fatty acid-induced insulin resistance: decreased muscle PI3K activation but unchanged Akt phosphorylation. J Clin Endocrinol Metab 87:226–234[Abstract/Free Full Text]
  84. Dresner A, Laurent D, Marcucci M, Griffin ME, Dufour S, Cline SW, Slezak LA, Andersen DK, Hundal RS, Rothman DL, Petersen KF, Shulman GI 1999 Effects of free fatty acids on glucose transport and IRS-1 associated phosphatidylinositol 3-kinase activity. J Clin Invest 103:253–259[Abstract/Free Full Text]
  85. Kashyap S, Belfort R, Finlayson J, Barrentine A, Mandarino L, DeFronzo RA, Cusi K 2002 A chronic physiologic increase in FFA in healthy subjects causes insulin signaling defects similar to those of insulin-resistant non-diabetic subjects with a strong family history of T2DM. Diabetes 51(Suppl 2):A24
  86. Thompson AL, Cooney GJ 2000 Acyl-CoA inhibition of hexokinase in rat and human skeletal muscle is a potential mechanism of lipid-induced insulin resistance. Diabetes 49:1761–1764[Abstract]
  87. Tippett PS, Neet KE 1982 An allosteric model for the inhibition of glucokinase by long chain acyl coenzyme A. J Biol Chem 257:14846–14852
  88. Schmitz-Peiffer C, Craig DL, Biden TJ 1999 Ceramide generation is sufficient to account for the inhibition of the insulin-stimulated PKB pathway in C2C12 skeletal muscle cells pretreated with palmitate. J Biol Chem 274:24202–24210[Abstract/Free Full Text]
  89. Chavez JA, Knotts TA, Wang LP, Lit G, Dobrowsky RT, Florant GL, Summers SA 2003 A role for ceramide, but not diacylglycerol, in the antagonism of insulin signal transduction by saturated fatty acids. J Biol Chem 278:10297–10303[Abstract/Free Full Text]
  90. DeFronzo RA, Ferrannini E, Hendler R, Wahren J, Felig P 1978 Influence of hyperinsulinemia, hyperglycemia, and the route of glucose administration on splanchnic glucose exchange. Proc Natl Acad Sci USA 75:5173–5177[Abstract]
  91. DeFronzo RA, Ferrannini E, Hendler R, Felig P, Wahren J 1983 Regulation of splanchnic and peripheral glucose uptake by insulin and hyperglycemia. Diabetes 32:35–45[Medline]
  92. Cherrington AD 1999 Banting Lecture. Control of glucose uptake and release by the liver in vivo. Diabetes 48:1198–1214[Free Full Text]
  93. Exton JH, Corbin JG, Park CR 1969 Control of gluconeogenesis in liver. IV. Differential effects of fatty acids and glucagon on ketogenesis and gluconeogenesis in the perfused rat liver. J Biol Chem 244:4095–4102[Abstract/Free Full Text]
  94. Bahl JJ, Matsuda M, DeFronzo RA, Bressler R 1997 In vitro and in vivo suppression of gluconeogenesis by inhibition of pyruvate carboxylase. Biochem Pharmacol 53:67–74[CrossRef][Medline]
  95. Williamson JR, Kreisberg RA, and Felts PW 1966 Mechanisms for the stimulation of gluconeogenesis by fatty acids in perfused rat liver. Proc Natl Acad Sci USA 56:247–254[Medline]
  96. Massillon D, Barzailai N, Hawkins M, Prus-Wetheimer D, Rossetti L 1997 Induction of hepatic glucose-6-phosphatase gene expression by lipid infusion. Diabetes 46:153–157[Abstract]
  97. Chen X, Iqbal N, Boden G 1999 The effect of free fatty acids on gluconeogenesis and glycogenolysis in normal subjects. J Clin Invest 103:365–372[Abstract/Free Full Text]
  98. Roden M, Stingl H, Chandramouli V, Schumann WC, Hofer A, Landau BR, Nowotny P, Waldhausl W, Shulman GI 2000 Effects of free fatty acid elevation on postabsorptive endogenous glucose production and gluconeogenesis in humans. Diabetes 49:701–707[Abstract]
  99. Puhakainen I, Yki-Jarvinen H 1993 Inhibition of lipolysis decreases lipid oxidation and gluconeogenesis from lactate but not fasting hyperglycemia or total hepatic glucose production in NIDDM. Diabetes 42:1694–1699[Abstract]
  100. Mittelman SD, Bergman RN 2000 Inhibition of lipolysis causes suppression of endogenous glucose production independent of changes in insulin. Am J Physiol 279:E630–E637
  101. Ferrannini E, Barrett EJ, Bevilacqua S, DeFronzo RA 1983 Effect of fatty acids on glucose production and utilization in man. J Clin Invest 72:1737–1747[Medline]
  102. Bevilacqua S, Bonadonna R, Buzzigoli G, Boni C, Ciociaro D, Maccari F, Giorico MA, Ferrannini E 1987 Acute elevation of free fatty acid levels leads to hepatic insulin resistance in obese subjects. Metabolism 36:502–506[Medline]
  103. Golay A, Swislocki ALM, Chen Y-DI, Reaven GM 1987 Relationships between plasma free fatty acid concentration, endogenous glucose production, and fasting hyperglycemia in normal and non-insulin-dependent diabetic individuals. Metabolism 36:692–696[Medline]
  104. Boden G, Chen X, Capulong E, Mozzoli M 2001 Effects of free fatty acids on gluconeogenesis and autoregulation of glucose production in type 2 diabetes. Diabetes 50:810–816[Abstract/Free Full Text]
  105. Groop LC, Saloranta C, Shank M, Bonadonna RC, Ferrannini E, DeFronzo RA 1991 The role of free fatty acid metabolism in the pathogenesis of insulin resistance in obesity and noninsulin-dependent diabetes mellitus. J Clin Endocrinol Metab 72:96–107[Abstract]
  106. Seppala-Lindroos A, Vehkavaara S, Hakkinen A-M, Goto T, Westerbacka J, Sovijarvi A, Halavaara J, Yki-Jarvinen H 2002 Fat accumulation in the liver is associated with defects in insulin suppression of glucose production and serum free fatty acids independent of obesity in normal men. J Clin Endocrinol Metab 87:3023–3028[Abstract/Free Full Text]
  107. Lam TKT, Carpentier A, Lewis GF, van de Werve G, Fantus IG, Giacca A 2003 Mechanisms of the free fatty acid-induced increase in hepatic glucose production. Am J Physiol 284:E863–E873
  108. Kim JK, Fillmore JJ, Chen Y, Yu C, Moore IK, Pypaert M, Lutz EP, Kako Y, Velez-Carrasco W, Goldberg IJ, Breslow JL, Shulman GI 2001 Tissue-specific overexpression of lipoprotein lipase causes tissue-specific insulin resistance. Proc Natl Acad Sci USA 98:7522–7527[Abstract/Free Full Text]
  109. Baron AD, Schaeffer L, Shragg P, Kolterman OG 1987 Role of hyperglucagonemia in maintenance of increased rates of hepatic glucose output in type II diabetics. Diabetes 36:274–283[Abstract]
  110. Matsuda M, DeFronzo RA, Consoli A, Bressler P, Del Prato S 2002 Dose response curve relating plasma glucagon to hepatic glucose production and glucose disposal in type 2 diabetes mellitus. Metabolism 51:1111–1119[CrossRef][Medline]
  111. Waldhausl W, Bratusch-Marrain P, Gasic S, Korn A, Nowotny P 1982 Insulin production rate, hepatic insulin retention, and splanchnic carbohydrate metabolism after oral glucose ingestion in hyperinsulinemic type II (non-insulin dependent) diabetes mellitus. Diabetologia 23:6–15[Medline]
  112. Fruhbeck G, Gomez-Ambrosi J, Muruzabal FJ, Burrell MA 2001 The adipocyte: a model for integration of endocrine and metabolic signaling in energy metabolism regulation. Am J Physiol 280:E827–E847
  113. Berg AH, Combs TP, Du X, Brownlee M, Scherer PE 2001 The adipocyte-secreted protein Acrp30 enhances hepatic insulin action. Nat Med 7:947–953[CrossRef][Medline]
