Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Apr 28.
Published in final edited form as: Curr Diab Rep. 2008 Jun;8(3):243–248. doi: 10.1007/s11892-008-0042-x

The Complexity of Diabetic Cardiomyopathy: Lessons from Patients and Animal Models

Romain Harmancey 1, Heinrich Taegtmeyer 1
PMCID: PMC3637972  NIHMSID: NIHMS453871  PMID: 18625124

Abstract

Cardiovascular diseases are foreseeable complications of diabetes mellitus. Prominent among those complications is heart failure. Diabetic cardiomyopathy is a distinct entity independent of coronary artery disease and hypertension. Most of our knowledge on the pathogenesis of diabetic cardiomyopathy comes from studies performed on various animal models. The recent advances in the domain confirm that the disease is above all a maladaptation of the heart mostly driven by the metabolic derangements that accompany diabetes mellitus.

Introduction

Diabetes mellitus is considered one of the major causes of premature death and disability, mainly due to cardiovascular complications. With the depressing forecast of 366 millions people suffering from diabetes by 2030, the epidemic represents a sword of Damocles for health care in most countries [1]. The increasing prevalence of obesity is considered a “firestarter” for type 2 diabetes.

Prominent among the complications of diabetes is heart failure. Diabetic cardiomyopathy (DCM) was historically described as a disease process that affects the myocardium independently of macrovascular complications currently associated with common risk factors like arterial hypertension, hypercholesterolemia, and coronary artery disease [2]. Although many of the structural and functional abnormalities of the heart which characterize DCM have been described, the origins of the disease are still unclear. The pathogenesis of DCM is an active field of investigation, and we wish to refer the reader to other excellent reviews published recently on the matter [35]. We report here recent advances in research supporting the idea that DCM may originate from a failure of the heart to adapt to chronic changes in cardiac metabolism.

Evidence for Diabetic Cardiomyopathy in Humans

Structural and functional features of diabetic cardiomyopathy

In the 1970s the Framingham Heart Study demonstrated an increased incidence of congestive heart failure in diabetic patients of both sexes, which was independent of age, obesity, hypertension, coronary artery disease, and hypercholesterolemia [6]. Echocardiographic studies on independent populations and the more recent use of transmitral inflow with tissue Doppler analysis have permitted to establish the typical pattern of alterations characterizing DCM [7]. Diabetic cardiomyopathy is a combination of left ventricular structure abnormalities (concentric remodeling/hypertrophy) with diastolic dysfunction, which may precede the development of systolic dysfunction and eventually progress to overt heart failure. Heart failure is defined as a clinical syndrome in which exercise intolerance is associated with left ventricular (LV) dysfunction of the heart [8].

Systemic and myocardial metabolic shifts

When considering heart failure in patients with diabetes one must also consider the metabolic derangements of diabetes mellitus: hyperglycemia, insulin resistance (for type 2 diabetes), and dyslipidemia with increased levels of circulating nonesterified fatty acids (NEFA). The resulting changes in nutrient supply and uptake at the level of the cardiac myocyte dramatically affect the way the heart converts chemical to mechanical energy. Exact mechanisms still need to be worked out. According to the studies performed in animal models, a main feature of DCM is intramyocardial lipid accumulation [35], and the concept of cardiac lipotoxicity will be discussed further below. While cardiac steatosis was already considered as a possible cause of sudden death during the nineteenth century [9], both its presence and its involvement in the development of DCM in humans have been ignored for a long time.

We have observed intramyocardial lipid accumulation in the failing heart of diabetic patients that resembles the lipotoxic Zucker diabetic fatty (ZDF) rat heart [10]. More recent work by others has shown that cardiac steatosis is found in patients with impaired glucose tolerance and precedes the onset of type 2 diabetes mellitus and LV systolic dysfunction [11]. Intramyocardial lipid accumulation in type 2 diabetic patients directly also correlates with impaired contractility and decreased capability of myocytes to adapt following infarction [12]. Both increased plasma NEFA levels and myocardial triglyceride accumulation are correlated to impaired diastolic function in healthy young people [13]. Lastly, abnormal left ventricular energy metabolism in obese insulin resistant men precedes functional and structural remodeling of the heart [14]. Thus a metabolic derangement of the heart characterized by an intramyocardial lipid accumulation is likely to contribute to DCM development in humans.

