Open access peer-reviewed chapter

Thiazolidinediones Cause Cardiotoxicity via PPARγ- Independent Mechanism

Written By

Jing-Bo Jiang, James A. Balschi, Francis X. McGowan Jr and Huamei He

Submitted: 17 March 2018 Reviewed: 21 May 2018 Published: 05 November 2018

DOI: 10.5772/intechopen.78957

From the Edited Volume

Cardiotoxicity

Edited by Wenyong Tan

Chapter metrics overview

1,416 Chapter Downloads

View Full Metrics

Abstract

Thiazolidinediones (TZDs), peroxisome proliferator-activated receptor gamma (PPARγ) agonists, are highly effective antidiabetic drugs that are widely used to treat type 2 diabetes mellitus (T2DM) due to their unique beneficial actions, such as a renoprotective effect, amelioration of glucose homeostasis, and blood pressure lowering, that other antidiabetic drugs do not have. Those beneficial actions, however, are shadowed by the increased risks of cardiovascular adverse events, including mitochondrial dysfunction, oxidative stress and myocardial energy deficiency, fluid retention, congestive heart failure, and myocardial infarction. Except PPARγ, TZDs also have affinity to numerous non-PPARγ targets in mitochondria, cytosol, and cytoplasm, including MitoNEET, mitochondrial pyruvate carrier, dehydrogenases involved in tricarboxylic acid cycle and electron transport, cytoplasmic ion channels, Na-K-pump, and other unknown enzymes. By binding to these targets, TZDs produce off-target effects and potentially increase cardiotoxicity. In this chapter, we review recent studies, both experimental and clinical, on the myocardial adverse effects associated with TZDs and their underlying mechanisms. We focus our review in large part on the relationship between these myocardial adverse effects and PPARγ.

Keywords

  • thiazolidinedione
  • myocardial energy metabolism
  • mitochondria
  • oxidative stress
  • peroxisome proliferator-activated receptors
  • heart failure
  • myocardial infarction

1. Introduction

Thiazolidinediones (TZDs) including ciglitazone, pioglitazone, rosiglitazone, and troglitazone, also known as glitazones after the prototypical drug ciglitazone, are a class of heterocyclic compounds consisting of a five-membered C3NS ring. Among them, pioglitazone and rosiglitazone were approved for clinical use in the United States and Canada. Through activation of peroxisome proliferator-activated receptor gamma (PPARγ), these compounds improve insulin sensitivity, reduce hyperglycemia, and afford unique beneficial actions, such as a renoprotective effect and blood pressure lowering that other antidiabetic drugs do not have [1]. Therefore, they have been widely used to treat type 2 diabetes mellitus (T2DM) as monotherapy or in combination with other types of oral antidiabetic agents (sulfonylureas, metformin, and acarbose). Their original approvals were based on the ability to reduce insulin resistance, increase peripheral glucose utilization, and decrease hepatic glycogen output, accordingly, lower blood glucose concentration [2]. TZDs provide robust improvement in glycemic control that is comparable to other established agents, such as metformin and the sulfonylureas [3, 4]. More importantly, since the progressive failure and loss of β-cells are ultimately responsible for the onset and progression of T2DM, the potential of TZDs to preserve β-cells is an extremely desirable function in glucose-lowing medicine [5, 6, 7]. According to ADOPT (A diabetes Outcome Prevention Study), the rate of monotherapy failure with TZDs is lower than other antidiabetic agents such as metformin and glyburide [8].

The ultimate value of TZDs and any other glucose-lowing drugs should rely on not only the improvement of acute hyperglycemic crises and their serious consequences, but also the reduction of long-term complications associated with diabetes. Theoretically, reducing hyperglycemia over the long term should decrease the possibility of the complications, but this is not the case for rosiglitazone. Instead, its beneficial actions are shadowed by the increased risks of cardiovascular adverse events [9].

The coexistence of heart failure (HF) and T2DM is common and has a strong impact on clinical management and prognosis. The action of any antidiabetic therapy on cardiovascular system is particularly important because more than 70% of deaths in diabetic patients are from cardiovascular causes [10], and clinical courses of cardiovascular events and T2DM frequently progress in parallel [11, 12]. Glucose-lowering drugs, including the TZDs, have complex organ-specific effects on diverse biological processes that may determine the effects on cardiovascular events end point. Unfortunately, the potential for unexpected cardiovascular side effects when rosiglitazone is administered to patients was not fully assessed before the approval from U.S. Food and Drug Administration (FDA) in 1999 and from the European Medicines Evaluation Agency (EMEC) in 2000. According to over 40 clinical trials conducted from 1999 to 2007, rosiglitazone has been reported to increase risks of heart failure [13, 14] and myocardial infarction in T2DM patients [15, 16, 17]. Additionally, rosiglitazone was associated with a significant increase in the risk of death from cardiovascular causes that had borderline significance [13, 16, 18, 19]. Rosiglitazone may also worsen the clinical course in patients with pre-existing left ventricular dysfunction [20]. Approximately 10 years after the introduction of rosiglitazone, EMEC required two post-marketing studies on long-term adverse effects and recommended that rosiglitazone be suspended from the European market because the benefits no longer outweighed the risk. Similarly, pioglitazone therapy was also associated with an increased risk of major adverse cardiovascular events in patients with pre-diabetes or insulin resistance and diabetes [21]. In this chapter, we review recent studies, both experimental and clinical, on the myocardial adverse effects associated with TZDs, rosiglitazone in particular, and discuss their underlying mechanisms. We focus our discussion in large part on the relationship between these myocardial adverse effects and PPARγ.

Advertisement

2. On-target effects of TZDs

Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors of nuclear hormone receptor superfamily comprising three subtypes such as PPARα, PPARβ/δ, and PPARγ [22]. PPARα is predominantly expressed in metabolically active tissues such as the adipose tissue, liver, heart, kidney and skeletal muscle [23], and mainly influences fatty acid metabolism and its activation lowers lipid levels [24]. PPARβ/δ is ubiquitously expressed with the highest levels in the intestine, colon, skin, adipose tissue, skeletal muscle and brain and kidney [25] and is involved in fatty acid oxidation in muscle [26]. PPARγ is mostly expressed in adipose tissue, but also found in the skeletal muscle, liver, kidney, colon, intestine, pancreas, brain, immune cells, and retina and throughout the cardiovascular system at relatively low levels [24, 25, 27]. Activation of PPARγ causes lipogenesis, adipocyte differentiation, and insulin sensitization and enhances glucose metabolism and also decreases plasma free fatty acid level [26]. Functionally, this receptor controls the expression of networks of genes involved in adipogenesis, lipid and glucose metabolism, inflammation and maintenance of metabolic homeostasis. TZDs act by activating PPARγ. When activated, PPARγ and retinoid X receptor (RXR) form heterodimeric complex PPARγ /RXR, which then binds to a specific DNA sequence element termed peroxisome proliferator response element (PPRE), increasing transcription of various involved genes and decreasing transcription of others [28, 29]. The major effects of expression and repression of the aforementioned genes are to increase the storage of fatty acids in adipocytes, thereby decreasing the amount of fatty acids present in circulation. Accordingly, cells become more dependent on the oxidation of carbohydrates, more specifically glucose, in order to yield energy for other cellular processes. Therefore, TZDs improve insulin sensitivity and reduce hyperglycemia [30]. PPARγ on-target effects of TZDs are summarized in Table 1.

