Lipid metabolism and signaling in cardiac lipotoxicity Kenneth D’Souza, Carine Nzirorera, Petra C. Kienesberger PII: DOI: Reference:

S1388-1981(16)30043-9 doi: 10.1016/j.bbalip.2016.02.016 BBAMCB 57911

To appear in:

BBA - Molecular and Cell Biology of Lipids

Received date: Revised date: Accepted date:

30 December 2015 19 February 2016 19 February 2016

Please cite this article as: Kenneth D’Souza, Carine Nzirorera, Petra C. Kienesberger, Lipid metabolism and signaling in cardiac lipotoxicity, BBA - Molecular and Cell Biology of Lipids (2016), doi: 10.1016/j.bbalip.2016.02.016

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT Heart Lipid Metabolism

NU

SC

RI

PT

BBA Molecular and Cell Biology of Lipids

MA

Lipid metabolism and signaling in

AC CE P

TE

D

cardiac lipotoxicity

Kenneth D’Souza1, Carine Nzirorera1, and Petra C. Kienesberger1,*

1

Dalhousie Medicine New Brunswick, Department of Biochemistry and Molecular Biology, Dalhousie University, 100 Tucker Park Road, Saint John, New Brunswick, Canada

*

Address correspondence to: Petra C. Kienesberger, Dalhousie Medicine New Brunswick,

Department of Biochemistry and Molecular Biology, Dalhousie University, 100 Tucker Park Road, Saint John E2L 4L5, New Brunswick, Canada. Tel: +1-506-636-6971; Fax: 506-6366001. E-mail: [email protected]

1

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

Abstract The heart balances uptake, metabolism and oxidation of fatty acids (FAs) to maintain ATP

PT

production, membrane biosynthesis and lipid signaling. Under conditions where FA uptake

RI

outpaces FA oxidation and FA sequestration as triacylglycerols in lipid droplets, toxic FA metabolites such as ceramides, diacylglycerols, long-chain acyl-CoAs, and acylcarnitines can

SC

accumulate in cardiomyocytes and cause cardiomyopathy. Moreover, studies using mutant mice

NU

have shown that dysregulation of enzymes involved in triacylglycerol, phospholipid, and sphingolipid metabolism in the heart can lead to the excess deposition of toxic lipid species that

MA

adversely affect cardiomyocyte function. This review summarizes our current understanding of lipid uptake, metabolism and signaling pathways that have been implicated in the development

Highlights

AC CE P

ischemia-reperfusion.

TE

D

of lipotoxic cardiomyopathy under conditions including obesity, diabetes, aging, and myocardial



Cardiomyocyte lipid overload can have toxic effects and cause cardiac dysfunction.



Cardiac lipotoxicity is observed during pathophysiological conditions including obesity, insulin resistance, diabetes, aging, and myocardial ischemia-reperfusion.



Studies using animal models have uncovered mechanisms of cardiac lipotoxicity.



Proteins involved in FA uptake and oxidation as well as TAG, sphingolipid, and phospholipid metabolism have been implicated in cardiac lipotoxicity.

Keywords Heart; Cardiomyopathy; Lipids; Lipotoxicity; Obesity; Diabetes

2

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

Abbreviations ACSL1, long-chain acyl-CoA synthetase 1

PT

ATGL, adipose triglyceride lipase

RI

CerS5, ceramide synthase 5 CPT, carnitine palmitoyltransferase

SC

DAG, diacylglycerol

ER, endoplasmic reticulum

MA

ERK, extracellular signal regulated kinase

NU

DGAT, diacylglycerol acyltransferase

FA, fatty acid

TE

D

FABPpm, plasma membrane isoform of fatty acid binding protein FoxO1, forkhead box protein O1

AC CE P

HSL, hormone-sensitive lipase IκB, Inhibitor of NFκB IKK, IκB kinase

JNK, c-Jun N-terminal kinase

iPLA2, calcium-independent phospholipase A2 LPA, lysophosphatidic acid LPC, lysophosphatidylcholine LPL, lipoprotein lipase MAPK, mitogen activated protein kinases MLK3, mixed lineage kinase-3 MFBD, milk fat-based high fat diet NFκB, nuclear factor-κB 3

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

PGC1, peroxisome proliferator-activated receptor γ-coactivator 1 PIP2, phosphatidylinositol-4,5-bisphosphate

PT

PKC, protein kinase C

RI

PP2A, protein phosphatase 2A PPAR, peroxisome proliferator-activated receptor

SC

ROS, reactive oxygen species

NU

SAPK, stress activated protein kinase S1P, sphingosine-1-phosphate

MA

TAG, triacylglycerol TNFα, tumor necrosis factor α

D

TLR4, toll-like receptor 4

AC CE P

TE

VLDLR, very low density lipoprotein receptor

4

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

1. Introduction Lipotoxicity is defined as the process where excess accumulation of lipids and over-

PT

activation of lipid signaling pathways trigger cellular distress and dysfunction, which may

RI

manifest as insulin resistance, defective mitochondria, energy starvation, and endoplasmic reticulum stress and may ultimately lead to apoptotic cell death or lipoapoptosis (Figure 1) (1).

SC

When this process occurs in cardiomyocytes, the consequences can be cardiac dysfunction and

NU

heart failure, which is also referred to as lipotoxic cardiomyopathy (1,2). The archetypal conditions triggering a lipotoxic milieu within cardiomyocytes are metabolic disorders such as

MA

obesity, insulin resistance, and diabetes mellitus (3-6). In addition, cardiac lipotoxicity has also been observed during other patho/physiological conditions, e.g. myocardial ischemia-

TE

D

reperfusion/infarction and aging (7-9). Increased myocardial lipid deposition develops when cardiomyocyte fatty acid (FA) uptake outpaces FA oxidation, leading to an increased availability

AC CE P

of FAs for non-oxidative metabolic pathways and accumulation of cardiotoxic FA metabolites (3). In the case of obesity, insulin resistance, and diabetes the main culprit is excess FA delivery due to augmented dietary fat intake and adipose tissue lipolysis (10). In addition, myocardial insulin resistance and impaired glucose utilization may further increase myocardial FA uptake and accumulation of toxic lipid species (11). While insulin resistance is likely a key contributing factor to obesity and diabetes-induced lipotoxic cardiomyopathy by causing a shift in cardiac substrate utilization from glucose to FAs and concomitant FA overload, it is also a consequence of cardiac lipotoxicity since toxic FA metabolites can impair myocardial insulin signaling (12,13). Due to its limited ability to synthesize FAs, the myocardium relies heavily on the uptake of FAs in the form of albumin-bound free FAs or lipoproteins - chylomicrons and very-low-

5

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

density-lipoproteins - from the circulation to sustain cardiomyocyte lipid homeostasis and energy metabolism (14). In fact, uptake of FAs is a major determinant of FA oxidation and therefore

PT

mitochondrial ATP production (15-17). Although FAs are the principal energy substrate for the

RI

healthy adult heart, cardiomyocytes are omnivorous, deriving their ATP for contractile work from a variety of sources including glucose, lactate, and amino acids, allowing for metabolic

SC

flexibility to adjust substrate utilization to circulating substrate concentrations, work load,

NU

hormonal stimuli and oxygen supply (3,18,19). In contrast, diseased hearts often have decreased metabolic flexibility, which can contribute to cardiac dysfunction and lipotoxic cardiomyopathy

MA

(19-21).

To date, numerous rodent models have been generated and studied that were instrumental

TE

D

for the characterization of lipotoxic cardiomyopathy and using which lipid metabolic and signaling pathways were identified that mediate cardiomyocyte lipotoxicity (for a list of mouse

AC CE P

models of cardiac lipotoxicity see Goldberg et al. (3)). In addition, it has also been revealed that the fruit fly can exhibit lipotoxic cardiomyopathy when fed a high fat diet, suggesting that the molecular mechanisms that lead to toxic lipid overload of the myocardium are evolutionarily conserved (22). It has become evident that chronically increased accumulation of certain FA metabolites in cardiomyocytes and the ensuing pathological sequelae are sufficient to precipitate cardiac dysfunction in these animal models. The FA metabolites linked to cardiac lipotoxicity include

ceramides,

diacylglycerols

(DAGs),

long-chain

acyl-CoAs,

acylcarnitines,

lysophospholipids, and triacylglycerols (TAG), although it is believed that the latter represents a marker of lipotoxicity rather than being directly cytotoxic (Figure 2) (4,23). Changes in FA uptake, storage, and/or oxidation in animal models of lipotoxic cardiomyopathy influence more than one lipid metabolism and signaling pathway, which renders it difficult to pinpoint a specific

6

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

FA metabolite causative for cardiomyocyte dysfunction. It is unlikely to be the excess deposition of a single FA metabolite and over-activation of a single lipid signaling pathway, but rather a

PT

combination of these that triggers cardiac dysfunction in animal models of lipotoxic

RI

cardiomyopathy.

The etiology and functional relevance of cardiac lipotoxicity is less clear in the human

SC

heart when compared to animal models of lipotoxic cardiomyopathy. In this regard, the

NU

development of proton magnetic resonance spectroscopy for the noninvasive quantification of intramyocardial TAG in vivo was an important contribution towards the understanding of the

MA

relationship between increased cardiac lipid deposition and cardiac function in humans (24-27). Using this technique, it has been demonstrated that increased myocardial TAG deposition is not

TE

D

only associated with but precedes cardiac dysfunction in humans with type 2 diabetes mellitus (28) and correlates with body mass index (29,30). These findings indicate that there is a

AC CE P

causative relationship between cardiac lipid accumulation and impaired myocardial contractility. Moreover, it has been suggested that cardiac steatosis and ensuing lipotoxic cardiomyopathy are widespread and clinically highly relevant given the global rise in the prevalence of obesity and metabolic complications (30).

It is currently poorly understood whether the cardiac accumulation of lipids other than TAGs, which are believed to contribute to cardiac lipotoxicity in animal models, are altered in the human heart during obesity, insulin resistance, diabetes, and other conditions associated with lipotoxic cardiomyopathy. Nevertheless, it is likely that increased TAG levels in the human heart are a marker for overall increased cardiac content of toxic lipid metabolites and the dysregulation of lipid metabolism and signaling pathways negatively impacting cardiac metabolism and function. The goal of this review is to provide an overview of metabolic pathways and proteins

7

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

involved in the regulation of cardiomyocyte lipid levels and signaling that have been implicated in the development of lipotoxic cardiomyopathy.

PT

2. Cardiac fatty acid delivery and uptake

RI

The major sources of FAs for cardiomyocytes are albumin-bound free FAs and FAs released from lipoprotein TAGs either at the coronary lumen via lipoprotein lipase (LPL) or following

SC

receptor-mediated endocytosis. Uptake of these FAs occurs either via passive diffusion or

NU

protein carrier-mediated transport by fatty acid translocase (FAT)/CD36, the plasma membrane isoform of fatty acid binding protein (FABPpm), and fatty acid transport protein (FATP) 1/4/6

MA

(Figure 2)(18). Using positron emission tomography imaging, it has been demonstrated that increased postprandial FA uptake in hearts from overweight and obese subjects with impaired

TE

D

glucose tolerance is associated with early impairment of left ventricular function and increased myocardial oxidative metabolism (31). The latter findings indicate that chronically enhanced

AC CE P

myocardial FA uptake in the setting of obesity precipitates lipotoxic cardiomyopathy in humans. A number of studies using rodent models have provided more mechanistic insight into how increased myocardial FA uptake can lead to excess accumulation of lipid metabolites and trigger lipotoxic cardiomyopathy (32-34). Cardiac FA transport was enhanced in obese-diabetic Zucker rats, which was paralleled by an increase in sarcolemmal FA transporters CD36 and FABPpm (35). Cardiomyocytes from obese-diabetic Zucker rats displayed increased CD36-mediated longchain FA uptake and TAG accumulation that preceded contractile dysfunction, suggesting that enhanced sarcolemmal expression of CD36 promotes diabetes-mediated cardiac lipotoxicity (36,37). Moreover, adult rat cardiomyocytes displayed increased sarcolemmal CD36 protein expression upon incubation with media mimicking obese/diabetic conditions (high insulin, high palmitate concentrations), which was associated with enhanced basal palmitate uptake, lipid

8

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

accumulation, and decreased sarcomere shortening (38). Inhibition of CD36 in these cells protected from increased FA uptake, TAG deposition, and contractile dysfunction (38),

PT

demonstrating that CD36-mediated FA uptake plays a key role in cardiomyocyte lipotoxicity

RI

triggered by an obese/diabetic milieu.

CD36 has also been implicated in aging-induced lipotoxic cardiomyopathy (8). Aged

SC

mice displayed increased cardiac CD36 expression when compared to young mice, which was

NU

paralleled by increased cardiac lipid accumulation, decreased cardiac function, and hypertrophic remodeling (8). CD36 deficient mice were resistant to aging-induced lipotoxic cardiomyopathy

MA

(8). A role of CD36 in facilitating lipotoxic cardiomyopathy has also been shown in mice with cardiomyocyte-specific over-expression of peroxisome proliferator-activated receptor α

D

(PPARα) (39), a lipostat which increases transcription of an array of genes involved in FA

TE

transport and utilization in the diabetic heart (40). Chronic activation of PPARα in hearts of these

AC CE P

mice augmented FA uptake that exceeded FA oxidation resulting in increased myocardial lipid (TAG) deposition and systolic dysfunction, which was exacerbated by feeding a high fat diet rich in long-chain FAs or induction of diabetes (33,40). CD36 deficiency protected against cardiac steatosis and cardiomyopathy in PPARα over-expressing mice, suggesting that inhibition of CD36 may be an effective therapeutic intervention for diabetes-related lipotoxic cardiomyopathy (39). Indeed, because of its key role in cardiac FA uptake and lipotoxicity, CD36 is being explored as therapeutic target to ameliorate lipotoxic cardiomyopathy and insulin resistance via immunochemical inhibition of sarcolemmal CD36 and interference with the subcellular recycling of CD36 (41). Similar to the deletion of CD36, deficiency of LPL rescued myocardial TAG accumulation and contractile dysfunction in PPARα over-expressing mice (42). While these studies show that CD36 and LPL are required for lipotoxic cardiomyopathy in PPARα

9

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

over-expressing hearts, the identity of the toxic FA metabolite(s) mediating these effects remains unclear.

PT

In addition to PPARα over-expression, forced expression of FATP1 specifically in

RI

cardiomyocytes resulted in increased FA uptake and lipotoxic cardiomyopathy that was associated with increased accumulation of free FAs but not TAGs, and diastolic dysfunction

SC

(32). Cardiac FA uptake was also upregulated indirectly by over-expressing long-chain acyl-CoA

NU

synthetase 1 (ACSL1), leading to increased deposition of TAGs, ceramides, and certain phospholipid species, lipoapoptosis and systolic dysfunction (2). Moreover, an important role in

MA

lipotoxicity following myocardial infarction has been ascribed to the very-low-density lipoprotein receptor (7). Specifically, decreasing FA uptake via very-low-density lipoprotein

TE

D

receptor deficiency protected from ischemia-induced TAG accumulation, endoplasmic reticulum stress, and apoptosis and improved survival in mice subjected to myocardial infarction (7).

AC CE P

Taken together, these findings show that multiple proteins involved in cardiomyocyte FA uptake are implicated in the development of cardiac lipotoxicity and cardiomyopathy during conditions including obesity/diabetes, aging, and myocardial infarction.