The Pathogenesis of Diabetic Cardiomyopathy

Animal models of diabetic cardiomyopathy

Most of the molecular mechanisms involved in the pathogenesis of DCM have been (and are) investigated with rodent animal models of type 1 or type 2 diabetes mellitus (see Table). Because DCM is a complex multifactorial disease, it is unlikely that there will ever be an animal model which presents all the various aspects of the human syndrome. The mechanisms of the disease are therefore investigated separately in a wide spectrum of different models [15]. The models consist either of animals with defective insulin actions or signaling, with an altered cardiac glucose and/or fatty acid utilization, with enhanced oxidative stress, or with enhanced cardiac fibrosis. The most popular animal models include the chemical ablation of the β cells of the pancreas (Streptozotocin (STZ)-treated rodents), animals spontaneously deficient in the leptin signaling (ob/ob and db/db mice, ZDF rats), and transgenic animals with a cardiac-specific lipotoxicity. All these models are not without limitations. In contrast to humans with diabetes mellitus, rodents are resistant to the development of atherosclerotic lesions and ischemia. At the same time animals unlike human patients are often not treated for diabetes. The rodent models have nonetheless served to identify a number of defects which are likely to be responsible for the development of DCM in humans. Recent studies using broad transcriptomic or proteomic approaches give a good overview of the profound modifications that affect the heart in diabetes mellitus [16; 17]. Besides the dysregulation of circulating factors which contributes to heart fibrosis and hypertrophy, the major intramyocellular changes focus on cardiac metabolism and a subsequent series of defects in mitochondrial function, reactive oxygen species (ROS) generation or dissipation, and in calcium homeostasis.

Table.

Proposed mechanisms in the pathogenesis of diabetic cardiomyopathy according to animal studies.

Mechanism Pathology Animal Model
Cell apoptosis and cardiac remodeling
      Overactivity of the renin-angiotensin system T1DM STZ-treated SD rats [20]
      AGE and AGE receptors activation T1DM STZ-treated Wistar rats [21]
      Intramyocardial inflammation T1DM STZ-treated SD rats [22; 23]
      Switch in contractile protein expression T1DM STZ-treated Wistar rats [38; 58]
Metabolic derangements
      Increased fatty acid uptake Lipotoxic CM HFD-fed Wistar rats [33], CD36 deficiency in mice with cardiac PPARα overexpression [34]
      Hyperglycemia-related alterations T1DM, T2DM Zucker diabetic fatty rats [36], STZ-treated FVB and db/db mice [37]
Impaired mitochondrial function
      Impaired ATP production T1DM, T2DM STZ-treated SD rats [43], db/db mice [48]
      Alterations in cardiolipin content and composition T1DM, T2DM STZ-treated C57BL/6 and ob/ob mice [45]
      Activation of the intrinsic pathway of apoptosis T2DM Zucker diabetic fatty rats [44]
Increased oxidative stress T1DM, T2DM OVE26 mice [49], ob/ob mice [51; 52], db/db mice [52], STZ-treated Wistar rats [21]
Impaired Ca2+ homeostasis T1DM, T2DM db/db mice [55; 63], STZ-treated Wistar rats [56; 57]

A brief overview of the results obtained from recent animal studies which are discussed further in the text. AGEs, advanced glycation end products; CM, cardiomyopathy; HFD, high fat diet; SD, Sprague Dawley; STZ, streptozotocin; T1DM, type 1 diabetes mellitus; T2DM, type 2 diabetes mellitus.

Circulating factors, apoptosis and cardiac fibrosis

Impaired contractile function seems to be partly caused by cell necrosis or apoptosis and myocardial fibrosis, although a recent study using noninvasive echocardiography and invasive LV catheterization rather reported a reduction of LV mass in STZ-treated rats [18]. Several circulating factors have been shown to trigger the myocardial remodeling and constitute potential therapeutic targets for treating DCM. Thus, the upregulation of the renin-angiotensin system in the diabetic heart enhances oxidative stress and myocardial cell death [19], and its blockade attenuates myofibrillar remodeling in the heart [20]. The deleterious effects of hyperglycemia on advanced glycated end products (AGEs) receptor activation and increase of intracardiac profibrogenic factors are alleviated by dehydroepiandrosterone (DHEA) treatment [21]. Leukocyte infiltration of the heart is also likely to play a pivotal role in the pathophysiology of DCM, and both, treatment with Pralnacasan and Atorvastatin, aiming to decrease intramyocardial inflammation, improved cardiac function in STZ-treated rats [22; 23].

Cardiac metabolism

In metabolic terms the mammalian heart is an omnivore which burns both fatty acids and glucose. Under normal conditions, 60% to 90% of the cardiac ATP comes from oxidative phosphorylation of ADP and oxidation of fatty acids [24]. However, cardioprotective innate mechanisms in normal heart include an increase of carbohydrate oxidation in acute and chronic stress conditions like ischemia or hypertrophy [25]. But in the diabetic state, the heart relies essentially on fatty acid oxidation for energy provision, even under stress [26]. The enhanced fatty acid metabolism may be the consequence of the Randle’s “glucose fatty-acid cycle” [27], which stresses the inhibition of glucose oxidation by fatty acids to a greater extent than glycolysis and the inhibition of glycolysis to a greater extent than glucose uptake, but it may be also due to a decrease in glucose uptake triggered by insulin resistance. Thus, the metabolic flexibility of the heart is impaired with diabetes, which we consider a major contributing factor to cardiac maladaptation [28].