Cells Target genes Gene expression On-target effects Reference
Adipocyte Acetyl-CoA synthetase ↑ triglyceride [31]
Adipocyte Adipocyte-specific fatty acid binding protein ↑ lipid oxidation [32]
Adipocyte Cbl-associated protein ↑ insulin sensitivity [33]
Adipocyte Fatty acid translocase ↑ fatty acid uptake [34]
Hepatocyte, β-cell Glucokinase ↑ glucose homeostasis [35, 36]
Hepatocyte, β-cell Glucose transporter 2 ↑ glucose sensing [35]
Adipocyte Glucose transporter 4 ↑ glucose uptake [37]
Adipocyte Glycerol kinase ↓ free fatty acid [38, 39]
Adipocyte Insulin receptor substrate 2 ↑ insulin sensitivity [40]
Adipocyte, hepatocyte Interleukin-6 ↑ insulin sensitivity [41, 42]
Adipocyte Leptin ↑ insulin sensitivity [43, 44]
Adipocyte, muscle cell Lipoprotein lipase ↓ triglyceride [45, 46]
Adipocyte Perilipin ↓ free fatty acid [47, 48]
Adipocyte, hepatocyte Phosphoenolpyruvate carboxykinase ↑ triglyceride
↓ lipid oxidation
[30, 32, 49]
Adipocyte, hepatocyte Tumor necrosis factor-α ↑ insulin sensitivity [41]

Table 1.

Selected PPARγ on-target effects of TZDs.

Besides, TZDs selectively augment or partially mimic certain actions of insulin, causing a slowly generated hypoglycemic effect without increasing pancreatic insulin secretion in noninsulin-dependent diabetic patients via the activation of PPARγ, which increases transcription of certain insulin-sensitive genes [2]. Thus, the action of TZDs is often accompanied by a reduction in circulating concentrations of insulin, triglycerides and non-esterified fatty acids while the β-cell function is largely restored. The PPARγ on-target effects of TZDs, however, are still not completely clear.

Advertisement

3. Off-target effects of TZDs

Drugs exert desired and undesired effects based on their binding interactions with protein target(s) and off-target(s), providing evidence for its efficacy and toxicity. Many different and seemingly unrelated side effects have emerged during the development of TZDs, such as fatal hepatotoxicity, rhabdomyolysis, nephrotoxicity, multisystem organ failure, etc. In isolated hearts [50] or in vivo [25], rosiglitazone suppressed Jun NH2-terminal kinase and activated the adenosine monophosphate-activated protein kinase and protein kinase B (AKT) pathways, and these effects could not be fully blocked by a PPARγ antagonist [51]. Similarly, studies in the pig demonstrated rapid effects of troglitazone in the recovery of left ventricular function after ischemia/reperfusion injury [52, 53]. The effect of troglitazone in this model was found to be imparted not by the TZD moiety but by its tocopherol moiety, which does not activate PPARγ [53]. Together these data suggest that in addition to PPARγ-dependent (on-target) effects, TZDs also exert PPARγ-independent (off-target) effects.

Using [3H]pioglitazone, a structurally related iodinated photoaffinity probe, mass spectrometry analysis and amino-terminal sequencing, a 17-kDa mitochondrial protein mitoNEET has been identified as a saturable and specific binding site for [3H]pioglitazone [54]. MitoNEET is broadly expressed in insulin-sensitive tissues including the liver, muscle, adipose, and heart [55]. MitoNEET is an integral iron-sulfur-cluster transfer protein in the outer mitochondrial membrane that has been shown to inhibit mitochondrial iron transport, which may in turn decrease mitochondrial respiratory activity [56, 57], oxidative capacity [55] and redox-sensitive signaling [58]. Overexpressing mitoNEET in adipocytes decreased the levels of reactive oxygen species [57]. In contrast, knocking down mitoNEET in adipocytes increased reactive oxygen species-induced protein damage [57].

Similarly, the mitochondrial pyruvate carrier 2 (Mpc-2) has also been identified as a direct mitochondrial target of the TZDs (mTOT) using photoaffinity and mass spectrometry-based proteomics approaches [59]. Two mTOT-binding TZDs with little effect on PPARγ (MSDC-0160 and MSDC-0602) were shown to enhance brown adipose tissue formation and improve insulin sensitivity in mice, whereas the deletion of the Mpc-2/mTOT gene resulted in a loss of brown adipose tissue formation [59]. A phase IIb study in patients with diabetes suggested that MSDC-0160 may have similar glucose-lowering efficacy to pioglitazone, with preliminary hints of fewer side effects [60]. MSDC-0160 was associated with a lower level of fluid retention [60]. These data suggest that specifically targeting Mpc-2/mTOT may have potential as a therapy for diabetes, and that both on-target and off-target effects may contribute to efficacy of the drugs, but off-target effects potentially increase cardiotoxicity.

Pioglitazone and rosiglitazone possess a common functional core, glitazone, which is considered a privileged scaffold upon which to build a drug selective for a given target-in this case, PPARγ. A retrospective analysis of pioglitazone and rosiglitazone has identified numerous non-PPARγ proteins as high affinity binders of TZDs in the rat heart, including mitochondrial and cytoplasmic dehydrogenases, ion channels, modulators and enzymes involved in glucose homeostasis, mitochondrial energy production and synaptic transduction [61].

Defining the off-target effects of TZDs and determining whether their cardiovascular adverse effects are mediated through PPARγ-dependent or -independent mechanisms will be critical in developing new therapeutic agents. From this point of view, we discuss recent studies, both experimental and clinical, on the myocardial adverse effects associated with TZDs, particularly rosiglitazone and their underlying mechanisms, focusing in large part on PPARγ-independent (off-target) mechanism in the following context.

Advertisement

4. Rosiglitazone causes myocardial energy deficiency and mitochondrial dysfunction via PPARγ-independent mechanism

Using 31P-nuclear magnetic resonance (NMR) spectroscopy, we measured intracellular phosphocreatine (PCr), adenosine triphosphate (ATP), and calculated free energy of ATP hydrolysis (ΔGATP) in isolated beating hearts perfused in Langendorff mode with regular Krebs-Henseleit buffer containing 10 mM glucose and 0.5 mM pyruvate. At baseline, all hearts from cardiomyocyte-specific PPARγ deficient (PPARγ−/−) mice and their littermate control (PPARγ+/+) mice showed similar PCr and ATP resonance areas, and PCr/ATP ratio, indicating the loss of regulatory action of cardiomyocyte PPARγ on myocardial energy metabolism can be compensated in vivo. At the human therapeutic concentrations of 1 and 3 μM, rosiglitazone showed no marked effects on the resonance areas and concentrations of intracellular PCr ([PCr]) and ATP ([ATP]). At the supratherapeutic concentrations of 10 and 30 μM, however, rosiglitazone decreased myocardial [PCr], [ATP], and ΔGATP in both PPARγ−/− and PPARγ+/+ mice in parallel compared with their vehicle controls [62]. To confirm the results from 31P-NMR spectroscopy, we freeze-clamped hearts from those mice at the end of each experiment and then measured total ATP, ADP, AMP content using HPLC and calculated energy charge. Consistent with the abovementioned results, total ATP content, ATP to ADP ratio and energy charge decreased following acute treatment with rosiglitazone at 10 and 30 μM in hearts from both PPARγ−/− and PPARγ+/+ mice compared with vehicle control [62].

Since mitochondrial oxidation of fatty acid and glucose is a major source of ATP in cardiomyocytes, we measured glucose and palmitate oxidation rates in fresh tissue homogenates using [1-14C]-glucose and [1-14C]-palmitic acid, respectively. At the therapeutic concentrations of 1 and 3 μM, incubation of rosiglitazone with myocardial homogenates for 60 min did not change glucose and palmate oxidation rates. At the supratherapeutic concentrations of 10 and 30 μM, however, it decreased oxidation rates of glucose and palmitate in myocardial homogenate from both PPARγ−/− and PPARγ+/+ mice to the same extent. Consistently, rosiglitazone decreased also mitochondrial respiration rate at these supratherapeutic concentrations in both homogenates [62].