  114. Combs TP, Berg AH, Obici S, Scherer PE, Rossetti L 2001 Endogenous glucose production is inhibited by the adipose-derived protein Acrp30. J Clin Invest 108:1875–1881[Abstract/Free Full Text]
  115. Yamauchi T, Kamon J, Waki H, Terauchi Y, Kubota N, Hara K, Mori Y, Ide T, Murakami K, Tsuboyama-Kasaoka N, Ezaki O, Akanuma Y, Gavrilova O, Vinson C, Reitman ML, Kagechika H, Shudo K, Yoda M, Nakano Y, Tobe K, Nagai R, Kimura S, Tomita M, Froguel P, Kadowaki T 2001 The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nat Med 7:941–946[CrossRef][Medline]
  116. Weyer C, Funahashi T, Tanaka S, Hotta K, Matsuzawa Y, Pratley RE, Tataranni PA 2001 Hypoadiponectinemia in obesity and type 2 diabetes: close association with insulin resistance and hyperinsulinemia. J Clin Endocrinol Metab 86:1930–1935[Abstract/Free Full Text]
  117. Hotta K, Funahashi T, Bodkin NL, Ortmeyer HK, Arita Y, Hansen BC, Matsuzawa Y 2001 Circulating concentrations of the adipocyte protein adiponectin are decreased in parallel with reduced insulin sensitivity during the progression to type 2 diabetes in rhesus monkeys. Diabetes 50:1126–1133[Abstract/Free Full Text]
  118. Arita Y, Kihara S, Ouchi N, Takahashi M, Maeda K, Miyagawa J, Hotta K, Shimomura I, Nakamura T, Miyaoka K, Kuriyama H, Nishida M, Yamashita S, Okubo K, Matsubara K, Muraguchi M, Ohmoto Y, Funahashi T, Matsuzawa Y 1999 Paradoxical decrease of an adipose-specific protein, adiponectin, in obesity. Biochem Biophys Res Commun 257:79–83[CrossRef][Medline]
  119. Hotta K, Funahashi T, Arita Y, Takahashi M, Matsuda M, Okamoto Y, Iwahashi H, Kuriyama H, Ouchi N, Maeda K, Nishida M, Kihara S, Sakai N, Nakajima T, Hasegawa K, Muraguchi M, Ohmoto Y, Nakamura T, Yamashita S, Hanafusa T, Matsuzawa Y 2000 Plasma concentrations of a novel, adipose-specific protein, adiponectin, in type 2 diabetic patients. Arterioscler Thromb Vasc Biol 20:1595–1599[Abstract/Free Full Text]
  120. Bajaj M, Suraamornkul S, Pratipanawatr T, Hardies LJ, Pratipanawatr W, Glass L, Miyazaki Y, DeFronzo RA 2003 Pioglitazone reduces hepatic fat content and augments splanchnic glucose uptake in patients with type 2 diabetes mellitus. Diabetes 52:1364–1370[Abstract/Free Full Text]
  121. Bajaj M, Suraamornkul S, Piper P, Hardies LJ, Glass L, Cersosimo E, Pratipanawatr, T, Miyazaki Y, DeFronzo RA 2004 Decreased plasma adiponectin concentrations are closely related to hepatic fat content and hepatic insulin resistance in pioglitazone-treated type 2 diabetic patients. J Clin Endocrinol Metab 89:200–206[Abstract/Free Full Text]
  122. Krakoff J, Funahashi T, Stehouwer CDA, Schalkwijk CG, Tanaka S, Matsuzawa Y, Kobes S, Tataranni PA, Hanson RL, Knowler WC, Lindsay RS 2003 Inflammatory markers, adiponectin, and risk of type 2 diabetes in the Pima Indian. Diabetes Care 26:1745–1751[Abstract/Free Full Text]
  123. Yang W-S, Jeng C-Y, Wu T-J, Tanaka S, Funahashi T, Matsuzawa Y, Wang J-P, Chen C-L, Tai T-Y, Chuang L-M 2002 Synthetic peroxisome proliferators-activated receptor-{gamma} agonist, rosiglitazone, increases plasma levels of adiponectin in type 2 diabetic patients. Diabetes Care 25:376–380[Abstract/Free Full Text]
  124. Scherer PE, Williams S, Fogliano M, Baldini G, Lodish HF 1995 A novel serum protein similar to C1q, produced exclusively in adipocytes. J Biol Chem 270:26746–26749[Abstract/Free Full Text]
  125. Hunter SJ, Garvey WT 1998 Insulin action and insulin resistance: diseases involving defects in insulin receptors, signal transduction, and the glucose transport effector system. Am J Med 105:331–345[CrossRef][Medline]
  126. Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee RR, Wright CM, Patel HR, Ahima RS, Lazar MA 2001 The hormone resistin links obesity to diabetes. Nature 409:307–312[CrossRef][Medline]
  127. Lazar MA 2002 Resistin and obesity-associated insulin resistance. Trends Endocrinol Metab 13:18–23[CrossRef][Medline]
  128. Savage DB, Sewter CP, Klenk ES, Segal DG, Vidal-Puig A, Considine RV, O’Rahilly S 2001 Resistin/Fizz3 expression in relation to obesity and peroxisome proliferator-activated receptor-{gamma} action in humans. Diabetes 50:2199–2202[Abstract/Free Full Text]
  129. Wang H, Chu WS, Hemphill C, Elbein SC 2002 Human resistin gene: molecular scanning and evaluation of association with insulin sensitivity and type 2 diabetes in Caucasians. J Clin Endocrinol Metab 87:2520–2524[Abstract/Free Full Text]
  130. McTernan PG, McTernan CL, Chetty R, Jenner K, Fisher FM, Lauer MN, Crocker J, Barnett AH, Kumar S 2002 Increased resistin gene and protein expression in human abdominal adipose tissue. J Clin Endocrinol Metab 87:2407[Abstract/Free Full Text]
  131. Vidal-Puig A, O’Rahilly S 2001 Resistin: a new link between obesity and insulin resistance? Clin Endocrinol (Oxf) 55:437–438[CrossRef][Medline]
  132. McTernan CL, McTernan PG, Harte AL, Levick PL, Barnett AH, Kumar S 2002 Resistin, central obesity, and type 2 diabetes. Lancet 359:46–47[CrossRef][Medline]
  133. McTernan PG, Fisher FM, McTernan CL, Chetty R, Valsamakis G, Harte AL, Lauer MN, Smith SA, Barnett AH, Kumar S 2002 Depot-specific expression of resistin in human adipose tissue and regulation of resistin secretion by insulin and rosiglitazone in human subcutaneous adipocytes in vitro. Diabetologia 45(Suppl 2):A66–A67
  134. Smith SR, Bai F, Charbonneau C, Janderova L, Argyropoulos G 2003 A promoter genotype and oxidative stress potentially link resistin to human insulin resistance. Diabetes 52:1611–1618[Abstract/Free Full Text]
  135. Rajala MW, Obici S, Scherer PE, Rossetti L 2003 Adipose-derived resistin and gut-derived resistin-like molecule-ß selectively impair insulin action on glucose production. J Clin Invest 111:225–230[Abstract/Free Full Text]
  136. Steppan CM, Brown EJ, Wright CM, Bhat S, Banerjee RR, Dai CY, Enders GH, Silberg DG, Wen X, Wu GD, Lazar MA 2001 A family of tissue-specific resistin-like molecules. Proc Natl Acad Sci USA 98:502–506[Abstract/Free Full Text]
  137. Haugen F, Jorgenson A, Drevon CA, Trayhurn P 2001 Inhibition by insulin of resistin gene expression in 3T3–L1 adipocytes. FEBS Lett 507:105–108[CrossRef][Medline]
  138. Hartman HB, Xu X, Tyler KX, Dalal CK, Lazar MA 2002 Mechanisms regulating adipocyte expression of resistin. J Biol Chem 277:19754–19761[Abstract/Free Full Text]
  139. Shojima N, Sakoda FL, Ogihara T, Fujishiro M, Katagiri H, Anai M, Onishi Y, Ono H, Inukai K, Abe M, Fukushima Y, Kikuchi M, Oka Y, Asano T 2002 Humoral regulation of resistin expression in 3T3–L1 and mouse adipose cells. Diabetes 51:1737–1744[Abstract/Free Full Text]
  140. Saye JA, Cassis LA, Sturgill TW, Lynch KR, Peach MJ 1989 Angiotensinogen gene expression in 3T3–L1 cells. Am J Physiol 256:C448–C451
  141. Karlsson C, Lindell K, Ottosson M, Sjostrom L, Carlsson B, Carlsson LM 1998 Human adipose tissue expresses angiotensinogen and enzymes required for its conversion to angiotensin II. J Clin Endocrinol Metab 83:3925–3929[Abstract/Free Full Text]
