You are on page 1of 15

Prostaglandins and Other Lipid Mediators 164 (2023) 106698

Contents lists available at ScienceDirect

Prostaglandins and Other Lipid Mediators


journal homepage: www.elsevier.com/locate/prostaglandins

Lipoxin and glycation in SREBP signaling: Insight into diabetic


cardiomyopathy and associated lipotoxicity
Muskan Thakur, Rashmi S. Tupe *
Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune 412115, Maharashtra State, India

A R T I C L E I N F O A B S T R A C T

Keywords: Diabetes and cardiovascular diseases are the leading cause of morbidity and mortality worldwide. Diabetes in­
Diabetic cardiomyopathy creases cardiovascular risk through hyperglycemia and atherosclerosis. Chronic hyperglycemia accelerates gly­
Lipid metabolism cation reaction, which forms advanced glycation end products (AGEs). Additionally, hyperglycemia with
Glycation
enhanced levels of cholesterol, native and oxidized low-density lipoproteins, free fatty acids, and oxidative stress
Lipoxin
SREBP signaling
induces lipotoxicity. Accelerated glycation and disturbed lipid metabolism are characteristic features of diabetic
heart failure. SREBP signaling plays a significant role in lipid and glucose homeostasis. AGEs increase lipotoxicity
in diabetic cardiomyopathy by inhibiting SREBP signaling. While anti-inflammatory lipid mediators, lipoxins
resolve inflammation caused by lipotoxicity by upregulating the PPARγ expression and regulating CD36. PPARγ
connects the bridge between glycation and lipoxin in SREBP signaling. A summary of treatment modalities
against diabetic cardiomyopathy is given in brief. This review indicates the novel therapeutic approach in the
crosstalk between glycation and lipoxin in SREBP signaling.

1. Introduction systolic and diastolic dysfunctions, thereby causing diabetic cardiomy­


opathy (DCM) [4]. They also activate multiple intracellular signaling
Cardiovascular diseases (CVDs) impact a significant part of the pathways and trigger inflammation [10].
global population, with a 31.4% death rate from 50% of global mortality Uninterrupted cardiac muscle contraction needs high energy inputs
caused due to non-communicable diseases cumulatively [1]. Most recent from biomolecules like carbohydrates, lipids, proteins, ketones, and
figures show the logarithmic increase in the mortality rate due to CVDs fatty acids [11]. Lipids are the most abundant source of energy for the
which was 18.6 million in 2019, indicating that CVDs are a global topic heart as the normal heart generates about 70–90% ATP from fatty acid
of concern [2]. Various risk factors and stimulators increase the severity oxidation [12] (Fig. 1). Moreover, lipids serve many critical roles in
of CVDs; one of them is diabetes, a highly prevalent disease worldwide biological systems as they are the signaling molecules, substrates for
[3,4]. Globally, CVDs affect approximately 32.2% of persons with type 2 β-oxidation, and target moieties for post-translational protein modifi­
diabetes mellitus [5]. Likewise, type 2 diabetes is associated with a cations [13]. Various lipid mediators produced from arachidonic acid
2–4-fold increase in the risk of sudden cardiac arrest or heart failure [6]. have a wide range of therapeutic advantages (discussed later in this
One of the significant risk factors of diabetic-CVDs is glycation, a reac­ review) in DCM. Importantly, α, β- polyunsaturated lipid aldehydes act
tion between the carbonyl group of reducing sugar in the bloodstream as lipid electrophiles that modify lipids, proteins, and nucleic acids [14].
and the free amino group of proteins and lipids occurring during chronic A heart can switch its substrate selection based on the energy demand to
hyperglycemia [7–9]. It gives rise to various advanced glycation end cope with critical physiological and pathological conditions. Disturbed
products (AGEs), having receptor-dependent and independent effects on metabolism of cardiac energy is the primary characteristic of a failing
cells. AGEs disrupt extracellular matrix proteins’ structural, functional, heart where in pathological situations, impaired insulin signaling,
and enzymatic properties and hamper their degradation, leading to lipolysis, and fatty acid release from adipose tissue are augmented

Abbreviations: CVDs, Cardiovascular diseases; DCM, Diabetic cardiomyopathy; LDL, Low-density lipoprotein; Ox-LDL, Oxidized LDL; AGEs, Advanced glycation
end products; RAGE, Receptor for AGEs; SREBP, Sterol regulator element binding protein; LX, Lipoxin; LO, Lipoxygenase; ROS, Reactive oxygen species; CD36,
Cluster of differentiation 36; PPARγ,, Peroxisome proliferator-activated receptors gamma.
* Corresponding author.
E-mail address: rashmi.tupe@ssbs.edu.in (R.S. Tupe).