The most common perception on cardiac metabolism in diabetes mellitus is that NEFA supply is overwhelming the oxidative capacities of the heart, therefore leading to the intrasarcoplasmic accumulation of neutral lipids and their intermediates. The excess of lipid intermediates is then directed to signaling pathways whose overactivity will affect critical mechanisms like ATP production, insulin sensitivity, cell contractility, and apoptosis. In short, DCM is a lipotoxic cardiomyopathy. Thus, ceramide is a multifunctional regulatory molecule, at the crossroads of the inflammatory response, insulin resistance, and apoptosis [29]. Diacylglycerol induces insulin resistance by activating distinct isoforms of protein kinase C [30]. The long-chain fatty acyl CoA themselves can inhibit glycolysis and are suspected to regulate glucose uptake [31]. Excess palmitoyl-CoA is also suspected to trigger intrinsic apoptosis by enhancing endoplasmic reticulum stress and calcium release [32].

Although several checkpoints regulate fatty acid metabolism, recent results have stressed the importance of fatty acid uptake in the regulation of intramyocardial lipid content. Cardiomyocytes from rats fed a high fat diet for 8 weeks display an increased localization of the fatty acid translocase FAT/CD36 at the sarcolemma, which is preceding the onset of cardiac contractile dysfunction [33]. Moreover, CD36 deficiency is sufficient to reverse the lipotoxic phenotype of mice with cardiac-specific overexpression of the nuclear receptor PPARα [34].

While much emphasis has been given to the lipotoxic aspect of DCM, dysregulated carbohydrate metabolism and glucotoxicity must also be considered. Hyperglycemia and insulin resistance are further contributors to cardiac maladaptation in the diabetic state. According to the Randle hypothesis, glucose may accumulate into the cells and be redirected to alternate metabolic pathways with additional regulatory consequences, namely the pentose phosphate pathway and the hexosamine biosynthetic pathway [35]. For example, increased intracellular levels of UDP-N-acetylglucosamine is suspected to alter excitation/contraction coupling by promoting the O-linked glycosylation of target proteins like the myosin heavy chain (MHC) isoforms or the sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA). Accordingly, impaired cardiomyocyte function in ZDF rats is associated with an increase in O-linked glycosylation of high-molecular-mass proteins [36]. Moreover, hyperglycemia is likely to be responsible for the decrease in protein levels of the essential myocardial survival factor GATA4 seen in several animal models of diabetes mellitus [37]. The expression levels of the glucose transporters GLUT1 and GLUT4 are downregulated quickly and in a sustained manner after the onset of insulin-deficient diabetes, possibly as an adaptative mechanism of the heart [38]. Given the deleterious effects of hyperglycemia, it is tempting to consider the hypothesis arguing that insulin resistance may be, at least on the short term, a physiological mechanism to protect the cell [39]. It should be noted that unlike PPARα, cardiac-specific overexpression of PPARβ/δ does not induce lipotoxic cardiomyopathy. In fact, while both nuclear receptors induce expression of genes involved in mitochondrial fatty acid oxidation, PPARβ/δ preferentially induces glucose utilization whereas PPARα represses it [40].

In short, DCM is primarily a metabolic cardiomyopathy. Defects in both fatty acid and glucose oxidation pathways are intimately related and are preceding the contractile dysfunctions.

Impaired mitochondrial function

Because the mitochondrion is the cradle of both glucose and fatty acids conversion to a usable form of energy (ATP), it was obvious to hypothesize that the impaired metabolism characterizing DCM may be related to mitochondrial dysfunction. Not surprisingly, impaired mitochondrial function has been demonstrated in type 1 and type 2 diabetic animals. Microarray analysis performed on the LV transcriptome of STZ-treated rats revealed that a disproportionate number of the genes whose expression is significantly altered by diabetes are expressed in the mitochondrion [16]. Most of these genes encode for enzymes involved in β oxidation and are highly upregulated in accordance to the switch toward fatty acid utilization. There is also an increased number of mitochondria in cardiomyocytes of type 1 and type 2 models of diabetes [41; 42], which is probably due to the activation of mitochondrial biogenesis by the coactivator of PPARα, PGC-1α. However, the genes encoding for several components of the oxidative phosphorylation process (the “OXPHOS proteins”) are rather downregulated [43]. Lastly, ATP production in the diabetic heart is decreased. Therefore, the increase in mitochondrial number and volume density does not compensate for the decrease in energy coupling.