  142. Hainault I, Nebout G, Turban S, Ardouin B, Ferre P, Quignard-Boulange A 2002 Adipose tissue-specific increase in angiotensinogen expression and secretion in the obese (fa/fa) Zucker rat. Am J Physiol 282:E59–E66
  143. Hsueh WA, Law RE 1999 Insulin signaling in the arterial wall. Am J Cardiol 84:21J–24J[CrossRef]
  144. Goetze S, Xi XP, Graf K, Fleck E, Hsueh WA, Law RE 1999 Troglitazone inhibits angiotensin II-induced extracellular signal-regulated kinase 1/2 nuclear translocation and activation in vascular smooth muscle cells. FEBS Lett 452:277–282[CrossRef][Medline]
  145. Yudkin JS 1999 Abnormalities of coagulation and fibrinolysis in insulin resistance. Evidence for a common antecedent? Diabetes Care 22(Suppl 3):C25–C30
  146. Juhan-Vague I, Alessi MC 1997 PAI-I, obesity, insulin resistance and risk of cardiovascular events. Thromb Haemost 78:656–660[Medline]
  147. Alessi MC, Peiretti F, Morange P, Henry M, Nalbone G, Juhan-Vague I 1997 Production of plasminogen activator inhibitor 1 by human adipose tissue: possible link between visceral fat accumulation and vascular disease. Diabetes 46:860–867[Abstract]
  148. Alessi MC, Bastelica D, Morange P, Berthet B, Leduc I, Verdier M, Geel O, Juhan-Vague I 2000 Plasminogen activator inhibitor 1, transforming growth factor-ß1, and BMI are closely associated in human adipose tissue during morbid obesity. Diabetes 49:1374–1380[Abstract]
  149. Birgel M, Gottschling-Zeller H, Rohrig K, Hauner H 2000 Role of cytokines in the regulation of plasminogen activator inhibitor-1 expression and secretion in newly differentiated subcutaneous human adipocytes. Arterioscler Thromb Vasc Biol 20:1682–1687[Abstract/Free Full Text]
  150. McCall JL, Tuckey JA, Parry BR 1992 Serum tumour necrosis factor {alpha} and insulin resistance in gastrointestinal cancer. Br J Surg 79:1361–1363[Medline]
  151. Van der Poll T, Romijn JA, Endert E, Borm JJ, Buller HR, Sauerwein HP 1991 Tumor necrosis factor mimics the metabolic response to acute infection in healthy humans. Am J Physiol 261:E457–E465
  152. Ryden M, Dicker A, van Harmelen V, Hauner H, Brunnberg M, Perbeck L, Lonnqvist F, Arner P 2002 Mapping of early signaling events in tumor necrosis factor-{alpha}-mediated lipolysis in human fat cells. J Biol Chem 277:1085–1091[Abstract/Free Full Text]
  153. Hotamisligil GS, Shargill NS, Spiegelman BM 1993 Adipose expression of tumor necrosis factor-{alpha}: direct role in obesity-linked insulin resistance. Science 259:87–91[Medline]
  154. Feinstein R, Kanety H, Papa MZ, Lunenfeld B, Karasik A 1993 Tumor necrosis factor-{alpha} suppresses insulin-induced tyrosine phosphorylation of insulin receptor and its substrates. J Biol Chem 268:26055–26058[Abstract/Free Full Text]
  155. Hotamisligil GS, Peraldi P, Budavari A, Ellis R, White MF, Spiegelman BM 1996 IRS-1-mediated inhibition of insulin receptor tyrosine kinase activity in TNF-{alpha}- and obesity-induced insulin resistance. Science 271:665–668[Abstract]
  156. Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM 1995 Increased adipose tissue expression of tumor necrosis factor-{alpha} in human obesity and insulin resistance. J Clin Invest 95:2409–2415[Medline]
  157. Miyazaki Y, Pipek R, Mandarino LJ, DeFronzo RA 2003 Tumor necrosis factor {alpha} and insulin resistance in obese type 2 diabetic patients. Int J Obesity 27:88–94[CrossRef]
  158. Nilsson J, Jovinge S, Niemann A, Reneland R, Lithell H 1998 Relation between plasma tumor necrosis factor-{alpha} and insulin sensitivity in elderly men with non-insulin-dependent diabetes mellitus. Arterioscler Thromb Vasc Biol 18:1199–1202[Abstract/Free Full Text]
  159. Zinman B, Hanley AJ, Harris SB, Kwan J, Fantus IG 1999 Circulating tumor necrosis factor-{alpha} concentrations in a native Canadian population with high rates of type 2 diabetes mellitus. J Clin Endocrinol Metab 84:272–278[Abstract/Free Full Text]
  160. Spiegelman BM 1998 PPAR-{gamma}: adipogenic regulator and thiazolidinedione receptor. Diabetes 47:507–514[Abstract]
  161. Mohamed-Ali V, Goodrick S, Rawesh A, Katz DR, Miles JM, Yudkin JS, Klein S, Coppack SW 1997 Subcutaneous adipose tissue releases interleukin-6, but not tumor necrosis factor-{alpha}, in vivo. J Clin Endocrinol Metab 82:4196–200[Abstract/Free Full Text]
  162. Van Snick J 1990 Interleukin 6: an over-view. Annu Rev Immunol 8:253–278[CrossRef][Medline]
  163. Sandler S, Bendtzen K, Eizirik DL, Welsh M 1990 Interleukin-6 affects insulin secretion and glucose metabolism of rat pancreatic islets in vitro. Endocrinology 126:1288–1294[Abstract]
  164. Pradhan AD, Manson JE, Rifai N, Buring JE, Ridker PM 2001 C-Reactive protein, interleukin 6, and risk of developing type 2 diabetes mellitus. JAMA 286:327–334[Abstract/Free Full Text]
  165. Pickup JC, Chusney GD, Thomas SM, Burt D 2000 Plasma interleukin-6, tumour necrosis factor and blood cytokine production in type 2 diabetes. Life Sci 67:291–300[CrossRef][Medline]
  166. Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM 1994 Positional cloning of the mouse obese gene and its human homologue. Nature 372:425–432[CrossRef][Medline]
  167. Halaas JL, Gajiwala KS, Maffei M, Cohen SL, Chait BT, Rabinowitz D, Lallone RL, Burley SK, Friedman JM 1995 Weight-reducing effects of the plasma protein encoded by the obese gene. Science 269:543–546[Medline]
  168. Zierath JR, Frevert EU, Ryder JW, Berggren PO, Kahn BB 1998 Evidence against a direct effect of leptin on glucose transport in skeletal muscle and adipocytes. Diabetes 47:1–4[Abstract]
  169. Montague CT, Prins JB, Sanders L, Zhang J, Sewter CP, Digby J, Byrne CD, O’Rahilly S 1998 Depot-related gene expression in human subcutaneous and omental adipocytes. Diabetes 47:1384–1391[Abstract]
  170. Ceddia RB, William Jr WN, Curi R 1999 Comparing effects of leptin and insulin on glucose metabolism in skeletal muscle: evidence for an effect of leptin on glucose uptake and decarboxylation. Int J Obesity Related Metab Disord 23:75–82[CrossRef][Medline]
  171. Kamohara S, Burcelin R, Halaas JL, Friedman JM, Charron MJ 1997 Acute stimulation of glucose metabolism in mice by leptin treatment. Nature 389:374–377[CrossRef][Medline]
  172. Muoio DM, Dohm GL 2002 Peripheral metabolic actions of leptin. Best Practice Res Clin Endocrinol Metab 16:653–666[CrossRef][Medline]
  173. Ranganathan S, Ciaraldi TP, Henry RR, Mudaliar S, Kern PA 1998 Lack of effect of leptin on glucose transport, lipoprotein lipase, and insulin action in adipose and muscle cells. Endocrinology 139:2509–2513[Abstract/Free Full Text]
  174. Mantzoros CS, Flier JS 2000 Editorial: leptin as a therapeutic agent–trials and tribulations. J Clin Endocrinol Metab 85:4000–4002[Free Full Text]
  175. Clement K, Vaisse C, Lahlou N, Cabrol S, Pelloux V, Cassuto D, Gourmelen M, Dina C, Chambaz J, Lacorte JM, Basdevant A, Bougneres P, Lebouc Y, Froguel P, Guy-Grand B 1998 A mutation in the human leptin receptor gene causes obesity and pituitary dysfunction. Nature 392:398–401[CrossRef][Medline]