We then determined the effects of rosiglitazone on both mitochondrial and cytosolic rate-limiting enzymes controlling ATP synthesis. When incubated with fresh tissue homogenate or isolated mitochondria for 60 min, rosiglitazone at 1 and 3 μM did not affect the activities of cytosolic and mitochondrial enzymes tested as compared with vehicle treatment. At the supratherapeutic concentrations of 10 and 30 μM, however, rosiglitazone decreased the activities of myocardial mitochondrial complexes I and IV in both PPARγ−/− and PPARγ+/+ mice to the same extent, but did not alter the activities of other mitochondrial enzymes citrate synthase, creatine kinase, Complexes II, III, V and cytosolic enzymes phosphofructokinase, lactate dehydrogenase and glyceraldehyde 3-phosphate dehydrogenase [62]. These results indicate that the higher concentrations of rosiglitazone caused myocardial energy deficiency and mitochondrial dysfunction in the cardiomyocytes in a PPARγ-independent manner. Consistent with our study, Brunmair et al. reported that 10–100 μM TZDs rosiglitazone, troglitazone and pioglitazone inhibited mitochondrial complex I activity, respiratory control and glucose oxidation in the rat liver and skeletal muscles [63]; Rachek et al. found that troglitazone induced mitochondrial dysfunction and cell death in human hepatocytes [64]; results from Scatena et al. also suggested that TZDs induced a non-PPARγ-mediated effect: mitochondrial respiratory chain dysfunction [65].

The PPARγ-independence of rosiglitazone-induced energy deficiency and mitochondrial dysfunction are also supported by the following evidences: (1) Treatment with PPARγ agonist medium-chain triglyceride decanoic acid improved mitochondrial function as evidenced by increases in mitochondrial number, activities of mitochondrial enzyme citrate synthase, complex I, and catalase [66], whereas treatment with PPARγ agonist rosiglitazone induced mitochondrial dysfunction, suggesting rosiglitazone likely induces the mitochondrial dysfunction via PPARγ-independent mechanism [62]. (2) PPARγ-dependent effects are based upon altered transcription of genes involved in energy metabolism and usually require hours to days to take into effect. The myocardial energy deficiency was observed after short time (30–60 min) exposure to rosiglitazone in our study [62]. Such an acute treatment generally does not allow gene expression to change after transcriptional activation of PPARγ, indicating rosiglitazone likely induced myocardial energy deficiency via PPARγ-independent mechanism.

To rule out the possibility that rosiglitazone caused myocardial energy deficiency and mitochondrial dysfunction through activation of PPARγ in other cardiac cells including fibroblast, smooth muscle cells and endothelial cells, we examined the effects of GW9662, a specific PPARγ antagonist on the detrimental actions of rosiglitazone on myocardial energy metabolism and mitochondrial function. We found that perfusion of hearts from C57BL/6 mice with 10 μM GW9662 for 60 min affected neither total ATP content, nor ATP/ADP ratio, nor energy charge. This antagonist did not reverse the decreases in total ATP content, ATP/ADP ratio and energy charge induced by rosiglitazone at 10 μM in those hearts. Furthermore, 10 μM GW9662 showed no effects on the oxidation rates of glucose and palmitate, mitochondrial respiration rate, or the activities of mitochondrial complexes I and IV, it did not antagonize the downregulations of those parameters by rosiglitazone at the supratherapeutic concentration of 10 μM, either [62]. Additionally, treatments with rosiglitazone at the supratherapeutic concentrations of 10 and 30 μM for 60 min significantly decreased intracellular ATP content in cultured mouse cardiomyocytes. In contrast, treatment with 10 μM rosiglitazone showed no effect on intracellular ATP content in cultured mouse cardiac fibroblasts, treatment with 30 μM rosiglitazone only slightly decreased intracellular ATP content in these fibroblasts. Interestingly, pretreatment with 30 μM GW9662 did not prevent the decreases in intracellular ATP content induced by rosiglitazone in these cultured cardiomyocytes or cardiac fibroblasts [62]. These results further support that rosiglitazone induces myocardial energy deficiency via PPARγ-independent mechanism.

To maintain energy homeostasis, the capacities of ATP synthesis by mitochondrial oxidative phosphorylation, glycolysis, and phosphotransferase (i.e., creatine kinase, CK) reactions must match the demand for ATP utilization by the sarcomere, ion pumps, etc. [67]. Therefore, increased ATP utilization and decreased ATP synthesis, singly or in combination, can cause energy deficiency. The free energy of ATP hydrolysis GATP decreased following rosiglitazone treatment. Furthermore, heart mechanical work (assessed by rate pressure product, an indirect index of calcium cycling, metabolic demand, and ATP utilization) also decreased following acute treatment with rosiglitazone at 10–30 μM. These results suggest that decreased ATP synthesis may be responsible for myocardial energy deficiency induced by rosiglitazone. The main pathways for ATP synthesis in hearts are glycolysis, phosphoryltransfer reactions, and substrate oxidative phosphorylation. Rosiglitazone showed no effect on glycolytic rate-limiting enzymes and the product of CK activity and total creatine content [62], indicating that neither glycolysis nor phosphoryltransfer reaction is likely to be involved in rosiglitazone-induced myocardial energy deficiency.

The inhibition of complex I by rosiglitazone caused impaired oxidation of NADH and in turn decreased NAD content. As a result, NADH/NAD ratio increased. The impaired oxidation of NADH leads to decreased substrate oxidation and in turn decreased ATP synthesis. Complex IV acts as the terminus of mitochondrial electron transport by accepting four electrons to reduce a single oxygen molecule. The reaction is coupled with the transfer of four protons across the mitochondrial membrane, driving ATP synthesis. Thus, the inhibition of both complexes I and IV by rosiglitazone reduces ATP synthesis, which manifests as the myocardial energy deficiency induced by rosiglitazone.

Advertisement

5. Rosiglitazone induces myocardial mitochondrial oxidative stress via PPARγ-independent mechanism

To assess the in vitro effects of rosiglitazone on redox homeostasis, we determined enzyme (NADPH oxidase, xanthine oxidase and mitochondrial complexes I and III)-dependent reactive oxygen species (ROS) O2 production, the capacity of ROS elimination systems including superoxide dismutase (SOD), reduced glutathione (GSH), glutathione peroxidase and catalase, and biomarkers malondialdehyde (MDA), protein carbonyl and 8-hydroxy-2’-deoxyguanosine (8HOdG) of oxidative damage to lipids, proteins and DNAs, respectively, in isolated mitochondria and nuclei. We found that at 1 and 3 μM, rosiglitazone showed no effects on any of the aforementioned parameters. At 10 and 30 μM, however, rosiglitazone increased mitochondrial complexes I- and III-dependent O2 production, decreased the level of mitochondrial GSH and SOD activity, and increased the levels of mitochondrial MDA, protein carbonyl and 8-OHdG [62]. Interestingly, pretreatment with 30 μM GW9662 did not prevent rosiglitazone-induced changes in the above redox parameters. Furthermore, even at the supratherapeutic concentrations of 10 and 30 μM, rosiglitazone did not affect the activities of catalase and glutathione peroxidase, and changed neither the level of nuclear protein carbonyl nor the level of nuclear 8-OHdG [62]. Similar to our study, rosiglitazone at 50 and 60 μM induced apoptosis via oxidative stress in cultured H9c2 cells [68].

We also assessed the acute effects of rosiglitazone on mitochondrial oxidative stress in vivo. At 1 mg/kg, injection of rosiglitazone into mouse tail vein showed no effect on the levels of myocardial mitochondrial MDA, protein carbonyl and 8-OHdG. At 10 mg/kg, however, rosiglitazone increased the levels of these mitochondrial oxidative stress markers. Importantly, injection of antioxidant N-acetyl-L-cysteine 600 mg/kg into mouse tail vein prevented the above rosiglitazone-induced changes of mitochondrial oxidative stress markers in vivo [62]. Furthermore, intravenous injection of GW9662 at 1 mg/kg, previously demonstrated to interact selectively with PPARγ, acting as a potent and full PPARγ antagonist, did not prevent 10 mg/kg rosiglitazone induced myocardial oxidative stress [62]. Taken together, our in vitro and in vivo data support that rosiglitazone induces myocardial mitochondrial oxidative stress via PPARγ-independent mechanism, possibly by decreasing mitochondrial ROS-scavenging capacity.