The mitochondrion is also a central determinant of cell survival. The mitochondrion forms a converging node for apoptotic stimuli by promoting the release of pro-apoptotic factors like cytochrome c, Smac/DIABLO, and the apoptosis-inducing factor (AIF). This intrinsic pathway of apoptosis is overactivated in the heart of ZDF rats and may be involved in remodeling and cardiac dysfunction [44]. Moreover the loss of cardiolipin, an important component of the inner mitochondrial membrane necessary for oxidative phosphorylation, is an early event related to apoptosis in animal models of diabetes [45]. The mitochondrion is also an important source of ROS, which are a naturally-occurring byproduct of the respiratory process. ROS production is enhanced in the diabetic heart consequently to the increased reliance of the cells on β oxidation. Recent data support the activation of a fatty acid-mediated uncoupling system in the mitochondrion that may help to dissipate this oxidative stress. Evidences point toward the activation of a long-chain fatty acyl CoA generation and export system in the mitochondria of STZ-induced diabetic rats [46]. We have found that such a mechanism was activated in rats fed a high fat diet for 48 weeks and correlated to a maintained cardiac power, whereas rats fed a high carbohydrate and high fat diet (Western diet) did not show any activation of this system and elicited impaired cardiac function [47]. However, others think that uncoupling mechanisms may be detrimental on the long term by driving an ATP depletion [48]. Whether the fatty acid-mediated uncoupling system is beneficial or not in DCM will require further investigations. However, the chronic activation of this system may partly explain why the heart maladapts in diabetes mellitus.

Reactive oxygen species and oxidative stress

Oxidative stress in DCM may be caused by an increase in ROS production and a concomitant decrease in the antioxidant defenses of the cell [17]. Strategies that enhance mitochondrial ROS scavenging systems allow to reduce diabetes-induced cardiac dysfunction [4]. Even thought the respiratory chain of the mitochondrion is a primary source for ROS production in the diabetic cardiomyocyte [49], ROS can also be generated by other enzymatic systems, such as NADPH oxidase whose expression is increased by hyperglycemia, or xanthine oxidoreductase [50; 51]. ROS may be involved in the cardiac remodeling accompanying DCM by stimulating apoptosis [52], but also by increasing AGEs formation and the number of AGE-receptors, thus triggering the activation of profibrogenic factors [21]. Excessive oxidants may also activate PKC, therefore contributing to insulin resistance [53].

Impaired calcium homeostasis

Because the main feature of DCM is a contractile dysfunction, it seemed obvious to look for potential defects in ion homeostasis, and especially Ca2+ homeostasis [54]. In db/db mice, Ca2+ cycling is fully disturbed due to reduced Ca2+ influx via L-type Ca2+ channels, lowered Ca2+ release from sarcoplasmic reticulum, slowed Ca2+ reuptake, and increased Ca2+ efflux [55]. In STZ-induced diabetic rats, the main cause for impaired Ca2+ cycling consists of depressed sarcoplasmic reticulum function through decreased expression of SERCA2a and the ryanodine receptor, whereas the L-type Ca2+ current is not significantly modified [56; 57]. In contribution to impaired Ca2+ cycling, diabetes mellitus is also accompanied by a marked switch of MHC expression from the alpha to the beta isoform. The V3 isozymes formed with βMHC have a lower Ca2+-activated ATPase activity, which could mediate in part the diminished contractility of the diabetic heart [58]. Again, the switch in MHC isoforms and the decrease in SERCA2a expression in the diabetic state are rapid events which precede the venue of contractile function [38]. In humans, the heart of nonischemic heart failure patients shows a most important decrease in the expression of the myocyte enhancer factor 2C (MEF2C) and its regulated transcripts SERCA2a and αMHC when associated to diabetes mellitus [59].

As it could be expected, impaired Ca2+ homeostasis may be of major importance in the pathogenesis of DCM. It is of note that SERCA2a overexpression protected the heart of STZ-diabetic mice from severe contractile dysfunction [60]. Although this remains to be determined for DCM, cytoplasmic Ca2+ overload may participate to cardiac remodeling through a mitochondrial-dependent necrotic process [61]. Results suggest that impaired excitation/contraction coupling which results from inadequate Ca2+ homeostasis, rather than interstitial fibrosis, is linked to the early stages of diabetic diastolic dysfunction [62]. Interestingly, calcium transients are impaired in cardiomyocytes of normal mice treated with palmitate, whereas the same treatment improved the calcium transients in cardiomyocytes isolated from db/db mice [63]. The discrepancy in results is explained by an absence of depolarization of the mitochondrial membrane potential and no increase in mitochondrial ROS production for the ob/ob cardiomyocytes [63]. Besides establishing new evidences for a tight link between metabolism, mitochondrial function, and cardiac contractility, this study further strengthens the notion that DCM may originate from an improper adaptation of the heart to a modified metabolic environment.

Conclusions

Most of our present knowledge on the pathogenesis of DCM is actually based on scattered data obtained from a wide variety of animal models. Even though a few of the proposed mechanisms have been presently investigated in humans, a particular attention should be paid to the dramatic changes in nutrient supply to the heart and the impaired cardiac metabolic flexibility. The mitochondrion appears to play a central role in the regulation of the mechanisms proposed to be involved in the development of DCM. While the features of lipotoxic cardiomyopathy have received much attention, both glucose and fatty acid derivatives may co-jointly determine the fate of cardiac remodeling and contractility in diabetes mellitus. Strategies aiming to restore normal heart metabolism in DCM should be expected to restore normal cardiac function. Glucolipotoxicity as a concept requires further investigation.