  176. Farooqi IS, Jebb SA, Langmack G, Lawrence E, Cheetham CH, Prentice AM, Hughes IA, McCamish MA, O’Rahilly S 1999 Effects of recombinant leptin therapy in a child with congenital leptin deficiency. N Engl J Med 341:879–884[Free Full Text]
  177. Gainsford T, Willson TA, Metcalf D, Handman E, McFarlane C, Ng A, Nicola NA, Alexander WS, Hilton DJ 1996 Leptin can induce proliferation, differentiation, and functional activation of hemopoietic cells. Proc Natl Acad Sci USA 93:14564–14568[Abstract/Free Full Text]
  178. Fernandes G, Handwerger BS, Yunis EJ, Brown DM 1978 Immune response in the mutant diabetic C57BL/Ks-dt+ mouse. Discrepancies between in vitro and in vivo immunological assays. J Clin Invest 61:243–250[Medline]
  179. Chandra RK 1980 Cell-mediated immunity in genetically obese C57BL/6J ob/ob mice. Am J Clin Nutr 33:13–16[Abstract]
  180. Pickup JC, Crook MA 1998 Is type II diabetes mellitus a disease of the innate immune system? Diabetologia 41:1241–1248[CrossRef][Medline]
  181. Choy LN, Rosen BS, Spiegelman BM 1992 Adipsin and an endogenous pathway of complement from adipose cells. J Biol Chem 267:12736–12741[Abstract/Free Full Text]
  182. Cianflone K, Maslowska M, Sniderman AD 1999 Acylation stimulating protein (ASP), an adipocyte autocrine: new directions. Semin Cell Dev Biol 10:31–41[CrossRef][Medline]
  183. Maslowska M, Vu H, Phelis S, Sniderman AD, Rhode BM, Blank D, Cianflone K 1999 Plasma acylation stimulating protein, adipsin and lipids in non-obese and obese populations. Eur J Clin Invest 29:679–686[CrossRef][Medline]
  184. Festa A, D’Agostino Jr R, Howard G, Mykkanen L, Tracy RP, Haffner SM 2000 Chronic subclinical inflammation as part of the insulin resistance syndrome: the Insulin Resistance Atherosclerosis Study (IRAS). Circulation 102:42–47[Abstract/Free Full Text]
  185. Blake GJ, Ridker PM 2001 High sensitivity C-reactive protein for predicting cardiovascular disease: an inflammatory hypothesis. Eur Heart J 22:349–352[Free Full Text]
  186. DeFronzo RA, Ferrannini E 1991 Insulin resistance: a multifaceted syndrome responsible for NIDDM, obesity, hypertension, dyslipidemia, and ASCVD. Diabetes Care Rev 14:173–194
  187. Reaven GM 1988 Banting lecture 1988. Role of insulin resistance in human disease. Diabetes 37:1595–1607[Abstract]
  188. Loffreda S, Yang SQ, Lin HZ, Karp CL, Brengman ML, Wang DJ, Klein AS, Bulkley GB, Bao C, Noble PW, Lane MD, Diehl AM 1998 Leptin regulates proinflammatory immune responses. FASEB J 12:57–65[Abstract/Free Full Text]
  189. Luster AD 1998 Chemokines: chemotactic cytokines that mediate inflammation. N Engl J Med 338:436–445[Free Full Text]
  190. Gomez-Ambrosi J, Fruhbeck G 2001 Do resistin and resistin-like molecules also link obesity to inflammatory diseases? Ann Intern Med 135:306–307[Free Full Text]
  191. Yokota T, Oritani K, Takahashi I, Ishikawa J, Matsuyama A, Ouchi N, Kihara S, Funahashi T, Tenner AJ, Tomiyama Y, Matsuzawa Y 2000 Adiponectin, a new member of the family of soluble defense collagens, negatively regulates the growth of myelomonocytic progenitors and the functions of macrophages. Blood 96:1723–1732[Abstract/Free Full Text]
  192. Collins T, Cybulsky MI 2001 NF-{kappa}B: pivotal mediator or innocent bystander in atherogenesis? J Clin Invest 107:255–264[Free Full Text]
  193. Kim JK, Kim YJ, Fillmore JJ, Chen Y, Moore I, Lee J, Yuan M, Li ZW, Karin M, Perret P, Shoelson SE, Shulman GI 2001 Prevention of fat-induced insulin resistance by salicylate. J Clin Invest 108:437–446[Abstract/Free Full Text]
  194. Yuan M, Konstantopoulos N, Lee J, Hansen L, Li ZW, Karin M, Shoelson SE 2001 Reversal of obesity- and diet-induced insulin resistance with salicylates or targeted disruption of I{kappa}{kappa}ß. Science 293:1673–1677[Abstract/Free Full Text]
  195. Stevenson MA, Zhao MJ, Asea A, Coleman CN, Calderwood SK 1999 Salicylic acid and aspirin inhibit the activity of RSK2 kinase and repress RSK2-dependent transcription of cyclic AMP response element binding protein- and NF-{kappa}B-responsive genes. J Immunol 163:5608–5616[Abstract/Free Full Text]
  196. Osman A, Pendergrass M, Cusi K, Christ CY, Adams JM, DeFronzo RA, Mandarino LJ RKS2 (p90 rks) activity is increased in skeletal muscle in type 2 diabetes mellitus. Biochem Biophys Acta, in press
  197. Mora S, Pessin JE 2002 An adipocentric view of signaling and intracellular trafficking. Diabetes Metab Res Rev 18:345–356[CrossRef][Medline]
  198. Harris MI, Flegal KM, Cowie CC, Eberhardt MS, Goldstein DE, Little RR, Wiedmeyer HM, Byrd-Holt DD 1998 Prevalence of diabetes, impaired fasting glucose, and impaired glucose tolerance in U.S. adults. The Third National Health and Nutrition Examination Survey, 1988–1994. Diabetes Care 21:518–524[Abstract]
  199. Pi-Sunyer F 1996 Weight and non-insulin-dependent diabetes mellitus. Am J Clin Nutr 63(Suppl 3):426S–429S
  200. Knowler WC, Pettitt DJ, Savage PJ, Bennett PH 1981 Diabetes incidence in Pima Indians: contributions of obesity and parental diabetes. Am J Epidemiol 113:144–156[Abstract]
  201. Carey V, Walters E, Colditz G, Solomon C, Willett W, Rosner B, Speizer F, Manson J 1997 Body fat distribution and risk of non-insulin-dependent diabetes mellitus in women: the Nurses’ Health Study. Am J Epidemiol 45:614–619
  202. Wannamethee SG, Shaper AG 1999 Weight change and duration of overweight and obesity in the incidence of type 2 diabetes. Diabetes Care 22:1266–1272[Abstract]
  203. Perry IJ, Wannamethee SG, Shaper AG, Alberti KG 1999 Serum true insulin concentration and the risk of clinical non-insulin dependent diabetes during long-term follow-up. Int J Epidemiol 28:735–741[Abstract]
  204. Burke JP, Williams K, Haffner SM, Villalpando CG, Stern MP 2001 Elevated incidence of type 2 diabetes in San Antonio, Texas, compared with that of Mexico City, Mexico. Diabetes Care 24:1573–158[Abstract/Free Full Text]
  205. DeFronzo RA, Sherwin RS, Hendler R, Felig P 1978 Insulin binding to monocytes and insulin action in human obesity, starvation, and refeeding. J Clin Invest 62:204–213[Medline]
  206. Kolterman OG, Insel J, Saekow M, Olfesky JM 1980 Mechanisms of insulin resistance in human obesity: evidence for receptor and postreceptor defects. J Clin Invest 65:1272–1284[Medline]
  207. Ferrannini E, Natali A, Bell P, Cavallo-Perin P, Lalic N, Mingrone G, for the European Group for the Study of Insulin Resistance (EGIR) 1997 Insulin resistance and hypersecretion in obesity. J Clin Invest 100:1166–1173[Abstract/Free Full Text]
  208. Bjorntorp P 1991 Metabolic implications of body fat distribution. Diabetes Care 14:1132–1143[Abstract]
  209. Chan JM, Rimm EB, Colditz GA, Stapfer MJ, Willett WC 1994 Obesity, fat distribution, and weight gain as risk factors for clinical diabetes in men. Diabetes Care 17:961–969[Abstract]
  210. Rabinowitz D, Zierler KL 1962 Forearm metabolism in obesity and its response to intraarterial insulin. Characterization of insulin resistance and evidence for adaptive hyperinsulinism. J Clin Invest 41:2173–2181