3. TAG synthesis and mobilization It has been reported that a significant proportion, if not the majority, of FAs that enter the cardiomyocyte are shuttled through the intracellular TAG pool for temporary storage prior to oxidation (43). While TAGs per se are generally believed to be non-toxic, sequestration and release of FAs via intracellular TAG metabolism is likely a major determinant of toxic FA and FA metabolite accumulation (Figure 2). The concept that TAG accumulation can protect from FA-induced lipotoxicity by sequestering FAs that could otherwise stimulate adverse signaling

10

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

events was first established by Listenberger et al. (44) using non-cardiac cells. Specifically, while incubation of cells with excess palmitate, a saturated FA which poorly incorporates into

PT

TAGs, led to lipotoxic cell death, co-incubation with the unsaturated FA oleate mitigated

RI

palmitate-induced lipotoxicity by diverting palmitate away from apoptosis-stimulating pathways into the TAG pool (44). Furthermore, incubation of cells with oleate induced lipotoxicity when

SC

TAG synthesis was impaired (44). These studies show that sequestration of FAs in TAGs can

NU

rescue lipotoxic effects of saturated FAs. A recent study by Bosma et al. (45) highlights that this concept also applies to cardiomyocytes. In line with this notion, cardiac-specific over-expression

MA

of diacylglycerol acyltransferase (DGAT) 1, which catalyzes the final step in TAG synthesis, increased cardiac TAG content ~2-fold and decreased the accumulation of diacylglycerols

TE

D

(DAGs), ceramides, and free FAs without affecting heart function at baseline (46). The cardiac phenotype of DGAT1 over-expressing mice is reminiscent of changes in cardiac lipid

AC CE P

accumulation that occur following exercise (46). Furthermore, forced DGAT1 expression ameliorated lipotoxic cardiomyopathy in mice with cardiomyocyte-specific over-expression of either ACSL1 or PPARγ, which was associated with decreased cardiac DAG and ceramide levels, but unchanged TAG accumulation (46,47). However, a study using another cardiacspecific DGAT1 over-expression model reported that DGAT1-induced cardiac steatosis was associated with cardiac dysfunction, as was evidenced primarily by impaired diastolic function (48). Diastolic dysfunction was accompanied by increased cardiac fibrosis and decreased mitochondrial biogenesis (48). Despite increased myocardial TAG levels, cardiac content of free FAs and DAGs was unchanged in this model (48). The reasons for the divergent effects of DGAT1 over-expression on myocardial lipid levels, most notably free FAs and DAGs, and cardiac function are presently unclear, but may involve differences in background strain or

11

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

transgene construct design. Cardiomyocyte-specific DGAT1 knockout mice generated by Lui et al. (49) developed heart failure and died prematurely (49). Hearts from these mice exhibited

PT

severely increased cardiac DAG and ceramide levels (49), suggesting that decreased

RI

sequestration of toxic FA metabolites in TAGs due to DGAT1 deficiency in the myocardium produces a lipotoxic milieu that contributes to heart failure. Interestingly, this was associated

SC

with increased PPARα and CD36 expression leading to enhanced myocardial FA uptake (49),

NU

suggesting that cardiomyocyte-specific DGAT1 deficiency not only reduces the ability to sequester toxic lipid metabolites in TAGs but also leads to increased FA uptake which further

MA

exacerbates lipotoxic cardiomyopathy. Surprisingly, global DGAT1 knockout mice did not exhibit cardiac lipotoxicity or dysfunction (50). Expression of PPAR and genes involved in FA

TE

D

utilization, including CD36, was reduced in the heart from DGAT1 knockout mice, which was accompanied by decreased myocardial FA oxidation and increased glucose uptake (50). These

AC CE P

findings suggest that global DGAT1 deficiency leads to a shift in substrate utilization away from FAs to glucose, thus preventing lipotoxicity. The presence or absence of DGAT1 in the intestine may contribute to the differences in cardiac phenotypes observed between global versus cardiomyocyte-specific DGAT1 knockout mice (49,51). Intestinal DGAT1 stimulates dietary fat secretion out of enterocytes and therefore influences the availability of dietary fats in tissues including the heart (51). As such, mice expressing DGAT1 in the intestine only negated the protection from diet-induced hepatic steatosis and obesity in global DGAT1 knockout mice (51). Moreover, when cardiomyocyte-specific DGAT1 knockout mice were crossed with intestinal DGAT1 knockout mice, heart function and survival improved, which was associated with a reduction in the accumulation of DAGs and ceramides in the myocardium of double-knockout mice (49). DGAT1 expression in the heart is markedly decreased in humans with heart failure,

12

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

which is associated with increased cardiac accumulation of DAGs and ceramides - reminiscent of cardiac-specific DGAT1 knockout mice - and reduced levels of TAGs and FAs (52). Unlike

PT

the cardiomyocyte-specific DGAT1 knockout mouse, expression of PGC1α, CD36, and other

RI

proteins involved in lipid metabolism was decreased in the failing human heart (52). It is possible that the degree of DGAT1 inhibition (knockout in mice versus partial deficiency in

SC

humans), the duration of DGAT1 inhibition, developmental effects of DGAT1 deficiency in

NU

knockout mice, and/or species-dependant mechanisms contribute to this discrepancy. Compared to DGAT1, relatively little information is currently available about the role of

MA

glycerol-3-phosphate acyltransferases (53) and lipins/phosphatidate phosphatases (54), which are key enzymes in glycerolipid biosynthesis upstream of DGAT, in modulating myocardial lipid

D

levels and lipotoxic cardiomyopathy. Lipin1 was identified as the major lipin isoform in the heart

energy

metabolism

and

lipotoxicity.

Lipin1

expression

and

phosphatidate

AC CE P

cardiac

TE

and was found to be regulated by PGC1α/estrogen related receptor (55), important modulators of

phosphohydrolase activity were downregulated in the failing heart from TG9 mice (55). Interestingly, lipin mRNA expression and phosphatidate phosphohydrolase 1 activity was also reduced in Zucker diabetic fatty rats and type 2 diabetic patients (56). Insulin therapy led to increased lipin expression and phosphatidate phosphohydrolase 1 activity (56). Likewise, phosphatidate phosphohydrolase 1 and 2 activity was decreased in the heart from obese-insulin resistant JCR:LA corpulent rats (57). Taken together, these studies suggest that lipins may be involved in lipotoxic cardiomyopathy induced by insulin resistance-diabetes. The first and rate-limiting step in TAG hydrolysis in the heart is mediated by adipose triglyceride lipase (ATGL/PNPLA2) (58,59). Loss of ATGL function in mice either via deletion of ATGL or its co-activating protein CGI-58 led to massive cardiac TAG accumulation and

13

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

precipitated heart failure (58-60). Cardiac steatosis was accompanied by fibrotic and hypertrophic remodeling and a depression of mitochondrial FA oxidation in ATGL knockout

PT

mice (58,59). The latter was due to a reduction in PPARα and PPARγ-coactivator (PGC) 1 activation in hearts from constitutive whole body and muscle-specific ATGL deficient mice (58),

RI

although a drastic decrease in PPARα signaling was not obvious in inducible cardiomyocyte-

SC

specific ATGL deficient mice (59). It is likely that, irrespective of changes in lipid signaling,

NU

mitochondrial function and metabolism that compromise cardiac contractility, the excessive deposition of TAG-filled lipid droplets per se in ATGL deficient hearts is mechanically

MA

obstructing and impairs proper contractile functioning of cardiomyocytes. Mutations in the ATGL/PNPLA2 gene have also been linked to heart failure in humans, constituting a new clinical

TE

D

entity termed “triglyceride deposit cardiomyovasculopathy” (61,62). TAG content in hearts from humans with defective ATGL is also massively (~upwards of 30-times) increased (62), similar to

AC CE P

mice with ATGL deficiency. In contrast to ATGL knockout mice, patients with PNPLA2 mutations had increased expression of PPARα, PPARγ, and CD36 in the heart, suggesting that enhanced lipid uptake and TAG synthesis further augment myocardial TAG deposition and contribute to heart failure in these individuals (62). It is presently unclear whether levels of lipids other than TAGs, which are typically associated with lipotoxicity, are changed in ATGL deficient mice and patients with mutated PNPLA2. These studies suggest that although TAGs can sequester toxic FAs and lipid intermediates in lipid droplets, massive TAG accumulation in the heart beyond a certain threshold may also lead to insufficient mechanical contraction and contribute to heart failure. While lack of ATGL function leads to heart failure in mice and humans, mice with cardiac-specific over-expression of ATGL were protected from type 1 diabetes-induced

14

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

augmentation of cardiac TAG content and lipotoxic cardiomyopathy (63). Constitutive ATGL over-expression not only prevented an increase in myocardial TAG deposition in streptozotocin-

PT

diabetic mice, but also prevented diabetes-induced increases in palmitoyl-CoA and ceramide

RI

accumulation and reduced PPARα and PPARα target gene expression (63). Interestingly, in human cardiomyocyte-derived AC16 cells, adenoviral overexpression of ATGL reduced

SC

intracellular TAG storage and increased free FA levels, which was associated with stimulation of

NU

ER stress (45); these findings suggest that acute ATGL activation can trigger lipotoxicity in cardiomyocytes due to the inability of cells to sequester FAs in lipid droplets (45) while chronic

MA

ATGL activation in vivo protects from lipotoxicity likely due to counter-regulatory downregulation of PPARα and overall lipid utilization (63-65). Similar to chronic ATGL

TE

D

overexpression in type 1 diabetic mice, cardiac-specific over-expression of hormone-sensitive lipase (HSL), which hydrolyzes predominantly DAGs and, to a lesser extent, TAGs, prevented

AC CE P

cardiac steatosis, fibrosis, and cardiomyopathy due to streptozotocin-induced diabetes (66). Moreover, it has been demonstrated using a high fat diet-fed Drosophila obesity model that cardiac-specific over-expression of the ATGL ortholog, Brummer lipase, prevents obesityinduced cardiac TAG accumulation and cardiac dysfunction (22). A similar effect was also achieved upon inhibition of lipogenesis and stimulation of lipolysis via impairment of the insulin-target of rapamycin pathway in hearts from obese Drosophila (22). These findings suggest that regulation of cardiac TAG catabolism via ATGL and HSL modulates cardiac lipotoxicity under conditions of obesity and diabetes, although the precise mechanisms for this effect remain to be fully uncovered. Recent studies also showed that cardiomyocyte-specific over-expression of perilipin 5, a lipid droplet coating protein that connects lipid droplets with mitochondria and limits

15

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

intracellular TAG catabolism, resulted in cardiac steatosis (67,68), reduced PPARα activation (67,68), mitochondrial dysfunction (68), left ventricular hypertrophy (67,68), and mild

PT

impairment of cardiac function (68). The absence of more severe cardiac TAG accumulation and

RI

dysfunction, similar to that observed following ATGL ablation, in mice with perilipin 5 overexpression has been ascribed to increased protein kinase A-mediated perilipin 5

SC

phosphorylation, allowing for some lipolysis to occur (69). Deficiency of perilipin 5 led to the

NU

depletion of cardiac TAGs due to excess lipolysis, increased cardiac FA oxidation, and formation of reactive oxygen species (ROS) that contributed to age-related cardiomyopathy, suggesting that

MA

perilipin 5 function needs to be finely balanced to maintain cardiac TAG homeostasis and protect from lipotoxicity (70).

TE

D

Apart from the aforementioned TAG metabolic pathways that are centered on the formation and degradation of cytosolic lipid droplets, the heart is also able to synthesize and

AC CE P

secrete TAGs in the form of lipoproteins (14,71,72). This TAG secretion pathway is upregulated in the heart in diet-induced obesity via increased microsomal triglyceride transfer protein expression (71), which has been suggested to limit cardiac TAG accumulation. Moreover, activation of cardiomyocyte lipoprotein secretion via over-expression of apolipoprotein B abolished cardiac TAG accumulation in high fat diet-fed and streptozotocin-diabetic mice and protected against obesity/diabetes-induced cardiac dysfunction (71). Similarly, apolipoprotein B overexpression protected against lipotoxic cardiomyopathy induced by cardiomyocte-specific over-expression of glycosylphosphatidylinositol-anchored human lipoprotein lipase (LpLGPI) (73). Taken together, these studies suggest that the metabolism of TAGs in lipid droplets and TAG secretion via lipoproteins play a major role in the pathogenesis of lipotoxic cardiomyopathy.

16

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

4. Mitochondrial FA metabolism

PT

To enable the transport of long-chain acyl-CoAs through the mitochondrial double-membrane,

RI

they are converted to acylcarnitines by carnitine palmitoyltransferase (CPT) 1 at the outer mitochondrial membrane (Figure 2). After passing the inner mitochondrial membrane,

SC

acylcarnitines are converted back to acyl-CoAs by CPT2 and subjected to β-oxidation for ATP

NU

production. CPT1β is the predominant CPT1 isoform in the heart (74). Although mice with heterozygous CPT1β deficiency exhibited unchanged myocardial FA oxidation and no overt

MA

cardiac phenotype at baseline, decreased FA oxidation and increased TAG and ceramide accumulation were evidenced following transverse aortic constriction-induced pressure overload

D

(74). Additionally, cardiac hypertrophy, cardiomyocyte apoptosis, and cardiac dysfunction were

TE

exacerbated in these mice, suggesting that partial CPT1β deficiency causes myocardial

AC CE P

lipotoxicity following hemodynamic stress (74). Mice with homozygous cardiac- and skeletal muscle-specific CPT1β deficiency died prematurely from heart failure even in the absence of hemodynamic stress (75). Hearts from these mice also exhibited massive hypertrophy and increased neutral lipid accumulation as was assessed by Oil Red O staining. Although this increase in lipid accumulation is indicative of lipotoxicity, levels of specific toxic lipids have not been assessed (75). While targeted CPT1β deletion in mice appears to cause lipotoxic cardiomyopathy (75), inhibition of CPT1 and CPT2 using the antianginal drug, perhexiline, led to improved left ventricular ejection fraction, resting and peak stress myocardial function, oxygen consumption and improved skeletal muscle energetics in patients with chronic heart failure (76). The proposed mechanism for this effect is that by inhibiting FA utilization, perhexiline induces a metabolic shift toward utilization of glucose and lactate for ATP

17

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

production (76). Perhexiline treatment may thereby improve insulin sensitivity and oxygenefficiency in the failing heart (76). The discrepancy between the detrimental effect of CPT1β

PT

deletion in mice and the beneficial effect of pharmacological targeting of CPT1/2 using

RI

perhexiline in humans with heart failure could be due to differing degrees and duration of CPT1 inhibition, species-specific effects of CPT1 inhibition, non-specific effects of perhexiline and/or

SC

pre-existing cardiac morbidity (63). Notably, while short-term (8 days) inhibition of CPT1 using

NU

etomoxir did not result in excess cardiac lipid accumulation in rats, 10-day administration of etomoxir precipitated cardiomyopathy in mice (75,77,78).