Acknowledgements

Work from the authors’ laboratory has been supported, in part, by a grant from the NHLBI of the NIH (RO1-HL/AG61483). We thank Roxy A. Tate for expert editorial help.

References

Papers of particular interest, published recently, have been highlighted as:

* Of importance

** Of major Importance

  • 1.Smyth S, Heron A. Diabetes and obesity: the twin epidemics. Nat Med. 2006;12:75–80. doi: 10.1038/nm0106-75. [DOI] [PubMed] [Google Scholar]
  • 2.Rubler S, Dlugash J, Yuceoglu YZ, et al. New type of cardiomyopathy associated with diabetic glomerulosclerosis. Am J Cardiol. 1972;30:595–602. doi: 10.1016/0002-9149(72)90595-4. [DOI] [PubMed] [Google Scholar]
  • 3.An D, Rodrigues B. Role of changes in cardiac metabolism in development of diabetic cardiomyopathy. Am J Physiol Heart Circ Physiol. 2006;291:H1489–H1506. doi: 10.1152/ajpheart.00278.2006. [DOI] [PubMed] [Google Scholar]
  • 4. Boudina S, Abel ED. Diabetic cardiomyopathy revisited. Circulation. 2007;115:3213–3223. doi: 10.1161/CIRCULATIONAHA.106.679597. A thorough review on diabetic cardiomyopathy. It reports on many original studies investigating diabetic cardiomyopathy.
  • 5.Poornima IG, Parikh P, Shannon RP. Diabetic cardiomyopathy: the search for a unifying hypothesis. Circ Res. 2006;98:596–605. doi: 10.1161/01.RES.0000207406.94146.c2. [DOI] [PubMed] [Google Scholar]
  • 6.Kannel WB, McGee DL. Diabetes and cardiovascular disease. The Framingham study. Jama. 1979;241:2035–2038. doi: 10.1001/jama.241.19.2035. [DOI] [PubMed] [Google Scholar]
  • 7.Galderisi M. Diastolic dysfunction and diabetic cardiomyopathy: evaluation by Doppler echocardiography. J Am Coll Cardiol. 2006;48:1548–1551. doi: 10.1016/j.jacc.2006.07.033. [DOI] [PubMed] [Google Scholar]
  • 8.Cohn JN. Pathophysiology and Clinical Recognition of Heart Failure. In: Willerson JT, Cohn JN, Wellens HJJ, Holmes DR Jr, editors. Cardiovascular Medicine. New York: Springer; 2007. pp. 1379–1396. [Google Scholar]
  • 9.Bedford E. The story of fatty heart. A disease of Victorian times. Br Heart J. 1972;34:23–28. doi: 10.1136/hrt.34.1.23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Sharma S, Adrogue JV, Golfman L, et al. Intramyocardial lipid accumulation in the failing human heart resembles the lipotoxic rat heart. Faseb J. 2004;18:1692–1700. doi: 10.1096/fj.04-2263com. [DOI] [PubMed] [Google Scholar]
  • 11. McGavock JM, Lingvay I, Zib I, et al. Cardiac steatosis in diabetes mellitus: a 1H-magnetic resonance spectroscopy study. Circulation. 2007;116:1170–1175. doi: 10.1161/CIRCULATIONAHA.106.645614. The article reports that cardiac steatosis precedes the onset of cardiomyopathy in type 2 diabetic patients. In complement to the previous study from Sharma et al. (ref 10), this article establishes a strong connection between impaired cardiac metabolism and cardiac dysfunction in humans.
  • 12.Borisov AB, Ushakov AV, Zagorulko AK, et al. Intracardiac lipid accumulation, lipoatrophy of muscle cells and expansion of myocardial infarction in type 2 diabetic patients. Micron. 2007 doi: 10.1016/j.micron.2007.11.002. [DOI] [PubMed] [Google Scholar]
  • 13.van der Meer RW, Hammer S, Smit JW, et al. Short-term caloric restriction induces accumulation of myocardial triglycerides and decreases left ventricular diastolic function in healthy subjects. Diabetes. 2007;56:2849–2853. doi: 10.2337/db07-0768. [DOI] [PubMed] [Google Scholar]
  • 14.Perseghin G, Ntali G, De Cobelli F, et al. Abnormal left ventricular energy metabolism in obese men with preserved systolic and diastolic functions is associated with insulin resistance. Diabetes Care. 2007;30:1520–1526. doi: 10.2337/dc06-2429. [DOI] [PubMed] [Google Scholar]
  • 15.Hsueh W, Abel ED, Breslow JL, et al. Recipes for creating animal models of diabetic cardiovascular disease. Circ Res. 2007;100:1415–1427. doi: 10.1161/01.RES.0000266449.37396.1f. [DOI] [PubMed] [Google Scholar]
  • 16.Glyn-Jones S, Song S, Black MA, et al. Transcriptomic analysis of the cardiac left ventricle in a rodent model of diabetic cardiomyopathy: molecular snapshot of a severe myocardial disease. Physiol Genomics. 2007;28:284–293. doi: 10.1152/physiolgenomics.00204.2006. [DOI] [PubMed] [Google Scholar]
  • 17.Hamblin M, Friedman DB, Hill S, et al. Alterations in the diabetic myocardial proteome coupled with increased myocardial oxidative stress underlies diabetic cardiomyopathy. J Mol Cell Cardiol. 2007;42:884–895. doi: 10.1016/j.yjmcc.2006.12.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Wichi R, Malfitano C, Rosa K, et al. Noninvasive and invasive evaluation of cardiac dysfunction in experimental diabetes in rodents. Cardiovasc Diabetol. 2007;6:14. doi: 10.1186/1475-2840-6-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Frustaci A, Kajstura J, Chimenti C, et al. Myocardial cell death in human diabetes. Circ Res. 2000;87:1123–1132. doi: 10.1161/01.res.87.12.1123. [DOI] [PubMed] [Google Scholar]
  • 20.Machackova J, Liu X, Lukas A, Dhalla NS. Renin-angiotensin blockade attenuates cardiac myofibrillar remodelling in chronic diabetes. Mol Cell Biochem. 2004;261:271–278. doi: 10.1023/b:mcbi.0000028765.89855.73. [DOI] [PubMed] [Google Scholar]
  • 21.Aragno M, Mastrocola R, Alloatti G, et al. Oxidative stress triggers cardiac fibrosis in the heart of diabetic rats. Endocrinology. 2008;149:380–388. doi: 10.1210/en.2007-0877. [DOI] [PubMed] [Google Scholar]
  • 22.Van Linthout S, Riad A, Dhayat N, et al. Anti-inflammatory effects of atorvastatin improve left ventricular function in experimental diabetic cardiomyopathy. Diabetologia. 2007;50:1977–1986. doi: 10.1007/s00125-007-0719-8. [DOI] [PubMed] [Google Scholar]
  • 23.Westermann D, Van Linthout S, Dhayat S, et al. Cardioprotective and anti-inflammatory effects of interleukin converting enzyme inhibition in experimental diabetic cardiomyopathy. Diabetes. 2007;56:1834–1841. doi: 10.2337/db06-1662. [DOI] [PubMed] [Google Scholar]
  • 24.Taegtmeyer H, Hems R, Krebs HA. Utilization of energy-providing substrates in the isolated working rat heart. Biochem J. 1980;186:701–711. doi: 10.1042/bj1860701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Rajabi M, Kassiotis C, Razeghi P, Taegtmeyer H. Return to the fetal gene program protects the stressed heart: a strong hypothesis. Heart Fail Rev. 2007;12:331–343. doi: 10.1007/s10741-007-9034-1. [DOI] [PubMed] [Google Scholar]
  • 26.Stanley WC, Lopaschuk GD, McCormack JG. Regulation of energy substrate metabolism in the diabetic heart. Cardiovasc Res. 1997;34:25–33. doi: 10.1016/s0008-6363(97)00047-3. [DOI] [PubMed] [Google Scholar]
  • 27.Randle PJ, Garland PB, Hales CN, Newsholme EA. The glucose fatty-acid cycle. Its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus. Lancet. 1963;1:785–789. doi: 10.1016/s0140-6736(63)91500-9. [DOI] [PubMed] [Google Scholar]
  • 28.Taegtmeyer H, McNulty P, Young ME. Adaptation and maladaptation of the heart in diabetes: Part I: general concepts. Circulation. 2002;105:1727–1733. doi: 10.1161/01.cir.0000012466.50373.e8. [DOI] [PubMed] [Google Scholar]
  • 29.Summers SA. Ceramides in insulin resistance and lipotoxicity. Prog Lipid Res. 2006;45:42–72. doi: 10.1016/j.plipres.2005.11.002. [DOI] [PubMed] [Google Scholar]
  • 30.Xiang H, McNeill JH. Protein kinase C activity is altered in diabetic rat hearts. Biochem Biophys Res Commun. 1992;187:703–710. doi: 10.1016/0006-291x(92)91252-l. [DOI] [PubMed] [Google Scholar]
  • 31.Cooney GJ, Thompson AL, Furler SM, et al. Muscle long-chain acyl CoA esters and insulin resistance. Ann N Y Acad Sci. 2002;967:196–207. doi: 10.1111/j.1749-6632.2002.tb04276.x. [DOI] [PubMed] [Google Scholar]
  • 32.Borradaile NM, Han X, Harp JD, et al. Disruption of endoplasmic reticulum structure and integrity in lipotoxic cell death. J Lipid Res. 2006;47:2726–2737. doi: 10.1194/jlr.M600299-JLR200. [DOI] [PubMed] [Google Scholar]
  • 33.Ouwens DM, Diamant M, Fodor M, et al. Cardiac contractile dysfunction in insulin-resistant rats fed a high-fat diet is associated with elevated CD36-mediated fatty acid uptake and esterification. Diabetologia. 2007;50:1938–1948. doi: 10.1007/s00125-007-0735-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Yang J, Sambandam N, Han X, et al. CD36 deficiency rescues lipotoxic cardiomyopathy. Circ Res. 2007;100:1208–1217. doi: 10.1161/01.RES.0000264104.25265.b6. [DOI] [PubMed] [Google Scholar]
  • 35.Young ME, McNulty P, Taegtmeyer H. Adaptation and maladaptation of the heart in diabetes: Part II: potential mechanisms. Circulation. 2002;105:1861–1870. doi: 10.1161/01.cir.0000012467.61045.87. [DOI] [PubMed] [Google Scholar]
  • 36.Fulop N, Mason MM, Dutta K, et al. Impact of Type 2 diabetes and aging on cardiomyocyte function and O-linked N-acetylglucosamine levels in the heart. Am J Physiol Cell Physiol. 2007;292:C1370–C1378. doi: 10.1152/ajpcell.00422.2006. [DOI] [PubMed] [Google Scholar]
  • 37.Kobayashi S, Mao K, Zheng H, et al. Diminished GATA4 protein levels contribute to hyperglycemia-induced cardiomyocyte injury. J Biol Chem. 2007;282:21945–21952. doi: 10.1074/jbc.M703048200. [DOI] [PubMed] [Google Scholar]
  • 38.Depre C, Young ME, Ying J, et al. Streptozotocin-induced changes in cardiac gene expression in the absence of severe contractile dysfunction. J Mol Cell Cardiol. 2000;32:985–996. doi: 10.1006/jmcc.2000.1139. [DOI] [PubMed] [Google Scholar]
  • 39.Erol A. Insulin resistance is an evolutionarily conserved physiological mechanism at the cellular level for protection against increased oxidative stress. Bioessays. 2007;29:811–818. doi: 10.1002/bies.20618. [DOI] [PubMed] [Google Scholar]
  • 40.Burkart EM, Sambandam N, Han X, et al. Nuclear receptors PPARbeta/delta and PPARalpha direct distinct metabolic regulatory programs in the mouse heart. J Clin Invest. 2007;117:3930–3939. doi: 10.1172/JCI32578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Boudina S, Sena S, O'Neill BT, et al. Reduced mitochondrial oxidative capacity and increased mitochondrial uncoupling impair myocardial energetics in obesity. Circulation. 2005;112:2686–2695. doi: 10.1161/CIRCULATIONAHA.105.554360. [DOI] [PubMed] [Google Scholar]
  • 42.Shen X, Zheng S, Thongboonkerd V, et al. Cardiac mitochondrial damage and biogenesis in a chronic model of type 1 diabetes. Am J Physiol Endocrinol Metab. 2004;287:E896–E905. doi: 10.1152/ajpendo.00047.2004. [DOI] [PubMed] [Google Scholar]
  • 43.Turko IV, Murad F. Quantitative protein profiling in heart mitochondria from diabetic rats. J Biol Chem. 2003;278:35844–35849. doi: 10.1074/jbc.M303139200. [DOI] [PubMed] [Google Scholar]
  • 44.Lu MC, Tzang BS, Kuo WW, et al. More Activated Cardiac Mitochondrial-dependent Apoptotic Pathway in Obese Zucker Rats. Obesity (Silver Spring) 2007;15:2634–2642. doi: 10.1038/oby.2007.315. [DOI] [PubMed] [Google Scholar]
  • 45.Han X, Yang J, Yang K, et al. Alterations in myocardial cardiolipin content and composition occur at the very earliest stages of diabetes: a shotgun lipidomics study. Biochemistry. 2007;46:6417–6428. doi: 10.1021/bi7004015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Gerber LK, Aronow BJ, Matlib MA. Activation of a novel long-chain free fatty acid generation and export system in mitochondria of diabetic rat hearts. Am J Physiol Cell Physiol. 2006;291:C1198–C1207. doi: 10.1152/ajpcell.00246.2006. [DOI] [PubMed] [Google Scholar]
  • 47. Wilson CR, Tran MK, Salazar KL, et al. Western diet, but not high fat diet, causes derangements of fatty acid metabolism and contractile dysfunction in the heart of Wistar rats. Biochem J. 2007;406:457–467. doi: 10.1042/BJ20070392. This study demonstrates that the composition of the diet is of major importance for determining the genetic program regulating cardiac metabolism. The results will help to understand why the heart should adapt or maladapt in a disturbed metabolic environment.
  • 48.Boudina S, Sena S, Theobald H, et al. Mitochondrial energetics in the heart in obesity-related diabetes: direct evidence for increased uncoupled respiration and activation of uncoupling proteins. Diabetes. 2007;56:2457–2466. doi: 10.2337/db07-0481. [DOI] [PubMed] [Google Scholar]
  • 49.Song Y, Du Y, Prabhu SD, Epstein PN. Diabetic Cardiomyopathy in OVE26 Mice Shows Mitochondrial ROS Production and Divergence Between In Vivo and In Vitro Contractility. Rev Diabet Stud. 2007;4:159–168. doi: 10.1900/RDS.2007.4.159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Li L, Renier G. Activation of nicotinamide adenine dinucleotide phosphate (reduced form) oxidase by advanced glycation end products links oxidative stress to altered retinal vascular endothelial growth factor expression. Metabolism. 2006;55:1516–1523. doi: 10.1016/j.metabol.2006.06.022. [DOI] [PubMed] [Google Scholar]
  • 51.Saraiva RM, Minhas KM, Zheng M, et al. Reduced neuronal nitric oxide synthase expression contributes to cardiac oxidative stress and nitroso-redox imbalance in ob/ob mice. Nitric Oxide. 2007;16:331–338. doi: 10.1016/j.niox.2006.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Barouch LA, Gao D, Chen L, et al. Cardiac myocyte apoptosis is associated with increased DNA damage and decreased survival in murine models of obesity. Circ Res. 2006;98:119–124. doi: 10.1161/01.RES.0000199348.10580.1d. [DOI] [PubMed] [Google Scholar]
  • 53.Koya D, King GL. Protein kinase C activation and the development of diabetic complications. Diabetes. 1998;47:859–866. doi: 10.2337/diabetes.47.6.859. [DOI] [PubMed] [Google Scholar]
  • 54.Cesario DA, Brar R, Shivkumar K. Alterations in ion channel physiology in diabetic cardiomyopathy. Endocrinol Metab Clin North Am. 2006;35:601–610. ix–x. doi: 10.1016/j.ecl.2006.05.002. [DOI] [PubMed] [Google Scholar]
  • 55.Pereira L, Matthes J, Schuster I, et al. Mechanisms of [Ca2+]i transient decrease in cardiomyopathy of db/db type 2 diabetic mice. Diabetes. 2006;55:608–615. doi: 10.2337/diabetes.55.03.06.db05-1284. [DOI] [PubMed] [Google Scholar]
  • 56.Choi KM, Zhong Y, Hoit BD, et al. Defective intracellular Ca(2+) signaling contributes to cardiomyopathy in Type 1 diabetic rats. Am J Physiol Heart Circ Physiol. 2002;283:H1398–H1408. doi: 10.1152/ajpheart.00313.2002. [DOI] [PubMed] [Google Scholar]
  • 57.Lacombe VA, Viatchenko-Karpinski S, Terentyev D, et al. Mechanisms of impaired calcium handling underlying subclinical diastolic dysfunction in diabetes. Am J Physiol Regul Integr Comp Physiol. 2007;293:R1787–R1797. doi: 10.1152/ajpregu.00059.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Dillmann WH. Diabetes mellitus induces changes in cardiac myosin of the rat. Diabetes. 1980;29:579–582. doi: 10.2337/diab.29.7.579. [DOI] [PubMed] [Google Scholar]
  • 59.Razeghi P, Young ME, Cockrill TC, et al. Downregulation of myocardial myocyte enhancer factor 2C and myocyte enhancer factor 2C-regulated gene expression in diabetic patients with nonischemic heart failure. Circulation. 2002;106:407–411. doi: 10.1161/01.cir.0000026392.80723.dc. [DOI] [PubMed] [Google Scholar]
  • 60.Trost SU, Belke DD, Bluhm WF, et al. Overexpression of the sarcoplasmic reticulum Ca(2+)-ATPase improves myocardial contractility in diabetic cardiomyopathy. Diabetes. 2002;51:1166–1171. doi: 10.2337/diabetes.51.4.1166. [DOI] [PubMed] [Google Scholar]
  • 61.Nakayama H, Chen X, Baines CP, et al. Ca2+- and mitochondrial-dependent cardiomyocyte necrosis as a primary mediator of heart failure. J Clin Invest. 2007;117:2431–2444. doi: 10.1172/JCI31060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Dent CL, Bowman AW, Scott MJ, et al. Echocardiographic characterization of fundamental mechanisms of abnormal diastolic filling in diabetic rats with a parameterized diastolic filling formalism. J Am Soc Echocardiogr. 2001;14:1166–1172. doi: 10.1067/mje.2001.115124. [DOI] [PubMed] [Google Scholar]
  • 63.Fauconnier J, Andersson DC, Zhang SJ, et al. Effects of palmitate on Ca(2+) handling in adult control and ob/ob cardiomyocytes: impact of mitochondrial reactive oxygen species. Diabetes. 2007;56:1136–1142. doi: 10.2337/db06-0739. [DOI] [PubMed] [Google Scholar]

RESOURCES