  211. Gastaldelli A, Baldi S, Pettiti M, Toschi E, Camastra S, Natali A, Landau BR, Ferrannini E 2000 Influence of obesity and type 2 diabetes on gluconeogenesis and glucose output in humans: a quantitative study. Diabetes 49:1367–1373[Abstract]
  212. Kashiwagi A, Verso MA, Andrews J, Vasquez B, Reavan GM, Foley JE 1983 In vitro insulin resistance of human adipocytes isolated from subjects with noninsulin-dependent diabetes mellitus. J Clin Invest 72:1246–1254[Medline]
  213. Groop L, Bonadonna R, Simonson DC, Petrides A, Hasan S, DeFronzo RA 1992 Effect of insulin on oxidative and non-oxidative pathways of glucose and free fatty acid metabolism in human obesity. Am J Physiol 263:E79–E84
  214. Clausen JO, Borch-Johnsen K, Ibsen H, Bergman RN, Hougaard P, Winther K, Pedersen O 1996 Insulin sensitivity index, acute insulin response, and glucose effectiveness in a population-based sample of 380 young healthy Caucasians. Analysis of the impact of gender, body fat, physical fitness, and life-style factors. J Clin Invest 98:1195–1209[Abstract/Free Full Text]
  215. Kissebah AH, Peiris AN 1989 Biology of regional body fat distribution: relationship to non-insulin-dependent diabetes mellitus. Diabetes Metab Rev 5:83–109[Medline]
  216. Despres JP, Lemieux S, Lamarche B, Prud’homme D, Moorjani S, Brun LD, Gagne C, Lupien PJ 1995 The insulin resistance-dyslipidemic syndrome: contribution of visceral obesity and therapeutic implications. Int J Obes Related Metab Disord 19(Suppl 1):S76–S86
  217. Banerji MA, Chaiken RL, Gordon D, Kral JG, Lebovitz HE 1995 Does intra-abdominal adipose tissue in black men determine whether NIDDM is insulin-resistant or insulin-sensitive? Diabetes 44:141–146[Abstract]
  218. Bonora E, Del Prato S, Bonadonna RC, Gulli G, Solini A, Shank ML, Ghiatas AA, Lancaster JL, Kilcoyne RF, Alyassin AM, DeFronzo RA 1992 Total body fat content and fat topography are associated differently with in vivo glucose metabolism in nonobese and obese nondiabetic women. Diabetes 41:1151–1159[Abstract]
  219. Albu JB, Kovera AJ, Johnson JA 2000 Fat distribution and health in obesity. Ann NY Acad Sci 904:491–501[Abstract/Free Full Text]
  220. Arner P 1997 Regional adipocity in man. J Endocrinol 155:191–192[Free Full Text]
  221. Zierath JR, Livingston JN, Thorne A, Bolinder J, Reynisdottir S, Lonnqvist F, Arner P 1998 Regional difference in insulin inhibition of non-esterified fatty acid release from human adipocytes: relation to insulin receptor phosphorylation and intracellular signaling through the insulin receptor substrate-1-pathway. Diabetologia 41:1343–1354[CrossRef][Medline]
  222. Bjorntorp P 1990 "Portal" adipose tissue as a generator of risk factors for cardiovascular disease and diabetes. Arteriosclerosis 10:493–496[Medline]
  223. Arner P 2001 Regional differences in protein production by human adipose tissue. Biochem Soc Trans 29:72–75[CrossRef][Medline]
  224. Motoshima H, Wu X, Sinha MK, Hardy VE, Rosato EL, Barbot DJ, Rosato FE, Goldstein BJ 2002 Differential regulation of adiponectin secretion from cultured human omental and subcutaneous adipocytes: effects of insulin and rosiglitazone. J Clin Endocrinol Metab 87:5662–5667[Abstract/Free Full Text]
  225. Sewter CP, Blows F, Vidal-Puig A, O’Rahilly S 2002 Regional differences in the response of human pre-adipocytes to PPAR{gamma} and RXR{alpha} agonists. Diabetes 51:7218–723
  226. Pan DA, Lillioja S, Kriketos AD, Milner MR, Baur LA, Bogardus C, Jenkins AB, Storlien LH 1997 Skeletal muscle triglyceride levels are inversely related to insulin action. Diabetes 46:983–988[Abstract]
  227. Szczepaniak LS, Babcock EE, Schick F, Dobbins RL, Garg A, Burns DK, McGarry JD, Stein DT 1999 Measurement of intracellular triglyceride stores by H spectroscopy: validation in vivo. Am J Physiol 276:E977–E989
  228. Perseghin G, Scifo P, De Cobelli F, Pagliato E, Battezzati A, Arcelloni C, Vanzulli A, Testolin G, Pozza G, Del Maschio A, Luzi L 1999 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 48:1600–1606[Abstract]
  229. Krssak M, Falk Petersen K, Dresner A, DiPietro L, Vogel SM, Rothman DL, Roden M, Shulman GI 1999 Intramyocellular lipid concentrations are correlated with insulin sensitivity in humans: a 1H NMR spectroscopy study. Diabetologia 42:113–116[CrossRef][Medline]
  230. Stein DT, Szczepanik L, Dobbins R, McGarry JD 1999 Increasing intramyocellular triglyceride stores are associated with impaired glucose tolerance and NIDDM. Diabetes 48(Suppl 1):A287
  231. Goodpaster BH, Thaete FL, Simoneau JA, Kelley DE 1997 Subcutaneous abdominal fat and thigh muscle composition predict insulin sensitivity independently of visceral fat. Diabetes 46:1579–1585[Abstract]
  232. Goodpaster BH, Kelley DE, Wing RR, Meier A, Thaete FL 1999 Effects of weight loss on regional fat distribution and insulin sensitivity in obesity. Diabetes 48:839–847[Abstract]
  233. Kelley DE, Goodpaster B, Wing RR, Simoneau JA 1999 Skeletal muscle fatty acid metabolism in association with insulin resistance, obesity, and weight loss. Am J Physiol 277:E1130–E1141
  234. Greco AV, Mingrone G, Giancaterini A, Manco M, Morroni M, Cinti S, Granzotto M, Vettor R, Camastra S, Ferrannini E 2002 Insulin resistance in morbid obesity: reversal with intramyocellular fat depletion. Diabetes 51:144–151[Abstract/Free Full Text]
  235. Ryysy L, Hakkinen AM, Goto T, Vehkavaara S, Westerbacka J, Halavaara J, Yki-Jarvinen H 2000 Hepatic fat content and insulin action on free fatty acids and glucose metabolism rather than insulin absorption are associated with insulin requirements during insulin therapy in type 2 diabetic patients. Diabetes 49:749–758[Abstract]
  236. Robbins DC, Horton ES, Tulp O, Sims EA 1982 Familial partial lipodystrophy: complications of obesity in the non-obese? Metabolism 31:445–452[Medline]
  237. Arioglu E, Duncan-Morin D, Sebring N, Rother KI, Gottlieb N, Lieberman J, Herion D, Kleiner DE, Reynolds J, Premkumar A, Sumner AE, Hoofnagle J, Reitman ML, Taylor SI 2000 Efficacy and safety of troglitazone in the treatment of lipodystrophy syndromes. Ann Intern Med 133:263–274[Abstract/Free Full Text]
  238. Tansey JT, Sztalryd C, Gruia-Gray J, Roush DL, Zee JV, Gavrilova O, Reitman ML, Deng CX, Li C, Kimmel AR, Londos C 2001 Perilipin ablation results in a lean mouse with aberrant adipocyte lipolysis, enhanced leptin production, and resistance to diet-induced obesity. Proc Natl Acad Sci USA 98:6494–6499[Abstract/Free Full Text]
  239. Petersen KF, Oral EA, Dufour S, Befroy D, Ariyan C, Yu C, Cline GW, DePaoli AM, Taylor SI, Gorden P, Shulman GI 2002 Leptin reverses insulin resistance and hepatic steatosis in patients with severe lipodystrophy. J Clin Invest 109:1345–1350[Abstract/Free Full Text]
  240. Oral EA, Simha V, Ruiz E, Andewelt A, Premkumar A, Snell P, Wagner AJ, DePaoli AM, Reitman ML, Taylor SI, Gorden P, Garg A 2002 Leptin-replacement therapy for lipodystrophy. N Engl J Med 346:570–578[Abstract/Free Full Text]
  241. Reitman ML, Mason MM, Moitra J, Gavrilova O, Marcus-Samuels B, Eckhaus M, Vinson C 1999 Transgenic mice lacking white fat: models for understanding human lipoatrophic diabetes. Ann NY Acad Sci 892:289–296[Abstract/Free Full Text]
  242. Shimomura I, Hammer RE, Ikemoto S, Brown MS, Goldstein JL 1999 Leptin reverses insulin resistance and diabetes mellitus in mice with congenital lipodystrophy. Nature 401:73–76[CrossRef][Medline]
  243. Kim JK, Gavrilova O, Chen Y, Reitman ML, Shulman GI 2000 Mechanism of insulin resistance in A-ZIP/F-1 fatless mice. J Biol Chem 275:8456–8460[Abstract/Free Full Text]
  244. Reue K, Xu P, Wang XP, Slavin BG 2000 Adipose tissue deficiency, glucose intolerance, and increased atherosclerosis result from mutation in the mouse fatty liver dystrophy (fld) gene. J Lipid Res 41:1067–1076[Abstract/Free Full Text]
  245. Peterfy M, Phan J, Xu P, Reue K 2001 Lipodystrophy in the fld mouse results from mutation of a new gene encoding a nuclear protein, lipin. Nat Genet 27:121–124[CrossRef][Medline]
  246. Gavrilova O, Marcus-Samuels B, Graham D, Kim JK, Shulman GI, Castle AL, Vinson C, Eckhaus M, Reitman ML 2000 Surgical implantation of adipose tissue reverses diabetes in lipoatrophic mice. J Clin Invest 105:271–278[Abstract/Free Full Text]
  247. Weber RV, Buckley MC, Fried SK, Kral JG 2000 Subcutaneous lipectomy causes a metabolic syndrome in hamsters. Am J Physiol 279:R936–R943
  248. Danforth Jr E 2000 Failure of adipocyte differentiation causes type II diabetes mellitus? Nat Genet 26:13
  249. Bjorntorp P, Berchtold P, Tibblin G 1971 Insulin secretion in relation to adipose tissue in men. Diabetes 20:65–70[Medline]
  250. Schneider BS, Faust IM, Hemmes R, Hirsch J 1981 Effects of altered adipose tissue morphology on plasma insulin levels in the rat. Am J Physiol 240:E358–E362
  251. Salans LB, Knittle JL, Hirsch J 1968 Role of adipose cell size and adipose tissue insulin sensitivity in the carbohydrate intolerance of human obesity. J Clin Invest 47:153–165
  252. Weyer C, Wolford JK, Hanson RL, Foley JE, Tataranni PA, Bogardus C, Pratley RE 2001 Subcutaneous abdominal adipocyte size, a predictor of type 2 diabetes, is linked to chromosome 1q21–q23 and is associated with a common polymorphism in LMNA in Pima Indians. Mol Genet Metab 72:231–238[CrossRef][Medline]
  253. Weyer C, Foley JE, Bogardus C, Tataranni PA, Pratley RE 2000 Enlarged subcutaneous abdominal adipocyte size, but not obesity itself, predicts type II diabetes independent of insulin resistance. Diabetologia 43:1498–1506[CrossRef][Medline]
  254. Kahsyap S, Suraamornkul S, Bajaj M, Belfort R, Berria R, Mandarino L, DeFronzo R, Cusi K 2003 Effect of sustained reduction in plasma FFA concentration on insulin secretion in non-diabetic subjects with a strong family history of T2DM. Diabetes 52(Suppl 1):A336
  255. McGarry JD 1992 What if Minkowski had been ageusic? An alternative angle on diabetes. Sicence 258:766–770
  256. Mudaliar S, Henry RR 2001 New oral therapies for type 2 diabetes mellitus: the glitazones or insulin sensitizers. Annu Rev Med 52:239–257[CrossRef][Medline]
  257. Fonseca VA, Valiquett TR, Huang SM, Ghazzi MN, Whitcomb RW 1998 Troglitazone monotherapy improves glycemic control in patients with type 2 diabetes mellitus: a randomized, controlled study. The Troglitazone Study Group. J Clin Endocrinol Metab 83:3169–3176[Abstract/Free Full Text]
  258. Saltiel AR, Olefsky JM 1996 Thiazolidinediiones in the treatment of insulin resistance in type 2 diabetes. Diabetes 45:1661–1669[Abstract]
  259. Aronoff S, Rosenblatt S, Braithwaite S, Egan JW, Mathisen AL, Schneider RL 1996 Pioglitazone hydrochloride monotherapy improves glycemic control in the treatment of patients with type 2 diabetes: a 6-month randomized placebo-controlled dose-response study. The Pioglitazone 001 Study Group. Diabetes Care 23:1605–1611
  260. Wolffenbuttel BH, Sels JP, Huijberts MS 2001 Rosiglitazone. Expert Opin Pharmacother 2:467–478[Medline]
  261. Miyazaki Y, Mahankali A, Matsuda M, Glass L, Mahankali S, Ferranini E, Cusi K, Mandarino L, DeFronzo RA 2001 Improved glycemic control and enhanced insulin sensitivity in liver and muscle in type 2 diabetic subjects treated with pioglitazone. Diabetes Care 24:710–719[Abstract/Free Full Text]
  262. Miyazaki Y, Mahankali A, Matsuda M, Mahankali S, Hardies J, Cusi K, Mandarino LJ, DeFronzo RA 2002 Effect of pioglitazone on abdominal fat distribution and insulin sensitivity in type 2 diabetic patients. J Clin Endocrinol Metab 87:2784–2791[Abstract/Free Full Text]
  263. Miyazaki Y, Glass L, Triplitt C, Matsuda M, Cusi K, Mandarino L, DeFronzo RA 2001 Effect of rosiglitazone on glucose and free fatty acid metabolism in type 2 diabetic patients. Diabetologia 44:2210–2219[CrossRef][Medline]
  264. Inzucchi SE, Maggs DG, Spollett GR, Page SL, Rife FS, Walton V, Shulman GI 1998 Efficacy and metabolic effects of metformin and troglitazone in type II diabetes mellitus. N Engl J Med 338:867–872[Abstract/Free Full Text]
  265. Maggs DG, Buchanan TA, Burant CF, Cline G, Gumbiner B, Hsueh WA, Inzucchi S, Kelley D, Nolan J, Olefsky JM, Polonsky KS, Silver D, Valiquett TR, Shulman GI 1998 Metabolic effects of troglitazone monotherapy in type 2 diabetes mellitus. A randomized, double-blind, placebo-controlled trial. Ann Intern Med 128:176–185[Abstract/Free Full Text]
  266. Olfesky JM 2000 Treatment of insulin resistance with peroxisome proliferator-activated receptor {gamma} agonists. J Clin Invest 106:467–427[Free Full Text]
  267. Kim YB, Ciaraldi TP, Kong A, Kim D, Chu N, Mohideen P, Mudaliar S, Henry RR, Kahn BB 2002 Troglitazone but not metformin restores insulin-stimulated phosphoinositide 3-kinase activity and increases p 110b protein levels in skeletal muscle of type 2 diabetic subjects. Diabetes 51:443–448[Abstract/Free Full Text]
  268. Mayerson AB, Hundal RS, Dufour S, Lebon V, Befroy D, Cline GW, Enocksson S, Inzucchi SE, Shulman GI, Petersen KF 2002 The effects of rosiglitazone on insulin sensitivity, lipolysis, and hepatic and skeletal muscle triglyceride content in patients with type 2 diabetes. Diabetes 51:797–802[Abstract/Free Full Text]
  269. Miyazaki Y, Matsuda M, DeFronzo RA 2002 Dose-response effect of pioglitazone on insulin sensitivity and insulin secretion in type 2 diabetes mellitus. Diabetes Care 25:517–523[Abstract/Free Full Text]
  270. Berkowitz K, Peters R, Kjos SL, Goico J, Marroquin A, Dunn ME, Xiang A, Azen S, Buchanan TA 1996 Effect of troglitazone on insulin sensitivity and pancreatic ß-cell function in women at high risk for NIDDM. Diabetes 45:1572–1579[Abstract]
  271. Finegood DT, McArthur MD, Kojwang D, Thomas MJ, Topp BG, Leonard T, Buckingham RE 2001 ß-Cell mass dynamics in Zucker diabetic fatty rats. Rosiglitazone prevents the rise in net cell death. Diabetes 50:1021–1029[Abstract/Free Full Text]
  272. Higa M, Zhou YT, Ravazzola M, Baetens D, Orci L, Unger RH 1999 Troglitazone prevents mitochondrial alterations, ß cell destruction, and diabetes in obese prediabetic rats. Proc Natl Acad Sci USA 96:11513–11518[Abstract/Free Full Text]
  273. Iwamoto Y, Kosaka K, Kuzuya T, Akanuma Y, Shigeta Y, Kaneko T 1996 Effects of troglitazone: a new hypoglycemic agent in patients with NIDDM poorly controlled by diet therapy. Diabetes Care 19:151–156[Abstract]
  274. Horton ES, Whitehouse F, Ghazzi MN, Venable TC, Troglitazone Study Group, Whitecomb RW 1998 Troglitazone in combination with sulfonylurea restores glycemic control in patients with type 2 diabetes. Diabetes Care 21:1462–1469[Abstract]
  275. Mori Y, Murakawa Y, Okada K, Horikoshi H, Yokoyama J, Yajima N, Ikeda Y 1999 Effects of troglitazone on body fat distribution in type 2 diabetic patients. Diabetes Care 22:908–912[Abstract]
  276. Fonseca V, Foyt HL, Shen K, Whitcomb R 2000 Long-term effects of troglitazone: open-label extension studies in type 2 diabetic patients. Diabetes Care 23:354–359[Abstract]
  277. Patel J, Anderson RJ, Rappaport EB 1999 Rosiglitazone monotherapy improve glycaemic control in patients with type 2 diabetes: a twelve-week, randomized, placebo-controlled study. Diabetes Obesity Metab 1:165–172[CrossRef]
  278. Lebovitz HE, Dole JF, Patwardhan R, Rappaport EB, Freed MI 2001 The Rosiglitazone Clinical Trials Study Group. Rosiglitazone monotherapy is effective in patients with type 2 diabetes. J Clin Endocrinol Metab 86:280–288[Abstract/Free Full Text]
  279. Chilcott J, Tappenden P, Jones ML, Wight JP 2001 A systematic review of the clinical effectiveness of pioglitazone in the treatment of type 2 diabetes mellitus. Clin Ther 23:1792–1823[CrossRef][Medline]
  280. Sims EA, Donforth Jr E, Horton ES, Bray GA, Glennon JA, Salans LB 1973 Endocrine and metabolic effects of experimental obesity in man. Recent Prog Horm Res 29:457–496[Medline]
  281. Henry RR, Wallace P, Olefsky JM 1986 Effects of weight loss on mechanisms of hyperglycemia in obese non-insulin-dependent diabetes mellitus. Diabetes 35:990–998[Abstract]
  282. Chilcott J, Tappenden P, Jones ML, Wight JP 2001 A systematic review of the effectiveness of pioglitazone in the treatment of type 2 diabetes mellitus. Clin Ther 23:1792–1823[CrossRef][Medline]
  283. Willson TM, Brown PJ, Sternbach DD, Henke BR 2000 The PPARs: from orphan receptors to drug discovery. J Med Chem 43:527–550[CrossRef][Medline]
  284. Berger J, Moller DE 2002 The mechanisms of action of PPARs. Annu Rev Med 53:409–435[CrossRef][Medline]
  285. Kahn CR, Chen L, Cohen SE 2000 Unraveling the mechanism of action of thiazolidinediones. J Clin Invest 106:1305–1307[Free Full Text]
  286. Willson TM, Cobb JE, Cowan DJ, Wiethe RW, Correa ID, Prakash SR, Beck KD, Moore LB, Kliewer SA, Lehmann JM 1996 The structure-activity relationship between peroxisome proliferator-activated receptor {gamma} agonism and the antihyperglycemic activity of thiazolidinediones. J Med Chem 39:665–668[CrossRef][Medline]
  287. Vidal-Puig AJ, Considine RV, Jimenez-Linan M, Werman A, Pories WJ, Caro JF, Flier JS 1997 Peroxisome proliferator-activated receptor gene expression in human tissues: effects of obesity, weight loss and regulation by insulin and glucocorticoids. J Clin Invest 99:2416–2422[Abstract/Free Full Text]
  288. Fajas L, Auboeuf D, Raspe E, Schoonjans K, Lefebvre AM, Saladin R, Najib J, laville M, Fruchart JC, Deeb S, Vidal-Puig A, Flier J, Briggs MR, Staels B, Vidal H, Auwerx J 1997 The organization promoter analysis, and expression of the human PPAR{gamma} gene. J Biol Chem 272:18779–18789[Abstract/Free Full Text]
  289. Mukherjee R, Jow L, Croston GE, Paterniti Jr JR 1997 Identification, characterization, and tissue distribution of human peroxisome proliferator-activated receptor (PPAR) isoforms PPAR{gamma}2 versus PPAR{gamma}1 and activation with retinoid X receptor agonists and antagonists. J Biol Chem 272:8071–8076[Abstract/Free Full Text]
  290. Xu HE, Lambert MH, Montana VG, Parks DJ, Blanchard SG, Brown PJ, Sternbach DD, Lehmann JM, Wisley GB, Willson TM, Kliewer SA, Milburn MV 1999 Molecular recognition of fatty acids by peroxisome proliferator-activated receptors. Mol Cell 3:397–403[Medline]
  291. Nagy L, Tontonoz P, Alvarez JGA, Chen H, Evans RM 1998 Oxidized LDL regulates macrophage gene expression through ligand activation of PPAR{gamma}. Cell 93:229–240[Medline]
  292. Ren D, Collingwood TN, Rebar EJ, Wolffe AP, Camp HS 2002 PPAR{gamma} knockdown by engineered transcription factors: exogenous PPAR{gamma}2 but not PPAR{gamma}1 reactivates adipogenesis. Genes Dev 16:27–32[Abstract/Free Full Text]
  293. Tontonoz P, Hu E, Spiegelman BM 1994 Stimulation of adipogenesis in fibroblasts by P, PAR{gamma}2, a lipid-activated transcription factor. Cell 79:1147–1156[Medline]
  294. Rosen ED, Sarraf P, Troy AE, Bradwin G, Moore K, Milstone DS, Spiegelman BM, Mortensen RM 1999 PPAR{gamma} is required for the differentiation of adipose tissue in vivo and in vitro. Mol Cell 4:611–617[Medline]
  295. Hallakou S, Doare F, Foufelle F, Kergost M, Guerre-Millo, Berthault MF, Dugail I, Mortin J, Auwerx J, Farre P 1997 Pioglitazone induces in vivo adipocyte differentiation in obese Zucker fa/fa rats. Diabetes 46:1393–1399[Abstract]
  296. Morrison RF, Farmer SF 2000 Hormonal signaling and transcriptional control of adipocyte differentiation. J Nutr 130:3116S–3121S
  297. Rosen Ed, Walkey CJ, Puigserver P, Spiegelman BM 2000 Transcriptional regulation of adipogenesis. Genes Dev 14:1293–1307[Free Full Text]
  298. Fajas L, Fruchart JC, Auwerx J 1998 Transcriptional control of adipogenesis. Curr Opin Cell Biol 10:165–173[CrossRef][Medline]
  299. Spiegelman BM, Flier JS 1996 Adipogenesis and obesity: rounding out the big picture. Cell 87:377–389[Medline]
  300. Rosen ED, Hsu CH, Wang X, Sakai S, Freeman MW, Gonzalez FJ, Spiegelman BM 2002 C/EBP{alpha} induces adipogenesis through PPAR{gamma}: a unified pathway. Genes Dev 16:22–26[Abstract/Free Full Text]
  301. Wu Z, Rosen ED, Brun R, Hauser S, Adelmant G, Troy AE, McKeon C, Darlington GJ, Spiegelman BM 1999 Cross-regulation of C/EBP {alpha} and PPAR {gamma} controls the transcriptional pathway of adipogenesis and insulin sensitivity. Mol Cell 3:151–158[Medline]
  302. El-Jack AK, Hamm JK, Pilch PF, Farmer SR 1999 Reconstitution of insulin-sensitive glucose transport in fibroblasts requires expression of both PPAR{gamma} and C/EBP{alpha}. J Biol Chem 274:7946–7951[Abstract/Free Full Text]
  303. Tontonoz P, Hu E, Grraves RA, Budavari AI, Spiegelman BM 1994 mmPPAR{gamma}2: tissue-specific regulator of an adipocyte enhancer. Genes Dev 8:1224–1234[Abstract]
  304. Tontonoz P, Hu E, Devine J, Beale EG, Spiegelman BM 1995 PPAR-{gamma}2 regulates adipose expression of the phosphoenolpyruvate carboxykinase gene. Mol Cell Biol 15:351–357[Abstract]
  305. Schoonjans K, Watanabe M, Suzuki H, Mahfoudi A, Krey G, Wahli W, Grimaldi P, Staels B, Yamamoto T, Auwerx J 1995 Induction of the acyl-coenzyme A synthetase gene by fibrates and fatty acids is mediated by a peroxisome proliferator response element in the C promoter. J Biol Chem 270:19269–19276[Abstract/Free Full Text]
  306. Schoonjans K, Peinado-Onsurbe J, Lefebvre AM, Heyman RA, Briggs M, Deeb S, Staels B, Auwerx J 1996 PPAR{alpha} and PPAR{gamma} activators direct a distinct tissue-specific transcriptional response via PPRE in thelipoprotein lipase gene. EMBO J 15:5336–5348[Abstract]
  307. Martin G, Schoonjans K, Lefebvre AM, Staels B, Auwerx J 1997 Coordinate regulation of the expression of the fatty acid transport protein and acyl-CoA synthetase genes by PPAR{alpha} and PPAR{gamma} activators. J Biol Chem 272:25210–25217[Abstract/Free Full Text]
  308. Sfeir Z, Ibrahimi A, Amri E, Grimaldi P, Abumrad N 1997 Regulation of FAT/CD36 gene expression: further evidence in support of a role of the protein in fatty acid binding/transport. Prostaglandins Leukotrienes Essent Fatty Acids 57:17–21[Medline]
  309. Ribon V, Johnson JH, Camp HS, Saltiel AR 1998 Thiazolidinediones and insulin resistance: peroxisome proliferator-activated receptor {gamma} activation stimulates expression of the CAP gene. Proc Natl Acad Sci USA 95:14751–14756[Abstract/Free Full Text]
  310. Adams M, Montague CT, Prins JB, Holder JC, Smith SA, Sanders L, Digby JE, Sewter CP, Lazer MA, Chatterjee VKK, O’Rahilly S 1997 Activators of peroxisome proliferators-activated receptor {gamma} have depot-specific effects on human preadipocyte differentiation. J Clin Invest 100:3149–3153[Abstract/Free Full Text]
  311. Okuno A, Tmemoto H, Tobe K, Ueki K, Mori Y, Iwamoto K, Umesono K, Akanuma Y, Fijiwara T, Horikoshi H, Yazaki Y, Kadowaki T 1998 Troglitazone increase the number of small adipocytes without the change of white adipose tissue mass in obese Zucker rats. J Clin Invest 101:1354–1361[Abstract/Free Full Text]
  312. de Souza CJ, Eckhardt M, Gagen K, Dong M, Chen W, Laurent D, Burkey BF 2001 Effects of pioglitazone on adipose tissue remodeling within the setting of obesity and insulin resistance. Diabetes 50:1863–1871[Abstract/Free Full Text]
  313. Gurnell M, Wentworth JM, Agostini M, Adams M, Collingwood TN, Provenzano C, Browne PO, Rajanayagam O, Burris TP, Schwabe JW, Lazar MA, Chatterjee VK 2000 A dominant-negative peroxisome proliferator-activated receptor {gamma} (PPAR{gamma}) mutant is a constitutive repressor and inhibits PPAR{gamma}-mediated adipogenesis. J Biol Chem 275:5754–5759[Abstract/Free Full Text]
  314. Boden G, Cheung P, Mozzoli M, Fried SK 2003 Effect of thiazolidinediones on glucose and fatty acid metabolism in patients with type 2 diabetes. Metabolism 52:753–759[CrossRef][Medline]
  315. Ciaraldi TP, Kong APS, Chu NV, Kim DD, Baxi S, Loviscach M, Plodkowski R, Reitz R, Caulfield M, Mudaliar S, Henry RR 2002 Regulation of glucose transport and insulin signaling by troglitazone or metformin in adipose tissue of type 2 diabetic subjects. Diabetes 51:30–36[Abstract/Free Full Text]
  316. Kelley IE, Walsh K, Han TS, Lean MEJ 1999 Effects of a thiazolidinedione compound on body fat and fat distribution of patients with type 2 diabetes. Diabetes Care 22:288–293[Abstract]
  317. Kawai T, Takei I, Oguma Y, Ohashi N, Tokui M, Oguchi S, Katsukawa F, Hirose H, Shimada A, Watanabe K, Saruta T 1999 Effects of troglitazone on fat distribution in the treatment of male type 2 diabetes. Metabolism 48:1102–1107[Medline]
  318. Barzilai N, She L, Liu BQ, Vuguin P, Cohen P, Wang J, Rossetti L 1999 Surgical removal of visceral fat reverses hepatic insulin resistance. Diabetes 48:94–98[Abstract]
  319. Rebuffe-Scrive M, Andersson B, Olbe L, Bjorntorp P 1989 Metabolism of adipose tissue in intraabdominal depots of nonobese men and women. Metabolism 38:453–458[Medline]
  320. Smith SR, Lovejoy JC, Greenway F, Ryan D, deJone L, de la Bretonne J, Volafova J, Bray GA 2001 Contributions of total body fat, abdominal subcutaneous adipose tissue compartments, and visceral adipose tissue to the metabolic complications of obesity. Metabolism 50:425–435[Medline]
  321. Frayn KN, Smara JS, Summers LK 1997 Visceral fat in relation to health: is it a major culprit or simply an innocent bystander? Int J Obes Relat Metab Disord 21:1191–1192[CrossRef][Medline]
  322. Engfeldt P, Arner P 1988 Lipolysis in human adipocytes, effects of cell size, age, and of regional differences. Horm Metab Res 19(Suppl):26–29
  323. Colberg SR, Simoneau JA, Thaete FL, Kelley DE 1995 Skeletal muscle utilization of free fatty acids in women with visceral obesity. J Clin Invest 95:1846–1853[Medline]
  324. Miyazaki Y, He H, Mandarino LJ, DeFronzo RA 2003 Rosiglitazone improves downstream insulin-receptor signaling in type 2 diabetic patients. Diabetes 52:1943–1950[Abstract/Free Full Text]
  325. Song H, Shojima N, Sakoda H, Ogihara T, Fujishoro M, Katagiri H, Anai M, Onishi Y, Ono H, Inukai K, Fukushima Y, Kikuchi M, Shimano H, Yamada N, Oka Y, Asano T 2002 Resistin is regulated by C/EBPs, PPARs, and signal-transducing molecules. Biochem Biophy Res Commun 299:291–298[CrossRef][Medline]
  326. Fonseca VA, Reynolds T, Hemphill D 1998 Effect of troglitazone on fibrinolysis and activated coagulation in patients with non-insulin-dependent diabetes mellitus. J Diabetes Complications 12:181–186[CrossRef][Medline]
  327. Yu JG, Javorschi S, Hevener Al, Norman RA, Sinha M, Olefsky JM 2002 The effect of thiazolidinediones on plasma adiponectin levels in normal, obese, and type 2 diabetic subjects. Diabetes 51:2968–2974[Abstract/Free Full Text]
  328. Sreenan S, Sturis J, Pugh W, Burant CF, Polonsky KS 1996 Prevention of hyperglycemia in the Zucker diabetic fatty rat by treatment with metformin or troglitazone. Am J Physiol 271:E742–E747
  329. Smith SA, Lister CA, Toseland CD, Buckingham RE 2000 Rosiglitazone prevent the onset of hyperglycemia and proteinuria in the Zucker diabetic fatty rat. Diabetes Obesity Metab 2:363–372[CrossRef]
  330. Finegood DT, McArthur D, Kojwang M, Thomas J, Topp BG, Leonard T, Buckingham RE 2001 ß-Cell mass dynamics in Zucker diabetic fatty rats: rosiglitazone prevents the rise in net cell death. Diabetes 50:1021–1029[Abstract/Free Full Text]
  331. Lupi R, Del Guerra S, Marselli L, Bugliani M, Boggi U, Mosca F, Marachetti P, Del Prato S 2002 PPAR{gamma} ligand-specificity in the modulation of insulin release from isolated human islets in response to fatty acid incubation. Diabetologia 45(Suppl 2):A161
  332. Marselli L, Bambini MC, Lupi R, Del Guerra S, Boggi U, Mosca F, Del Prato S, Marchetti P 2002 Rosiglitazone partially prevents free fatty acid-induced cytotoxicity of human pancreatic islet cells. Diabetologia 45(Suppl 2):A161
  333. Buchanan TA, Xiang AH, Peters RK 2002 Preservation of pancreatic ß cell function and prevention of type 2 diabetes by pharmacological treatment of insulin resistance in high-risk Hispanic women. Diabetes 51:2796–2803[Abstract/Free Full Text]
  334. Cavaghan MK, Ehrmann DA, Byrne MM, Polonsky KS 1997 Treatment with the oral antidiabetic agent troglitazone improves ß cell responses to glucose in subjects with impaired glucose tolerance. J Clin Invest 100:530–537[Abstract/Free Full Text]