MA

Cardiomyocyte-specific ablation of PPARδ, a transcriptional regulator of FA oxidation, resulted in decreased basal myocardial FA oxidation, which was accompanied by increased

TE

D

myocardial TAG deposition, cardiac hypertrophy and heart failure, suggestive of lipotoxic cardiomyopathy (79). Moreover, mice with deficiency of long-chain acyl-CoA dehydrogenase,

AC CE P

which initiates FA β-oxidation, had increased myocardial TAG content at baseline and, more so, following a 24-h fast, and exhibited elevated cardiac ceramide levels and systolic dysfunction in the fasted state (80). Conversely, cardiomyocyte-specific over-expression of pyruvate dehydrogenase kinase 4, an inhibitor of pyruvate dehydrogenase, increased myocardial FA oxidation and activated cardiac expression of PGC1α, a regulator of mitochondrial biogenesis and function, and prevented increased TAG accumulation following short-term (4 weeks) high fat diet feeding (81). Taken together, these studies suggest that insufficient FA oxidation can contribute to lipotoxic cardiomyopathy by producing an environment wherein uptake of FAs outstrips their degradation and oxidative metabolism, leading to cardiomyocyte lipid accumulation.

18

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

Apart from lipotoxic effects associated with impaired mitochondrial FA import and βoxidation, it has also been proposed that cardiac lipotoxicity can be induced by conditions where

PT

mitochondrial FA β-oxidation exceeds tricarboxylic acid cycle capacity and subsequent oxidative

RI

phosphorylation, leading to the accumulation of acylcarnitines (82,83). For instance, drastically increased myocardial acylcarnitine accumulation manifests during myocardial ischemia, wherein

SC

lack of oxygen supply and ensuing accrual of reducing equivalents prevent complete FA

NU

oxidation (82,83). While the increased conversion of mitochondrial FA β-oxidation intermediates may be initially protective, in that it prevents excess deposition of potentially cytotoxic acyl-

MA

CoAs and other lipid metabolites (84), acylcarnitines themselves have been shown to disrupt sarcolemmal integrity and electrophysiologic functions, possibly leading to myocardial

TE

D

arrhythmias (82,83), which suggests that acylcarnitines are potent cardiolipotoxins. It is important to note that during myocardial ischemia, accumulation of acylcarnitines reflects

AC CE P

impaired FA oxidation; specifically, long-chain acylcarnitines are increased while short-chain acylcarnitines are reduced in the ischemic heart (85). On the contrary, obesity/diabetes results in an increase in the accumulation of both short- and long-chain acylcarnitines caused by enhanced myocardial FA delivery and mitochondrial FA import (83,86,87). Su et al. (83) observed a several-fold increase in the concentrations of acylcarnitines, particularly long-chain acylcarnitines and 3-hydroxy acylcarnitines, in the streptozotocin-diabetic rat heart, which could be reversed by insulin treatment (83). 3-Hydroxy acylcarnitine accumulation in the diabetic heart is suggestive of incomplete FA β-oxidation due to substrate overload and demonstrates that acylCoA and acylcarnitine pools can be reversibly converted (83). These findings also suggest that 3hydroxy acyl-CoA dehydrogenase is rate-limiting for myocardial β-oxidation (83). Interestingly, calcium-independent phospholipase A2 (iPLA2) activity was increased in the ischemic and

19

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

diabetic heart and, by liberating FAs from phospholipids, was found to contribute substantially to the pool of FAs converted to myocardial acylcarnitines (83). Consistent with the notion that

PT

acylcarnitine accumulation is cardiotoxic, improvement of cardiac function in mice with cardiac-

RI

specific over-expression of PPARγ, which develop lipotoxic cardiomyopathy, by PPARα deletion was associated with decreased cardiac acylcarnitine levels (88). Surprisingly, these

SC

PPARα/PPARγ double-mutant mice exhibited increased FA oxidation and lipid droplet size,

NU

while cardiac TAG, DAG, and ceramide levels were unchanged when compared to the PPARγ over-expressors (88). The reasons for the paradoxical increase in FA oxidation and decrease in

MA

acylcarnitine concentrations upon superimposition of PPARα deficiency on the PPARγ transgenic background are puzzling. It is possible, however, that the double mutants have

TE

D

improved mitochondrial FA oxidation capacity, which limits incomplete FA oxidation events and acylcarnitine accumulation and thus ameliorates lipotoxic cardiomyopathy induced by

AC CE P

PPARγ over-expression.

Another transcription factor which regulates mitochondrial FA metabolism in the heart and which has more recently been linked to lipotoxic cardiomyopathy is forkhead box protein O1 (FoxO1) (89). FoxO1 activity is increased in the hearts from mice with diabetes induced either genetically or by high fat diet feeding (89). Cardiomyocyte-specific FoxO1 ablation prevented high fat diet-induced cardiomyopathy and cardiac insulin resistance, which was associated with reduced mitochondrial FA oxidation and increased glucose utilization (89). Deletion of FoxO1 also protected from myocardial lipid accumulation in mice with high fat diet-induced obesity, as was assessed by TAG measurements and Oil Red O staining of cardiac sections (89). In addition, overexpression of constitutively active FoxO1 in neonatal cardiomyocytes increased inhibitory serine phosphorylation of insulin receptor substrate 1, which impairs insulin signal transduction

20

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

(89). It is conceivable that FoxO1 contributes to obesity/diabetes-induced cardiac lipotoxicity by not only increasing myocardial insulin resistance and FA oxidation but overall FA uptake and

PT

utilization, leading to the accumulation of toxic lipid species.

RI

By producing ATP via oxidative phosphorylation, mitochondria are the principal source of ROS in cardiomyocytes (90). Leakage of electrons from complexes I and III in the electron

SC

transport chain leads to the continuous production of superoxide, which can react with nitric

NU

oxide to form the highly reactive peroxynitrite (90). Peroxynitrite and ROS then target phospholipids in the mitochondrial membrane to generate lipid peroxides (90). Cardiolipin, an

MA

abundant phospholipid in the mitochondrial membrane critically involved in mitochondrial function, is a primary target of lipid peroxidation since a large proportion of FAs contained in

TE

D

cardiolipin are unsaturated linoleic acid (90,91). Lipid peroxides that evade the antioxidant response can subsequently be converted to reactive aldehydes, which have the potential to

AC CE P

modulate membrane properties and impair the function of targeted proteins (90). Outcomes of lipid peroxidation product accumulation include calcium overload, increased mitochondrial uncoupling, and DNA mutagenesis, which can precipitate cardiac dysfunction. Increased accumulation of lipid peroxidation products has been observed in diabetic cardiomyopathy, myocardial infarction, and heart failure, suggesting that lipotoxicity induced by lipid peroxidation products may contribute to cardiomyopathies associated with increased oxidative stress (90). Further research is warranted to provide definitive evidence for a mechanistic link between myocardial lipid peroxidation product accumulation and lipotoxic cardiomyopathy.

5. Cardiomyocyte phospholipid metabolism and signaling

21

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

Phospholipids have vital biological roles as structural components of membranes, modulating the function of membrane-associated proteins, and signaling messengers. It is not surprising then

PT

that phospholipid and energy metabolism are interlinked and that alterations in phospholipid

RI

homeostasis can cause myocardial lipotoxicity (92-96). For example, the widely used antianginal and cardioprotective compound trimetazidine not only partially blocks FA oxidation by

SC

inhibiting 3-ketoacyl-coenzyme A thiolase, the final enzyme in β-oxidation, but also stimulates

NU

phospholipid synthesis and turnover (94). In doing so, trimetazidine may alter membrane properties and indirectly lower FA availability for mitochondrial oxidation by promoting

MA

sequestration of FAs in phospholipids (94). Interestingly, in the streptozotocin-diabetic rat heart, trimetazidine treatment protected from adverse remodeling of mitochondrial membrane FA

TE

D

composition and partially prevented a diabetes-induced decline in creatine kinase functional activity, suggesting that trimetazidine ameliorates diabetes-related mitochondrial dysfunction

AC CE P

and membrane lipotoxicity via sustained phospholipid homeostasis (94). A particularly bioactive phospholipid, lysophosphatidylcholine (LPC), has been identified as potently toxic upon its accumulation in the heart (93). LPCs are formed by the phospholipase A-mediated release of one of the two FAs contained in phospholipids either in the sn-1 or sn-2 position (97). LPCs have pro-inflammatory and pro-arrhythmogenic properties and myocardial accumulation of LPCs is associated with mitochondrial dysfunction (93). Increased LPC accumulation was evidenced during atherosclerosis and myocardial ischemia and hypoxia (93,97,98). By modulating cardiac ion channel function, LPC accumulation has been shown to contribute to cardiac arrhythmia and sudden cardiac death (93). Interestingly, mice lacking PGC1β, an important regulator of mitochondrial biogenesis and energy metabolism, exhibited increased cardiac concentrations of LPCs, presenting an example where mitochondrial

22

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

dysfunction is linked to alterations in (lyso)phospholipid metabolism (93). Lysophospholipid accumulation in PGC1β deficient hearts was associated with ion channel remodeling and

PT

deranged calcium handling, causing ventricular arrhythmia (93).

RI

Cardiac ischemia activates calcium-independent phospholipase A2 (iPLA2), which metabolizes phospholipids to produce FAs and lysophospholipids (99). When hearts from

SC

transgenic mice with cardiomyocyte-specific iPLA2β over-expression were subjected to

NU

ischemia, a drastic increase in free FAs released into the venous eluent and elevated LPC concentrations in ischemic zones were observed, which was accompanied by ventricular

MA

tachyarrhythmias (99). These findings suggest that excessive hydrolysis of membrane phospholipids by iPLA2β can trigger malignant ventricular tachyarrhythmias during acute

TE

D

cardiac ischemia (99). These changes were not observed in wild type hearts, which has been attributed to the fact that the murine myocardium generally contains extremely low amounts of

AC CE P

iPLA2 activity when compared to the human heart (99). Besides iPLA2β, iPLA2γ has also been linked to myocardial lipotoxicity and dysfunction (100). Mice with cardiomyocyte-specific iPLA2γ over-expression exhibited not only a drastic decrease in myocardial phospholipid content but also a pronounced increase in TAG levels and changes in TAG molecular species composition following brief caloric restriction, as well as fasting-induced hemodynamic dysfunction (100). Moreover, iPLA2γ over-expressing hearts had markedly increased lysophosphospholipid and acylcarnitine content (100), suggesting that over-active iPLA2γ and ensuing phospholipid breakdown in the myocardium leads to cardiac lipotoxicity. Arachidonic acid is sequestered in phospholipids and can be liberated upon activation of iPLA2s in the heart (95). In fact, the release of arachidonic acid by iPLA2s constitutes the ratelimiting step in the generation of eicosanoids, which is mediated by the subsequent arachidonic

23

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

acid oxidation via lipooxygenases, cyclooxygenases, and cytochrome P450 (95). Cardiac eicosanoid signaling plays important physiological and pathological roles by activating protein

PT

kinases and ion channels and is implicated in myocardial infarction, arrhythmogenesis, and

RI

hypertrophic remodeling (101). Therefore, it is likely that iPLA2 activation can cause cardiac lipotoxicity not only by producing excess lysophospholipids, but also by promoting adverse

NU

SC

eicosanoid accumulation and signaling.

6. Cardiac sphingolipid metabolism

MA

Ceramides are the most widely studied sphingolipids involved in lipotoxic cardiomyopathy. Cardiomyocytes can either synthesize ceramides or acquire these lipids from circulating

TE

D

lipoproteins. De novo synthesis of sphingolipids, including ceramides, is initiated by serine palmitoyltransferase (SPT) (Figure 2)(102). Alternatively, ceramides can also be generated via

AC CE P

sphingomyelinase or ceramide synthases (102), which can be modulated by Sirt3-mediated deacetylation (103). Cardiac ceramide levels were increased in mice with cardiomyocyte-specific over-expression of a glycosylphosphatidylinositol-anchored human lipoprotein lipase (LpLGPI), which develop dilated lipotoxic cardiomyopathy secondary to excess FA uptake (104). Consistent with the notion that increased ceramide accumulation is cytotoxic, incubation of a human cardiomyocyte cell line with C6-ceramide resulted in upregulation of the heart failure/hypertrophy markers, atrial natriuretic peptide and brain natriuretic peptide (104). A role of ceramide accumulation in lipotoxic cardiomyopathy in vivo was shown by inhibiting the ratelimiting enzyme of ceramide biosynthesis, serine palmitoyltransferase, either pharmacologically using myriocin or genetically using heterozygous deletion of a serine palmitoyltransferase subunit, which ameliorated systolic heart dysfunction in LpLGPI mice (104). This improvement

24

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

of in vivo heart function was also associated with reduced mortality rates, decreased expression of atrial natriuretic peptide and brain natriuretic peptide, and a shift in cardiac energy metabolism

PT

from FA oxidation towards increased glucose oxidation (104). While this study suggests that

RI

increased ceramide accumulation contributes to cardiomyopathy, it remains unclear whether other sphingolipids, which would be lowered with general inhibition of sphingolipid synthesis,

SC

also play a role in lipotoxic cardiomyopathy as observed in LpLGPI mice.

NU

A recent study implicated increased cardiac C14-ceramide concentrations in cardiac hypertrophy, dysfunction, and autophagy induced by a milk fat-based high fat diet (MFBD) rich

MA

in myristate (C14:0 FA) (105). Inhibition of de novo ceramide synthesis using myriocin restored cardiac function and prevented cardiac hypertrophy in MFBD-fed mice (105). Interestingly,

TE

D

myristate-induced cardiomyocyte autophagy and hypertrophy was dependent on ceramide synthase 5 (CerS5), which produces ceramide by N-acylation of sphingosine (ceramide salvage

AC CE P

pathway) (105). Consumption of a MFBD also resulted in increased de novo synthesis of noncanonical myristate-derived d16 sphingolipids, which were shown to promote cell death and cleavage of poly(ADP-ribose) polymerase (PARP) in cardiomyocytes (106). Taken together, these studies pinpointed specific sphingolipid species and metabolic pathways in promoting lipotoxic cardiomyopathy.

Besides inhibition of ceramide synthase, increasing ceramide catabolism via ceramidase may lower cardiomyocyte ceramide levels. A study by Holland et al. (107) found that adiponectin, an adipokine which promotes cell survival, insulin signaling, and inhibits inflammation, activates ceramide catabolism via ceramidase action and increases the subsequent formation

of

sphingosine-1-phosphate

(S1P),

a

potent

anti-apoptotic

signaling

sphingolipid/lysophospholipid (107). Mice with inducible cardiomyocyte-specific apoptosis via

25

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

caspase-8 activation exhibited improved survival upon inhibition of ceramide synthesis by myriocin treatment or increased adiponectin signaling (107). Conversely, adiponectin deficiency

PT

exacerbated cardiomyocyte apoptosis (107). Adiponectin over-expression was associated with an

RI

increase in circulating S1P and S1P administration was sufficient to completely prevent mortality in mice with induced cardiomyocyte apoptosis (107). These findings suggest that a reduction in

SC

ceramide concentrations and increased formation of S1P, which is subsequently secreted to

NU

activate S1P receptor signaling in an autocrine/paracrine manner, are key mechanisms by which adiponectin protects against cardiomyocyte apoptosis (107). Adiponectin levels decrease with

MA

obesity in rodents and humans (108,109), suggesting that increased ceramide and decreased S1P formation due to diminished adiponectin signaling and ceramidase activation contributes to

TE

D

obesity-induced lipotoxic cardiomyopathy.

A recent study suggested that lowering certain sphingolipid species in the normal heart

AC CE P

can precipitate cardiac dysfunction (110). Mice with cardiomyocyte-specific deficiency of the serine palmitoyltransferase subunit 2 (Sptlc2) exhibited reduced cardiac concentrations of C18:0 and very long-chain ceramides, dihydroceramides, as well as the sphingoid bases, sphingosine and sphinganine (110). These alterations in sphingolipid content were paralleled by increases in certain fatty acyl-CoA species, elevated cardiolipin content, and altered phospholipid FA composition (110). Interestingly, Sptlc2 deficiency led to increased cardiac ER stress and apoptosis, ventricular wall thinning, cardiac fibrosis, left ventricular dilatation and systolic heart dysfunction (110). These findings demonstrate that reduced cardiac sphingolipid synthesis via Sptlc2 not only decreases certain sphingolipid species but also leads to significant changes in concentrations and composition of other cardiac lipids and dilated cardiomyopathy (110). This

26

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

study also highlights that more research is required to elucidate the link between metabolism of ceramides/sphingolipids and other lipids that may have toxic effects.

PT

Although there is strong evidence from animal models that ceramide accumulation

RI

contributes to cardiomyocyte lipotoxicity and apoptosis in obesity and diabetes, it remains unclear whether similar relationships exist in the human heart (111). For example, although

SC

markers of apoptosis were increased in the atrial appendage of obese subjects with and without a

NU

history of diabetes, total ceramide and sphingoid base levels were unchanged, despite altered mRNA expression of several enzymes involved in sphingolipid metabolism (111). Additional

MA

studies are necessary to better understand the role of ceramides and other sphingolipids and their

TE

D

metabolizing enzymes in cardiac lipotoxicity in humans.

AC CE P

7. Extracellular lipid signaling

While studies on cardiac lipotoxicity generally examine the intracellular accumulation of lipids and ensuing activation of signaling pathways from within the cardiomyocyte, it should also be noted that it is plausible that signaling induced by extracellular lipids contributes to cardiac injury in conditions

such as obesity/diabetes and myocardial

ischemia-reperfusion.

Cardiomyocytes express sarcolemmal receptors through which circulating lipids, e.g. lysophosphatidic acid (LPA), S1P, and free FAs, induce downstream signaling. Signaling cascades initiated by these receptors can have profound effects on cell survival, structure, function, and metabolism (112-118). Toll-like receptor 4 (TLR4) is best known for binding to lipopolysaccharide from Gramnegative bacteria and thereby activating the innate immune system. It has been suggested that

27

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

FAs can also bind to TLR4 indirectly through Fetuin A and trigger an inflammatory response via TLR4 signaling, which contributes to FA-induced insulin resistance (119-121). Alternatively,

PT

FAs can be incorporated into lipid raft domains, to enhance TLR4 recruitment and dimerization (122). It has been proposed that lipid-induced insulin resistance mediated by TLR4 is dependent

RI

on ceramide synthesis through activation of IKKβ-NFκB signaling (117). Interestingly, TLR4

SC

expression was upregulated in cardiomyocytes from type 1 diabetic NOD mice (118). TLR4

NU

deficiency in these mice resulted in reduced myocardial TAG accumulation, which was accompanied by decreased LPL protein expression and ameliorated cardiac dysfunction (118).

MA

These findings suggest that elevated circulating FAs could mediate diabetes-induced lipotoxic cardiomyopathy in part through TLR4 signaling. However, additional studies are required to

TE

D

examine the role of FA-induced TLR4 activation in lipotoxic cardiomyopathy. Most of the LPA in blood is produced by autotaxin, a secreted lysophospholipase D

AC CE P

which catabolizes lysophospholipids such as LPC and lysophosphatidylserine (123). LPA signals through G protein-coupled receptors at the plasmamembrane, activating a variety of downstream signaling pathways, including PI3Kinase-Akt, adenylate cyclase-cAMP, Ras-p42/p44MAPK, Rho-Rho kinase, and phospholipase C signaling (123). Although our understanding of how signaling through extracellular LPA influences cardiomyocytes is still in its infancy, emerging evidence suggests that LPA receptor activation plays an important pathophysiologic role (113,123-125). Specifically, LPA signaling has been implicated in the development of cardiomyocyte hypertrophy in part through LPA1 and LPA3 receptors (126). LPA1/3 protein levels were increased significantly in rat hearts that were subjected to myocardial infarction (126). Interestingly, LPA1 and LPA3 may play opposite roles in mediating cardiomyocyte hypertrophy. Knockdown of LPA1 enhanced LPA-induced hypertrophy, whereas knockdown of

28

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

LPA3 ameliorated LPA-induced hypertrophy in neonatal rat cardiomyocytes (127). The mechanisms underlying LPA mediated hypertrophy are incompletely understood, however, it has

PT

been suggested that LPA can enhance the expression of miR-23a, a known inducer of cardiac

RI

hypertrophy (127-129). A study by Pulinilkunnil et al. (125) also suggested that LPA signaling can enhance FA uptake in cardiomyocytes via stimulation of LPL trafficking. By triggering F-

SC

actin polymerisation through RhoA and Rho kinase, LPA promoted the transport of LPL-

NU

containing vesicles from the Golgi apparatus to the cardiomyocyte surface, which was followed by translocation of LPL to the coronary lumen (125).

MA

Recent studies showed that circulating levels of autotaxin, which critically influences extracellular LPA concentrations, correlates with measures of insulin resistance in humans

TE

D

(130,131). Moreover, in animal models, autotaxin-mediated LPA signaling has been implicated in the development of obesity and insulin resistance (132,133). Therefore, it is tempting to

AC CE P

speculate that the autotaxin-LPA signaling axis contributes to cardiac lipotoxicity – via upregulation of LPL-mediated FA delivery – and hypertrophic remodeling under conditions of obesity-induced insulin resistance. In addition, LPA signaling has been shown to impair cardiomyocyte contractility, suggesting that increased circulating LPA, as observed under conditions such as myocardial ischemia and obesity-insulin resistance, could contribute to cardiac dysfunction (124). Future studies are required to examine the detailed contributions of cardiomyocyte signaling induced by extracellular LPA and autotaxin to cardiac lipotoxicity. While autotaxin is a relatively novel adipokine and its role in cardiac lipotoxicity remains unexplored, several other adipokines including adiponectin (see also section 6.) and leptin have been suggested to play an important role in lipotoxic cardiomyopathy by modulating cardiac energy metabolism (6,134). For example, leptin increases cardiac FA oxidation and decreases

29

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

lipid accumulation (135). Moreover, overexpression of leptin protected against lipotoxic cardiomyopathy in acyl CoA synthase transgenic mice, suggesting that hyperleptinemia as

PT

observed during obesity may mitigate obesity-induced cardiac lipotoxicity (136), although the

RI

development of leptin resistance during obesity (137) may counter the cardioprotective effects of

SC

leptin.

NU

8. Lipotoxic signaling induced by DAGs and ceramides

DAGs and ceramides are arguably the most intensely studied toxic lipid species to date.

MA

DAGs are potent lipid second messengers that can activate classical (α, βI, βII, and γ) and novel (δ, ε, ε, and ζ) isoforms of PKC, both of which have been implicated in the development of

D

myocardial disease including cardiac hypertrophy, fibrosis, inflammation, and diabetic

TE

cardiomyopathy (138-142) (Figure 3). PKCβ2 expression is increased in the myocardium of

AC CE P

diabetic patients and animals (139,141,143,144). Forced expression of PKCβ in the myocardium leads to a cardiomyopathy characterized by hypertrophy, necrosis and an expansion of the interstitial extracellular matrix (139,145,146), suggesting that PKCβ over-activation by DAGs leads to cardiac dysfunction and remodeling. PKCβ may activate pro-inflammatory and prohypertrophic cytokines such as tumor necrosis factor α (TNFα) through NFκB in cardiomyocytes under hyperglycemic conditions, thereby decreasing Akt phosphorylation and insulin signaling (141,147-149). PKCs can activate NFκB by directly phosphorylating its inhibitor IκB or through the generation of reactive oxygen species, which can secondarily activate IκB kinases (IKKβ) (150,151). PKCζ, a novel PKC isoform, can directly inhibit insulin signaling through phosphorylation of insulin receptor substrate 1 (IRS1) on Ser307 and Ser1101 in skeletal and possibly cardiac muscle (152). PKCε, another novel PKC isoform, is activated in the rat heart in

30

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

response to streptozotozin-induced diabetes (153). PKCε is known to promote DAG-induced insulin resistance through inhibition of IRS2 and insulin receptor tyrosine kinase in the liver

PT

(154).

RI

It has been suggested that the DAG acyl chain composition, DAG steroisomer type, and cellular localization play an important role in activating signaling proteins such as PKCs by

SC

DAG signaling (155). For example, DAGs generated through phospholipase C cleavage of

NU

phosphatidylinositol-4,5-bisphosphate (PIP2) differentially induce translocation and activation of several PKC isoforms when compared to DAGs generated by phospholipase D cleavage of

MA

phosphatidylcholine (156,157). It has been proposed that cellular DAGs exist in three distinct pools of stereoisomers (155). These consist of sn-1,2 DAG generated through de novo

TE

D

lipogenesis via DGAT at the endoplasmic reticulum, sn-1,2 and sn-2,3 DAG generated by ATGL on lipid droplets, and sn-1,2 DAG generated by phospholipase C enzymes via hydrolysis of PIP2

AC CE P

at plasma and ER membranes (155). PKC activation is believed to occur mainly by the latter DAG pool and it has been suggested that TAG/ATGL-derived DAG would require isomerization prior to activation of PKC enzymes (155). Ceramides function as key components of lipotoxic signaling pathways linking lipidinduced inflammation and inhibition of insulin signaling (102) (Figure 3). For example, TLR4 signaling induces de novo ceramide synthesis through IKKβ-NFκB signaling (117). Moreover, activation of TLR4 signaling is dependent on the production of ceramide by acid sphingomyelinase (158). Ceramide is critically required for TLR4-induced insulin resistance (117) and has been suggested to induce insulin resistance by targeting activation of Akt through two independent mechanisms (159). First, ceramide can activate protein phosphatase 2A (PP2A), which dephosphorylates Akt and second, ceramide can prevent Akt translocation to the plasma

31

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

membrane through activation of PKCδ (159,160). Indeed, treatment of AC16 human cardiomyocytes with C6 ceramide decreased phosphorylation of Akt and its downstream target

PT

glycogen synthase kinase 3β (GSK3β) in a concentration dependent manner, which is consistent

RI

with findings in skeletal muscle cells and adipocytes (104). In addition to inhibiting insulin signaling via reducing Akt phosphorylation, ceramide can also inhibit phosphorylation of IRS1

SC

(161). Moreover, ceramide appears to activate mixed lineage kinase-3 (MLK3), which may

NU

inhibit IRS through stress activated protein kinases (SAPK) including p38 mitogen activated protein kinase (p38MAPK) and c-Jun N-terminal kinase (JNK) (162-166). Kinases in the MLK

MA

family regulate the stress response in cardiomyocytes, however, it remains to be determined whether ceramide also inhibits cardiomyocyte insulin signaling through these kinases (167).

TE

D

Apart from inhibiting insulin signaling, ceramides also signal through mitogen activated protein kinases (MAPKs), including extracellular signal regulated kinase (ERK), p38MAPK and

AC CE P

JNK to induce apoptosis (168). Specifically, ceramide triggers ERK dephosphorylation and stimulates phosphorylation of p38MAPK and JNK (168). MAPK signaling has been implicated in heart failure, cardiomyopathy, cardiac remodeling, and injury induced by hypertrophy and ischemia/reperfusion (169,170), suggesting that ceramide-MAPK signaling may play a key role in various forms of heart disease. Indeed, ceramide-induced apoptosis in rat cardiomyocytes could be attenuated by inhibiting JNK1 using antisense oligonucleotides (169). JNK can directly activate the mitochondrial apoptosis machinery in adult cardiac myocytes through the release of cytochrome c from mitochondria and activation of caspase 9 (171). It has also been proposed that ceramides induce apoptosis in neonatal rat cardiomyocytes through caspase 3 and 8-related proteases (172). Inhibition of ceramide synthesis using fumonisin B1 impaired palmitic acidinduced cytochrome c release, apoptosis and myofibril degeneration in adult rat cardiomyocytes

32

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

(173). Interestingly, incubation with sphinogomyelinase, which metabolizes sphingomyeline to ceramide, had no adverse effects on cardiomyocytes, suggesting that palmitate-induced

RI

PT

cardiomyocyte apoptosis is primarily mediated by de novo ceramide synthesis (173).

9. Concluding remarks

SC

Intense research and the generation of numerous animal models in the last fifteen years have

NU

been instrumental in improving our understanding of cardiac lipotoxicity, mostly in the context of obesity, insulin resistance, and diabetes. Certain lipids are believed to be particularly

MA

cardiotoxic when they accumulate in excess. These include ceramides, DAGs, long chain acylCoAs, acylcarnitines, and lysophospholipids. TAGs, on the other hand, are generally considered

D

to be non-toxic and likely serve as marker for increased lipid deposition in the heart.

TE

Nevertheless, enzymes involved in the synthesis and catabolism of TAGs do play a role in

AC CE P

cardiac lipotoxicity by regulating the sequestration of FAs in the TAG pool. A major limitation of most studies examining cardiac lipotoxicity in animal models is that only select lipids and lipid metabolism and signaling pathways were examined. More comprehensive lipidomics analysis will be necessary to uncover how accumulation and metabolism of toxic lipids in the myocardium are interconnected. Moreover, research examining cardiac lipotoxicity in the human heart is still in its infancy and future studies need to determine whether findings from animal models of lipotoxic cardiomyopathy translate to the human myocardium. Based on our understanding of the etiology of cardiac lipotoxicity from the study of animal models, it is apparent that limiting FA delivery and uptake produce the most consistent results in protecting from cardiac lipotoxicity not only during obesity/insulin resistance/diabetes, but also aging and myocardial ischemia/infarction (7,8,38,39,42). This is not entirely surprising since FA overload due to a mismatch between cardiomyocyte FA transport and FA metabolism 33

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

through oxidation or TAG synthesis is the primary culprit in lipotoxic cardiomyopathy. Therefore, targeting FA uptake, for example through CD36 inhibitors (41), may currently be the

PT

most promising avenue for the development of therapies for lipotoxic cardiomyopathy of

SC

RI

different etiologies.

NU

Acknowledgements

This work was supported by grants from the Natural Sciences and Engineering Research

Conflicts of Interest

TE

D

Research Foundation (NBHRF) to P.K.

MA

Council of Canada (NSERC), the Banting Research Foundation, and the New Brunswick Health

References 1. 2.

3. 4. 5.

6. 7.

AC CE P

We have no conflict of interest to declare.

Unger, R. H., and Orci, L. (2002) Lipoapoptosis: its mechanism and its diseases. Biochim Biophys Acta 1585, 202-212 Chiu, H. C., Kovacs, A., Ford, D. A., Hsu, F. F., Garcia, R., Herrero, P., Saffitz, J. E., and Schaffer, J. E. (2001) A novel mouse model of lipotoxic cardiomyopathy. J Clin Invest 107, 813-822 Goldberg, I. J., Trent, C. M., and Schulze, P. C. (2012) Lipid metabolism and toxicity in the heart. Cell Metab 15, 805-812 Birse, R. T., and Bodmer, R. (2011) Lipotoxicity and cardiac dysfunction in mammals and Drosophila. Crit Rev Biochem Mol Biol 46, 376-385 Brindley, D. N., Kok, B. P., Kienesberger, P. C., Lehner, R., and Dyck, J. R. (2010) Shedding light on the enigma of myocardial lipotoxicity: the involvement of known and putative regulators of fatty acid storage and mobilization. Am J Physiol Endocrinol Metab 298, E897-908 Wende, A. R., and Abel, E. D. (2010) Lipotoxicity in the heart. Biochim Biophys Acta 1801, 311-319 Perman, J. C., Bostrom, P., Lindbom, M., Lidberg, U., StAhlman, M., Hagg, D., Lindskog, H., Scharin Tang, M., Omerovic, E., Mattsson Hulten, L., Jeppsson, A., 34

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

14. 15.

16.

17.

18. 19.

20.

PT

D

13.

TE

12.

AC CE P

11.

MA

NU

10.

RI

9.

Petursson, P., Herlitz, J., Olivecrona, G., Strickland, D. K., Ekroos, K., Olofsson, S. O., and Boren, J. (2011) The VLDL receptor promotes lipotoxicity and increases mortality in mice following an acute myocardial infarction. J Clin Invest 121, 2625-2640 Koonen, D. P., Febbraio, M., Bonnet, S., Nagendran, J., Young, M. E., Michelakis, E. D., and Dyck, J. R. (2007) CD36 expression contributes to age-induced cardiomyopathy in mice. Circulation 116, 2139-2147 Dewald, O., Sharma, S., Adrogue, J., Salazar, R., Duerr, G. D., Crapo, J. D., Entman, M. L., and Taegtmeyer, H. (2005) Downregulation of peroxisome proliferator-activated receptor-alpha gene expression in a mouse model of ischemic cardiomyopathy is dependent on reactive oxygen species and prevents lipotoxicity. Circulation 112, 407-415 Girousse, A., Tavernier, G., Valle, C., Moro, C., Mejhert, N., Dinel, A. L., Houssier, M., Roussel, B., Besse-Patin, A., Combes, M., Mir, L., Monbrun, L., Bezaire, V., PrunetMarcassus, B., Waget, A., Vila, I., Caspar-Bauguil, S., Louche, K., Marques, M. A., Mairal, A., Renoud, M. L., Galitzky, J., Holm, C., Mouisel, E., Thalamas, C., Viguerie, N., Sulpice, T., Burcelin, R., Arner, P., and Langin, D. (2013) Partial inhibition of adipose tissue lipolysis improves glucose metabolism and insulin sensitivity without alteration of fat mass. PLoS Biol 11, e1001485 Gray, S., and Kim, J. K. (2011) New insights into insulin resistance in the diabetic heart. Trends Endocrinol Metab 22, 394-403 Borradaile, N. M., and Schaffer, J. E. (2005) Lipotoxicity in the heart. Curr Hypertens Rep 7, 412-417 Abel, E. D., O'Shea, K. M., and Ramasamy, R. (2012) Insulin resistance: metabolic mechanisms and consequences in the heart. Arterioscler Thromb Vasc Biol 32, 20682076 Kienesberger, P. C., Pulinilkunnil, T., Nagendran, J., and Dyck, J. R. (2013) Myocardial triacylglycerol metabolism. J Mol Cell Cardiol 55, 101-110 Heather, L. C., Pates, K. M., Atherton, H. J., Cole, M. A., Ball, D. R., Evans, R. D., Glatz, J. F., Luiken, J. J., Griffin, J. L., and Clarke, K. (2013) Differential translocation of the fatty acid transporter, FAT/CD36, and the glucose transporter, GLUT4, coordinates changes in cardiac substrate metabolism during ischemia and reperfusion. Circulation. Heart failure 6, 1058-1066 Nagendran, J., Pulinilkunnil, T., Kienesberger, P. C., Sung, M. M., Fung, D., Febbraio, M., and Dyck, J. R. (2013) Cardiomyocyte-specific ablation of CD36 improves postischemic functional recovery. J Mol Cell Cardiol 63C, 180-188 Augustus, A. S., Buchanan, J., Park, T. S., Hirata, K., Noh, H. L., Sun, J., Homma, S., D'Armiento, J., Abel, E. D., and Goldberg, I. J. (2006) Loss of lipoprotein lipase-derived fatty acids leads to increased cardiac glucose metabolism and heart dysfunction. J Biol Chem 281, 8716-8723 Lopaschuk, G. D., Ussher, J. R., Folmes, C. D., Jaswal, J. S., and Stanley, W. C. (2010) Myocardial fatty acid metabolism in health and disease. Physiol Rev 90, 207-258 Kolwicz, S. C., Jr., Purohit, S., and Tian, R. (2013) Cardiac metabolism and its interactions with contraction, growth, and survival of cardiomyocytes. Circ Res 113, 603616 Iozzo, P. (2010) Metabolic toxicity of the heart: insights from molecular imaging. Nutr Metab Cardiovasc Dis 20, 147-156

SC

8.

Heart Lipid Metabolism

35

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

26.

27.

28.

29. 30.

31.

32.

33.

34.

PT

RI

SC

NU

25.

MA

24.

D

23.

TE

22.

Taegtmeyer, H., Golfman, L., Sharma, S., Razeghi, P., and van Arsdall, M. (2004) Linking gene expression to function: metabolic flexibility in the normal and diseased heart. Ann N Y Acad Sci 1015, 202-213 Birse, R. T., Choi, J., Reardon, K., Rodriguez, J., Graham, S., Diop, S., Ocorr, K., Bodmer, R., and Oldham, S. (2010) High-fat-diet-induced obesity and heart dysfunction are regulated by the TOR pathway in Drosophila. Cell Metab 12, 533-544 Drosatos, K., and Schulze, P. C. (2013) Cardiac lipotoxicity: molecular pathways and therapeutic implications. Current heart failure reports 10, 109-121 Szczepaniak, L. S., Dobbins, R. L., Metzger, G. J., Sartoni-D'Ambrosia, G., Arbique, D., Vongpatanasin, W., Unger, R., and Victor, R. G. (2003) Myocardial triglycerides and systolic function in humans: in vivo evaluation by localized proton spectroscopy and cardiac imaging. Magn Reson Med 49, 417-423 Utz, W., Engeli, S., Haufe, S., Kast, P., Hermsdorf, M., Wiesner, S., Pofahl, M., Traber, J., Luft, F. C., Boschmann, M., Schulz-Menger, J., and Jordan, J. (2011) Myocardial steatosis, cardiac remodelling and fitness in insulin-sensitive and insulin-resistant obese women. Heart 97, 1585-1589 Hammer, S., van der Meer, R. W., Lamb, H. J., Schar, M., de Roos, A., Smit, J. W., and Romijn, J. A. (2008) Progressive caloric restriction induces dose-dependent changes in myocardial triglyceride content and diastolic function in healthy men. J Clin Endocrinol Metab 93, 497-503 Schrauwen-Hinderling, V. B., Hesselink, M. K., Meex, R., van der Made, S., Schar, M., Lamb, H., Wildberger, J. E., Glatz, J., Snoep, G., Kooi, M. E., and Schrauwen, P. (2010) Improved ejection fraction after exercise training in obesity is accompanied by reduced cardiac lipid content. J Clin Endocrinol Metab 95, 1932-1938 McGavock, J. M., Lingvay, I., Zib, I., Tillery, T., Salas, N., Unger, R., Levine, B. D., Raskin, P., Victor, R. G., and Szczepaniak, L. S. (2007) Cardiac steatosis in diabetes mellitus: a 1H-magnetic resonance spectroscopy study. Circulation 116, 1170-1175 McGavock, J. M., Victor, R. G., Unger, R. H., and Szczepaniak, L. S. (2006) Adiposity of the heart, revisited. Ann Intern Med 144, 517-524 Szczepaniak, L. S., Victor, R. G., Orci, L., and Unger, R. H. (2007) Forgotten but not gone: the rediscovery of fatty heart, the most common unrecognized disease in America. Circ Res 101, 759-767 Labbe, S. M., Grenier-Larouche, T., Noll, C., Phoenix, S., Guerin, B., Turcotte, E. E., and Carpentier, A. C. (2012) Increased myocardial uptake of dietary fatty acids linked to cardiac dysfunction in glucose-intolerant humans. Diabetes 61, 2701-2710 Chiu, H. C., Kovacs, A., Blanton, R. M., Han, X., Courtois, M., Weinheimer, C. J., Yamada, K. A., Brunet, S., Xu, H., Nerbonne, J. M., Welch, M. J., Fettig, N. M., Sharp, T. L., Sambandam, N., Olson, K. M., Ory, D. S., and Schaffer, J. E. (2005) Transgenic expression of fatty acid transport protein 1 in the heart causes lipotoxic cardiomyopathy. Circ Res 96, 225-233 Finck, B. N., Lehman, J. J., Leone, T. C., Welch, M. J., Bennett, M. J., Kovacs, A., Han, X., Gross, R. W., Kozak, R., Lopaschuk, G. D., and Kelly, D. P. (2002) The cardiac phenotype induced by PPARalpha overexpression mimics that caused by diabetes mellitus. J Clin Invest 109, 121-130 Yagyu, H., Chen, G., Yokoyama, M., Hirata, K., Augustus, A., Kako, Y., Seo, T., Hu, Y., Lutz, E. P., Merkel, M., Bensadoun, A., Homma, S., and Goldberg, I. J. (2003)

AC CE P

21.

Heart Lipid Metabolism

36

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

39.

40.

41.

42.

43.

44.

45.

46.

PT

RI

SC

NU

MA

38.

D

37.

TE

36.

Lipoprotein lipase (LpL) on the surface of cardiomyocytes increases lipid uptake and produces a cardiomyopathy. J Clin Invest 111, 419-426 Luiken, J. J., Arumugam, Y., Dyck, D. J., Bell, R. C., Pelsers, M. M., Turcotte, L. P., Tandon, N. N., Glatz, J. F., and Bonen, A. (2001) Increased rates of fatty acid uptake and plasmalemmal fatty acid transporters in obese Zucker rats. J Biol Chem 276, 4056740573 Coort, S. L., Luiken, J. J., van der Vusse, G. J., Bonen, A., and Glatz, J. F. (2004) Increased FAT (fatty acid translocase)/CD36-mediated long-chain fatty acid uptake in cardiac myocytes from obese Zucker rats. Biochem Soc Trans 32, 83-85 Coort, S. L., Hasselbaink, D. M., Koonen, D. P., Willems, J., Coumans, W. A., Chabowski, A., van der Vusse, G. J., Bonen, A., Glatz, J. F., and Luiken, J. J. (2004) Enhanced sarcolemmal FAT/CD36 content and triacylglycerol storage in cardiac myocytes from obese zucker rats. Diabetes 53, 1655-1663 Angin, Y., Steinbusch, L. K., Simons, P. J., Greulich, S., Hoebers, N. T., Douma, K., van Zandvoort, M. A., Coumans, W. A., Wijnen, W., Diamant, M., Ouwens, D. M., Glatz, J. F., and Luiken, J. J. (2012) CD36 inhibition prevents lipid accumulation and contractile dysfunction in rat cardiomyocytes. Biochem J 448, 43-53 Yang, J., Sambandam, N., Han, X., Gross, R. W., Courtois, M., Kovacs, A., Febbraio, M., Finck, B. N., and Kelly, D. P. (2007) CD36 deficiency rescues lipotoxic cardiomyopathy. Circ Res 100, 1208-1217 Finck, B. N., Han, X., Courtois, M., Aimond, F., Nerbonne, J. M., Kovacs, A., Gross, R. W., and Kelly, D. P. (2003) A critical role for PPARalpha-mediated lipotoxicity in the pathogenesis of diabetic cardiomyopathy: modulation by dietary fat content. Proc Natl Acad Sci U S A 100, 1226-1231 Glatz, J. F., Angin, Y., Steinbusch, L. K., Schwenk, R. W., and Luiken, J. J. (2013) CD36 as a target to prevent cardiac lipotoxicity and insulin resistance. Prostaglandins, leukotrienes, and essential fatty acids 88, 71-77 Duncan, J. G., Bharadwaj, K. G., Fong, J. L., Mitra, R., Sambandam, N., Courtois, M. R., Lavine, K. J., Goldberg, I. J., and Kelly, D. P. (2010) Rescue of cardiomyopathy in peroxisome proliferator-activated receptor-alpha transgenic mice by deletion of lipoprotein lipase identifies sources of cardiac lipids and peroxisome proliferatoractivated receptor-alpha activators. Circulation 121, 426-435 Banke, N. H., Wende, A. R., Leone, T. C., O'Donnell, J. M., Abel, E. D., Kelly, D. P., and Lewandowski, E. D. (2010) Preferential oxidation of triacylglyceride-derived fatty acids in heart is augmented by the nuclear receptor PPARalpha. Circ Res 107, 233-241 Listenberger, L. L., Han, X., Lewis, S. E., Cases, S., Farese, R. V., Jr., Ory, D. S., and Schaffer, J. E. (2003) Triglyceride accumulation protects against fatty acid-induced lipotoxicity. Proc Natl Acad Sci U S A 100, 3077-3082 Bosma, M., Dapito, D. H., Drosatos-Tampakaki, Z., Huiping-Son, N., Huang, L. S., Kersten, S., Drosatos, K., and Goldberg, I. J. (2014) Sequestration of fatty acids in triglycerides prevents endoplasmic reticulum stress in an in vitro model of cardiomyocyte lipotoxicity. Biochim Biophys Acta 1841, 1648-1655 Liu, L., Shi, X., Bharadwaj, K. G., Ikeda, S., Yamashita, H., Yagyu, H., Schaffer, J. E., Yu, Y. H., and Goldberg, I. J. (2009) DGAT1 expression increases heart triglyceride content but ameliorates lipotoxicity. J Biol Chem 284, 36312-36323

AC CE P

35.

Heart Lipid Metabolism

37

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

48.

54.

55.

56.

57.

58.

AC CE P

53.

TE

D

52.

NU

51.

MA

50.

SC

RI

49.

Liu, L., Yu, S., Khan, R. S., Homma, S., Schulze, P. C., Blaner, W. S., Yin, Y., and Goldberg, I. J. (2012) Diacylglycerol acyl transferase 1 overexpression detoxifies cardiac lipids in PPARgamma transgenic mice. J Lipid Res 53, 1482-1492 Glenn, D. J., Wang, F., Nishimoto, M., Cruz, M. C., Uchida, Y., Holleran, W. M., Zhang, Y., Yeghiazarians, Y., and Gardner, D. G. (2011) A murine model of isolated cardiac steatosis leads to cardiomyopathy. Hypertension 57, 216-222 Liu, L., Trent, C. M., Fang, X., Son, N. H., Jiang, H., Blaner, W. S., Hu, Y., Yin, Y. X., Farese, R. V., Jr., Homma, S., Turnbull, A. V., Eriksson, J. W., Hu, S. L., Ginsberg, H. N., Huang, L. S., and Goldberg, I. J. (2014) Cardiomyocyte-specific loss of diacylglycerol acyltransferase 1 (DGAT1) reproduces the abnormalities in lipids found in severe heart failure. J Biol Chem 289, 29881-29891 Liu, L., Yu, S., Khan, R. S., Ables, G. P., Bharadwaj, K. G., Hu, Y., Huggins, L. A., Eriksson, J. W., Buckett, L. K., Turnbull, A. V., Ginsberg, H. N., Blaner, W. S., Huang, L. S., and Goldberg, I. J. (2011) DGAT1 deficiency decreases PPAR expression and does not lead to lipotoxicity in cardiac and skeletal muscle. J Lipid Res 52, 732-744 Lee, B., Fast, A. M., Zhu, J., Cheng, J. X., and Buhman, K. K. (2010) Intestine-specific expression of acyl CoA:diacylglycerol acyltransferase 1 reverses resistance to dietinduced hepatic steatosis and obesity in Dgat1-/- mice. J Lipid Res 51, 1770-1780 Chokshi, A., Drosatos, K., Cheema, F. H., Ji, R., Khawaja, T., Yu, S., Kato, T., Khan, R., Takayama, H., Knoll, R., Milting, H., Chung, C. S., Jorde, U., Naka, Y., Mancini, D. M., Goldberg, I. J., and Schulze, P. C. (2012) Ventricular assist device implantation corrects myocardial lipotoxicity, reverses insulin resistance, and normalizes cardiac metabolism in patients with advanced heart failure. Circulation 125, 2844-2853 Lewin, T. M., de Jong, H., Schwerbrock, N. J., Hammond, L. E., Watkins, S. M., Combs, T. P., and Coleman, R. A. (2008) Mice deficient in mitochondrial glycerol-3-phosphate acyltransferase-1 have diminished myocardial triacylglycerol accumulation during lipogenic diet and altered phospholipid fatty acid composition. Biochim Biophys Acta 1781, 352-358 Kok, B. P., Kienesberger, P. C., Dyck, J. R., and Brindley, D. N. (2012) Relationship of glucose and oleate metabolism to cardiac function in lipin-1 deficient (fld) mice. J Lipid Res 53, 105-118 Mitra, M. S., Schilling, J. D., Wang, X., Jay, P. Y., Huss, J. M., Su, X., and Finck, B. N. (2011) Cardiac lipin 1 expression is regulated by the peroxisome proliferator activated receptor gamma coactivator 1alpha/estrogen related receptor axis. J Mol Cell Cardiol 51, 120-128 Burgdorf, C., Hansel, L., Heidbreder, M., Johren, O., Schutte, F., Schunkert, H., and Kurz, T. (2009) Suppression of cardiac phosphatidate phosphohydrolase 1 activity and lipin mRNA expression in Zucker diabetic fatty rats and humans with type 2 diabetes mellitus. Biochem Biophys Res Commun 390, 165-170 Jamal, Z., Martin, A., Gomez-Munoz, A., Hales, P., Chang, E., Russell, J. C., and Brindley, D. N. (1992) Phosphatidate phosphohydrolases in liver, heart and adipose tissue of the JCR:LA corpulent rat and the lean genotypes: implications for glycerolipid synthesis and signal transduction. Int J Obes Relat Metab Disord 16, 789-799 Haemmerle, G., Moustafa, T., Woelkart, G., Buttner, S., Schmidt, A., van de Weijer, T., Hesselink, M., Jaeger, D., Kienesberger, P. C., Zierler, K., Schreiber, R., Eichmann, T., Kolb, D., Kotzbeck, P., Schweiger, M., Kumari, M., Eder, S., Schoiswohl, G.,

PT

47.

Heart Lipid Metabolism

38

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

64.

65.

66.

67.

68.

PT

AC CE P

63.

TE

D

62.

MA

61.

NU

SC

60.

Wongsiriroj, N., Pollak, N. M., Radner, F. P., Preiss-Landl, K., Kolbe, T., Rulicke, T., Pieske, B., Trauner, M., Lass, A., Zimmermann, R., Hoefler, G., Cinti, S., Kershaw, E. E., Schrauwen, P., Madeo, F., Mayer, B., and Zechner, R. (2011) ATGL-mediated fat catabolism regulates cardiac mitochondrial function via PPAR-alpha and PGC-1. Nat Med 17, 1076-1085 Kienesberger, P. C., Pulinilkunnil, T., Nagendran, J., Young, M. E., Bogner-Strauss, J. G., Hackl, H., Khadour, R., Heydari, E., Haemmerle, G., Zechner, R., Kershaw, E. E., and Dyck, J. R. (2013) Early structural and metabolic cardiac remodelling in response to inducible adipose triglyceride lipase ablation. Cardiovasc Res 99, 442-451 Zierler, K. A., Jaeger, D., Pollak, N. M., Eder, S., Rechberger, G. N., Radner, F. P., Woelkart, G., Kolb, D., Schmidt, A., Kumari, M., Preiss-Landl, K., Pieske, B., Mayer, B., Zimmermann, R., Lass, A., Zechner, R., and Haemmerle, G. (2013) Functional cardiac lipolysis in mice critically depends on comparative gene identification-58. J Biol Chem 288, 9892-9904 Hirano, K., Ikeda, Y., Zaima, N., Sakata, Y., and Matsumiya, G. (2008) Triglyceride deposit cardiomyovasculopathy. N Engl J Med 359, 2396-2398 Hirano, K., Tanaka, T., Ikeda, Y., Yamaguchi, S., Zaima, N., Kobayashi, K., Suzuki, A., Sakata, Y., Sakata, Y., Kobayashi, K., Toda, T., Fukushima, N., Ishibashi-Ueda, H., Tavian, D., Nagasaka, H., Hui, S. P., Chiba, H., Sawa, Y., and Hori, M. (2014) Genetic mutations in adipose triglyceride lipase and myocardial up-regulation of peroxisome proliferated activated receptor-gamma in patients with triglyceride deposit cardiomyovasculopathy. Biochem Biophys Res Commun 443, 574-579 Pulinilkunnil, T., Kienesberger, P. C., Nagendran, J., Waller, T. J., Young, M. E., Kershaw, E. E., Korbutt, G., Haemmerle, G., Zechner, R., and Dyck, J. R. (2013) Myocardial adipose triglyceride lipase overexpression protects diabetic mice from the development of lipotoxic cardiomyopathy. Diabetes 62, 1464-1477 Kienesberger, P. C., Pulinilkunnil, T., Sung, M. M., Nagendran, J., Haemmerle, G., Kershaw, E. E., Young, M. E., Light, P. E., Oudit, G. Y., Zechner, R., and Dyck, J. R. (2012) Myocardial ATGL overexpression decreases the reliance on fatty acid oxidation and protects against pressure overload-induced cardiac dysfunction. Mol Cell Biol 32, 740-750 Pulinilkunnil, T., Kienesberger, P. C., Nagendran, J., Sharma, N., Young, M. E., and Dyck, J. R. (2013) Cardiac-specific adipose triglyceride lipase overexpression protects from cardiac steatosis and dilated cardiomyopathy following diet-induced obesity. Int J Obes (Lond) 38, 205-215 Ueno, M., Suzuki, J., Zenimaru, Y., Takahashi, S., Koizumi, T., Noriki, S., Yamaguchi, O., Otsu, K., Shen, W. J., Kraemer, F. B., and Miyamori, I. (2008) Cardiac overexpression of hormone-sensitive lipase inhibits myocardial steatosis and fibrosis in streptozotocin diabetic mice. Am J Physiol Endocrinol Metab 294, E1109-1118 Pollak, N. M., Schweiger, M., Jaeger, D., Kolb, D., Kumari, M., Schreiber, R., Kolleritsch, S., Markolin, P., Grabner, G. F., Heier, C., Zierler, K. A., Ruelicke, T., Zimmermann, R., Lass, A., Zechner, R., and Haemmerle, G. (2013) Cardiac-specific overexpression of perilipin 5 provokes severe cardiac steatosis via the formation of a lipolytic barrier. J Lipid Res 54, 1092-1102 Wang, H., Sreenivasan, U., Gong, D. W., O'Connell, K. A., Dabkowski, E. R., Hecker, P. A., Ionica, N., Konig, M., Mahurkar, A., Sun, Y., Stanley, W. C., and Sztalryd, C. (2013)

RI

59.

Heart Lipid Metabolism

39

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

72.

73.

76.

77.

78.

79.

80.

PT

RI

AC CE P

75.

TE

D

74.

SC

71.

NU

70.

Cardiomyocyte specific perilipin 5 over expression leads to myocardial steatosis, and modest cardiac dysfunction. J Lipid Res 54, 953-965 Pollak, N. M., Jaeger, D., Kolleritsch, S., Zimmermann, R., Zechner, R., Lass, A., and Haemmerle, G. (2015) The interplay of protein kinase A and perilipin 5 regulates cardiac lipolysis. J Biol Chem 290, 1295-1306 Kuramoto, K., Okamura, T., Yamaguchi, T., Nakamura, T. Y., Wakabayashi, S., Morinaga, H., Nomura, M., Yanase, T., Otsu, K., Usuda, N., Matsumura, S., Inoue, K., Fushiki, T., Kojima, Y., Hashimoto, T., Sakai, F., Hirose, F., and Osumi, T. (2012) Perilipin 5, a lipid droplet-binding protein, protects the heart from oxidative burden by sequestering fatty acid from excessive oxidation. J Biol Chem 287, 23852-23863 Bartels, E. D., Nielsen, J. M., Hellgren, L. I., Ploug, T., and Nielsen, L. B. (2009) Cardiac expression of microsomal triglyceride transfer protein is increased in obesity and serves to attenuate cardiac triglyceride accumulation. PLoS One 4, e5300 Nielsen, L. B., Bartels, E. D., and Bollano, E. (2002) Overexpression of apolipoprotein B in the heart impedes cardiac triglyceride accumulation and development of cardiac dysfunction in diabetic mice. J Biol Chem 277, 27014-27020 Yokoyama, M., Yagyu, H., Hu, Y., Seo, T., Hirata, K., Homma, S., and Goldberg, I. J. (2004) Apolipoprotein B production reduces lipotoxic cardiomyopathy: studies in heartspecific lipoprotein lipase transgenic mouse. J Biol Chem 279, 4204-4211 He, L., Kim, T., Long, Q., Liu, J., Wang, P., Zhou, Y., Ding, Y., Prasain, J., Wood, P. A., and Yang, Q. (2012) Carnitine palmitoyltransferase-1b deficiency aggravates pressure overload-induced cardiac hypertrophy caused by lipotoxicity. Circulation 126, 17051716 Haynie, K. R., Vandanmagsar, B., Wicks, S. E., Zhang, J., and Mynatt, R. L. (2014) Inhibition of carnitine palymitoyltransferase1b induces cardiac hypertrophy and mortality in mice. Diabetes Obes Metab 16, 757-760 Lee, L., Campbell, R., Scheuermann-Freestone, M., Taylor, R., Gunaruwan, P., Williams, L., Ashrafian, H., Horowitz, J., Fraser, A. G., Clarke, K., and Frenneaux, M. (2005) Metabolic modulation with perhexiline in chronic heart failure: a randomized, controlled trial of short-term use of a novel treatment. Circulation 112, 3280-3288 Luiken, J. J., Niessen, H. E., Coort, S. L., Hoebers, N., Coumans, W. A., Schwenk, R. W., Bonen, A., and Glatz, J. F. (2009) Etomoxir-induced partial carnitine palmitoyltransferase-I (CPT-I) inhibition in vivo does not alter cardiac long-chain fatty acid uptake and oxidation rates. Biochem J 419, 447-455 Cabrero, A., Merlos, M., Laguna, J. C., and Carrera, M. V. (2003) Down-regulation of acyl-CoA oxidase gene expression and increased NF-kappaB activity in etomoxirinduced cardiac hypertrophy. J Lipid Res 44, 388-398 Cheng, L., Ding, G., Qin, Q., Huang, Y., Lewis, W., He, N., Evans, R. M., Schneider, M. D., Brako, F. A., Xiao, Y., Chen, Y. E., and Yang, Q. (2004) Cardiomyocyte-restricted peroxisome proliferator-activated receptor-delta deletion perturbs myocardial fatty acid oxidation and leads to cardiomyopathy. Nat Med 10, 1245-1250 Bakermans, A. J., Geraedts, T. R., van Weeghel, M., Denis, S., Joao Ferraz, M., Aerts, J. M., Aten, J., Nicolay, K., Houten, S. M., and Prompers, J. J. (2011) Fasting-induced myocardial lipid accumulation in long-chain acyl-CoA dehydrogenase knockout mice is accompanied by impaired left ventricular function. Circ Cardiovasc Imaging 4, 558-565

MA

69.

Heart Lipid Metabolism

40

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

82.

88.

89.

90.

91. 92. 93.

AC CE P

87.

TE

D

86.

NU

85.

MA

84.

SC

RI

83.

Chambers, K. T., Leone, T. C., Sambandam, N., Kovacs, A., Wagg, C. S., Lopaschuk, G. D., Finck, B. N., and Kelly, D. P. (2011) Chronic inhibition of pyruvate dehydrogenase in heart triggers an adaptive metabolic response. J Biol Chem 286, 11155-11162 Ford, D. A., Han, X., Horner, C. C., and Gross, R. W. (1996) Accumulation of unsaturated acylcarnitine molecular species during acute myocardial ischemia: metabolic compartmentalization of products of fatty acyl chain elongation in the acylcarnitine pool. Biochemistry 35, 7903-7909 Su, X., Han, X., Mancuso, D. J., Abendschein, D. R., and Gross, R. W. (2005) Accumulation of long-chain acylcarnitine and 3-hydroxy acylcarnitine molecular species in diabetic myocardium: identification of alterations in mitochondrial fatty acid processing in diabetic myocardium by shotgun lipidomics. Biochemistry 44, 5234-5245 Koves, T. R., Ussher, J. R., Noland, R. C., Slentz, D., Mosedale, M., Ilkayeva, O., Bain, J., Stevens, R., Dyck, J. R., Newgard, C. B., Lopaschuk, G. D., and Muoio, D. M. (2008) Mitochondrial overload and incomplete fatty acid oxidation contribute to skeletal muscle insulin resistance. Cell Metab 7, 45-56 Idell-Wenger, J. A., Grotyohann, L. W., and Neely, J. R. (1978) Coenzyme A and carnitine distribution in normal and ischemic hearts. J Biol Chem 253, 4310-4318 Thyfault, J. P., Cree, M. G., Zheng, D., Zwetsloot, J. J., Tapscott, E. B., Koves, T. R., Ilkayeva, O., Wolfe, R. R., Muoio, D. M., and Dohm, G. L. (2007) Contraction of insulin-resistant muscle normalizes insulin action in association with increased mitochondrial activity and fatty acid catabolism. Am J Physiol Cell Physiol 292, C729739 Ussher, J. R., Koves, T. R., Jaswal, J. S., Zhang, L., Ilkayeva, O., Dyck, J. R., Muoio, D. M., and Lopaschuk, G. D. (2009) Insulin-stimulated cardiac glucose oxidation is increased in high-fat diet-induced obese mice lacking malonyl CoA decarboxylase. Diabetes 58, 1766-1775 Son, N. H., Yu, S., Tuinei, J., Arai, K., Hamai, H., Homma, S., Shulman, G. I., Abel, E. D., and Goldberg, I. J. (2010) PPARgamma-induced cardiolipotoxicity in mice is ameliorated by PPARalpha deficiency despite increases in fatty acid oxidation. J Clin Invest 120, 3443-3454 Battiprolu, P. K., Hojayev, B., Jiang, N., Wang, Z. V., Luo, X., Iglewski, M., Shelton, J. M., Gerard, R. D., Rothermel, B. A., Gillette, T. G., Lavandero, S., and Hill, J. A. (2012) Metabolic stress-induced activation of FoxO1 triggers diabetic cardiomyopathy in mice. J Clin Invest 122, 1109-1118 Anderson, E. J., Katunga, L. A., and Willis, M. S. (2012) Mitochondria as a source and target of lipid peroxidation products in healthy and diseased heart. Clin Exp Pharmacol Physiol 39, 179-193 Lesnefsky, E. J., Minkler, P., and Hoppel, C. L. (2009) Enhanced modification of cardiolipin during ischemia in the aged heart. J Mol Cell Cardiol 46, 1008-1015 Lim, H. Y., and Bodmer, R. (2011) Phospholipid homeostasis and lipotoxic cardiomyopathy: a matter of balance. Fly (Austin) 5, 234-236 Gurung, I. S., Medina-Gomez, G., Kis, A., Baker, M., Velagapudi, V., Neogi, S. G., Campbell, M., Rodriguez-Cuenca, S., Lelliott, C., McFarlane, I., Oresic, M., Grace, A. A., Vidal-Puig, A., and Huang, C. L. (2011) Deletion of the metabolic transcriptional coactivator PGC1beta induces cardiac arrhythmia. Cardiovasc Res 92, 29-38

PT

81.

Heart Lipid Metabolism

41

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

94.

Ovide-Bordeaux, S., Bescond-Jacquet, A., and Grynberg, A. (2005) Cardiac mitochondrial alterations induced by insulin deficiency and hyperinsulinaemia in rats: targeting membrane homeostasis with trimetazidine. Clin Exp Pharmacol Physiol 32, 1061-1070 Cedars, A., Jenkins, C. M., Mancuso, D. J., and Gross, R. W. (2009) Calciumindependent phospholipases in the heart: mediators of cellular signaling, bioenergetics, and ischemia-induced electrophysiologic dysfunction. J Cardiovasc Pharmacol 53, 277289 Lim, H. Y., Wang, W., Wessells, R. J., Ocorr, K., and Bodmer, R. (2011) Phospholipid homeostasis regulates lipid metabolism and cardiac function through SREBP signaling in Drosophila. Genes Dev 25, 189-200 Schmitz, G., and Ruebsaamen, K. (2010) Metabolism and atherogenic disease association of lysophosphatidylcholine. Atherosclerosis 208, 10-18 Sobel, B. E., Corr, P. B., Robison, A. K., Goldstein, R. A., Witkowski, F. X., and Klein, M. S. (1978) Accumulation of lysophosphoglycerides with arrhythmogenic properties in ischemic myocardium. J Clin Invest 62, 546-553 Mancuso, D. J., Abendschein, D. R., Jenkins, C. M., Han, X., Saffitz, J. E., Schuessler, R. B., and Gross, R. W. (2003) Cardiac ischemia activates calcium-independent phospholipase A2beta, precipitating ventricular tachyarrhythmias in transgenic mice: rescue of the lethal electrophysiologic phenotype by mechanism-based inhibition. J Biol Chem 278, 22231-22236 Mancuso, D. J., Han, X., Jenkins, C. M., Lehman, J. J., Sambandam, N., Sims, H. F., Yang, J., Yan, W., Yang, K., Green, K., Abendschein, D. R., Saffitz, J. E., and Gross, R. W. (2007) Dramatic accumulation of triglycerides and precipitation of cardiac hemodynamic dysfunction during brief caloric restriction in transgenic myocardium expressing human calcium-independent phospholipase A2gamma. J Biol Chem 282, 9216-9227 Jenkins, C. M., Cedars, A., and Gross, R. W. (2009) Eicosanoid signalling pathways in the heart. Cardiovasc Res 82, 240-249 Bikman, B. T., and Summers, S. A. (2011) Ceramides as modulators of cellular and whole-body metabolism. J Clin Invest 121, 4222-4230 Novgorodov, S. A., Riley, C. L., Keffler, J. A., Yu, J., Kindy, M. S., Macklin, W. B., Lombard, D. B., and Gudz, T. I. (2015) SIRT3 Deacetylates Ceramide Synthases: Implications for Mitochondrial Dysfunction and Brain Injury. J Biol Chem ahead of print Park, T. S., Hu, Y., Noh, H. L., Drosatos, K., Okajima, K., Buchanan, J., Tuinei, J., Homma, S., Jiang, X. C., Abel, E. D., and Goldberg, I. J. (2008) Ceramide is a cardiotoxin in lipotoxic cardiomyopathy. J Lipid Res 49, 2101-2112 Russo, S. B., Baicu, C. F., Van Laer, A., Geng, T., Kasiganesan, H., Zile, M. R., and Cowart, L. A. (2012) Ceramide synthase 5 mediates lipid-induced autophagy and hypertrophy in cardiomyocytes. J Clin Invest 122, 3919-3930 Russo, S. B., Tidhar, R., Futerman, A. H., and Cowart, L. A. (2013) Myristate-derived d16:0 sphingolipids constitute a cardiac sphingolipid pool with distinct synthetic routes and functional properties. J Biol Chem 288, 13397-13409 Holland, W. L., Miller, R. A., Wang, Z. V., Sun, K., Barth, B. M., Bui, H. H., Davis, K. E., Bikman, B. T., Halberg, N., Rutkowski, J. M., Wade, M. R., Tenorio, V. M., Kuo, M.

99.

101. 102. 103.

104.

105.

106.

107.

SC

AC CE P

TE

100.

NU

98.

MA

97.

D

96.

RI

PT

95.

Heart Lipid Metabolism

42

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

109.

115. 116.

117.

118.

119.

120.

AC CE P

114.

TE

D

113.

NU

112.

MA

111.

SC

RI

110.

S., Brozinick, J. T., Zhang, B. B., Birnbaum, M. J., Summers, S. A., and Scherer, P. E. (2011) Receptor-mediated activation of ceramidase activity initiates the pleiotropic actions of adiponectin. Nat Med 17, 55-63 Abel, E. D., Litwin, S. E., and Sweeney, G. (2008) Cardiac remodeling in obesity. Physiol Rev 88, 389-419 Park, M., and Sweeney, G. (2013) Direct effects of adipokines on the heart: focus on adiponectin. Heart Fail Rev 18, 631-644 Lee, S. Y., Kim, J. R., Hu, Y., Khan, R., Kim, S. J., Bharadwaj, K. G., Davidson, M. M., Choi, C. S., Shin, K. O., Lee, Y. M., Park, W. J., Park, I. S., Jiang, X. C., Goldberg, I. J., and Park, T. S. (2012) Cardiomyocyte specific deficiency of serine palmitoyltransferase subunit 2 reduces ceramide but leads to cardiac dysfunction. J Biol Chem 287, 1842918439 Baranowski, M., Blachnio-Zabielska, A., Hirnle, T., Harasiuk, D., Matlak, K., Knapp, M., Zabielski, P., and Gorski, J. (2010) Myocardium of type 2 diabetic and obese patients is characterized by alterations in sphingolipid metabolic enzymes but not by accumulation of ceramide. J Lipid Res 51, 74-80 Karliner, J. S., Honbo, N., Summers, K., Gray, M. O., and Goetzl, E. J. (2001) The lysophospholipids sphingosine-1-phosphate and lysophosphatidic acid enhance survival during hypoxia in neonatal rat cardiac myocytes. J Mol Cell Cardiol 33, 1713-1717 Hilal-Dandan, R., Means, C. K., Gustafsson, A. B., Morissette, M. R., Adams, J. W., Brunton, L. L., and Heller Brown, J. (2004) Lysophosphatidic acid induces hypertrophy of neonatal cardiac myocytes via activation of Gi and Rho. J Mol Cell Cardiol 36, 481493 Ishii, I., Fukushima, N., Ye, X., and Chun, J. (2004) Lysophospholipid receptors: signaling and biology. Annu Rev Biochem 73, 321-354 Means, C. K., and Brown, J. H. (2009) Sphingosine-1-phosphate receptor signalling in the heart. Cardiovasc Res 82, 193-200 Hofmann, U., Burkard, N., Vogt, C., Thoma, A., Frantz, S., Ertl, G., Ritter, O., and Bonz, A. (2009) Protective effects of sphingosine-1-phosphate receptor agonist treatment after myocardial ischaemia-reperfusion. Cardiovasc Res 83, 285-293 Holland, W. L., Bikman, B. T., Wang, L. P., Yuguang, G., Sargent, K. M., Bulchand, S., Knotts, T. A., Shui, G., Clegg, D. J., Wenk, M. R., Pagliassotti, M. J., Scherer, P. E., and Summers, S. A. (2011) Lipid-induced insulin resistance mediated by the proinflammatory receptor TLR4 requires saturated fatty acid-induced ceramide biosynthesis in mice. J Clin Invest 121, 1858-1870 Dong, B., Qi, D., Yang, L., Huang, Y., Xiao, X., Tai, N., Wen, L., and Wong, F. S. (2012) TLR4 regulates cardiac lipid accumulation and diabetic heart disease in the nonobese diabetic mouse model of type 1 diabetes. Am J Physiol Heart Circ Physiol 303, H732-742 Shi, H., Kokoeva, M. V., Inouye, K., Tzameli, I., Yin, H., and Flier, J. S. (2006) TLR4 links innate immunity and fatty acid-induced insulin resistance. J Clin Invest 116, 30153025 Pal, D., Dasgupta, S., Kundu, R., Maitra, S., Das, G., Mukhopadhyay, S., Ray, S., Majumdar, S. S., and Bhattacharya, S. (2012) Fetuin-A acts as an endogenous ligand of TLR4 to promote lipid-induced insulin resistance. Nat Med 18, 1279-1285

PT

108.

Heart Lipid Metabolism

43

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

126.

127.

128.

129. 130.

131.

132.

133.

134.

PT

RI

SC

NU

125.

MA

124.

D

123.

TE

122.

Huang, S., Rutkowsky, J. M., Snodgrass, R. G., Ono-Moore, K. D., Schneider, D. A., Newman, J. W., Adams, S. H., and Hwang, D. H. (2012) Saturated fatty acids activate TLR-mediated proinflammatory signaling pathways. J Lipid Res 53, 2002-2013 Wong, S. W., Kwon, M. J., Choi, A. M., Kim, H. P., Nakahira, K., and Hwang, D. H. (2009) Fatty acids modulate Toll-like receptor 4 activation through regulation of receptor dimerization and recruitment into lipid rafts in a reactive oxygen species-dependent manner. J Biol Chem 284, 27384-27392 Lin, M. E., Herr, D. R., and Chun, J. (2010) Lysophosphatidic acid (LPA) receptors: signaling properties and disease relevance. Prostaglandins Other Lipid Mediat 91, 130138 Cremers, B., Flesch, M., Kostenis, E., Maack, C., Niedernberg, A., Stoff, A., Sudkamp, M., Wendler, O., and Bohm, M. (2003) Modulation of myocardial contractility by lysophosphatidic acid (LPA). J Mol Cell Cardiol 35, 71-80 Pulinilkunnil, T., An, D., Ghosh, S., Qi, D., Kewalramani, G., Yuen, G., Virk, N., Abrahani, A., and Rodrigues, B. (2005) Lysophosphatidic acid-mediated augmentation of cardiomyocyte lipoprotein lipase involves actin cytoskeleton reorganization. Am J Physiol Heart Circ Physiol 288, H2802-2810 Chen, J., Chen, Y., Zhu, W., Han, Y., Han, B., Xu, R., Deng, L., Cai, Y., Cong, X., Yang, Y., Hu, S., and Chen, X. (2008) Specific LPA receptor subtype mediation of LPAinduced hypertrophy of cardiac myocytes and involvement of Akt and NFkappaB signal pathways. J Cell Biochem 103, 1718-1731 Yang, J., Nie, Y., Wang, F., Hou, J., Cong, X., Hu, S., and Chen, X. (2013) Reciprocal regulation of miR-23a and lysophosphatidic acid receptor signaling in cardiomyocyte hypertrophy. Biochim Biophys Acta 1831, 1386-1394 Lin, Z., Murtaza, I., Wang, K., Jiao, J., Gao, J., and Li, P. F. (2009) miR-23a functions downstream of NFATc3 to regulate cardiac hypertrophy. Proc Natl Acad Sci U S A 106, 12103-12108 Wang, K., Lin, Z. Q., Long, B., Li, J. H., Zhou, J., and Li, P. F. (2012) Cardiac hypertrophy is positively regulated by MicroRNA miR-23a. J Biol Chem 287, 589-599 Rachakonda, V. P., Reeves, V. L., Aljammal, J., Wills, R. C., Trybula, J. S., DeLany, J. P., Kienesberger, P. C., and Kershaw, E. E. (2015) Serum autotaxin is independently associated with hepatic steatosis in women with severe obesity. Obesity (Silver Spring) 23, 965-972 Reeves, V. L., Trybula, J. S., R.C., W., Goodpaster, B. H., J.J., D., Kienesberger, P. C., and Kershaw, E. E. (2015) Serum Autotaxin/ENPP2 correlates with insulin resistance in older humans with obesity. Obesity 23, 2371–2376 Nishimura, S., Nagasaki, M., Okudaira, S., Aoki, J., Ohmori, T., Ohkawa, R., Nakamura, K., Igarashi, K., Yamashita, H., Eto, K., Uno, K., Hayashi, N., Kadowaki, T., Komuro, I., Yatomi, Y., and Nagai, R. (2014) ENPP2 contributes to adipose tissue expansion in dietinduced obesity. Diabetes 63, 4154-4164 Rancoule, C., Dusaulcy, R., Treguer, K., Gres, S., Attane, C., and Saulnier-Blache, J. S. (2013) Involvement of autotaxin/lysophosphatidic acid signaling in obesity and impaired glucose homeostasis. Biochimie 96, 140-143 Unger, R. H., Scherer, P. E., and Holland, W. L. (2013) Dichotomous roles of leptin and adiponectin as enforcers against lipotoxicity during feast and famine. Mol Biol Cell 24, 3011-3015

AC CE P

121.

Heart Lipid Metabolism

44

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

139.

140.

143.

144.

145.

146.

147.

148.

PT

AC CE P

142.

TE

D

141.

RI

138.

SC

137.

NU

136.

Atkinson, L. L., Fischer, M. A., and Lopaschuk, G. D. (2002) Leptin activates cardiac fatty acid oxidation independent of changes in the AMP-activated protein kinase-acetylCoA carboxylase-malonyl-CoA axis. J Biol Chem 277, 29424-29430 Lee, Y., Naseem, R. H., Duplomb, L., Park, B. H., Garry, D. J., Richardson, J. A., Schaffer, J. E., and Unger, R. H. (2004) Hyperleptinemia prevents lipotoxic cardiomyopathy in acyl CoA synthase transgenic mice. Proc Natl Acad Sci U S A 101, 13624-13629 Myers, M. G., Jr., Leibel, R. L., Seeley, R. J., and Schwartz, M. W. (2010) Obesity and leptin resistance: distinguishing cause from effect. Trends Endocrinol Metab 21, 643-651 Churchill, E., Budas, G., Vallentin, A., Koyanagi, T., and Mochly-Rosen, D. (2008) PKC isozymes in chronic cardiac disease: possible therapeutic targets? Annu Rev Pharmacol Toxicol 48, 569-599 Way, K. J., Isshiki, K., Suzuma, K., Yokota, T., Zvagelsky, D., Schoen, F. J., Sandusky, G. E., Pechous, P. A., Vlahos, C. J., Wakasaki, H., and King, G. L. (2002) Expression of connective tissue growth factor is increased in injured myocardium associated with protein kinase C beta2 activation and diabetes. Diabetes 51, 2709-2718 Xia, Z., Kuo, K. H., Nagareddy, P. R., Wang, F., Guo, Z., Guo, T., Jiang, J., and McNeill, J. H. (2007) N-acetylcysteine attenuates PKCbeta2 overexpression and myocardial hypertrophy in streptozotocin-induced diabetic rats. Cardiovasc Res 73, 770-782 Lei, S., Li, H., Xu, J., Liu, Y., Gao, X., Wang, J., Ng, K. F., Lau, W. B., Ma, X. L., Rodrigues, B., Irwin, M. G., and Xia, Z. (2013) Hyperglycemia-induced protein kinase C beta2 activation induces diastolic cardiac dysfunction in diabetic rats by impairing caveolin-3 expression and Akt/eNOS signaling. Diabetes 62, 2318-2328 Ferreira, J. C., Brum, P. C., and Mochly-Rosen, D. (2011) betaIIPKC and epsilonPKC isozymes as potential pharmacological targets in cardiac hypertrophy and heart failure. J Mol Cell Cardiol 51, 479-484 Guo, M., Wu, M. H., Korompai, F., and Yuan, S. Y. (2003) Upregulation of PKC genes and isozymes in cardiovascular tissues during early stages of experimental diabetes. Physiol genomics 12, 139-146 Bowling, N., Walsh, R. A., Song, G., Estridge, T., Sandusky, G. E., Fouts, R. L., Mintze, K., Pickard, T., Roden, R., Bristow, M. R., Sabbah, H. N., Mizrahi, J. L., Gromo, G., King, G. L., and Vlahos, C. J. (1999) Increased protein kinase C activity and expression of Ca2+-sensitive isoforms in the failing human heart. Circulation 99, 384-391 Wakasaki, H., Koya, D., Schoen, F. J., Jirousek, M. R., Ways, D. K., Hoit, B. D., Walsh, R. A., and King, G. L. (1997) Targeted overexpression of protein kinase C beta2 isoform in myocardium causes cardiomyopathy. Proc Natl Acad Sci U S A 94, 9320-9325 Bowman, J. C., Steinberg, S. F., Jiang, T., Geenen, D. L., Fishman, G. I., and Buttrick, P. M. (1997) Expression of protein kinase C beta in the heart causes hypertrophy in adult mice and sudden death in neonates. J Clin Invest 100, 2189-2195 Min, W., Bin, Z. W., Quan, Z. B., Hui, Z. J., and Sheng, F. G. (2009) The signal transduction pathway of PKC/NF-kappa B/c-fos may be involved in the influence of high glucose on the cardiomyocytes of neonatal rats. Cardiovasc Diabetol 8, 8 Kawamura, N., Kubota, T., Kawano, S., Monden, Y., Feldman, A. M., Tsutsui, H., Takeshita, A., and Sunagawa, K. (2005) Blockade of NF-kappaB improves cardiac function and survival without affecting inflammation in TNF-alpha-induced cardiomyopathy. Cardiovasc Res 66, 520-529

MA

135.

Heart Lipid Metabolism

45

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

151.

156.

157.

158.

159.

160.

161.

AC CE P

TE

155.

MA

154.

D

153.

NU

SC

152.

PT

150.

Higuchi, Y., Chan, T. O., Brown, M. A., Zhang, J., DeGeorge, B. R., Jr., Funakoshi, H., Gibson, G., McTiernan, C. F., Kubota, T., Jones, W. K., and Feldman, A. M. (2006) Cardioprotection afforded by NF-kappaB ablation is associated with activation of Akt in mice overexpressing TNF-alpha. Am J Physiol Heart Circ Physiol 290, H590-598 Itani, S. I., Ruderman, N. B., Schmieder, F., and Boden, G. (2002) Lipid-induced insulin resistance in human muscle is associated with changes in diacylglycerol, protein kinase C, and IkappaB-alpha. Diabetes 51, 2005-2011 Ghosh, S., and Baltimore, D. (1990) Activation in vitro of NF-kappa B by phosphorylation of its inhibitor I kappa B. Nature 344, 678-682 Yu, C., Chen, Y., Cline, G. W., Zhang, D., Zong, H., Wang, Y., Bergeron, R., Kim, J. K., Cushman, S. W., Cooney, G. J., Atcheson, B., White, M. F., Kraegen, E. W., and Shulman, G. I. (2002) Mechanism by which fatty acids inhibit insulin activation of insulin receptor substrate-1 (IRS-1)-associated phosphatidylinositol 3-kinase activity in muscle. J Biol Chem 277, 50230-50236 Kang, N., Alexander, G., Park, J. K., Maasch, C., Buchwalow, I., Luft, F. C., and Haller, H. (1999) Differential expression of protein kinase C isoforms in streptozotocin-induced diabetic rats. Kidney Int 56, 1737-1750 Galbo, T., Perry, R. J., Jurczak, M. J., Camporez, J. P., Alves, T. C., Kahn, M., Guigni, B. A., Serr, J., Zhang, D., Bhanot, S., Samuel, V. T., and Shulman, G. I. (2013) Saturated and unsaturated fat induce hepatic insulin resistance independently of TLR-4 signaling and ceramide synthesis in vivo. Proc Natl Acad Sci U S A 110, 12780-12785 Eichmann, T. O., Kumari, M., Haas, J. T., Farese, R. V., Jr., Zimmermann, R., Lass, A., and Zechner, R. (2012) Studies on the substrate and stereo/regioselectivity of adipose triglyceride lipase, hormone-sensitive lipase, and diacylglycerol-O-acyltransferases. J Biol Chem 287, 41446-41457 Pettitt, T. R., Martin, A., Horton, T., Liossis, C., Lord, J. M., and Wakelam, M. J. (1997) Diacylglycerol and phosphatidate generated by phospholipases C and D, respectively, have distinct fatty acid compositions and functions. Phospholipase D-derived diacylglycerol does not activate protein kinase C in porcine aortic endothelial cells. J Biol Chem 272, 17354-17359 Madani, S., Hichami, A., Legrand, A., Belleville, J., and Khan, N. A. (2001) Implication of acyl chain of diacylglycerols in activation of different isoforms of protein kinase C. Faseb J 15, 2595-2601 Cuschieri, J., Bulger, E., Billgrin, J., Garcia, I., and Maier, R. V. (2007) Acid sphingomyelinase is required for lipid Raft TLR4 complex formation. Surg Infect (Larchmt) 8, 91-106 Stratford, S., Hoehn, K. L., Liu, F., and Summers, S. A. (2004) Regulation of insulin action by ceramide: dual mechanisms linking ceramide accumulation to the inhibition of Akt/protein kinase B. J Biol Chem 279, 36608-36615 Powell, D. J., Hajduch, E., Kular, G., and Hundal, H. S. (2003) Ceramide disables 3phosphoinositide binding to the pleckstrin homology domain of protein kinase B (PKB)/Akt by a PKCzeta-dependent mechanism. Mol Cell Biol 23, 7794-7808 Miura, A., Kajita, K., Ishizawa, M., Kanoh, Y., Kawai, Y., Natsume, Y., Sakuma, H., Yamamoto, Y., Yasuda, K., and Ishizuka, T. (2003) Inhibitory effect of ceramide on insulin-induced protein kinase Czeta translocation in rat adipocytes. Metabolism 52, 1924

RI

149.

Heart Lipid Metabolism

46

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

166.

167.

168.

170.

171.

172.

173.

PT

RI

AC CE P

TE

169.

SC

165.

NU

164.

MA

163.

Jaeschke, A., and Davis, R. J. (2007) Metabolic stress signaling mediated by mixedlineage kinases. Mol Cell 27, 498-508 Galadari, S., Rahman, A., Pallichankandy, S., Galadari, A., and Thayyullathil, F. (2013) Role of ceramide in diabetes mellitus: evidence and mechanisms. Lipids Health Dis 12, 98 Sathyanarayana, P., Barthwal, M. K., Kundu, C. N., Lane, M. E., Bergmann, A., Tzivion, G., and Rana, A. (2002) Activation of the Drosophila MLK by ceramide reveals TNFalpha and ceramide as agonists of mammalian MLK3. Mol Cell 10, 1527-1533 Xu, Z., Maroney, A. C., Dobrzanski, P., Kukekov, N. V., and Greene, L. A. (2001) The MLK family mediates c-Jun N-terminal kinase activation in neuronal apoptosis. Mol Cell Biol 21, 4713-4724 Kanety, H., Hemi, R., Papa, M. Z., and Karasik, A. (1996) Sphingomyelinase and ceramide suppress insulin-induced tyrosine phosphorylation of the insulin receptor substrate-1. J Biol Chem 271, 9895-9897 Ola, A., Kerkela, R., Tokola, H., Pikkarainen, S., Skoumal, R., Vuolteenaho, O., and Ruskoaho, H. (2010) The mixed-lineage kinase 1-3 signalling pathway regulates stress response in cardiac myocytes via GATA-4 and AP-1 transcription factors. Br J Pharmacol 159, 717-725 Chen, C. L., Lin, C. F., Chang, W. T., Huang, W. C., Teng, C. F., and Lin, Y. S. (2008) Ceramide induces p38 MAPK and JNK activation through a mechanism involving a thioredoxin-interacting protein-mediated pathway. Blood 111, 4365-4374 Hreniuk, D., Garay, M., Gaarde, W., Monia, B. P., McKay, R. A., and Cioffi, C. L. (2001) Inhibition of c-Jun N-terminal kinase 1, but not c-Jun N-terminal kinase 2, suppresses apoptosis induced by ischemia/reoxygenation in rat cardiac myocytes. Mol Pharmacol 59, 867-874 Liang, Q., and Molkentin, J. D. (2003) Redefining the roles of p38 and JNK signaling in cardiac hypertrophy: dichotomy between cultured myocytes and animal models. J Mol Cell Cardiol 35, 1385-1394 Aoki, H., Kang, P. M., Hampe, J., Yoshimura, K., Noma, T., Matsuzaki, M., and Izumo, S. (2002) Direct activation of mitochondrial apoptosis machinery by c-Jun N-terminal kinase in adult cardiac myocytes. J Biol Chem 277, 10244-10250 Wang, J., Zhen, L., Klug, M. G., Wood, D., Wu, X., and Mizrahi, J. (2000) Involvement of caspase 3- and 8-like proteases in ceramide-induced apoptosis of cardiomyocytes. J Card Fail 6, 243-249 Dyntar, D., Eppenberger-Eberhardt, M., Maedler, K., Pruschy, M., Eppenberger, H. M., Spinas, G. A., and Donath, M. Y. (2001) Glucose and palmitic acid induce degeneration of myofibrils and modulate apoptosis in rat adult cardiomyocytes. Diabetes 50, 21052113

D

162.

Heart Lipid Metabolism

47

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

Lipid peroxides Free fatty acids Acylcarnitines Ceramides

Lipotoxicity

Diacylglycerols

Long-chain acyl-CoAs

Apoptotic cell death Cardiac dysfunction Cardiac remodeling Arrhythmia Heart failure

RI

PT

Insulin resistance Mitochondrial damage ATP starvation ER stress

SC

Obesity Insulin resistance Diabetes Aging Myocardial ischemia/ reperfusion etc.

NU

Lysophospholipids

Figure 1. Causes and consequences of cardiac lipotoxicity. Stressors including metabolic

MA

diseases, aging, and myocardial ischemia-reperfusion can lead to increased accumulation of lipids and lipid metabolites in the heart that have toxic effects on cardiomyocytes. Cardiac

AC CE P

TE

D

lipotoxicity can trigger cell stress and impair myocardial function.

48

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

PIP2 PLC

Lipoprotein

Lipid droplet PL

DAG DAG

PT

Lyso-PL, eicosanoids

HSL

TAG

SC

RI

FA/acylATGL CoA CGI-58 PLIN5

TAG

Acyl-CoA

Lipid peroxides

NU

Acyl-CoA

Acylcarnitines

ROS, peroxynitrite

Ceramides

FA uptake FA oxidation Lipid storage

CerS5 SPT1 ER

FoxO1 PPARα/δ/γ Nucleus

AC CE P

Mitochondria

TE

D

β-oxidation

MA

S1P

DGAT1

DAG

Figure 2. Schematic representation of lipids and proteins involved in lipid uptake and metabolism that have been implicated in cardiac lipotoxicity. Excess FA delivery and uptake in cardiomyocytes, impaired or incomplete FA oxidation, lipid peroxidation, as well as ceramide, glycerolipid, and phospholipid metabolism have been linked to cardiac lipotoxicity. Lipid classes and metabolites that have been associated with lipotoxicity in the heart include long chain acylCoAs, FAs, acylcarnitines, lipid peroxides, ceramides and other sphingolipids, DAGs, lyso-PLs, and eicosanoids (highlighted in bold letters).

49

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

Insulin

FAs

TNFα IR

SMase

p38 MAPK

IRS

MLK3

NFκB

JNK

SC

PI3K

PKCδ

NFκB

PP2A Caspase 9

Akt

Apoptosis

MA

Ceramide

NU

Caspase 3/8

RI

classical/novel PKCs

Ceramide JNK

Sarcolemma Insulin resistance

IκB Nucleus

NFκB

D

Mitochondria

TNFR

DAG

PT

TLR4

TE

Figure 3. Proposed mechanisms of cardiac lipotoxicity induced by DAGs and ceramides.

AC CE P

DAGs induce lipotoxicity through the activation of classical and novel protein kinase C (PKC) isoforms and subsequent inhibition of insulin signaling at the level of insulin receptor (IRS) as well as through activation of nuclear factor κB (NFκB) and inflammatory cytokine signaling. Additionally, NFκB activation can lead to increased ceramide synthesis. Ceramides are also mediators of toll-like receptor 4 signaling and can induce insulin resistance via the activation of MAPK signaling and IRS inhibition or inhibition of Akt via protein kinase Cδ (PKCδ) and protein phosphatase 2A (PP2A). By stimulating caspases ceramides can also induce cellular apoptosis.

50

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

Conflicts of Interest

AC CE P

TE

D

MA

NU

SC

RI

PT

We have no conflicts of interest to declare.

51

ACCEPTED MANUSCRIPT BBA Molecular and Cell Biology of Lipids

Heart Lipid Metabolism

Highlights Cardiomyocyte lipid overload can have toxic effects and cause cardiac dysfunction.



Cardiac lipotoxicity is observed during pathophysiological conditions including obesity,

PT



RI

insulin resistance, diabetes, aging, and myocardial ischemia-reperfusion. Studies using animal models have uncovered mechanisms of cardiac lipotoxicity.



Proteins involved in FA uptake and oxidation as well as TAG, sphingolipid, and

SC



AC CE P

TE

D

MA

NU

phospholipid metabolism have been implicated in cardiac lipotoxicity.

52

Lipid metabolism and signaling in cardiac lipotoxicity.

The heart balances uptake, metabolism and oxidation of fatty acids (FAs) to maintain ATP production, membrane biosynthesis and lipid signaling. Under ...
735KB Sizes 6 Downloads 9 Views