https://doi.org/10.1016/j.prostaglandins.2022.106698
Received 26 September 2022; Received in revised form 8 November 2022; Accepted 10 November 2022
Available online 13 November 2022
1098-8823/© 2022 Elsevier Inc. All rights reserved.
M. Thakur and R.S. Tupe Prostaglandins and Other Lipid Mediators 164 (2023) 106698

Fig. 1. Glucose and fatty acid metabolism in a healthy heart. In cardiomyocytes, glucose and free fatty acids are taken in through their respective receptors, CD36
and GLUT4. In the cytosol, acetyl CoA is produced from free fatty acids, and glucose produces pyruvate through glycolysis. Once pyruvate is transported to the
mitochondria, it produces acetyl CoA. On the other hand, the carnitine receptor facilitates the transport of long-chain fatty acids into mitochondria where it is
modified to acetyl-CoA on β-oxidation. Acetyl-CoA produced from fatty acid and glucose metabolism makes FADH2 and NADH through the TCA cycle and generates
ATP after entering into ETC, thereby, maintaining cardiac energy metabolism. Abbreviations: CoA, coenzyme A, TCA, tricarboxylic acid cycle; FADH2, Flavin adenine
dinucleotide; NADH, nicotinamide adenine dinucleotide; ECT, electron transport chain; ATP, adenosine triphosphate; CPT-1, carnitine palmitoyltransferase-1; CD36,
cluster of differentiation 36; ACS, acyl-CoA synthetases; CACT, carnitine-acylcarnitine translocase; CPT-2, carnitine palmitoyltransferase-2; MPC, mitochondrial
pyruvate carrier; ADP, adenosine diphosphate; GLUT4, glucose transporter type 4.

[15–17]. A decrease in the oxidation rate of fatty acids and more reli­ glucose toxicity to lipotoxicity in diabetic hearts. Similarly, another
ability to glycolysis for ATP generation indicate impaired cardiac study described miR-320 as a late-responding miRNA that exacerbates
functioning. Disturbed substrate switch and selection are also mainly apoptosis and cardiac dysfunctions in the diabetic milieu, concluding
associated with deleterious consequences [18]. Gradually, the heart gets that knocking out miR-320 would be an effective treatment strategy
used to the rapid adoption of fatty acids for ATP generation. Chronic against DCM [24]. Furthermore, cumulative pieces of evidence indi­
situations often fatigue the left ventricle for adequate cardiac output and cated that cardiac lipotoxicity could affect heart functions through
lead to DCM [19] (Fig. 2). modulating miRNA profiling as they can limit senescence (miR-195),
Due to the great importance of lipid metabolism in DCM and other regulate microvascular complications (miR-93), decrease fibrosis and
metabolic disorders, researchers are keen to explore signaling molecules vascular permeability (miR-200a-3p, miR-200b) and help the patients to
or lipid mediators associated with the pathogenesis of DCM. Recently, it recover from hyperglycemia, diabetes mellitus, and associated diabetic
has been reported that sarcolemmal substrate transporters for fatty complications. However, many other miRNAs are linked with the dis­
acids, glucose transporter type 4, and a cluster of differentiation 36 ease progression (miR-351 [25], miR-34a, miR-146, miR-195, etc.). This
(CD36) play a crucial role in cardiac energy metabolism [20], as they hints that miRNAs can be explored more in DCM-related pathogenesis
can shift lipid metabolism to lipotoxicity. Thus, they can be targeted for [26].
metabolic modulation therapy against DCM. Inspecting CD36 levels can While research continues to study lipid metabolism through various
regulate cardiac energy, lipid metabolism and repair cardiac contrac­ aspects, this review revises the role of lipid metabolism in yet another set
tions [21]. Upregulated expression of CD36 was found in the diabetic of inflammation mediators of DCM. Our work focuses on glycation,
heart; later, it was discussed that miR-320 could suppress CD36 tran­ hampered lipid metabolism in DCM, cellular signaling events, and the
scriptions and improve cardiac functions [22]. In the context of lipid mediators involved in DCM pathogenesis. This review also dis­
miR-320, Li and colleagues have explained how miR-320 induces car­ cusses the need for translation studies, a lab-to-bed approach, with po­
diac lipotoxicity by enhancing CD36 gene expressions [23]. Their tential therapeutics for improved or novel diagnostic or prognostic
research showed upregulated miR-320 expressions in the nucleus of biomarkers for alleviating DCM.
cardiomyocytes of db/db mice and diabetic hearts from patients with
cardiac dysfunctions, suggesting the role of miR-320 as a small acti­ 1.1. Multifactorial etiology of CVDs in diabetes
vating RNA at the transcriptional level. Also, CD36 was found to be the
target gene of this miRNA. As a result, the translocation of miR-320 to One of the drastic consequences of diabetes is CVDs. In addition to
the nucleus upregulated CD36 expressions, disrupting cardiac lipid biochemical pathways connecting diabetes to CVDs, some lifestyle-
metabolism. Thus, the miR-320/CD36 pathway was summarized to link related factors also take part in diabetes leading to CVDs [27].

2
M. Thakur and R.S. Tupe Prostaglandins and Other Lipid Mediators 164 (2023) 106698

Multiple factors associated with diabetes participate in CVDs patho­


genesis; some are illustrated in Fig. 3.

1.1.1. Aging and senescence


It is also believed that the prevalence of diabetes increases with
aging. It is the most common incidence in adults aged 65 years and older
[28]. Aging is responsible for reduced insulin sensitivity in patients with
diabetes. Age-associated elevated visceral adiposity results in high levels
of pro-inflammatory cytokines in the blood that interfere with insulin
signaling [29]. Insulin resistance in people with diabetes impairs lipid
uptake and storage, leading to elevated plasma levels of atherogenic
low-density lipoprotein (LDL), causing CVDs-associated mortality [30].
The aging of cells in a diabetic milieu triggers senescence [31]. Type 2
diabetes is often described as a disease of premature immunosenescence
with a loss of naïve CD4+ and CD8+ T cells with elevated late-stage T cell
differentiation [32–34]. In 2019, it was demonstrated that senescent T
cells cause hyperglycemia and inflammation [34,35], involving cardiac
hypertrophy, fibrosis, arterial stiffness, altered calcium signaling,
mitochondrial and endothelial dysfunctions, excess reactive oxygen
species (ROS) generation, impaired cell signaling [36]. Studies have
shown senescence accumulation in most cardiac cell populations that
modifies cell phenotypes and enhances inflammation, ending in heart
failure in the diabetic population [37,38].

1.1.2. Sex difference and body mass index


There are well-established differences between women and men in
terms of epidemiology, pathophysiology, effects of treatment therapies,
and clinical outcomes [39]. These differences are called sex differences
due to differential gene expression from sex chromosomes and the car­
diovascular system. It is established that ovulating women without
Fig. 2. Disturbed cardiac energy metabolism leading to the DCM. Impaired diabetes experience significantly lower cardiovascular incidences than
glucose and fatty acid metabolism enhance cardiac overload and pressure diabetic women and men, hinting at the role of estrogen hormone in
through oxidation, and metabolic inflammation, leading to cardiac hypertro­
reducing CVDs events [40]. However, in the diabetic population, men
phy, contractile dysfunctions, and, ultimately, DCM. Abbreviation: ATP, aden­
are twice, and females are five times at risk of developing CVDs
osine triphosphate.
compared to the general population [41]. Women show end-systolic
ventricular pressures at a much higher rate than men [39]. This sug­
gests that estrogen replacement therapy may help ameliorate CVDs.

Fig. 3. Factors associating diabetes with CVD. Formation of AGEs in hyperglycemia, promotion of atherosclerosis and dyslipidemia, macrovascular defects,
endothelial dysfunction, leukocyte recruitment and aggregation, augmented oxidative stress, and mitochondrial dysfunction is some of the paramount factors
participating in DCM pathogenesis. In addition, ruptured plaque leads to thrombotic cell death resulting in insufficient inflammatory resolution.

3
M. Thakur and R.S. Tupe Prostaglandins and Other Lipid Mediators 164 (2023) 106698

However, clinical studies have challenged this notion [42–45]. On the individuals [60], progressing to various diabetic complications such as
other hand, body mass index is tightly associated with a higher risk of DCM.
diabetes mellitus, hypertension, and CVDs. Increased risk of diabetes
and associated cardiac complications were observed at higher BMI levels 1.1.5. ROS production, lipid metabolism, and mitochondrial stress
in men than in women [46]. A higher body mass index defines obesity, Small changes or modifications in lipids’ location or composition can
the apparent reason behind diabetes and CVDs related mortality. significantly affect cell functions and their survival. Oxidative stress
Obesity can participate in low-grade systemic inflammation, abrupt occurs when the cellular environment becomes excessively oxidized
immune response, maladaptive immune metabolism, and cytokine because of ROS overproduction. Enhanced oxidative stress in diabetes
release, further developing cardiac hypertrophy, fibrosis, and cardio­ can directly damage lipids and promotes their lipid peroxidation, which
myopathy [47,48]. has various effects on cellular functions, such as increased apoptosis,
decreased autophagy, disturbed cell proliferation, transcriptional
1.1.3. Atherosclerosis and dyslipidemia deregulation [14,61], enhanced oxidative stress-mediated inflammatory
Diabetes can align itself with the state of chronic, low-level inflam­ signaling and leukocytes activation, cytokine secretion and chronic
mation where increased ROS production in hyperglycemia accelerates inflammation, leading to DCM.
the process of atherosclerosis, a focal, progressive, and stable disease Almost all cell organelles produce ROS, and mitochondria generating
where arterial lesions are caused. Diabetes may only have one pathway hydrogen peroxide is believed to be the primary source of oxidants [62].
to cause atherosclerosis which is dyslipidemia. It is highly related to High glucose flux in diabetes helps in excess ROS generation, enhancing
diabetes pathogenesis, as 97% of patients who have diabetes are dysli­ mitochondrial dysfunction [63]. Furthermore, hyperglycemia-induced
pidemic [49,50]. These lesions are promoted by LDL and the accumu­ mitochondrial ROS has been found to accelerate almost all the path­
lation of fatty substances, calcium, cholesterol, and fibrin, which fill up ways by which hyperglycemia causes diabetic complications, such as
the inner lining of the arteries or other vessel walls and block the protein kinase C activation, positive regulation of hexosamine, and
oxygenated blood flow to the heart and other body parts. The de­ polyol pathways [58]. A class of cytotoxic lipids directly affects mito­
positions build up over time and narrow the arterial diameter [51]. As chondrial stress, resulting in cell apoptosis. In the inner mitochondrial
the atherosclerotic plaques grow, they come in contact with circulating membrane, bisphosphatidyl glycerol phospholipid cardiolipin is local­
platelets. As a result, platelets get aggregated into the atherosclerotic ized which is highly susceptible to lipid peroxidation and generates
plaque, ultimately leading to thrombotic cell death. Due to increased hydroperoxides, one of them is 4-hydroxynonenal, known for disrupting
ROS production, hyperactive platelets also elevate inflammation and mitochondria and their functioning [64,65]. 4-hydroxynonenal also acts
thrombosis [52]. Atherosclerosis might cause problems like angina, as a signaling molecule and regulates several signaling pathways
strokes, or myocardial infarctions. The plaques form in the smaller ar­ involved in oxidative stress, cell proliferation, senescence, autophagy,
teries causing microvascular disorder [53]. Macrovascular defects apoptosis, and necrosis, such as nuclear factor erythroid 2-related factor
caused due to diabetes in CVDs mainly focus on atherosclerosis. One of 2, activating protein-1, NF-κB, and peroxisome-proliferator-activated
the prominent LDL-cholesterol in atherosclerosis is its small and dense receptors (PPAR) [66]. In addition, the oxidation of cardiolipin can
form. They can bond strongly with extracellular adhesion molecules hamper the activity of complexes I and IV, leading to impaired mito­
[54]. They are more susceptible to oxidation and can penetrate the chondrial respiration and energy production. Reduced cardiolipin and
arterial wall and accelerate atherosclerosis. Oxidized LDL (ox-LDL) is its oxidation mediated-mitochondrial dysfunction are well connected
more prone to acquire new properties and is considered “foreign” by the with CVDs pathogenesis, diabetes, atherosclerosis, and many more
immune system [55]. Structural and functional modifications in ox-LDL metabolic disorders [67].
attract leukocytes to the injured endothelial. Leukocytes’ enhanced
ability to take up ox-LDL promotes their differentiation into foam cells 1.1.6. ROS production, lipid metabolism, and endoplasmic reticulum stress
[56]. The endoplasmic reticulum is the primary cellular organelle for
Contrastingly, high-density lipoproteins are known for their athe­ synthesizing lipids, protein folding and maturation, and calcium ho­
roprotective nature as they perform reverse cholesterol transport. In this meostasis [68]. A significant part of the integral and secretory proteins
process, cell cholesterol is removed by efflux to apoproteinA-I and high- translocate into the endoplasmic reticulum lumen as it provides space
density lipoprotein as extracellular acceptors, transporting the choles­ and environment for posttranslational protein modifications and their
terol to the liver for excretion or its utilization in the form of bile acids foldings [69]. Misleading endoplasmic reticulum functioning causes
[57]. oxidative stress, disturbed calcium homeostasis, ischemia, and over­
expression of misfolded proteins, leading to endoplasmic reticulum
1.1.4. Inflammatory cytokines stress and activation of unfolding protein response. Such a response
Activation of multiple intracellular pathways, including NF-κB dur­ either halts further posttranslational stage of misfolded proteins or ac­
ing diabetes and CVDs, facilitates the secretion of pro-inflammatory tivates signaling pathways to produce molecular chaperons that correct
cytokines. As soon as cytokines are released from the inflamed cells, protein folding. If nothing happens, this response switches on apoptotic
leukocytes get activated and start accumulating at the injury site. Acti­ signaling, playing an essential role in DCM [70,71]. Li et al. (2007)
vated leukocytes also produce ROS in excess, anchoring the activation of showed the interference of endoplasmic reticulum stress in the apoptosis
inflammatory pathways and contributing to CVDs pathophysiology of cardiomyocytes in streptozotocin-induced diabetic rats [72]. Studies
[58]. This immune response from leukocytes enhances chronic inflam­ have pointed out a close association between endoplasmic reticulum
mation conditions when incoming leukocytes cannot dispose of the site stress and dysregulated lipid metabolism in various cells having meta­
of inflammation, injuring endothelial cells. Accelerated cytokine secre­ bolic importance [73]. Other studies have shown that the crosstalk be­
tion from inflamed endothelial enhances the lipolysis of adipocytes. tween hyperglycemia-induced glycation and oxidative stress can
Increased lipolysis increases the levels of circulating free fatty acids that progress diabetes, CVDs, and eventually DCM [74]. Thus, targeting the
deposit in the target tissues [59]. Abnormal levels and metabolism of endoplasmic reticulum and oxidative stress induced by glycation in the
lipids, fatty acids, cholesterol, and other metabolites present around the diabetic milieu can ameliorate diabetes-induced cardiac remodeling and
cells help in the secretion of inflammatory cytokines (C-reactive protein, cardiomyopathy.
plasminogen activator inhibitor-1, interleukin-1β, etc.) and activate
monocytes, which later turn into foam cells, accelerating atheroscle­ 2. Glycation and lipid metabolism: digging deep into DCM
rosis, myocytes’ stiffness and scarring, thus leading to DCM. Increased
secretion of cytokines often enhances insulin resistance in diabetic Glycation is the crucial player in DCM pathogenesis as it causes

4
M. Thakur and R.S. Tupe Prostaglandins and Other Lipid Mediators 164 (2023) 106698

Fig. 4. Glycation reaction leading to DCM. AGEs, through their receptor-dependent or independent actions, hamper extracellular matrix proteins’ structural and
functional properties. It also disturbs glucose and lipid metabolism, and together these defects lead to DCM. Abbreviations: AGEs, advanced glycation end products;
RAGE, receptor for AGEs; ECM, extracellular matrix.

Table 1
Various types of AGEs associated with DCM.
Classification of AGEs Examples Role in DCM References

Cytotoxicity Toxic Crosslinking AGEs Vesperlysine, Pentosidine, Crossline, Increased vascular and myocardial stiffness, [79]
AGEs with fluorescence MRX, Vesper lysine A, GOLD, MOLD, endothelial dysfunction, atherosclerotic plaque
GOLDIC, MOLDIC, glucosepane. formation
Non-fluorescent IDL, Alkyl formyl glycosyl pyrrole,
cross-linking Arginine lysine imidazole,
Glucosepane
Non- Noncross-linking and Pyrraline, Argpyrimidine, MG- The amount of CML significantly increased in the [80]
toxic nonfluorescent AGEs imidazolones, 3-DG-imidizolones, arterial wall of AGE-albumin-treated animals. Even in
AGEs GA-pyridine, CML, CEL, FL the absence of DM, AGEs triggered atherogenesis in
dyslipidemic mice.
Classification based on the Modified protein HbA1c, