Peroxisome proliferator–activated receptors (PPARs) form a family of nuclear hormone receptors involved in energy hemostasis and lipid metabolism (1,2) and include three isotypes encoded by different genes: PPARα (chromosome 22q12–13.1), PPARβ/δ (chromosome 6p21.2–21.1), and PPARγ (chromosome 3p25). PPARα was the first discovered and causes cellular peroxisome proliferation in rodent livers (3), giving this receptor family its name. Upon activation, PPARs interact with retinoid X receptor to create heterodimers, which bind to a specific DNA sequence motif termed peroxisome proliferator response element (4). Peroxisome proliferator response element usually appears in promoter regions and is constructed from repeats of nucleotide sequence AGGTCA separated by a single nucleotide.

PPARα is widely expressed in tissues with high fatty acid catabolic activity: brown fat, heart, liver, kidney, and intestine (5). Upon activation by endogenous fatty acids and their derivatives, PPARα mediates fatty acid catabolism, gluconeogenesis, and ketone body synthesis, mainly in liver (69). In rodents, PPARα activation also influences immune modulation (10,11) and amino acid metabolism (12), reduces plasma triglyceride, reduces muscle and liver steatosis, and ameliorates insulin resistance (IR) (13,14). Pharmacologic PPARα activation is achieved by fibrates (7) and results in reduced (30–50%) triglyceride and VLDL levels by increasing lipid uptake, lipoprotein lipase–mediated lipolysis, and β-oxidation (15). This is accompanied by a modest increase in HDL cholesterol (5–20%), secondary to transcriptional induction of apolipoprotein A-I/A-II synthesis in liver (15). In man, the primary effect of PPARα is to reduce plasma triglyceride concentration; effects on plasma free fatty acid (FFA) concentration/FFA oxidation, muscle/liver fat content, and muscle/hepatic insulin sensitivity have not been demonstrated with current PPARα agonists such as fenofibrate (16,17). Fibrates are used to treat severe hypertriglyceridemia and combined hyperlipidemia (1820). Clinical trials to establish a role for PPARα agonists (fenofibrate, gemfibrozil) in primary or secondary cardiovascular prevention in patients with hypertriglyceridemia or diabetes have been disappointing (21,22). Clinically significant effects of fibrates on glucose homeostasis, IR, and insulin secretion in man have not been demonstrated (16,17,23).

PPARβ/δ is expressed ubiquitously, correlating with the level of cellular proliferation exhibited in different tissues (24). In rodents, PPARβ/δ activation exerts metabolic effects in skin, gut, skeletal muscle, adipose tissue, and brain (25,26). Several PPARβ/δ agonists are in clinical trials because of their beneficial effects on dyslipidemia (27,28) and other components of metabolic syndrome (29,30).

PPARγ has two splice variants, PPARγ1 and PPARγ2, differing by 30 amino acids in the N′ terminal end. While PPARγ1 is widely expressed in tissues (skeletal muscle heart, liver) at low levels, both are highly expressed in adipose tissue (31,32). PPARγ is considered the “master” regulator of adipogenesis (33). PPARγ overexpression in cultured fibroblasts transforms them into adipocytes (34), while selective adipose deletion of PPARγ results in lipodystrophy and IR (3537). Dominant negative PPARγ mutations are associated with lipodystrophy (in the limbs and gluteal region), dyslipidemia, hypertension, and severe IR (3840). PPARγ polymorphisms (specifically, Pro12Ala) are associated with increased risk of developing type 2 diabetes (T2DM) (4143). PPARγ agonists, thiazolidinediones (2,44,45), are potent insulin sensitizers, enhance insulin secretion, improve glucose tolerance, and are the focus of this review.

Troglitazone was the first thiazolidinedione approved by the U.S. Food and Drug Administration (FDA) and shown to improve insulin sensitivity and β-cell function in T2DM, impaired glucose tolerance (IGT), and nondiabetic individuals (4650). Troglitazone also was shown to improve endothelial dysfunction in obesity and T2DM (49,51), induce ovulation in PCOS (52), and effectively treat lipodystrophy (53). Troglitazone also caused fat redistribution from visceral to subcutaneous adipose tissue (54,55) and reduced circulating levels of inflammatory adipocytokines and FFAs, while increasing plasma adiponectin levels (2). Thus, troglitazone shares many beneficial effects with pioglitazone and rosiglitazone. However, because of hepatotoxicity troglitazone was removed from the U.S. market by the FDA in 1997 (56). However, the idiosyncratic liver toxicity observed with troglitazone does not appear to be a class effect. In a review of the literature, alanine aminotransferase levels >10 times the upper limit of normal were observed in 0.68% of diabetic patients treated with troglitazone versus no individuals treated with pioglitazone or rosiglitazone (57). (See subsequent discussion on nonalcoholic steatohepatitis [NASH].)

Rosiglitazone shares similar beneficial effects with pioglitazone and troglitazone on insulin sensitivity, β-cell function, glycemic control, endothelial function, and adipocyte metabolism (see subsequent discussion). However, because of concerns about cardiovascular safety rosiglitazone has been severely restricted in the U.S. and has been removed from the market in Europe and many other countries. In 2007, a meta-analysis by Nissen and Wolski (58) suggested an increased incidence of cardiovascular events in diabetic patients treated with rosiglitazone. In 2010, a patient-level analysis by FDA statisticians of data supplied by GlaxoSmithKline gave hazard ratio (HR) 1.4 for composite MACE end point (cardiovascular death, myocardial infarction [MI], stroke) and 1.80 for MI (59), leading to removal of rosiglitazone from the U.S. market for all practical purposes. In a recent literature review, Schernthaner and Chilton found that rosiglitazone consistently was associated with HR >1.0 for cardiovascular events, while pioglitazone was associated with HR <1.0 (60).

In subsequent sections, we will focus on the pleotrophic effect of thiazolidinediones, with emphasis on pioglitazone and rosiglitazone.

Pleotrophic effects of PPARγ agonists

PPARγ agonists exert pleotrophic effects on glucose and lipid metabolism in multiple tissues and have become an important therapeutic agent for treating T2DM (45,61,62).

Glycemic control.

Thiazolidinediones are potent insulin sensitizers in liver/muscle/adipocytes (14,6167), augment/preserve β-cell function (68), and produce durable HbA1c reduction in T2DM. In eight of eight long-term (>1.5 years), double-blind, or active comparator studies (Fig. 1), thiazolidinediones caused durable HbA1c reduction (rev. in 61) lasting up to 5–6 years (69). Their durable effect on glycemic control results from combined action to both augment β-cell function and enhance insulin sensitivity. In T2DM patients with starting HbA1c 8.0–8.5%, one can expect a 1.0–1.5% decrease in HbA1c (7076). Thiazolidinediones are approved for monotherapy and add-on therapy to all oral hypoglycemic agents, glucagon-like peptide-1 analogs, and insulin (76).

Figure 1

Thiazolidinediones produce a sustained long-term reduction in HbA1c in eight of eight double-blind or placebo- or active-comparator controlled studies. (See text for a more detailed discussion.) Reprinted with permission from DeFronzo (61).

Figure 1

Thiazolidinediones produce a sustained long-term reduction in HbA1c in eight of eight double-blind or placebo- or active-comparator controlled studies. (See text for a more detailed discussion.) Reprinted with permission from DeFronzo (61).

Close modal

Insulin sensitivity in liver and muscle.

In liver, thiazolidinediones augment insulin sensitivity and inhibit gluconeogenesis, leading to reduction in fasting plasma glucose concentration (63,64). In muscle, thiazolidinediones are the only true insulin sensitizers, producing a decline in postprandial glucose levels (61,66,67). Metformin is a weak insulin sensitizer in muscle, and it has been difficult to demonstrate a muscle insulin-sensitizing effect in absence of weight loss (77,78). Thiazolidinedione-mediated improvement in insulin sensitivity in T2DM is mediated via multiple mechanisms: PPARγ activation, enhanced insulin signaling, increased glucose transport, enhanced glycogen synthesis, improved mitochondrial function, and fat mobilization out of muscle/liver, i.e., reversal of lipotoxicity (45,62,7982). Recent studies suggest that metabolic effects of thiazolidinediones are mediated by mitochondrial target of thiazolidinediones, mtot1 and mtot2, which represent the pyruvate transporter (83,84).

For insulin to exert its metabolic effects, it must first bind to and activate insulin receptor by phosphorylating three key tyrosine molecules on β chain (Fig. 2). This causes insulin receptor substrate (IRS)-1 translocation to plasma membrane, where it undergoes tyrosine phosphorylation, leading to phosphatidylinositol 3-kinase (PI3 kinase) and Akt activation. This causes glucose transport into cell, activation of nitric oxide synthase with arterial vasodilation (8587), and stimulation of multiple intracellular metabolic processes (45).

Figure 2

Insulin signal transduction in healthy nondiabetic (left panel) and T2DM (right panel) subjects. Thiazolidinediones improve insulin signaling through the PI-3 kinase pathway, while inhibiting insulin signaling through the MAP kinase pathway. Reprinted with permission from DeFronzo (61).

Figure 2

Insulin signal transduction in healthy nondiabetic (left panel) and T2DM (right panel) subjects. Thiazolidinediones improve insulin signaling through the PI-3 kinase pathway, while inhibiting insulin signaling through the MAP kinase pathway. Reprinted with permission from DeFronzo (61).

Close modal

In humans, we demonstrated that insulin-stimulated tyrosine phosphorylation of IRS-1 in muscle is severely impaired in lean T2DM (81,88,89), in obese normal glucose tolerant (NGT) individuals (89), and in insulin-resistant NGT offspring of two T2DM parents (90,91) (Fig. 2); similar results have been reported by others (9295). This insulin-signaling defect leads to reduced glucose transport, impaired nitric oxide release (explaining endothelial dysfunction), and multiple defects in intramyocellular glucose metabolism.

In contrast to the defect in IRS-1 activation, the mitogen-activated protein (MAP) kinase pathway, which can be activated by Shc, is normally responsive to insulin (61,62,88,89) (Fig. 2). Stimulation of MAP kinase activates multiple intracellular pathways involved in inflammation, cellular proliferation, and atherogenesis (62,9698).

The defect in IRS-1 tyrosine phosphorylation impairs glucose transport, and resultant hyperglycemia stimulates fasting/postprandial insulin secretion. Because MAP kinase retains normal sensitivity to insulin (62,88,89,94), hyperinsulinemia causes excessive stimulation of this pathway and activation of multiple intracellular pathways involved in inflammation and atherogenesis. This provides a pathogenic link that, in part, can explain the strong association between IR and atherosclerotic cardiovascular disease in nondiabetic and T2DM individuals (99102).

Thiazolidinediones are the only antidiabetes drugs that simultaneously augment insulin signaling through IRS-1 and inhibit MAP kinase pathway (61,77,81), providing a molecular mechanism to explain results from CHICAGO (104) and Pioglitazone Effect on Regression of Intravascular Sonographic Coronary Obstruction Prospective Evaluation (PERISCOPE) (105) studies, in which pioglitazone reduced progression of carotid intima-media thickness (IMT) and coronary atherosclerosis in T2DM. Consistent with these anatomical studies, pioglitazone in PROactive (106) decreased (P = 0.027) MACE end point (death, MI, stroke) by 16%.

Adipocyte insulin sensitivity.

In adipose tissue, thiazolidinediones are potent insulin sensitizers, inhibiting lipolysis and release of inflammatory cytokines, while increasing adiponectin secretion (67,79,80,107109). In T2DM and obese NGT individuals, adipocytes are resistant to insulin’s antilipolytic effect, resulting in accelerated triglyceride breakdown with release of FFA. Elevated plasma FFAs enhance FFA flux into cells, leading to accumulation of toxic lipid metabolites (fatty acyl CoAs, diacylglycerol, ceramides), which inhibit insulin action in muscle/liver (62,110112) and impair β-cell function (113). Thus, these lipotoxic molecules antagonize the core defects that characterize T2DM. By improving insulin sensitivity in adipocytes and inhibiting lipolysis, thiazolidinediones reduce plasma FFA, leading to enhanced insulin sensitivity in muscle/liver and improved β-cell function in T2DM.

In T2DM, adipocytes are in a state of chronic inflammation, as evidenced by monocyte infiltration (114). Inflamed adipocytes release adipocytokines (tumor necrosis factor-α, resistin, angiotensinogen, plasminogen activator inhibitor 1, interleukin-6, and others), which cause IR, impair β-cell function, promote inflammation in distant tissues, augment thrombosis, and accelerate atherogenesis (79,80). Adipocytes from T2DM patients have reduced ability to secrete adiponectin (81,82), a potent vasodilator and antiatherogenic molecule. Thiazolidinediones suppress inflammation in adipose tissue, inhibit release of inflammatory and prothrombotic adipokines, and augment adiponectin secretion.

Thiazolidinediones reverse lipotoxicity

The current diabetes epidemic is being driven by the obesity epidemic. Both obesity and T2DM are characterized by tissue fat overload (Fig. 3). Accumulation of intracellular toxic lipid metabolites causes IR in muscle/liver by inhibiting insulin signaling, glycogen synthesis, and glucose oxidation (rev. in 61,62). Fat accumulation in liver causes nonalcoholic fatty liver disease (NAFLD) and NASH (115), which has become the leading cause of cirrhosis in Westernized countries. Fat accumulation in β-cells impairs insulin secretion and promotes apoptosis (113). Fat deposition in arteries promotes atherogenesis (62), while fat accumulation in visceral depots is associated with coronary arterial disease (116).

Figure 3

Body fat distribution in T2DM patients and its redistribution with thiazolidinediones (TZD). (See text for a detailed discussion.) TG, triglyceride. Reprinted with permission from DeFronzo and colleagues (79).

Figure 3

Body fat distribution in T2DM patients and its redistribution with thiazolidinediones (TZD). (See text for a detailed discussion.) TG, triglyceride. Reprinted with permission from DeFronzo and colleagues (79).

Close modal

Thiazolidinediones reverse lipotoxicity by mobilizing fat out of muscle/liver/β-cells/arteries and relocating fat to subcutaneous adipose depots where it is metabolically “benign” (62,79,80) (Fig. 3). After binding to PPARγ, thiazolidinediones stimulate subcutaneous adipocytes to divide and induce multiple genes involved in lipogenesis (117). Newly formed subcutaneous adipocytes take up FFA, leading to marked reduction in plasma FFA and decreased FFA flux into liver/muscle/β-cells/arteries. Thiazolidinediones also increase expression of PPARγ coactivator (PGC-1), the master regulator of mitochondrial biogenesis (118,119). Increased PGC-1 upregulates multiple mitochondrial oxidative phosphorylation genes, increasing fat oxidation and decreasing levels of intracellular toxic lipid metabolites.

Thiazolidinediones and β-cell function

Thiazolidinediones exert potent effects to improve/preserve β-cell function (68) and demonstrate durability of glycemic control for up to 5–6 years in eight of eight studies (rev. in 61). This is in contrast to sulfonylureas and metformin, which, after initial HbA1c decline, are associated with progressive HbA1c rise, resulting from progressive β-cell failure (120122).

In addition to studies performed in T2DM, six studies demonstrate that thiazolidinediones prevent IGT progression to T2DM (123128). In Diabetes Reduction Assessment with Ramipril and Rosiglitazone Medication (DREAM), T2DM was reduced by 62% with rosiglitazone (124), while in Actos Now for the prevention of diabetes (ACT NOW) (127) pioglitazone decreased IGT conversion to T2DM by 72%. All six studies demonstrated that, in addition to their insulin-sensitizing effect, thiazolidinediones preserved β-cell function. β-Cells respond to increased plasma glucose levels with an increase in insulin secretion, and ΔIG is modulated by severity of IR (128). The insulin secretion/IR index (ΔI/ΔG ÷ IR) represents the gold standard for β-cell function and should not be equated with plasma insulin response. In ACT NOW, improvement in insulin secretion/IR index was the strongest predictor of diabetes prevention in IGT subjects and reversion to NGT. Similar results have been demonstrated in TRoglitazone In the Prevention Of Diabetes (TRIPOD) and Pioglitazone In Prevention Of Diabetes (PIPOD) (123,126), in which development of diabetes in Hispanic women with GDM was decreased by 52 and 62%. In Canadian Normoglycemia Outcomes Evaluation (CANOE) (128), low-dose rosiglitazone (4 mg/day), combined with low-dose metformin (1,000 mg/day), reduced IGT conversion to T2DM by 70%. In vivo and in vitro studies with human/rodent islets demonstrate that thiazolidinediones exert protective effects on β-cell function (129131). Studies from our group using insulin secretion/IR index have shown that thiazolidinediones markedly augment β-cell function in T2DM patients (68) (Fig. 4).

Figure 4

Thiazolidinediones enhance β-cell function (insulin secretion/IR index) in new-onset, drug-naïve T2DM patients and in long-standing, sulfonylurea-treated T2DM individuals (69). *P < 0.01.

Figure 4

Thiazolidinediones enhance β-cell function (insulin secretion/IR index) in new-onset, drug-naïve T2DM patients and in long-standing, sulfonylurea-treated T2DM individuals (69). *P < 0.01.

Close modal

Improved β-cell function with thiazolidinediones results from 1) stimulatory effect of PPARγ to increase GLUT2, glucokinase (132), and Pdx (133) in β-cells; 2) reduced intracellular levels of toxic lipid metabolites (129,132,134,135); 3) muscle/liver insulin-sensitizing effect of thiazolidinediones, which reduce insulin and, therefore, amylin secretion (amylin degradation products are toxic to β-cells [136,137]; the ability of thiazolidinediones to protect human islets from amylin toxicity is mediated via PI3 kinase–dependent pathway [138]); and 4) studies in β-cell insulin receptor knockout (BIRKO) mice suggest that defective insulin signaling through IRS-1/PI3 kinase impairs insulin secretion (139) and that thiazolidinediones correct this insulin signaling defect (129), resulting in enhanced insulin secretion.

Summary

Thiazolidinediones improve multiple defects (IR in liver/muscle/adipocytes and β-cell dysfunction) that comprise the Ominous Octet (61) (Fig. 5), cause durable HbA1c reduction, and can be used as monotherapy or in combination with any other antidiabetes agent. Pioglitazone and rosiglitazone similarly reduce HbA1c, improve insulin sensitivity in muscle/liver/adipocytes, and enhance β-cell function.

Figure 5

Pioglitazone corrects four of the eight pathophysiologic components of the Ominous Octet. Modified with permission from DeFronzo (61). TZD, thiazolidinediones.

Figure 5

Pioglitazone corrects four of the eight pathophysiologic components of the Ominous Octet. Modified with permission from DeFronzo (61). TZD, thiazolidinediones.

Close modal

IR (metabolic) syndrome represents a cluster of metabolic and cardiovascular disorders, each of which represents a major cardiovascular risk factor (62). A common thread linking all IR syndrome components is the basic molecular etiology of IR (61,62,81,88,89), which not only promotes inflammation and atherogenesis but also aggravates other components of the syndrome. Pioglitazone and rosiglitazone ameliorate the molecular defect in insulin signaling, enhance muscle/hepatic/adipocyte insulin sensitivity, correct hyperinsulinemia, improve glucose tolerance and endothelial dysfunction, reduce blood pressure, decrease plasma FFA levels, increase HDL cholesterol, transform small dense LDL particles into larger less atherogenic ones, shift body fat from visceral to subcutaneous depots, mobilize fat out of muscle/liver, reduce plasminogen activator inhibitor 1/tumor necrosis factor-α levels, and increase plasma adiponectin (rev. in 62). Rosiglitazone produces metabolic effects similar to those of pioglitazone with two notable exceptions: rosiglitazone increases both plasma LDL cholesterol and triglycerides (140). Concerns about cardiovascular safety (58) have led to removal of rosiglitazone from U.S. (56) and European markets.

Pioglitazone reduces cardiovascular events

Pioglitazone is the only antidiabetes medication shown, in a large prospective placebo-controlled outcome study, to reduce cardiovascular events. In PROactive, 5,238 T2DM patients with prior cardiovascular event or multiple CVD risk factors were randomized to pioglitazone or placebo plus standard of care for all cardiovascular risk factors (106). Compared with placebo, pioglitazone reduced the second principal MACE end point (cardiovascular mortality, MI, stroke) by 16% (P < 0.02) (Fig. 6A). Cardiovascular benefit most likely resulted from combined improvements in dyslipidemia (increased HDL cholesterol), endothelial dysfunction, blood pressure, HbA1c, other inflammatory markers that were not measured, and direct effect on arterial wall to inhibit atherogenesis (141). In a subgroup of 2,445 patients with previous MI, pioglitazone reduced (HR 0.72, P = 0.04) likelihood of subsequent MI by 16% (142) (Fig. 6C). In 984 patients with previous stroke, pioglitazone caused 47% reduction (HR 0.53, P = 0.008) in recurrent stroke (3,143) (Fig. 6D).

Figure 6

A: Kaplan-Meier plot of time to MACE end point (mortality, MI, stroke) in T2DM patients treated with pioglitazone (PIO) or placebo (Plc) in PROactive. Redrawn with permission from Dormandy et al. (106). B: Pioglitazone reduces recurrent MI in diabetic patients with a previous MI in PROactive. Redrawn with permission from Erdmann et al. (142). C: Pioglitazone reduces recurrent stroke in diabetic patients with a previous stroke or PROactive. Redrawn with permission from Wilcox et al. (143). D: Meta-analysis of all published studies (excluding PROactive) in which the effect of pioglitazone versus placebo or active comparator on cardiovascular events is examined. Redrawn with permission from Lincoff et al. (145).

Figure 6

A: Kaplan-Meier plot of time to MACE end point (mortality, MI, stroke) in T2DM patients treated with pioglitazone (PIO) or placebo (Plc) in PROactive. Redrawn with permission from Dormandy et al. (106). B: Pioglitazone reduces recurrent MI in diabetic patients with a previous MI in PROactive. Redrawn with permission from Erdmann et al. (142). C: Pioglitazone reduces recurrent stroke in diabetic patients with a previous stroke or PROactive. Redrawn with permission from Wilcox et al. (143). D: Meta-analysis of all published studies (excluding PROactive) in which the effect of pioglitazone versus placebo or active comparator on cardiovascular events is examined. Redrawn with permission from Lincoff et al. (145).

Close modal

The composite primary end point (mortality, nonfatal MI, silent MI, stroke, acute coronary syndrome, coronary artery bypass grafting/percutaneous coronary intervention, leg amputation, leg revascularization) did not reach significance (HR 0.90, P = 0.09) because of increased number of leg revascularization procedures in the pioglitazone group. Leg revascularization is not a MACE end point and typically is excluded from cardiovascular intervention trials, i.e., with statins, because the major risk factors for peripheral vascular disease are gravity (i.e., subject’s height) and smoking, which are not influenced by antidiabetes therapy. Subsequent PROactive analyses confirmed that pioglitazone has no beneficial effect on peripheral vascular disease (144). Consistent with PROactive, a meta-analysis of all pioglitazone studies published (excluding PROactive) and reported to the FDA demonstrated a 25% decrease in cardiovascular events (145) (Fig. 6B), and a recent review recommended that pioglitazone should be considered in diabetic patients with cardiovascular disease (146).

Two additional studies demonstrated that pioglitazone slows anatomical progression of atherosclerotic cardiovascular disease. In PERISCOPE (105), T2DM patients with established coronary artery disease were randomized to pioglitazone or glimepiride for 1.5 years. In the glimeperide-treated group, percent atheroma volume progressed, while percent atheroma volume regressed in the pioglitazone-treated group. In CHICAGO, pioglitazone halted progression of carotid IMT, whereas carotid IMT progressed in the glimepiride-treated group (P = 0.008) (104). Results of these two anatomical trials (104,105), when viewed in concert with cardiovascular outcome trials (106,145), strongly suggest that pioglitazone provides cardiovascular protection, especially in individuals with established cardiovascular disease.

The different effects of pioglitazone and rosiglitazone on cardiovascular outcomes remains unexplained. One obvious explanation is rise in plasma LDL cholesterol and triglyceride observed with rosiglitazone (140). Another explanation involves differential regulation of gene expression by rosiglitazone and pioglitazone. In muscle (147) and adipocytes (148), multiple genes are differentially stimulated or inhibited by the two thiazolidinediones, and the function of these genes is largely unknown.

Thiazolidinediones prevent T2DM in high-risk individuals

Six large prospective, randomized, double-blind, placebo-controlled studies (TRIPOD [126], PIPOD [123], DPP [125], DREAM [124], CANOE [128], and ACT NOW [127]) have provided conclusive evidence that thiazolidinediones dramatically reduce by 52–72% conversion of prediabetes (IGT and/or IFG) to T2DM. In ACT NOW, IGT conversion to T2DM was reduced by 72% and carotid IMT progression was diminished by >50% versus placebo (127). Increased β-cell function (insulin secretion/IR index) was the strongest predictor of diabetes prevention. In ACT NOW and other prevention trials reductions in HbA1c, blood pressure, triglycerides, inflammatory cytokines, and rise in HDL cholesterol also have been observed (127).

In T2DM hepatic fat accumulation, NAFLD is common and represents a precursor for NASH. NASH is associated with hepatic/muscle IR (115) and accelerated atheogenesis (148). Several large, placebo-controlled studies have demonstrated that pioglitazone mobilizes fat from liver, reduces hepatic injury, and causes histologic improvement in inflammation/fibrosis in NASH (149151). Pioglitazone also reduces liver fat and improves IR in lipodystrophic patients (152). Studies examining effect of rosiglitazone in NASH have shown an initial beneficial effect on liver histologic parameters with no benefit from prolonged continuous treatment (153).

Diabetic rodents develop renal insufficiency and histologic lesions analogous to those in man, and thiazolidinediones reduce mesangial matrix (hallmark lesion of diabetic nephropathy) volume, decrease urinary protein excretion, and prevent renal failure (154,155). PPARγ is expressed diffusely throughout kidney, and PPARγ agonists inhibit mesangial cell proliferation and reduce mRNA expression of matrix proteins (collagen, fibronectin) and transforming growth factor-β, which has been implicated in glomerular injury (156). In diabetic humans, pioglitazone (157) and rosiglitazone (158) reduce albuminuria, although long-term studies examining effect of thiazolidinediones on GFR have not been performed. Beneficial effect of thiazolidinediones to reduce albuminuria cannot be explained by improved glycemic control and is closely correlated with improved insulin sensitivity (159).

Diabetic individuals with renal insufficiency are at increased risk for cardiovascular disease/mortality (159). In PROactive, pioglitazone significantly reduced MACE end point in patients with and without reduced GFR (160). Thiazolidinediones also reduced all-cause mortality in hemodialysis-treated patients (161).

Benefits of pioglitazone on glycemic control and prevention of cardiovascular disease are well established. However, physicians must be cognizant of potential side effects to maximize benefit and minimize risk. The majority of pioglitazone’s beneficial effects on glucose metabolism, insulin sensitivity, insulin secretion, and cardiovascular risk factors are observed with a dose of 30 mg/day (70,162). At this dose, side effects are mild and manageable. Increasing dose to 45 mg/day provides little more efficacy and substantially increases risk of side effects (70). Therefore, we recommend a starting dose of 7.5–15 mg/day, tritiated to 30 mg/day (163165). Combined pioglitazone/metformin therapy (166,167) is particularly effective in reducing HbA1c, does not cause hypoglycemia, and minimizes side effects. Moreover, both pioglitazone (106,145) and metformin (121) reduce cardiovascular events, although the number (n = 344) of subjects in the metformin arm of the UK Prospective Diabetes Study (UKPDS) was small and would not satisfy current standards for a cardiovascular intervention study.

Fat weight gain

On average, pioglitazone-treated subjects gain ∼2–3 kg of fat weight after 1 year (70,76,106,168), which results from PPARγ stimulation of hunger centers in hypothalamus (169). Simultaneously, PPARγ activation redistributes fat from visceral to subcutaneous depots (55,79,170), mobilizes fat out of muscle/liver/β-cells (79,80,149,150,171), inhibits lipolysis/reduces plasma FFA (79,80,109), and stimulates PGC-1/other mitochondrial genes involved in lipid oxidation (118). The net result is a metabolically more favorable fat distribution from visceral to subcutaneous depots where it is metabolically benign (79,80) and depletion of toxic lipid metabolites in muscle/liver/β-cells (62). Of note, the greater the weight gain, the greater the improvements in β-cell function and insulin sensitivity and the greater the reduction in HbA1c (68,170,172). On a short-term basis, i.e., up to 3 years (106), no adverse effects of thiazolidinedione-associated weight gain have been observed. Long-term effects, if any, of thiazolidinedione-associated weight gain remain unknown. Weight gain, if excessive, should be managed with reinforcement of dietary advice and exercise, reduction in pioglitazone dose, or use of pharmacologic agents approved for weight loss.

Bone fractures

T2DM patients treated with thiazolidinediones have increased risk of fracture (173176), which primarily occurs in distal long bones of upper (forearm, hand, wrist) and lower (foot, ankle, fibula, tibia) limbs and is related to trauma. Excess fracture risk is 0.8 fractures per 100 patient-years (1.9 in pioglitazone treated vs. 1.1 in comparator treated) (173176). This represents a small but significant risk. Since increased fracture risk primarily occurs in postmenopausal females and not in premenopausal women or men, pioglitazone should be used with caution in postmenopausal women or not at all.

Fluid retention and congestive heart failure

Thiazolidinediones may cause fluid retention, which can exacerbate heart failure in diabetic patients who do not uncommonly have underlying diastolic dysfunction (106). When used as monotherapy, edema occurs in 3–5% of individuals and is dose related (177). Edema most commonly occurs when thiazolidinediones are used with sulfonylureas and especially with insulin (177180). Fluid retention occurs secondary to peripheral vasodilation (181) and stimulation of ENac (epithelial sodium) channel in collecting duct (182). Sodium retention responds well to distally acting diuretics, spironolactone or triamterene (183). Pedal edema identifies individuals at risk to develop congestive heart failure (CHF) and who should be treated with a diuretic or reduction in pioglitazone dose. In PROactive, incidence of CHF was 6%. However, cases were not adjudicated, and mortality and cardiovascular events tended to be decreased in pioglitazone-treated individuals who developed CHF (106,184). These results suggest that after excess fluid has been diuresed, the cardioprotective effect of pioglitazone becomes evident. Lastly, pioglitazone has no negative impact on cardiac function (185) and improves endothelial dysfunction (186).

In PROactive (106), incidence of malignancy was similar in pioglitazone (3.7%) and placebo (3.8%) groups. However, two imbalances were noted. There were more cases of bladder cancer in pioglitazone (n = 16) versus placebo (n = 6) groups (P = 0.069). Prior to unblinding, external experts adjudicated that 11 cases could not plausibly be related to treatment. Of the remaining nine case subjects, six were treated with pioglitazone and three with placebo (P = 0.309). The other imbalance was related to breast cancer; there were fewer breast cancers in the pioglitazone versus placebo group (3 vs. 11, P = 0.034). Thus, the nonsignificant increase in bladder cancer was numerically offset by the statistically significant decrease in breast cancer.

In 2003, the FDA requested that a safety study be conducted to assess whether pioglitazone increased bladder cancer risk. After 4 years of a 10-year longitudinal cohort study of 193,099 patients (187), ever use of pioglitazone was not associated with increased bladder cancer risk (HR 1.2 [95% CI 0.9–1.5]). However, in patients receiving pioglitazone for ≥24 months, there was slight increased bladder cancer risk (1.4 [1.03–2.0]); 95% of cancers were detected at an early in situ stage, and authors acknowledged that this could have been attributed to the fact that pioglitazone-treated patients underwent greater surveillance for bladder cancer. Bladder cancer risk increased from 7/10,000 patient-treatment years (no pioglitazone) to 10/10,000 (with pioglitazone)—an increase of 3 cases per 10,000 patient-treatment years. Overall, there was no increase in total cancers in pioglitazone-treated patients (187,188), and risk of some cancers (colon, kidney/renal pelvis, breast) was decreased (188). In a recent 8-year analysis of the same study population, HR for bladder cancer was 0.98 (95% CI 0.81–1.18) (189). If pioglitazone actually increased bladder cancer risk, one would have expected HR to increase—not decrease—after 8 years. These results argue against a putative role for pioglitazone in development of bladder cancer. Further, overall incidence of malignancy has been reported not to increase (106) or decrease in certain cancer types (breast and liver) in pioglitazone-treated patients (188,190192). Lastly, any increased bladder cancer risk must be viewed in the context of protection against all-cause death, MI, and stroke, i.e., MACE end point in PROactive. It has been estimated that treatment of 10,000 patients with pioglitazone would avoid 210 MIs, stroke, or deaths over 3 years (193) compared with a potential increase of three cases of bladder cancer per 10,000 patients over the same period. Moreover, even this increase of 3/10,000 disappeared after 8 years (189).

Based upon the body of evidence reviewed above (not including 8-year follow-up data reported by Lewis), the FDA recommended that pioglitazone not be used in patients with active bladder cancer or prior bladder cancer history. We recommend that any hematuria be evaluated to exclude bladder cancer before starting pioglitazone.

As reviewed in preceding sections, the benefit-to-risk ratio for pioglitazone is very favorable. Importantly, if physicians are aware of potential risks associated with thiazolidinediones and if the pioglitazone dose does not exceed 30 mg/day, side effects can be reduced even further (Table 1).

Table 1

Benefits and risks associated with thiazolidinedione therapy

Benefits and risks associated with thiazolidinedione therapy
Benefits and risks associated with thiazolidinedione therapy

This publication is based on the presentations from the 4th World Congress on Controversies to Consensus in Diabetes, Obesity and Hypertension (CODHy). The Congress and the publication of this supplement were made possible in part by unrestricted educational grants from Abbott, AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Eli Lilly, Ethicon Endo-Surgery, Janssen, Medtronic, Novo Nordisk, Sanofi, and Takeda.

R.A.D. is a member of the Advisory Board of Takeda, Bristol-Myers Squibb, Janssen, Boehringer Ingelheim, Novo Nordisk, Lexicon, and Amylin. R.A.D. is a member of the Speakers Bureau of Novo Nordisk, Amylin, Bristol-Myers Squibb, and Janssen. No other potential conflicts of interest relevant to this article were reported.

R.E., R.A.D., and M.A.-G. contributed to writing, revising, and reviewing the manuscript. R.A.D. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

1.
Michalik
L
,
Auwerx
J
,
Berger
JP
, et al
.
International Union of Pharmacology. LXI. Peroxisome proliferator-activated receptors
.
Pharmacol Rev
2006
;
58
:
726
741
[PubMed]
2.
Spiegelman
BM
.
PPAR-gamma: adipogenic regulator and thiazolidinedione receptor
.
Diabetes
1998
;
47
:
507
514
[PubMed]
3.
Issemann
I
,
Green
S
.
Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators
.
Nature
1990
;
347
:
645
650
[PubMed]
4.
Kliewer
SA
,
Umesono
K
,
Noonan
DJ
,
Heyman
RA
,
Evans
RM
.
Convergence of 9-cis retinoic acid and peroxisome proliferator signalling pathways through heterodimer formation of their receptors
.
Nature
1992
;
358
:
771
774
[PubMed]
5.
Mandard
S
,
Müller
M
,
Kersten
S
.
Peroxisome proliferator-activated receptor alpha target genes
.
Cell Mol Life Sci
2004
;
61
:
393
416
[PubMed]
6.
Lefebvre
P
,
Chinetti
G
,
Fruchart
JC
,
Staels
B
.
Sorting out the roles of PPAR alpha in energy metabolism and vascular homeostasis
.
J Clin Invest
2006
;
116
:
571
580
[PubMed]
7.
Vu-Dac
N
,
Schoonjans
K
,
Kosykh
V
, et al
.
Fibrates increase human apolipoprotein A-II expression through activation of the peroxisome proliferator-activated receptor
.
J Clin Invest
1995
;
96
:
741
750
[PubMed]
8.
Kersten
S
,
Seydoux
J
,
Peters
JM
,
Gonzalez
FJ
,
Desvergne
B
,
Wahli
W
.
Peroxisome proliferator-activated receptor alpha mediates the adaptive response to fasting
.
J Clin Invest
1999
;
103
:
1489
1498
[PubMed]
9.
Reddy
JK
,
Hashimoto
T
.
Peroxisomal beta-oxidation and peroxisome proliferator-activated receptor alpha: an adaptive metabolic system
.
Annu Rev Nutr
2001
;
21
:
193
230
[PubMed]
10.
Devchand
PR
,
Keller
H
,
Peters
JM
,
Vazquez
M
,
Gonzalez
FJ
,
Wahli
W
.
The PPARalpha-leukotriene B4 pathway to inflammation control
.
Nature
1996
;
384
:
39
43
[PubMed]
11.
Staels
B
,
Koenig
W
,
Habib
A
, et al
.
Activation of human aortic smooth-muscle cells is inhibited by PPARalpha but not by PPARgamma activators
.
Nature
1998
;
393
:
790
793
[PubMed]
12.
Kersten
S
,
Mandard
S
,
Escher
P
, et al
.
The peroxisome proliferator-activated receptor alpha regulates amino acid metabolism
.
FASEB J
2001
;
15
:
1971
1978
[PubMed]
13.
Chou
CJ
,
Haluzik
M
,
Gregory
C
, et al
.
WY14,643, a peroxisome proliferator-activated receptor alpha (PPARalpha) agonist, improves hepatic and muscle steatosis and reverses insulin resistance in lipoatrophic A-ZIP/F-1 mice
.
J Biol Chem
2002
;
277
:
24484
24489
[PubMed]
14.
Guerre-Millo
M
,
Gervois
P
,
Raspé
E
, et al
.
Peroxisome proliferator-activated receptor alpha activators improve insulin sensitivity and reduce adiposity
.
J Biol Chem
2000
;
275
:
16638
16642
[PubMed]
15.
Staels
B
,
Dallongeville
J
,
Auwerx
J
,
Schoonjans
K
,
Leitersdorf
E
,
Fruchart
JC
.
Mechanism of action of fibrates on lipid and lipoprotein metabolism
.
Circulation
1998
;
98
:
2088
2093
[PubMed]
16.
Bajaj
M
,
Suraamornkul
S
,
Hardies
LJ
,
Glass
L
,
Musi
N
,
DeFronzo
RA
.
Effects of peroxisome proliferator-activated receptor (PPAR)-alpha and PPAR-gamma agonists on glucose and lipid metabolism in patients with type 2 diabetes mellitus
.
Diabetologia
2007
;
50
:
1723
1731
[PubMed]
17.
Avogaro
A
,
Piliego
T
,
Catapano
A
,
Miola
M
,
Tiengo
A
for the Gemfibrozil Study Group
.
The effect of gemfibrozil on lipid profile and glucose metabolism in hypertriglyceridaemic well-controlled non-insulin-dependent diabetic patients
.
Acta Diabetol
1999
;
36
:
27
33
[PubMed]
18.
Fruchart
JC
,
Brewer
HB
 Jr
,
Leitersdorf
E
Fibrate Consensus Group
.
Consensus for the use of fibrates in the treatment of dyslipoproteinemia and coronary heart disease
.
Am J Cardiol
1998
;
81
:
912
917
[PubMed]
19.
Ooi
TC
,
Heinonen
T
,
Alaupovic
P
, et al
.
Efficacy and safety of a new hydroxymethylglutaryl-coenzyme A reductase inhibitor, atorvastatin, in patients with combined hyperlipidemia: comparison with fenofibrate
.
Arterioscler Thromb Vasc Biol
1997
;
17
:
1793
1799
[PubMed]
20.
Brunzell
JD
.
Clinical practice. Hypertriglyceridemia
.
N Engl J Med
2007
;
357
:
1009
1017
[PubMed]
21.
Keech
A
,
Simes
RJ
,
Barter
P
, et al
FIELD study investigators
.
Effects of long-term fenofibrate therapy on cardiovascular events in 9795 people with type 2 diabetes mellitus (the FIELD study): randomised controlled trial
.
Lancet
2005
;
366
:
1849
1861
[PubMed]
22.
Ginsberg
HN
,
Elam
MB
,
Lovato
LC
, et al
ACCORD Study Group
.
Effects of combination lipid therapy in type 2 diabetes mellitus
.
N Engl J Med
2010
;
362
:
1563
1574
[PubMed]
23.
Belfort
R
,
Berria
R
,
Cornell
J
,
Cusi
K
.
Fenofibrate reduces systemic inflammation markers independent of its effects on lipid and glucose metabolism in patients with the metabolic syndrome
.
J Clin Endocrinol Metab
2010
;
95
:
829
836
[PubMed]
24.
Braissant
O
,
Foufelle
F
,
Scotto
C
,
Dauça
M
,
Wahli
W
.
Differential expression of peroxisome proliferator-activated receptors (PPARs): tissue distribution of PPAR-alpha, -beta, and -gamma in the adult rat
.
Endocrinology
1996
;
137
:
354
366
[PubMed]
25.
Michalik
L
,
Desvergne
B
,
Basu-Modak
S
,
Tan
NS
,
Wahli
W
.
Nuclear hormone receptors and mouse skin homeostasis: implication of PPARbeta
.
Horm Res
2000
;
54
:
263
268
[PubMed]
26.
Barak
Y
,
Liao
D
,
He
W
, et al
.
Effects of peroxisome proliferator-activated receptor delta on placentation, adiposity, and colorectal cancer
.
Proc Natl Acad Sci USA
2002
;
99
:
303
308
[PubMed]
27.
Choi
YJ
,
Roberts
BK
,
Wang
X
, et al
.
Effects of the PPAR-δ agonist MBX-8025 on atherogenic dyslipidemia
.
Atherosclerosis
2012
;
220
:
470
476
[PubMed]
28.
Bays
HE
,
Schwartz
S
,
Littlejohn
T
 3rd
, et al
.
MBX-8025, a novel peroxisome proliferator receptor-delta agonist: lipid and other metabolic effects in dyslipidemic overweight patients treated with and without atorvastatin
.
J Clin Endocrinol Metab
2011
;
96
:
2889
2897
[PubMed]
29.
Risérus
U
,
Sprecher
D
,
Johnson
T
, et al
.
Activation of peroxisome proliferator-activated receptor (PPAR)delta promotes reversal of multiple metabolic abnormalities, reduces oxidative stress, and increases fatty acid oxidation in moderately obese men
.
Diabetes
2008
;
57
:
332
339
[PubMed]
30.
Cariou
B
,
Zaïr
Y
,
Staels
B
,
Bruckert
E
.
Effects of the new dual PPAR α/δ agonist GFT505 on lipid and glucose homeostasis in abdominally obese patients with combined dyslipidemia or impaired glucose metabolism
.
Diabetes Care
2011
;
34
:
2008
2014
[PubMed]
31.
Tontonoz
P
,
Graves
RA
,
Budavari
AI
, et al
.
Adipocyte-specific transcription factor ARF6 is a heterodimeric complex of two nuclear hormone receptors, PPAR gamma and RXR alpha
.
Nucleic Acids Res
1994
;
22
:
5628
5634
[PubMed]
32.
Vidal-Puig
AJ
,
Considine
RV
,
Jimenez-Liñan
M
, et al
.
Peroxisome proliferator-activated receptor gene expression in human tissues. Effects of obesity, weight loss, and regulation by insulin and glucocorticoids
.
J Clin Invest
1997
;
99
:
2416
2422
[PubMed]
33.
Tontonoz
P
,
Spiegelman
BM
.
Fat and beyond: the diverse biology of PPARgamma
.
Annu Rev Biochem
2008
;
77
:
289
312
[PubMed]
34.
Tontonoz
P
,
Hu
E
,
Spiegelman
BM
.
Stimulation of adipogenesis in fibroblasts by PPAR gamma 2, a lipid-activated transcription factor
.
Cell
1994
;
79
:
1147
1156
[PubMed]
35.
He
W
,
Barak
Y
,
Hevener
A
, et al
.
Adipose-specific peroxisome proliferator-activated receptor gamma knockout causes insulin resistance in fat and liver but not in muscle
.
Proc Natl Acad Sci USA
2003
;
100
:
15712
15717
[PubMed]
36.
Imai
T
,
Takakuwa
R
,
Marchand
S
, et al
.
Peroxisome proliferator-activated receptor gamma is required in mature white and brown adipocytes for their survival in the mouse
.
Proc Natl Acad Sci USA
2004
;
101
:
4543
4547
[PubMed]
37.
Medina-Gomez
G
,
Gray
SL
,
Yetukuri
L
, et al
.
PPAR gamma 2 prevents lipotoxicity by controlling adipose tissue expandability and peripheral lipid metabolism
.
PLoS Genet
2007
;
3
:
e64
[PubMed]
38.
Barroso
I
,
Gurnell
M
,
Crowley
VE
, et al
.
Dominant negative mutations in human PPARgamma associated with severe insulin resistance, diabetes mellitus and hypertension
.
Nature
1999
;
402
:
880
883
[PubMed]
39.
Agostini
M
,
Schoenmakers
E
,
Mitchell
C
, et al
.
Non-DNA binding, dominant-negative, human PPARgamma mutations cause lipodystrophic insulin resistance
.
Cell Metab
2006
;
4
:
303
311
[PubMed]
40.
Semple
RK
,
Chatterjee
VK
,
O’Rahilly
S
.
PPAR gamma and human metabolic disease
.
J Clin Invest
2006
;
116
:
581
589
[PubMed]
41.
Ek
J
,
Urhammer
SA
,
Sørensen
TI
,
Andersen
T
,
Auwerx
J
,
Pedersen
O
.
Homozygosity of the Pro12Ala variant of the peroxisome proliferation-activated receptor-gamma2 (PPAR-gamma2): divergent modulating effects on body mass index in obese and lean Caucasian men
.
Diabetologia
1999
;
42
:
892
895
[PubMed]
42.
Deeb
SS
,
Fajas
L
,
Nemoto
M
, et al
.
A Pro12Ala substitution in PPARgamma2 associated with decreased receptor activity, lower body mass index and improved insulin sensitivity
.
Nat Genet
1998
;
20
:
284
287
[PubMed]
43.
Florez
JC
,
Jablonski
KA
,
Sun
MW
, et al
Diabetes Prevention Program Research Group
.
Effects of the type 2 diabetes-associated PPARG P12A polymorphism on progression to diabetes and response to troglitazone
.
J Clin Endocrinol Metab
2007
;
92
:
1502
1509
[PubMed]
44.
Lehmann
JM
,
Moore
LB
,
Smith-Oliver
TA
,
Wilkison
WO
,
Willson
TM
,
Kliewer
SA
.
An antidiabetic thiazolidinedione is a high affinity ligand for peroxisome proliferator-activated receptor gamma (PPAR gamma)
.
J Biol Chem
1995
;
270
:
12953
12956
[PubMed]
45.
Yki-Järvinen
H
.
Thiazolidinediones
.
N Engl J Med
2004
;
351
:
1106
1118
[PubMed]
46.
Nolan
JJ
,
Ludvik
B
,
Beerdsen
P
,
Joyce
M
,
Olefsky
J
.
Improvement in glucose tolerance and insulin resistance in obese subjects treated with troglitazone
.
N Engl J Med
1994
;
331
:
1188
1193
[PubMed]
47.
Suter
SL
,
Nolan
JJ
,
Wallace
P
,
Gumbiner
B
,
Olefsky
JM
.
Metabolic effects of new oral hypoglycemic agent CS-045 in NIDDM subjects
.
Diabetes Care
1992
;
15
:
193
203
[PubMed]
48.
Cavaghan
MK
,
Ehrmann
DA
,
Byrne
MM
,
Polonsky
KS
.
Treatment with the oral antidiabetic agent troglitazone improves beta cell responses to glucose in subjects with impaired glucose tolerance
.
J Clin Invest
1997
;
100
:
530
537
[PubMed]
49.
Caballero
AE
,
Saouaf
R
,
Lim
SC
, et al
.
The effects of troglitazone, an insulin-sensitizing agent, on the endothelial function in early and late type 2 diabetes: a placebo-controlled randomized clinical trial
.
Metabolism
2003
;
52
:
173
180
[PubMed]
50.
Inzucchi
SE
,
Maggs
DG
,
Spollett
GR
, et al
.
Efficacy and metabolic effects of metformin and troglitazone in type II diabetes mellitus
.
N Engl J Med
1998
;
338
:
867
872
[PubMed]
51.
Tack
CJ
,
Ong
MK
,
Lutterman
JA
,
Smits
P
.
Insulin-induced vasodilatation and endothelial function in obesity/insulin resistance. Effects of troglitazone
.
Diabetologia
1998
;
41
:
569
576
[PubMed]
52.
Azziz
R
,
Ehrmann
D
,
Legro
RS
, et al
PCOS/Troglitazone Study Group
.
Troglitazone improves ovulation and hirsutism in the polycystic ovary syndrome: a multicenter, double blind, placebo-controlled trial
.
J Clin Endocrinol Metab
2001
;
86
:
1626
1632
[PubMed]
53.
Arioglu
E
,
Duncan-Morin
J
,
Sebring
N
, et al
.
Efficacy and safety of troglitazone in the treatment of lipodystrophy syndromes
.
Ann Intern Med
2000
;
133
:
263
274
[PubMed]
54.
Okuno
A
,
Tamemoto
H
,
Tobe
K
, et al
.
Troglitazone increases the number of small adipocytes without the change of white adipose tissue mass in obese Zucker rats
.
J Clin Invest
1998
;
101
:
1354
1361
[PubMed]
55.
Kelly
IE
,
Han
TS
,
Walsh
K
,
Lean
ME
.
Effects of a thiazolidinedione compound on body fat and fat distribution of patients with type 2 diabetes
.
Diabetes Care
1999
;
22
:
288
293
[PubMed]
56.
Malik
AH
,
Prasad
P
,
Saboorian
MH
, et al
.
Hepatic injury due to troglitazone
.
Dig Dis Sci
2000
;
45
:
210
214
57.
Lebovitz
HE
,
Kreider
M
,
Freed
MI
.
Evaluation of liver function in type 2 diabetic patients during clinical trials: evidence that rosiglitazone does not cause hepatic dysfunction
.
Diabetes Care
2002
;
25
:
815
821
[PubMed]
58.
Nissen
SE
,
Wolski
K
.
Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes
.
N Engl J Med
2007
;
356
:
2457
2471
[PubMed]
59.
Callaghan F. Rosiglitazone cardiovascular safety meta-analysis [Internet]. Available from http://www.fda.gov/advisorycommittees/committeesmeetingmaterials/drugs/endocrinologicandmetabolicdrugsadvisorycommittee/ucm21895.htm. Accessed 13 July 2010
60.
Schernthaner
G
,
Chilton
RJ
.
Cardiovascular risk and thiazolidinediones—what do meta-analyses really tell us?
Diabetes Obes Metab
2010
;
12
:
1023
1035
[PubMed]
61.
DeFronzo
RA
.
Banting Lecture. From the triumvirate to the ominous octet: a new paradigm for the treatment of type 2 diabetes mellitus
.
Diabetes
2009
;
58
:
773
795
[PubMed]
62.
DeFronzo
RA
.
Insulin resistance, lipotoxicity, type 2 diabetes and atherosclerosis: the missing links. The Claude Bernard Lecture 2009
.
Diabetologia
2010
;
53
:
1270
1287
[PubMed]
63.
Gastaldelli
A
,
Miyazaki
Y
,
Pettiti
M
, et al
.
The effect of rosiglitazone on the liver: decreased gluconeogenesis in patients with type 2 diabetes
.
J Clin Endocrinol Metab
2006
;
91
:
806
812
[PubMed]
64.
Gastaldelli
A
,
Miyazaki
Y
,
Mahankali
A
, et al
.
The effect of pioglitazone on the liver: role of adiponectin
.
Diabetes Care
2006
;
29
:
2275
2281
[PubMed]
65.
Mayerson
AB
,
Hundal
RS
,
Dufour
S
, et al
.
The effects of rosiglitazone on insulin sensitivity, lipolysis, and hepatic and skeletal muscle triglyceride content in patients with type 2 diabetes
.
Diabetes
2002
;
51
:
797
802
[PubMed]
66.
Miyazaki
Y
,
Mahankali
A
,
Matsuda
M
, et al
.
Improved glycemic control and enhanced insulin sensitivity in type 2 diabetic subjects treated with pioglitazone
.
Diabetes Care
2001
;
24
:
710
719
[PubMed]
67.
Miyazaki
Y
,
DeFronzo
RA
.
Rosiglitazone and pioglitazone similarly improve insulin sensitivity and secretion, glucose tolerance and adipocytokines in type 2 diabetic patients
.
Diabetes Obes Metab
2008
;
10
:
1204
1211
[PubMed]
68.
Gastaldelli
A
,
Ferrannini
E
,
Miyazaki
Y
,
Matsuda
M
,
Mari
A
,
DeFronzo
RA
.
Thiazolidinediones improve beta-cell function in type 2 diabetic patients
.
Am J Physiol Endocrinol Metab
2007
;
292
:
E871
E883
[PubMed]
69.
Viberti
G
,
Kahn
SE
,
Greene
DA
, et al
.
A diabetes outcome progression trial (ADOPT): an international multicenter study of the comparative efficacy of rosiglitazone, glyburide, and metformin in recently diagnosed type 2 diabetes
.
Diabetes Care
2002
;
25
:
1737
1743
[PubMed]
70.
Aronoff
S
,
Rosenblatt
S
,
Braithwaite
S
,
Egan
JW
,
Mathisen
AL
,
Schneider
RL
.
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
2000
;
23
:
1605
1611
[PubMed]
71.
Lebovitz
HE
,
Dole
JF
,
Patwardhan
R
,
Rappaport
EB
,
Freed
MI
Rosiglitazone Clinical Trials Study Group
.
Rosiglitazone monotherapy is effective in patients with type 2 diabetes
.
J Clin Endocrinol Metab
2001
;
86
:
280
288
[PubMed]
72.
Schernthaner
G
,
Matthews
DR
,
Charbonnel
B
,
Hanefeld
M
,
Brunetti
P
Quartet [corrected] Study Group
.
Efficacy and safety of pioglitazone versus metformin in patients with type 2 diabetes mellitus: a double-blind, randomized trial
.
J Clin Endocrinol Metab
2004
;
89
:
6068
6076
[PubMed]
73.
Charbonnel
BH
,
Matthews
DR
,
Schernthaner
G
,
Hanefeld
M
,
Brunetti
P
QUARTET Study Group
.
A long-term comparison of pioglitazone and gliclazide in patients with Type 2 diabetes mellitus: a randomized, double-blind, parallel-group comparison trial
.
Diabet Med
2005
;
22
:
399
405
[PubMed]
74.
Hanefeld
M
,
Brunetti
P
,
Schernthaner
GH
,
Matthews
DR
,
Charbonnel
BH
QUARTET Study Group
.
One-year glycemic control with a sulfonylurea plus pioglitazone versus a sulfonylurea plus metformin in patients with type 2 diabetes
.
Diabetes Care
2004
;
27
:
141
147
[PubMed]
75.
Matthews
DR
,
Charbonnel
BH
,
Hanefeld
M
,
Brunetti
P
,
Schernthaner
G
.
Long-term therapy with addition of pioglitazone to metformin compared with the addition of gliclazide to metformin in patients with type 2 diabetes: a randomized, comparative study
.
Diabetes Metab Res Rev
2005
;
21
:
167
174
[PubMed]
76.
Pioglitazone (marketed as Actos, Actoplus Met, and Duetact) information [Internet]. Available from http://www.fda.gov/Drugs/DrugSafety/PostmarketDrugSafetyInformationforPatientsandProviders/ucm109136.htm. Accessed 30 April 2013
77.
Cusi
K
,
Consoli
A
,
DeFronzo
RA
.
Metabolic effects of metformin on glucose and lactate metabolism in noninsulin-dependent diabetes mellitus
.
J Clin Endocrinol Metab
1996
;
81
:
4059
4067
[PubMed]
78.
Natali
A
,
Ferrannini
E
.
Effects of metformin and thiazolidinediones on suppression of hepatic glucose production and stimulation of glucose uptake in type 2 diabetes: a systematic review
.
Diabetologia
2006
;
49
:
434
441
[PubMed]
79.
Bays
H
,
Mandarino
L
,
DeFronzo
RA
.
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
.
J Clin Endocrinol Metab
2004
;
89
:
463
478
[PubMed]
80.
Bays
HE
,
González-Campoy
JM
,
Bray
GA
, et al
.
Pathogenic potential of adipose tissue and metabolic consequences of adipocyte hypertrophy and increased visceral adiposity
.
Expert Rev Cardiovasc Ther
2008
;
6
:
343
368
[PubMed]
81.
Miyazaki
Y
,
He
H
,
Mandarino
LJ
,
DeFronzo
RA
.
Rosiglitazone improves downstream insulin receptor signaling in type 2 diabetic patients
.
Diabetes
2003
;
52
:
1943
1950
[PubMed]
82.
Coletta
DK
,
Sriwijitkamol
A
,
Wajcberg
E
, et al
.
Pioglitazone stimulates AMP-activated protein kinase signalling and increases the expression of genes involved in adiponectin signalling, mitochondrial function and fat oxidation in human skeletal muscle in vivo: a randomised trial
.
Diabetologia
2009
;
52
:
723
732
[PubMed]
83.
McDonald
WGCG
,
Cole
SL
,
Holewa
DD
,
Brightwell-Conrad
AS
,
Kletzien
RF
,
Colca
JR
.
Identification of a mitochondrial target of thiazolidinediones (mTOT)
.
Diabetes
2012
;
61
(
Suppl. 1
):
A28
84.
Colca
VJ
 Jr
,
Adams
WJ
,
Liang
J
,
Zhou
R
,
Orloff
DG
.
Clinical proof of concepts with a prototype mTOT modulating insulin sensitizer
.
Diabetes
2012
;
61
(
Suppl. 1
):
A246
85.
Kashyap
SR
,
DeFronzo
RA
.
The insulin resistance syndrome: physiological considerations
.
Diab Vasc Dis Res
2007
;
4
:
13
19
[PubMed]
86.
Kashyap
SR
,
Roman
LJ
,
Lamont
J
, et al
.
Insulin resistance is associated with impaired nitric oxide synthase activity in skeletal muscle of type 2 diabetic subjects
.
J Clin Endocrinol Metab
2005
;
90
:
1100
1105
[PubMed]
87.
Montagnani
M
,
Chen
H
,
Barr
VA
,
Quon
MJ
.
Insulin-stimulated activation of eNOS is independent of Ca2+ but requires phosphorylation by Akt at Ser(1179)
.
J Biol Chem
2001
;
276
:
30392
30398
[PubMed]
88.
Bajaj
M
,
DeFronzo
RA
.
Metabolic and molecular basis of insulin resistance
.
J Nucl Cardiol
2003
;
10
:
311
323
[PubMed]
89.
Cusi
K
,
Maezono
K
,
Osman
A
, et al
.
Insulin resistance differentially affects the PI 3-kinase- and MAP kinase-mediated signaling in human muscle
.
J Clin Invest
2000
;
105
:
311
320
[PubMed]
90.
Kashyap
SR
,
Belfort
R
,
Berria
R
, et al
.
Discordant effects of a chronic physiological increase in plasma FFA on insulin signaling in healthy subjects with or without a family history of type 2 diabetes
.
Am J Physiol Endocrinol Metab
2004
;
287
:
E537
E546
[PubMed]
91.
Pratipanawatr
W
,
Pratipanawatr
T
,
Cusi
K
, et al
.
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
2001
;
50
:
2572
2578
[PubMed]
92.
Rothman
DL
,
Magnusson
I
,
Cline
G
, et al
.
Decreased muscle glucose transport/phosphorylation is an early defect in the pathogenesis of non-insulin-dependent diabetes mellitus
.
Proc Natl Acad Sci USA
1995
;
92
:
983
987
[PubMed]
93.
Morino
K
,
Petersen
KF
,
Dufour
S
, et al
.
Reduced mitochondrial density and increased IRS-1 serine phosphorylation in muscle of insulin-resistant offspring of type 2 diabetic parents
.
J Clin Invest
2005
;
115
:
3587
3593
[PubMed]
94.
Krook
A
,
Björnholm
M
,
Galuska
D
, et al
.
Characterization of signal transduction and glucose transport in skeletal muscle from type 2 diabetic patients
.
Diabetes
2000
;
49
:
284
292
[PubMed]
95.
Bouzakri
K
,
Roques
M
,
Gual
P
, et al
.
Reduced activation of phosphatidylinositol-3 kinase and increased serine 636 phosphorylation of insulin receptor substrate-1 in primary culture of skeletal muscle cells from patients with type 2 diabetes
.
Diabetes
2003
;
52
:
1319
1325
[PubMed]
96.
Wang
CC
,
Goalstone
ML
,
Draznin
B
.
Molecular mechanisms of insulin resistance that impact cardiovascular biology
.
Diabetes
2004
;
53
:
2735
2740
[PubMed]
97.
Draznin
B
.
Molecular mechanisms of insulin resistance: serine phosphorylation of insulin receptor substrate-1 and increased expression of p85alpha: the two sides of a coin
.
Diabetes
2006
;
55
:
2392
2397
[PubMed]
98.
Hsueh
WA
,
Law
RE
.
Insulin signaling in the arterial wall
.
Am J Cardiol
1999
;
84
:
21J
24J
[PubMed]
99.
Hanley
AJ
,
Williams
K
,
Stern
MP
,
Haffner
SM
.
Homeostasis model assessment of insulin resistance in relation to the incidence of cardiovascular disease: the San Antonio Heart Study
.
Diabetes Care
2002
;
25
:
1177
1184
[PubMed]
100.
Isomaa
B
,
Almgren
P
,
Tuomi
T
, et al
.
Cardiovascular morbidity and mortality associated with the metabolic syndrome
.
Diabetes Care
2001
;
24
:
683
689
[PubMed]
101.
Rutter
MK
,
Meigs
JB
,
Sullivan
LM
,
D’Agostino
RB
 Sr
,
Wilson
PW
.
Insulin resistance, the metabolic syndrome, and incident cardiovascular events in the Framingham Offspring Study
.
Diabetes
2005
;
54
:
3252
3257
[PubMed]
102.
Bonora
E
,
Kiechl
S
,
Willeit
J
, et al
.
Insulin resistance as estimated by homeostasis model assessment predicts incident symptomatic cardiovascular disease in caucasian subjects from the general population: the Bruneck study
.
Diabetes Care
2007
;
30
:
318
324
[PubMed]
103.
Howard
G
,
Bergman
R
,
Wagenknecht
LE
, et al
Insulin Resistance Atherosclerosis Study (IRAS) Investigators
.
Ability of alternative indices of insulin sensitivity to predict cardiovascular risk: comparison with the “minimal model”
.
Ann Epidemiol
1998
;
8
:
358
369
[PubMed]
104.
Mazzone
T
,
Meyer
PM
,
Feinstein
SB
, et al
.
Effect of pioglitazone compared with glimepiride on carotid intima-media thickness in type 2 diabetes: a randomized trial
.
JAMA
2006
;
296
:
2572
2581
[PubMed]
105.
Nissen
SE
,
Nicholls
SJ
,
Wolski
K
, et al
PERISCOPE Investigators
.
Comparison of pioglitazone vs glimepiride on progression of coronary atherosclerosis in patients with type 2 diabetes: the PERISCOPE randomized controlled trial
.
JAMA
2008
;
299
:
1561
1573
[PubMed]
106.
Dormandy
JA
,
Charbonnel
B
,
Eckland
DJ
, et al
PROactive investigators
.
Secondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study (PROspective pioglitAzone Clinical Trial In macroVascular Events): a randomised controlled trial
.
Lancet
2005
;
366
:
1279
1289
[PubMed]
107.
Miyazaki
Y
,
Mahankali
A
,
Wajcberg
E
,
Bajaj
M
,
Mandarino
LJ
,
DeFronzo
RA
.
Effect of pioglitazone on circulating adipocytokine levels and insulin sensitivity in type 2 diabetic patients
.
J Clin Endocrinol Metab
2004
;
89
:
4312
4319
[PubMed]
108.
Bajaj
M
,
Suraamornkul
S
,
Piper
P
, et al
.
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
2004
;
89
:
200
206
[PubMed]
109.
Miyazaki
Y
,
Glass
L
,
Triplitt
C
, et al
.
Effect of rosiglitazone on glucose and non-esterified fatty acid metabolism in Type II diabetic patients
.
Diabetologia
2001
;
44
:
2210
2219
[PubMed]
110.
Abdul-Ghani
MA
,
Muller
FL
,
Liu
Y
, et al
.
Deleterious action of FA metabolites on ATP synthesis: possible link between lipotoxicity, mitochondrial dysfunction, and insulin resistance
.
Am J Physiol Endocrinol Metab
2008
;
295
:
E678
E685
[PubMed]
111.
Belfort
R
,
Mandarino
L
,
Kashyap
S
, et al
.
Dose-response effect of elevated plasma free fatty acid on insulin signaling
.
Diabetes
2005
;
54
:
1640
1648
[PubMed]
112.
Bajaj
M
,
Pratipanawatr
T
,
Berria
R
, et al
.
Free fatty acids reduce splanchnic and peripheral glucose uptake in patients with type 2 diabetes
.
Diabetes
2002
;
51
:
3043
3048
[PubMed]
113.
Kashyap
S
,
Belfort
R
,
Gastaldelli
A
, et al
.
A sustained increase in plasma free fatty acids impairs insulin secretion in nondiabetic subjects genetically predisposed to develop type 2 diabetes
.
Diabetes
2003
;
52
:
2461
2474
[PubMed]
114.
Wellen
KE
,
Hotamisligil
GS
.
Obesity-induced inflammatory changes in adipose tissue
.
J Clin Invest
2003
;
112
:
1785
1788
[PubMed]
115.
Yki-Järvinen
H
.
Thiazolidinediones and the liver in humans
.
Curr Opin Lipidol
2009
;
20
:
477
483
[PubMed]
116.
Després
JP
,
Moorjani
S
,
Lupien
PJ
,
Tremblay
A
,
Nadeau
A
,
Bouchard
C
.
Regional distribution of body fat, plasma lipoproteins, and cardiovascular disease
.
Arteriosclerosis
1990
;
10
:
497
511
[PubMed]
117.
Wang
YX
.
PPARs: diverse regulators in energy metabolism and metabolic diseases
.
Cell Res
2010
;
20
:
124
137
[PubMed]
118.
Patti
ME
,
Butte
AJ
,
Crunkhorn
S
, et al
.
Coordinated reduction of genes of oxidative metabolism in humans with insulin resistance and diabetes: Potential role of PGC1 and NRF1
.
Proc Natl Acad Sci USA
2003
;
100
:
8466
8471
[PubMed]
119.
Puigserver
P
,
Spiegelman
BM
.
Peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC-1 alpha): transcriptional coactivator and metabolic regulator
.
Endocr Rev
2003
;
24
:
78
90
[PubMed]
120.
UK Prospective Diabetes Study (UKPDS) Group
.
Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33)
.
Lancet
1998
;
352
:
837
853
[PubMed]
121.
UK Prospective Diabetes Study (UKPDS) Group
.
Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34)
.
Lancet
1998
;
352
:
854
865
[PubMed]
122.
Brown
JB
,
Conner
C
,
Nichols
GA
.
Secondary failure of metformin monotherapy in clinical practice
.
Diabetes Care
2010
;
33
:
501
506
[PubMed]
123.
Xiang
AH
,
Peters
RK
,
Kjos
SL
, et al
.
Effect of pioglitazone on pancreatic beta-cell function and diabetes risk in Hispanic women with prior gestational diabetes
.
Diabetes
2006
;
55
:
517
522
[PubMed]
124.
Gerstein
HC
,
Yusuf
S
,
Bosch
J
, et al
DREAM (Diabetes REduction Assessment with ramipril and rosiglitazone Medication) Trial Investigators
.
Effect of rosiglitazone on the frequency of diabetes in patients with impaired glucose tolerance or impaired fasting glucose: a randomised controlled trial
.
Lancet
2006
;
368
:
1096
1105
[PubMed]
125.
Knowler
WC
,
Hamman
RF
,
Edelstein
SL
, et al
Diabetes Prevention Program Research Group
.
Prevention of type 2 diabetes with troglitazone in the Diabetes Prevention Program
.
Diabetes
2005
;
54
:
1150
1156
[PubMed]
126.
Buchanan
TA
,
Xiang
AH
,
Peters
RK
, et al
.
Preservation of pancreatic beta-cell function and prevention of type 2 diabetes by pharmacological treatment of insulin resistance in high-risk hispanic women
.
Diabetes
2002
;
51
:
2796
2803
[PubMed]
127.
DeFronzo
RA
,
Tripathy
D
,
Schwenke
DC
, et al
ACT NOW Study
.
Pioglitazone for diabetes prevention in impaired glucose tolerance
.
N Engl J Med
2011
;
364
:
1104
1115
[PubMed]
128.
Zinman
B
,
Harris
SB
,
Neuman
J
, et al
.
Low-dose combination therapy with rosiglitazone and metformin to prevent type 2 diabetes mellitus (CANOE trial): a double-blind randomised controlled study
.
Lancet
2010
;
376
:
103
111
[PubMed]
129.
Lupi
R
,
Del Guerra
S
,
Marselli
L
, et al
.
Rosiglitazone prevents the impairment of human islet function induced by fatty acids: evidence for a role of PPARgamma2 in the modulation of insulin secretion
.
Am J Physiol Endocrinol Metab
2004
;
286
:
E560
E567
[PubMed]
130.
Finegood
DT
,
McArthur
MD
,
Kojwang
D
, et al
.
Beta-cell mass dynamics in Zucker diabetic fatty rats. Rosiglitazone prevents the rise in net cell death
.
Diabetes
2001
;
50
:
1021
1029
[PubMed]
131.
Masuda
K
,
Okamoto
Y
,
Tsuura
Y
, et al
.
Effects of Troglitazone (CS-045) on insulin secretion in isolated rat pancreatic islets and HIT cells: an insulinotropic mechanism distinct from glibenclamide
.
Diabetologia
1995
;
38
:
24
30
[PubMed]
132.
Kim
HI
,
Cha
JY
,
Kim
SY
, et al
.
Peroxisomal proliferator-activated receptor-gamma upregulates glucokinase gene expression in beta-cells
.
Diabetes
2002
;
51
:
676
685
[PubMed]
133.
Moibi
JA
,
Gupta
D
,
Jetton
TL
,
Peshavaria
M
,
Desai
R
,
Leahy
JL
.
Peroxisome proliferator-activated receptor-gamma regulates expression of PDX-1 and NKX6.1 in INS-1 cells
.
Diabetes
2007
;
56
:
88
95
[PubMed]
134.
Higa
M
,
Zhou
YT
,
Ravazzola
M
,
Baetens
D
,
Orci
L
,
Unger
RH
.
Troglitazone prevents mitochondrial alterations, beta cell destruction, and diabetes in obese prediabetic rats
.
Proc Natl Acad Sci USA
1999
;
96
:
11513
11518
[PubMed]
135.
Matsui
J
,
Terauchi
Y
,
Kubota
N
, et al
.
Pioglitazone reduces islet triglyceride content and restores impaired glucose-stimulated insulin secretion in heterozygous peroxisome proliferator-activated receptor-gamma-deficient mice on a high-fat diet
.
Diabetes
2004
;
53
:
2844
2854
[PubMed]
136.
Haataja
L
,
Gurlo
T
,
Huang
CJ
,
Butler
PC
.
Islet amyloid in type 2 diabetes, and the toxic oligomer hypothesis
.
Endocr Rev
2008
;
29
:
303
316
[PubMed]
137.
Huang
CJ
,
Lin
CY
,
Haataja
L
, et al
.
High expression rates of human islet amyloid polypeptide induce endoplasmic reticulum stress mediated beta-cell apoptosis, a characteristic of humans with type 2 but not type 1 diabetes
.
Diabetes
2007
;
56
:
2016
2027
[PubMed]
138.
Lin
CY
,
Gurlo
T
,
Haataja
L
,
Hsueh
WA
,
Butler
PC
.
Activation of peroxisome proliferator-activated receptor-gamma by rosiglitazone protects human islet cells against human islet amyloid polypeptide toxicity by a phosphatidylinositol 3′-kinase-dependent pathway
.
J Clin Endocrinol Metab
2005
;
90
:
6678
6686
[PubMed]
139.
Kulkarni
RN
,
Brüning
JC
,
Winnay
JN
,
Postic
C
,
Magnuson
MA
,
Kahn
CR
.
Tissue-specific knockout of the insulin receptor in pancreatic beta cells creates an insulin secretory defect similar to that in type 2 diabetes
.
Cell
1999
;
96
:
329
339
[PubMed]
140.
van Wijk
JP
,
de Koning
EJ
,
Martens
EP
,
Rabelink
TJ
.
Thiazoldinediones and blood lipids in type 2 diabetes
.
Arterioscler Thromb Vasc Biol
2003
;
23
:
1144
–17
49
141.
Ferrannini
E
,
Betteridge
DJ
,
Dormandy
JA
, et al
.
High-density lipoprotein-cholesterol and not HbA1c was directly related to cardiovascular outcome in PROactive
.
Diabetes Obes Metab
2011
;
13
:
759
764
[PubMed]
142.
Erdmann
E
,
Dormandy
JA
,
Charbonnel
B
,
Massi-Benedetti
M
,
Moules
IK
,
Skene
AM
PROactive Investigators
.
The effect of pioglitazone on recurrent myocardial infarction in 2,445 patients with type 2 diabetes and previous myocardial infarction: results from the PROactive (PROactive 05) Study
.
J Am Coll Cardiol
2007
;
49
:
1772
1780
[PubMed]
143.
Wilcox
R
,
Bousser
MG
,
Betteridge
DJ
, et al
PROactive Investigators
.
Effects of pioglitazone in patients with type 2 diabetes with or without previous stroke: results from PROactive (PROspective pioglitAzone Clinical Trial In macroVascular Events 04)
.
Stroke
2007
;
38
:
865
873
[PubMed]
144.
Dormandy
JA
,
Betteridge
DJ
,
Schernthaner
G
,
Pirags
V
,
Norgren
L
PROactive investigators
.
Impact of peripheral arterial disease in patients with diabetes—results from PROactive (PROactive 11)
.
Atherosclerosis
2009
;
202
:
272
281
[PubMed]
145.
Lincoff
AM
,
Wolski
K
,
Nicholls
SJ
,
Nissen
SE
.
Pioglitazone and risk of cardiovascular events in patients with type 2 diabetes mellitus: a meta-analysis of randomized trials
.
JAMA
2007
;
298
:
1180
1188
[PubMed]
146.
Ryder
REJ
.
Pioglitazone: an agent which reduces stroke, myocardial infarction and death and is also a key component of the modern paradigm for the optimum management of type 2 diabetes
.
Brit J Diabetes Vasc Disease
2011
;
11
:
113
120
147.
Sears
DD
,
Hsiao
A
,
Ofrecio
JM
,
Chapman
J
,
He
W
,
Olefsky
JM
.
Selective modulation of promoter recruitment and transcriptional activity of PPARgamma
.
Biochem Biophys Res Commun
2007
;
364
:
515
521
[PubMed]
148.
Musso
G
,
Cassader
M
,
Rosina
F
,
Gambino
R
.
Impact of current treatments on liver disease, glucose metabolism and cardiovascular risk in non-alcoholic fatty liver disease (NAFLD): a systematic review and meta-analysis of randomised trials
.
Diabetologia
2012
;
55
:
885
904
[PubMed]
149.
Belfort
R
,
Harrison
SA
,
Brown
K
, et al
.
A placebo-controlled trial of pioglitazone in subjects with nonalcoholic steatohepatitis
.
N Engl J Med
2006
;
355
:
2297
2307
[PubMed]
150.
Bajaj
M
,
Suraamornkul
S
,
Pratipanawatr
T
, et al
.
Pioglitazone reduces hepatic fat content and augments splanchnic glucose uptake in patients with type 2 diabetes
.
Diabetes
2003
;
52
:
1364
1370
[PubMed]
151.
Aithal
GP
,
Thomas
JA
,
Kaye
PV
, et al
.
Randomized, placebo-controlled trial of pioglitazone in nondiabetic subjects with nonalcoholic steatohepatitis
.
Gastroenterology
2008
;
135
:
1176
1184
[PubMed]
152.
Slama
L
,
Lanoy
E
,
Valantin
MA
, et al
.
Effect of pioglitazone on HIV-1-related lipodystrophy: a randomized double-blind placebo-controlled trial (ANRS 113)
.
Antivir Ther
2008
;
13
:
67
76
[PubMed]
153.
Ratziu
V
,
Charlotte
F
,
Bernhardt
C
, et al
LIDO Study Group
.
Long-term efficacy of rosiglitazone in nonalcoholic steatohepatitis: results of the fatty liver improvement by rosiglitazone therapy (FLIRT 2) extension trial
.
Hepatology
2010
;
51
:
445
453
[PubMed]
154.
McCarthy
KJ
,
Routh
RE
,
Shaw
W
,
Walsh
K
,
Welbourne
TC
,
Johnson
JH
.
Troglitazone halts diabetic glomerulosclerosis by blockade of mesangial expansion
.
Kidney Int
2000
;
58
:
2341
2350
[PubMed]
155.
Yoshimoto
T
,
Naruse
M
,
Nishikawa
M
, et al
.
Antihypertensive and vasculo- and renoprotective effects of pioglitazone in genetically obese diabetic rats
.
Am J Physiol
1997
;
272
:
E989
E996
[PubMed]
156.
Sarafidis
PA
,
Bakris
GL
.
Protection of the kidney by thiazolidinediones: an assessment from bench to bedside
.
Kidney Int
2006
;
70
:
1223
1233
[PubMed]
157.
Sarafidis
PA
,
Stafylas
PC
,
Georgianos
PI
,
Saratzis
AN
,
Lasaridis
AN
.
Effect of thiazolidinediones on albuminuria and proteinuria in diabetes: a meta-analysis
.
Am J Kidney Dis
2010
;
55
:
835
847
[PubMed]
158.
Miyazaki
Y
,
Cersosimo
E
,
Triplitt
C
,
DeFronzo
RA
.
Rosiglitazone decreases albuminuria in type 2 diabetic patients
.
Kidney Int
2007
;
72
:
1367
1373
[PubMed]
159.
Nag
S
,
Bilous
R
,
Kelly
W
,
Jones
S
,
Roper
N
,
Connolly
V
.
All-cause and cardiovascular mortality in diabetic subjects increases significantly with reduced estimated glomerular filtration rate (eGFR): 10 years’ data from the South Tees Diabetes Mortality study
.
Diabet Med
2007
;
24
:
10
17
[PubMed]
160.
Schneider
CA
,
Ferrannini
E
,
DeFronzo
R
,
Schernthaner
G
,
Yates
J
,
Erdmann
E
.
Effect of pioglitazone on cardiovascular outcome in diabetes and chronic kidney disease
.
J Am Soc Nephrol
2008
;
19
:
182
187
[PubMed]
161.
Brunelli
SM
,
Thadhani
R
,
Ikizler
TA
,
Feldman
HI
.
Thiazolidinedione use is associated with better survival in hemodialysis patients with non-insulin dependent diabetes
.
Kidney Int
2009
;
75
:
961
968
[PubMed]
162.
Miyazaki
Y
,
Matsuda
M
,
DeFronzo
RA
.
Dose-response effect of pioglitazone on insulin sensitivity and insulin secretion in type 2 diabetes
.
Diabetes Care
2002
;
25
:
517
523
[PubMed]
163.
Aso
Y
,
Hara
K
,
Ozeki
N
, et al
.
Low-dose pioglitazone increases serum high molecular weight adiponectin and improves glycemic control in Japanese patients with poorly controlled type 2 diabetes
.
Diabetes Res Clin Pract
2009
;
85
:
147
152
[PubMed]
164.
Majima
T
,
Komatsu
Y
,
Doi
K
, et al
.
Safety and efficacy of low-dose pioglitazone (7.5 mg/day) vs. standard-dose pioglitazone (15 mg/day) in Japanese women with type 2 diabetes mellitus
.
Endocr J
2006
;
53
:
325
330
[PubMed]
165.
Rajagopalan
R
,
Perez
A
,
Ye
Z
,
Khan
M
,
Murray
FT
.
Pioglitazone is effective therapy for elderly patients with type 2 diabetes mellitus
.
Drugs Aging
2004
;
21
:
259
271
[PubMed]
166.
Perez
A
,
Zhao
Z
,
Jacks
R
,
Spanheimer
R
.
Efficacy and safety of pioglitazone/metformin fixed-dose combination therapy compared with pioglitazone and metformin monotherapy in treating patients with T2DM
.
Curr Med Res Opin
2009
;
25
:
2915
2923
[PubMed]
167.
Panikar
V
,
Joshi
SR
,
Bukkawar
A
,
Nasikkar
N
,
Santwana
C
.
Induction of long-term glycemic control in type 2 diabetic patients using pioglitazone and metformin combination
.
J Assoc Physicians India
2007
;
55
:
333
337
[PubMed]
168.
Einhorn
D
,
Rendell
M
,
Rosenzweig
J
,
Egan
JW
,
Mathisen
AL
,
Schneider
RL
.
Pioglitazone hydrochloride in combination with metformin in the treatment of type 2 diabetes mellitus: a randomized, placebo-controlled study. The Pioglitazone 027 Study Group
.
Clin Ther
2000
;
22
:
1395
1409
[PubMed]
169.
Sarruf
DA
,
Yu
F
,
Nguyen
HT
, et al
.
Expression of peroxisome proliferator-activated receptor-gamma in key neuronal subsets regulating glucose metabolism and energy homeostasis
.
Endocrinology
2009
;
150
:
707
712
[PubMed]
170.
Miyazaki
Y
,
Mahankali
A
,
Matsuda
M
, et al
.
Effect of pioglitazone on abdominal fat distribution and insulin sensitivity in type 2 diabetic patients
.
J Clin Endocrinol Metab
2002
;
87
:
2784
2791
[PubMed]
171.
Bajaj
M
,
Baig
R
,
Suraamornkul
S
, et al
.
Effects of pioglitazone on intramyocellular fat metabolism in patients with type 2 diabetes mellitus
.
J Clin Endocrinol Metab
2010
;
95
:
1916
1923
[PubMed]
172.
Miyazaki
YDE
,
Bajaj
M
,
Wajcberg
E
, et al
.
Predictors of improved glycemic control with rosiglitazone therapy in type 2 diabetic patients: a practical approach for the primary care physician
.
Br J Diabetes Vasc Dis
2005
;
5
:
28
35
173.
Takeda Pharmaceuticals North America. Actos (pioglitazone) [article online], 2007. www.fda.gov/safety/medwatch/safetyinformation/safetyalertsforhumanmedicalproducts/ucm150451.htm. Accessed 30 April 2013
174.
Kahn
SE
,
Haffner
SM
,
Heise
MA
, et al
ADOPT Study Group
.
Glycemic durability of rosiglitazone, metformin, or glyburide monotherapy
.
N Engl J Med
2006
;
355
:
2427
2443
[PubMed]
175.
Betteridge
DJ
.
Thiazolidinediones and fracture risk in patients with Type 2 diabetes
.
Diabet Med
2011
;
28
:
759
771
[PubMed]
176.
Bodmer
M
,
Meier
C
,
Kraenzlin
ME
,
Meier
CR
.
Risk of fractures with glitazones: a critical review of the evidence to date
.
Drug Saf
2009
;
32
:
539
547
[PubMed]
177.
Nesto
RW
,
Bell
D
,
Bonow
RO
, et al
American Heart Association
American Diabetes Association
.
Thiazolidinedione use, fluid retention, and congestive heart failure: a consensus statement from the American Heart Association and American Diabetes Association. October 7, 2003
.
Circulation
2003
;
108
:
2941
2948
[PubMed]
178.
Charbonnel
B
,
DeFronzo
R
,
Davidson
J
, et al
PROactive investigators
.
Pioglitazone use in combination with insulin in the prospective pioglitazone clinical trial in macrovascular events study (PROactive19)
.
J Clin Endocrinol Metab
2010
;
95
:
2163
2171
[PubMed]
179.
Raskin
P
,
Rendell
M
,
Riddle
MC
,
Dole
JF
,
Freed
MI
,
Rosenstock
J
Rosiglitazone Clinical Trials Study Group
.
A randomized trial of rosiglitazone therapy in patients with inadequately controlled insulin-treated type 2 diabetes
.
Diabetes Care
2001
;
24
:
1226
1232
[PubMed]
180.
Hanefeld
M
,
Pfützner
A
,
Forst
T
,
Kleine
I
,
Fuchs
W
.
Double-blind, randomized, multicentre, and active comparator controlled investigation of the effect of pioglitazone, metformin, and the combination of both on cardiovascular risk in patients with type 2 diabetes receiving stable basal insulin therapy: the PIOCOMB study
.
Cardiovasc Diabetol
2011
;
10
:
65
[PubMed]
181.
Mudaliar
S
,
Chang
AR
,
Henry
RR
.
Thiazolidinediones, peripheral edema, and type 2 diabetes: incidence, pathophysiology, and clinical implications
.
Endocr Pract
2003
;
9
:
406
416
[PubMed]
182.
Guan
Y
,
Hao
C
,
Cha
DR
, et al
.
Thiazolidinediones expand body fluid volume through PPARgamma stimulation of ENaC-mediated renal salt absorption
.
Nat Med
2005
;
11
:
861
866
[PubMed]
183.
Karalliedde
J
,
Buckingham
R
,
Starkie
M
,
Lorand
D
,
Stewart
M
,
Viberti
G
Rosiglitazone Fluid Retention Study Group
.
Effect of various diuretic treatments on rosiglitazone-induced fluid retention
.
J Am Soc Nephrol
2006
;
17
:
3482
3490
[PubMed]
184.
Erdmann
E
,
Charbonnel
B
,
Wilcox
RG
, et al
PROactive investigators
.
Pioglitazone use and heart failure in patients with type 2 diabetes and preexisting cardiovascular disease: data from the PROactive study (PROactive 08)
.
Diabetes Care
2007
;
30
:
2773
2778
[PubMed]
185.
van der Meer
RW
,
Rijzewijk
LJ
,
de Jong
HW
, et al
.
Pioglitazone improves cardiac function and alters myocardial substrate metabolism without affecting cardiac triglyceride accumulation and high-energy phosphate metabolism in patients with well-controlled type 2 diabetes mellitus
.
Circulation
2009
;
119
:
2069
2077
[PubMed]
186.
Sourij
H
,
Zweiker
R
,
Wascher
TC
.
Effects of pioglitazone on endothelial function, insulin sensitivity, and glucose control in subjects with coronary artery disease and new-onset type 2 diabetes
.
Diabetes Care
2006
;
29
:
1039
1045
[PubMed]
187.
Lewis
JD
,
Ferrara
A
,
Peng
T
, et al
.
Risk of bladder cancer among diabetic patients treated with pioglitazone: interim report of a longitudinal cohort study
.
Diabetes Care
2011
;
34
:
916
922
[PubMed]
188.
Ferrara
A
,
Lewis
JD
,
Quesenberry
CP
 Jr
, et al
.
Cohort study of pioglitazone and cancer incidence in patients with diabetes
.
Diabetes Care
2011
;
34
:
923
929
[PubMed]
189.
Cohort study of pioglitazone and bladder cancer in patients with type II diabetes. Available from http://www.clinicaltrials.gov/ct2/show/nct01637935?term=kpnc&rank=1. Accessed 30 April 2013
190.
Dormandy
J
.
PROactive study
.
Lancet
2006
;
367
:
26
27
[PubMed]
191.
Chang
C-H
,
Lin
J-W
,
Wu
L-C
,
Lai
M-S
,
Chuang
L-M
,
Chan
KA
.
Association of thiazolidinediones with liver cancer and colorectal cancer in type 2 diabetes mellitus
.
Hepatology
2012
;
55
:
1462
1472
[PubMed]
192.
van Staa
TP
,
Patel
D
,
Gallagher
AM
,
de Bruin
ML
.
Glucose-lowering agents and the patterns of risk for cancer: a study with the General Practice Research Database and secondary care data
.
Diabetologia
2012
;
55
:
654
665
[PubMed]
193.
Betteridge
DJ
,
DeFronzo
RA
,
Chilton
RJ
.
PROactive: time for a critical appraisal
.
Eur Heart J
2008
;
29
:
969
983
[PubMed]
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. See http://creativecommons.org/licenses/by-nc-nd/3.0/ for details.