Advertisement

6. Rosiglitazone causes cardiac dysfunction via PPARγ-independent mechanism

Normal cardiac contractile function requires energy homeostasis. As we found that rosiglitazone caused energy deficiency, we therefore further determined the effects of rosiglitazone on cardiac function. In ex vivo Langendorff-perfused hearts, treatment with rosiglitazone at 1 and 3 μM for 24–30 min showed no obvious effects on cardiac systolic function as assessed by left ventricular systolic pressure (LVSP) and the rate of tension development (+dP/dt). Treatment with rosiglitazone at 10 and 30 μM for 24–30 min, however, decreased LVSP and + dP/dt in hearts from C57BL/6, PPARγ−/− and PPARγ+/+ mice, indicating acute treatment with rosiglitazone at the supratherapeutic concentrations causes cardiac systolic dysfunction [62]. Similarly, treatment with rosiglitazone at 1 and 3 μM for 24–30 min showed no obvious effects on cardiac diastolic function as assessed by left ventricular end diastolic pressure (EDP) and the rate of relaxation (–dP/dt). Treatment with rosiglitazone at 10 and 30 μM for 24–30 min, however, increased EDP and decreased –dP/dt in all hearts from the above three genotypes, indicating acute treatment with rosiglitazone at the supratherapeutic concentrations also causes cardiac diastolic dysfunction [62]. Interestingly, rosiglitazone-induced cardiac dysfunction was not distinguishable among C57BL/6, PPARγ−/− and PPARγ+/+ mice, indicating acute rosiglitazone treatment caused cardiac dysfunction independently of cardiomyocyte PPARγ [62]. Additionally, treatment of hearts with 10 μM GW9662 for 60 min, affected neither cardiac function, nor rosiglitazone-induced cardiac dysfunction. In contrast, treatment of hearts with 20 mM N-acetyl-L-cysteine (NAC) for 60 min did not affect baseline cardiac function, but prevented cardiac dysfunction induced by rosiglitazone at the supratherapeutic concentration of 10 μM [62]. These data further support that rosiglitazone induced cardiac dysfunction via a mechanism related to oxidative stress and independent of PPARγ.

We also evaluated the side effects of rosiglitazone on cardiac function in vivo setting by using echocardiography. Injection of rosiglitazone at the dose of 1 mg/kg into mouse tail vein showed no effect on cardiac function as assessed by fraction shorting [29] and ejection fraction EF). At 10 mg/kg, however, rosiglitazone decreased FS and EF, indicating rosiglitazone caused cardiac dysfunction at a higher dose. NAC at 600 mg/kg alone showed no effect on cardiac function. In combination with 10 mg/kg rosiglitazone, however, this antioxidant prevented rosiglitazone-induced cardiac dysfunction. In contrast, intravenous injection of PPARγ selective antagonist GW9662 at 1 mg/kg did not prevent 10 mg/kg rosiglitazone-induced cardiac dysfunction [62]. These in vivo studies also support that rosiglitazone induces cardiac dysfunction via a mechanism related to oxidative stress and independent of PPARγ.

Advertisement

7. TZDs induce cardiac hypertrophy via PPARγ-independent mechanism

TZDs are expected to inhibit cardiomyocyte growth in vitro and in pressure overload models via activation of PPARγ. Paradoxically, TZDs have also been reported to induce cardiac hypertrophy in mice, rats and dogs [69, 70]. This side effect may occur because TZDs expand blood volume. However, an essential question is whether or not this effect is directly attributable to cardiac PPARγ activation. Treatment with TZD rosiglitazone 10 mg/kg per day for 4 weeks induced cardiac hypertrophy in both PPARγ−/− and PPARγ+/+ mice. Rosiglitazone treatment increased cardiac phosphorylation of p38 mitogen-activated protein kinase (p38-MAPK), a MAPK pathway essential for cardiac hypertrophy, in PPARγ−/− mice. The effect of rosiglitazone on p38-MAPK persisted in PPARγ−/− mouse hearts indicated that activation of p38-MAPK by TZDs is independent of cardiomyocyte PPARγ [70]. Furthermore, phosphorylation of c-Jun N-terminal kinases was not affected by rosiglitazone or cardiomyocyte PPARγ deletion. Surprisingly, despite hypertrophy, AKT phosphorylation was suppressed in PPARγ−/− mouse hearts [70]. These data demonstrate that cardiomyocyte PPARγ suppresses cardiac growth and embryonic gene expression and inhibits nuclear factor κB activity in vivo, and that rosiglitazone causes cardiac hypertrophy at least partially independent of PPAR-γ in cardiomyocytes [70].

Advertisement

8. TZDs increase risks of heart failure

Congestive heart failure (CHF) is a major complication of diabetes and occurs as a result of both atherosclerotic coronary disease and non-ischemic diabetic cardiomyopathy. TZDs improve glycemic control and afford beneficial effects on many markers of cardiovascular risk including blood pressure, waist to hip ratio, HDL levels, endothelial reactivity, C-reactive protein, fibrinolysis, and microalbuminuria by improving peripheral insulin sensitivity. These antidiabetic agents, however, have been reported to worsen the existing CHF or precipitate new-onset failure in several reviews and meta-analyses of placebo-controlled randomized clinical trials (RCTs).

Bolen et al. found that the risk for CHF was higher with TZDs as either monotherapy or combination therapy than with metformin or sulfonylureas, with a range of 0.8–3.6% for TZDS and 0–2.6% for non-TZDs [4]. Lago et al. found an increased risk of CHF in use of TZDs in patients with diabetes and prediabetes compared with placebo and active-controls: relative risk 1.72, 95% confidence interval (CI) 1.21–2.42. The overall event rate for CHF with TZDs was 2.3% and with the comparison drugs 1.4% [14]. Singh et al. reported that the relative risk of CHF in use of rosiglitazone in patients with diabetes or prediabetes compared with various other antidiabetic drugs was 2.09 (95% CI 1.52–2.88) [17]. They also examined onset of CHF in both pioglitazone and rosiglitazone compared with placebo in three randomized controlled trials with subjects with either type 2 diabetes or prediabetes. The odds ratio (OR) for all heart failure adverse events was 2.10 (95% CI 1.08–4.08). Four observational studies produced an OR 1.55 (95% CI 1.33–1.80). These authors also examined case reports, including 162 case subjects with 99 analyzable cases. Among these cases, the median time to onset of CHF was 24 weeks, although failure could occur early and did not appear to relate to dosage. CHF was not limited to the elderly; 26% of cases were in subjects less than 60 years of age [71]. Hernandez et al. found that the TZD therapy was significantly and consistently associated with a higher risk of CHF: TZDs 360/6807 [5.3%] versus placebo 234/6328 [3.7%], OR 1.59; 95% CI 1.34–1.89; p < 0.00001. The risk of CHF was higher with rosiglitazone than with pioglitazone (OR 2.73; 95% CI 1.46–5.10) versus (OR 1.51; 95% CI 1.26–1.81; p = 0.06). Rosiglitazone and pioglitazone were associated with a similar risk of serious/severe CHF (OR 1.47; 95% CI 1.16–1.87; p = 0.002). The use of TZDs was also associated with edema (OR 2.04; 95% CI 1.85–2.26; p < 0.00001) [72]. The above increased risk of CHF was largely confirmed in other meta-analyses: the use of rosiglitazone for >4 weeks in 132 trials involving 41,743 patients with or without T2DM was associated with a 69% higher relative risk of serious CHF [73]; and the combined short- and long-term use of pioglitazone in 19 RCTs involving 16,390 patients with T2DM found a 41% higher relative risk of serious CHF [74]. Another meta-analysis of 26 RCTs found 126% higher odds of peripheral edema in 15,332 diabetics with short- and long-term use of TZDs [75].

One of the potential mechanisms responsible for increased risk in CHF with TZD treatment may be the fluid accumulation observed in large-scale studies on antidiabetic medications [74, 75, 76]. In spite of a weak beneficial effect on blood pressure [77], volume overload beyond a certain threshold induced by TZDs increases the myocardial energy demand of the left ventricular and triggers metabolic disorder. As a compensatory mechanism, the contractile function of myocardia is temporarily restored via cardiac hypertrophy, overtaking the growing amount of mitochondrial respiration and ATP production gradually.

In susceptible individuals, these pathophysiological responses likely explain why rosiglitazone precipitate clinical heart failure, and why ischemic events are easily provoked. Notably, the sodium-retentive actions of rosiglitazone within the renal tubules are dose and duration-dependent and insulin-independent, accordingly, it is likely that concurrent treatment with insulin and rosiglitazone mutually reinforces the risk of each agent, thus markedly increases the possibility of worsening heart failure. The reasons for fluid retention and peripheral edema with TZD use are not fully understood and are likely to be multifactorial. One possibility is the reduction in renal excretion of sodium and an increase in sodium and free water retention. Whether these actions are PPARγ-dependent or not warrants further study.

The other potential mechanism responsible for increased risk in CHF with TZDs treatment may be related to their direct adverse effects on myocardial energy deficiency, mitochondrial function and cardiac function observed in our previous study [62]. It is well known that altered energy metabolism and cardiac dysfunction are common features of heart failure resulted from different causes, including diabetes. We demonstrated that rosiglitazone induced myocardial energy deficiency, mitochondrial dysfunction and cardiac dysfunction in perfused mouse hearts at the supratherapeutic concentrations of 10 and 30 μM and induced cardiac dysfunction in vivo at a high dose of 10 mg/kg [62]. TZDs might be accumulated over a longer period of time in the cell or their effects are in some other way “cumulative” in some patients who need increased doses due to the tolerance during a long period of therapeutic time, and in diabetic patients with renal dysfunction. Therefore, it is likely that TZDs increase the risk of heart failure in T2DM patients through their PPARγ-independent adverse effects on the heart.

Advertisement

9. TZDs increase risks of myocardial infarction

Muraglitazar, an investigational dual PPARα and PPARγ agonist, was the first TZD agent halted because of increased adverse cardiovascular events, including myocardial infarction, transient ischemic attack, and stroke, during phase 2 and 3 trials [78]. In 2007, Nissen and Wolski performed the first large meta-analysis of 42 trials involving 27,847 patients with randomized control group not receiving rosiglitazone and found that rosiglitazone was associated with a significant increase in the risk of myocardial infarction (OR 1.43, 95% CI 1.03–1.98, P = 0.03) and with an increase in the risk of cardiovascular death (OR 1.64, 95% CI 0.98–2.74, P = 0.06) [16]. In 2010, Nissen and Wolski published an update including 56 trials with 35,531 randomized patients: 19,509 who received rosiglitazone and 16,022 who received control therapy. They continued to demonstrate that rosiglitazone therapy significantly increased the risk of myocardial infarction (OR 1.28, 95% CI 1.02–1.63, P = 0.04) [79]. Consistent with above analyses, Ontario study [80] and Taiwan study [81] also reported the increased risks in both myocardial infarction and cardiovascular death following the treatment with rosiglitazone. Several other meta-analyses by Psaty and Furberg, GlaxoSmithKline, U.S. FDA and Singh et al. found the increased risks in myocardial infarction but uncertainty in cardiovascular death in patients with rosiglitazone treatment [17, 82, 83], whereas meta-analysis by Shuster et al. reported the increased risk in cardiovascular death but uncertainty in myocardial infarction in subjects treated with rosiglitazone [84].

In contrast, the meta-analysis by Diamond et al. and Lago et al. reported that rosiglitazone was not associated with an increase in the risk of myocardial infarction and cardiovascular death [14, 85].

These discrepancies can be ascribed to the inconsistencies in trial design, eligibility, follow-up, sample size, analytical methodology, and endpoint criteria among analyses and studies.

The mechanisms responsible for increased risks in myocardial infarction and cardiovascular death related to TZDs are not fully characterized. Several contributing factors are possible: first, the reduction in hemoglobin. TZDs, including rosiglitazone, may produce a modest reduction in the hemoglobin level. In susceptible patients, a reduced hemoglobin level may result in increased physiological stress, thereby provoking myocardial ischemia [16]. The second is adverse effects on serum lipids. TZDs may produce detrimental influences on serum lipids. Rosiglitazone increased low-density lipoprotein cholesterol (LDL-C) concentration of 18.6% among T2DM patients treated for 26 weeks with an 8-mg daily dose via increasing serum paraoxonase activity, which protects LDL-C against lipid peroxidation. This TZD also significantly increased triglyceride levels in 50 patients who were given at 4 mg/day for 3 months in addition to their usual treatment compared to baseline levels [86, 87]. Higher LDL-C level was consistently and independently associated with higher incidences of major adverse cardiovascular events after controlling for conventional risk factors [88]. Third, overload of intravascular volume. TZDs may induce fluid retention and peripheral edema likely via the reduction in renal excretion of sodium and an increase in sodium and free water retention [13]. The volume overload increases stress on the left ventricular wall, a factor that determines myocardial oxygen demand. In susceptible patients, an increase in myocardial oxygen demand could theoretically provoke ischemic events.

Advertisement

10. Summary

There are two TZDs approved for prescription use in the United States: rosiglitazone maleate (Avandia) and pioglitazone hydrochloride (Actos). Both have been widely used to treat adult patients with T2DM, either as monotherapy or in combination with insulin, metformin, or sulfonylurea when diet, exercise, and a single agent does not result in adequate glycemic control. The mechanisms of action of TZDs in lowering plasma glucose among patients with T2DM are thought to include the following: increase insulin sensitivity, decrease endogenous glucose production and postprandial gluconeogenesis, increase fasting and postprandial glucose clearance, and have beneficial effects on beta-cell function. The glycemic effects of these agents are thought to be mediated by binding to PPARγ (Figure 1).

Figure 1.

PPARγ-dependent (on-target) and -independent (off-target) effects of thiazolidinediones (TZDs). TZDs produce on-target effects by binding to nucleus PPARγ, increasing insulin sensitivity and glucose oxidation and contributing to efficacy of the drugs. They produce off-target effects by binding to numerous non-PPARγ targets including MitoNEET, mitochondrial pyruvate carrier (MCP), dehydrogenases involved in TCA cycle and electron transport chain complexes, cytoplasmic ion channels, Na-K-pump and other unknown enzymes. Off-target effects potentially increase cardiotoxicity including mitochondrial (Mito) dysfunction, oxidative stress and myocardial energy deficiency, fluid retention, congestive heart failure and myocardial infarction. Paradoxically, Mito dysfunction and energy deficiency may also stimulate insulin sensitivity and glucose uptake in the heart and indirectly contributing to efficacy of the drugs. A-CoA, acetyl-coenzyme A; TCA, tricarboxylic acid; e−, electron; CI, CII, CIII, CIV and CV, mitochondrial respiratory chain complexes I, II, III, IV and V, respectively; Q, coenzyme Q.

Their efficacy and beneficial effects, however, are shadowed by the increased risks of cardiovascular adverse events. Evidences are accumulating that TZDs, particularly rosiglitazone, cause cardiotoxicity including myocardial energy deficiency, mitochondrial dysfunction, and oxidative stress with concomitant cardiac dysfunction in ex vivo perfused hearts. TZDs may also cause cardiac hypertrophy in whole animal model. Additionally, TZDs increase the risks of heart failure and myocardial infarction in patients with T2DM. Understanding whether the cardiotoxicity induced by TZDs is PPARγ independent or not is an important issue for designing more specific PPARγ agonists with fewer side effects. TZDs also have affinity to numerous non-PPARγ targets in mitochondria, cytosol and cytoplasm, including MitoNEET, mitochondrial pyruvate carrier (MCP), dehydrogenases involved in TCA cycle and electron transport, cytoplasmic ion channels, Na-K-pump and other unknown enzymes. By binding to these non-PPARγ targets, TZDs produce off-target effects and potentially increase cardiotoxicity including mitochondrial dysfunction, oxidative stress and myocardial energy deficiency, fluid retention, congestive heart failure and myocardial infarction. Paradoxically, mitochondrial dysfunction and energy deficiency may also stimulate insulin sensitivity and glucose uptake in the heart and indirectly contributing to efficacy of TZDs. Therefore, TZDs may produce antidiabetic effects via both PPARγ-dependent and PPARγ-independent mechanisms, and they may induce cardiotoxicity solely via PPARγ-independent mechanism (Figure 1). This chapter also raised concerns that the use of TZDs may lead to a significant increase in adverse cardiovascular effects. The benefit/risk profile of TZDs should be considered when treating diabetic patients with or without prior cardiovascular diseases.

Acknowledgments

This work was partially supported by the National Institutes of Health [R01 HL46033 and HL78634 to JAB, P50 HL074734 to FXM, and by Brigham and Women’s Fund 104401 to HH.

Conflict of interest

Authors declare no conflict of interest.

References

  1. 1. Horita S, Nakamura M, Satoh N, Suzuki M, Seki G. Thiazolidinediones and edema: Recent advances in the pathogenesis of thiazolidinediones-induced renal sodium retention. PPAR Research. 2015;2015:646423
  2. 2. Day C. Thiazolidinediones: A new class of antidiabetic drugs. Diabetic Medicine. 1999;16(3):179-192
  3. 3. Vasudevan AR, Balasubramanyam A. Thiazolidinediones: A review of their mechanisms of insulin sensitization, therapeutic potential, clinical efficacy, and tolerability. Diabetes Technology & Therapeutics. 2004;6(6):850-863
  4. 4. Bolen S, Feldman L, Vassy J, Wilson L, Yeh HC, Marinopoulos S, et al. Systematic review: Comparative effectiveness and safety of oral medications for type 2 diabetes mellitus. Annals of Internal Medicine. 2007;147(6):386-399
  5. 5. Ishida H, Takizawa M, Ozawa S, Nakamichi Y, Yamaguchi S, Katsuta H, et al. Pioglitazone improves insulin secretory capacity and prevents the loss of beta-cell mass in obese diabetic db/db mice: Possible protection of beta cells from oxidative stress. Metabolism. 2004;53(4):488-494
  6. 6. Zeender E, Maedler K, Bosco D, Berney T, Donath MY, Halban PA. Pioglitazone and sodium salicylate protect human beta-cells against apoptosis and impaired function induced by glucose and interleukin-1beta. The Journal of Clinical Endocrinology and Metabolism. 2004;89(10):5059-5066
  7. 7. Campbell IW, Mariz S. Beta-cell preservation with thiazolidinediones. Diabetes Research and Clinical Practice. 2007;76(2):163-176
  8. 8. Kahn SE, Haffner SM, Heise MA, Herman WH, Holman RR, Jones NP, et al. Glycemic durability of rosiglitazone, metformin, or glyburide monotherapy. The New England Journal of Medicine. 2006;355(23):2427-2443
  9. 9. Ahmadian M, Suh JM, Hah N, Liddle C, Atkins AR, Downes M, et al. PPARgamma signaling and metabolism: The good, the bad and the future. Nature Medicine. 2013;19(5):557-566
  10. 10. Laakso M. Cardiovascular disease in type 2 diabetes: Challenge for treatment and prevention. Journal of Internal Medicine. 2001 Mar;249(3):225-235
  11. 11. Levitt Katz L, Gidding SS, Bacha F, Hirst K, McKay S, Pyle L, et al. Alterations in left ventricular, left atrial, and right ventricular structure and function to cardiovascular risk factors in adolescents with type 2 diabetes participating in the TODAY clinical trial. Pediatric Diabetes. 2015;16(1):39-47
  12. 12. Rosano GM, Vitale C, Seferovic P. Heart failure in patients with diabetes mellitus. Cardiac Failure Review. 2017;3(1):52-55
  13. 13. Nesto RW, Bell D, Bonow RO, Fonseca V, Grundy SM, Horton ES, et al. Thiazolidinedione use, fluid retention, and congestive heart failure: A consensus statement from the American Heart Association and American Diabetes Association. Circulation. 2003;108(23):2941-2948
  14. 14. Lago RM, Singh PP, Nesto RW. Congestive heart failure and cardiovascular death in patients with prediabetes and type 2 diabetes given thiazolidinediones: A meta-analysis of randomised clinical trials. Lancet. 2007;370(9593):1129-1136
  15. 15. Home PD, Pocock SJ, Beck-Nielsen H, Gomis R, Hanefeld M, Jones NP, et al. Rosiglitazone evaluated for cardiovascular outcomes—An interim analysis. The New England Journal of Medicine. 2007;357(1):28-38
  16. 16. Nissen SE, Wolski K. Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. The New England Journal of Medicine. 2007;356(24):2457-2471
  17. 17. Singh S, Loke YK, Furberg CD. Long-term risk of cardiovascular events with rosiglitazone: A meta-analysis. Journal of the American Medical Association. 2007;298(10):1189-1195
  18. 18. Boussageon R, Bejan-Angoulvant T, Saadatian-Elahi M, Lafont S, Bergeonneau C, Kassai B, et al. Effect of intensive glucose lowering treatment on all cause mortality, cardiovascular death, and microvascular events in type 2 diabetes: Meta-analysis of randomised controlled trials. BMJ. 2011;343:244
  19. 19. Greene SJ, Vaduganathan M, Khan MS, Bakris GL, Weir MR, Seltzer JH, et al. Prevalent and incident heart failure in cardiovascular outcome trials of patients with type 2 diabetes. Journal of the American College of Cardiology. 2018;71(12):1379-1390
  20. 20. Krentz A. Thiazolidinediones: Effects on the development and progression of type 2 diabetes and associated vascular complications. Diabetes/Metabolism Research and Reviews. 2009;25(2):112-126
  21. 21. Liao HW, Saver JL, Wu YL, Chen TH, Lee M, Ovbiagele B. Pioglitazone and cardiovascular outcomes in patients with insulin resistance, pre-diabetes and type 2 diabetes: A systematic review and meta-analysis. BMJ Open. 2017;7(1):e013927
  22. 22. Berger J, Moller DE. The mechanisms of action of PPARs. Annual Review of Medicine. 2002;53:409-435
  23. 23. Kersten S, Desvergne B, Wahli W. Roles of PPARs in health and disease. Nature. 2000;405(6785):421-424
  24. 24. Grygiel-Gorniak B. Peroxisome proliferator-activated receptors and their ligands: Nutritional and clinical implications—A review. Nutrition Journal. 2014;13:17
  25. 25. Braissant O, Foufelle F, Scotto C, Dauca 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 Jan;137(1):354-366
  26. 26. Monsalve FA, Pyarasani RD, Delgado-Lopez F, Moore-Carrasco R. Peroxisome proliferator-activated receptor targets for the treatment of metabolic diseases. Mediators of Inflammation. 2013;2013:549627
  27. 27. Barger PM, Kelly DP. PPAR signaling in the control of cardiac energy metabolism. Trends in Cardiovascular Medicine. 2000;10(6):238-245
  28. 28. Michalik L, Auwerx J, Berger JP, Chatterjee VK, Glass CK, Gonzalez FJ, et al. International Union of Pharmacology. LXI. Peroxisome proliferator-activated receptors. Pharmacological Reviews. 2006;58(4):726-741
  29. 29. Jonker JW, Suh JM, Atkins AR, Ahmadian M, Li P, Whyte J, et al. A PPARgamma-FGF1 axis is required for adaptive adipose remodelling and metabolic homeostasis. Nature. 2012;485(7398):391-394
  30. 30. Desvergne B, Wahli W. Peroxisome proliferator-activated receptors: Nuclear control of metabolism. Endocrine Reviews. 1999;20(5):649-688
  31. 31. Martin G, Schoonjans K, Lefebvre AM, Staels B, Auwerx J. Coordinate regulation of the expression of the fatty acid transport protein and acyl-CoA synthetase genes by PPARalpha and PPARgamma activators. The Journal of Biological Chemistry. 1997;272(45):28210-28217
  32. 32. Tontonoz P, Hu E, Devine J, Beale EG, Spiegelman BM. PPAR gamma 2 regulates adipose expression of the phosphoenolpyruvate carboxykinase gene. Molecular and Cellular Biology.1995;15(1):351-357
  33. 33. Baumann CA, Chokshi N, Saltiel AR, Ribon V. Cloning and characterization of a functional peroxisome proliferator activator receptor-gamma-responsive element in the promoter of the CAP gene. Journal of Biological Chemistry. 2000;275(13):9131-9135
  34. 34. Motojima K, Passilly P, Peters JM, Gonzalez FJ, Latruffe N. Expression of putative fatty acid transporter genes are regulated by peroxisome proliferator-activated receptor alpha and gamma activators in a tissue- and inducer-specific manner. The Journal of Biological Chemistry. 1998;273(27):16710-16714
  35. 35. Kim HI, Cha JY, Kim SY, Kim JW, Roh KJ, Seong JK, et al. Peroxisomal proliferator-activated receptor-gamma upregulates glucokinase gene expression in beta-cells. Diabetes. 2002;51(3):676-685
  36. 36. Kim SY, Kim HI, Park SK, Im SS, Li T, Cheon HG, et al. Liver glucokinase can be activated by peroxisome proliferator-activated receptor-gamma. Diabetes. 2004;53(Suppl 1):S66-S70
  37. 37. Wu Z, Xie Y, Morrison RF, Bucher NL, Farmer SR. PPARgamma induces the insulin-dependent glucose transporter GLUT4 in the absence of C/EBPalpha during the conversion of 3T3 fibroblasts into adipocytes. The Journal of Clinical Investigation. 1998;101(1):22-32
  38. 38. Guan HP, Li Y, Jensen MV, Newgard CB, Steppan CM, Lazar MA. A futile metabolic cycle activated in adipocytes by antidiabetic agents. Nature Medicine. 2002;8(10):1122-1128
  39. 39. Lasar D, Rosenwald M, Kiehlmann E, Balaz M, Tall B, Opitz L, et al. Peroxisome proliferator activated receptor gamma controls mature brown adipocyte inducibility through glycerol kinase. Cell Reports. 2018 Jan 16;22(3):760-773
  40. 40. Smith U, Gogg S, Johansson A, Olausson T, Rotter V, Svalstedt B. Thiazolidinediones (PPARgamma agonists) but not PPARalpha agonists increase IRS-2 gene expression in 3T3-L1 and human adipocytes. The FASEB Journal. 2001 Jan;15(1):215-220
  41. 41. Sigrist S, Bedoucha M, Boelsterli UA. Down-regulation by troglitazone of hepatic tumor necrosis factor-alpha and interleukin-6 mRNA expression in a murine model of non-insulin-dependent diabetes. Biochemical Pharmacology. 2000;60(1):67-75
  42. 42. Saraf N, Sharma PK, Mondal SC, Garg VK, Singh AK. Role of PPARg2 transcription factor in thiazolidinedione-induced insulin sensitization. The Journal of Pharmacy and Pharmacology. 2012;64(2):161-171
  43. 43. Kallen CB, Lazar MA. Antidiabetic thiazolidinediones inhibit leptin (Ob) gene expression in 3T3-L1 adipocytes. Proceedings of the National Academy of Sciences of the United States of America. 1996;93(12):5793-5796
  44. 44. Toruner F, Akbay E, Cakir N, Sancak B, Elbeg S, Taneri F, et al. Effects of PPARgamma and PPARalpha agonists on serum leptin levels in diet-induced obese rats. Hormone and Metabolic Research. 2004;36(4):226-230
  45. 45. Schoonjans K, Peinado-Onsurbe J, Lefebvre AM, Heyman RA, Briggs M, Deeb S, et al. PPARalpha and PPARgamma activators direct a distinct tissue-specific transcriptional response via a PPRE in the lipoprotein lipase gene. The EMBO Journal. 1996;15(19):5336-5348
  46. 46. Gervois P, Torra IP, Fruchart JC, Staels B. Regulation of lipid and lipoprotein metabolism by PPAR activators. Clinical Chemistry and Laboratory Medicine. 2000;38(1):3-11
  47. 47. Dalen KT, Schoonjans K, Ulven SM, Weedon-Fekjaer MS, Bentzen TG, Koutnikova H, et al. Adipose tissue expression of the lipid droplet-associating proteins S3-12 and perilipin is controlled by peroxisome proliferator-activated receptor-gamma. Diabetes. 2004;53(5):1243-1252
  48. 48. Kim HJ, Jung TW, Kang ES, Kim DJ, Ahn CW, Lee KW, et al. Depot-specific regulation of perilipin by rosiglitazone in a diabetic animal model. Metabolism. 2007;56(5):676-85
  49. 49. Devine JH, Eubank DW, Clouthier DE, Tontonoz P, Spiegelman BM, Hammer RE, et al. Adipose expression of the phosphoenolpyruvate carboxykinase promoter requires peroxisome proliferator-activated receptor gamma and 9-cis-retinoic acid receptor binding to an adipocyte-specific enhancer in vivo. The Journal of Biological Chemistry. 1999;274(19):13604-13612
  50. 50. Khandoudi N, Delerive P, Berrebi-Bertrand I, Buckingham RE, Staels B, Bril A. Rosiglitazone, a peroxisome proliferator-activated receptor-gamma, inhibits the Jun NH(2)-terminal kinase/activating protein 1 pathway and protects the heart from ischemia/reperfusion injury. Diabetes. 2002;51(5):1507-1514
  51. 51. Morrison A, Yan X, Tong C, Li J. Acute rosiglitazone treatment is cardioprotective against ischemia-reperfusion injury by modulating AMPK, Akt, and JNK signaling in nondiabetic mice. American Journal of Physiology. Heart and Circulatory Physiology. 2011 Sep;301(3):H895-H902
  52. 52. Zhu P, Lu L, Xu Y, Schwartz GG. Troglitazone improves recovery of left ventricular function after regional ischemia in pigs. Circulation. 2000;101(10):1165-1171
  53. 53. Xu Y, Gen M, Lu L, Fox J, Weiss SO, Brown RD, et al. PPAR-gamma activation fails to provide myocardial protection in ischemia and reperfusion in pigs. American Journal of Physiology. Heart and Circulatory Physiology. 2005;288(3):H1314-H1323
  54. 54. Colca JR, McDonald WG, Waldon DJ, Leone JW, Lull JM, Bannow CA, et al. Identification of a novel mitochondrial protein (“mitoNEET”) cross-linked specifically by a thiazolidinedione photoprobe. American Journal of Physiology. Endocrinology and Metabolism. 2004;286(2):E252-E260
  55. 55. Wiley SE, Murphy AN, Ross SA, van der Geer P, Dixon JE. MitoNEET is an iron-containing outer mitochondrial membrane protein that regulates oxidative capacity. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(13):5318-5323
  56. 56. Zuris JA, Harir Y, Conlan AR, Shvartsman M, Michaeli D, Tamir S, et al. Facile transfer of [2Fe-2S] clusters from the diabetes drug target mitoNEET to an apo-acceptor protein. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(32):13047-13052
  57. 57. Kusminski CM, Holland WL, Sun K, Park J, Spurgin SB, Lin Y, et al. MitoNEET-driven alterations in adipocyte mitochondrial activity reveal a crucial adaptive process that preserves insulin sensitivity in obesity. Nature Medicine. 2012;18(10):1539-1549
  58. 58. Paddock ML, Wiley SE, Axelrod HL, Cohen AE, Roy M, Abresch EC, et al. MitoNEET is a uniquely folded 2Fe 2S outer mitochondrial membrane protein stabilized by pioglitazone. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(36):14342-14347
  59. 59. Colca JR, McDonald WG, Cavey GS, Cole SL, Holewa DD, Brightwell-Conrad AS, et al. Identification of a mitochondrial target of thiazolidinedione insulin sensitizers (mTOT)—Relationship to newly identified mitochondrial pyruvate carrier proteins. PLoS One. 2013;8(5):e61551
  60. 60. Colca JR, VanderLugt JT, Adams WJ, Shashlo A, McDonald WG, Liang J, et al. Clinical proof-of-concept study with MSDC-0160, a prototype mTOT-modulating insulin sensitizer. Clinical Pharmacology and Therapeutics. 2013;93(4):352-359
  61. 61. Hoffmann BR, El-Mansy MF, Sem DS, Greene AS. Chemical proteomics-based analysis of off-target binding profiles for rosiglitazone and pioglitazone: Clues for assessing potential for cardiotoxicity. Journal of Medicinal Chemistry. 2012;55(19):8260-8271
  62. 62. He H, Tao H, Xiong H, Duan SZ, McGowan FX Jr, Mortensen RM, et al. Rosiglitazone causes cardiotoxicity via peroxisome proliferator-activated receptor gamma-independent mitochondrial oxidative stress in mouse hearts. Toxicological Sciences. 2014;138(2):468-481
  63. 63. Brunmair B, Staniek K, Gras F, Scharf N, Althaym A, Clara R, et al. Thiazolidinediones, like metformin, inhibit respiratory complex I: A common mechanism contributing to their antidiabetic actions? Diabetes. 2004;53(4):1052-1059
  64. 64. Rachek LI, Yuzefovych LV, Ledoux SP, Julie NL, Wilson GL. Troglitazone, but not rosiglitazone, damages mitochondrial DNA and induces mitochondrial dysfunction and cell death in human hepatocytes. Toxicology and Applied Pharmacology. 2009;240(3):348-354
  65. 65. Scatena R, Bottoni P, Martorana GE, Ferrari F, De Sole P, Rossi C, et al. Mitochondrial respiratory chain dysfunction, a non-receptor-mediated effect of synthetic PPAR-ligands: Biochemical and pharmacological implications. Biochemical and Biophysical Research Communications. 2004;319(3):967-973
  66. 66. Hughes SD, Kanabus M, Anderson G, Hargreaves IP, Rutherford T, O'Donnell M, et al. The ketogenic diet component decanoic acid increases mitochondrial citrate synthase and complex I activity in neuronal cells. Journal of Neurochemistry. 2014;129(3):426-433
  67. 67. Ingwall JS. On the control of metabolic remodeling in mitochondria of the failing heart. Circulation. Heart Failure. 2009;2(4):275-277
  68. 68. Mishra P, Singh SV, Verma AK, Srivastava P, Sultana S, Rath SK. Rosiglitazone induces cardiotoxicity by accelerated apoptosis. Cardiovascular Toxicology 2014;14(2):99-119
  69. 69. Arakawa K, Ishihara T, Aoto M, Inamasu M, Kitamura K, Saito A. An antidiabetic thiazolidinedione induces eccentric cardiac hypertrophy by cardiac volume overload in rats. Clinical and Experimental Pharmacology & Physiology. 2004;31(1-2):8-13
  70. 70. Duan SZ, Ivashchenko CY, Russell MW, Milstone DS, Mortensen RM. Cardiomyocyte-specific knockout and agonist of peroxisome proliferator-activated receptor-gamma both induce cardiac hypertrophy in mice. Circulation Research. 2005;97(4):372-379
  71. 71. Singh S, Loke YK, Furberg CD. Thiazolidinediones and heart failure: A teleo-analysis. Diabetes Care. 2007;30(8):2148-2153
  72. 72. Hernandez AV, Usmani A, Rajamanickam A, Moheet A. Thiazolidinediones and risk of heart failure in patients with or at high risk of type 2 diabetes mellitus: A meta-analysis and meta-regression analysis of placebo-controlled randomized clinical trials. American Journal of Cardiovascular Drugs. 2011;11(2):115-128
  73. 73. Mannucci E, Monami M, Di Bari M, Lamanna C, Gori F, Gensini GF, et al. Cardiac safety profile of rosiglitazone: A comprehensive meta-analysis of randomized clinical trials. International Journal of Cardiology. 2010;143(2):135-140
  74. 74. 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. Journal of the American Medical Association. 2007;298(10):1180-1188
  75. 75. Berlie HD, Kalus JS, Jaber LA. Thiazolidinediones and the risk of edema: A meta-analysis. Diabetes Research and Clinical Practice. 2007;76(2):279-289
  76. 76. Lu Y, Ma D, Xu W, Shao S, Yu X. Effect and cardiovascular safety of adding rosiglitazone to insulin therapy in type 2 diabetes: A meta-analysis. Journal of Diabetes Investigation. 2015;6(1):78-86
  77. 77. Sarafidis PA, Nilsson PM. The effects of thiazolidinediones on blood pressure levels—A systematic review. Blood Pressure. 2006;15(3):135-150
  78. 78. Nissen SE, Wolski K, Topol EJ. Effect of muraglitazar on death and major adverse cardiovascular events in patients with type 2 diabetes mellitus. Journal of the American Medical Association. 2005;294(20):2581-2586
  79. 79. Nissen SE, Wolski K. Rosiglitazone revisited: An updated meta-analysis of risk for myocardial infarction and cardiovascular mortality. Archives of Internal Medicine. 2010;170(14):1191-1201
  80. 80. Lipscombe LL, Gomes T, Levesque LE, Hux JE, Juurlink DN, Alter DA. Thiazolidinediones and cardiovascular outcomes in older patients with diabetes. Journal of the American Medical Association. 2007;298(22):2634-2643
  81. 81. Hsiao FY, Huang WF, Wen YW, Chen PF, Kuo KN, Tsai YW. Thiazolidinediones and cardiovascular events in patients with type 2 diabetes mellitus: A retrospective cohort study of over 473,000 patients using the National Health Insurance database in Taiwan. Drug Safety. 2009;32(8):675-690
  82. 82. Psaty BM, Furberg CD. The record on rosiglitazone and the risk of myocardial infarction. The New England Journal of Medicine. 2007;357(1):67-69
  83. 83. Kaul S, Bolger AF, Herrington D, Giugliano RP, Eckel RH. Thiazolidinedione Drugs and Cardiovascular Risks: A science advisory from the American Heart Association and American College of Cardiology Foundation. Circulation. 2010;121(16):1868-1877
  84. 84. Shuster JJ, Jones LS, Salmon DA. Fixed vs random effects meta-analysis in rare event studies: The rosiglitazone link with myocardial infarction and cardiac death. Statistics in Medicine. 2007;26(24):4375-4385
  85. 85. Diamond GA, Bax L, Kaul S. Uncertain effects of rosiglitazone on the risk for myocardial infarction and cardiovascular death. Annals of Internal Medicine. 2007;147(8):578-581
  86. 86. Goldberg RB, Kendall DM, Deeg MA, Buse JB, Zagar AJ, Pinaire JA, et al. A comparison of lipid and glycemic effects of pioglitazone and rosiglitazone in patients with type 2 diabetes and dyslipidemia. Diabetes Care. 2005;28(7):1547-1554
  87. 87. Atamer Y, Atamer A, Can AS, Hekimoglu A, Ilhan N, Yenice N, et al. Effects of rosiglitazone on serum paraoxonase activity and metabolic parameters in patients with type 2 diabetes mellitus. Brazilian Journal of Medical and Biological Research. 2013;46(6):528-532
  88. 88. Shiiba M, Zhang B, Miura SI, Ike A, Nose D, Kuwano T, et al. Association between discordance of LDL-C and non-HDL-C and clinical outcomes in patients with stent implantation: From the FU-Registry. Heart and Vessels. 2018;33(2):102-112

Written By

Jing-Bo Jiang, James A. Balschi, Francis X. McGowan Jr and Huamei He

Submitted: 17 March 2018 Reviewed: 21 May 2018 Published: 05 November 2018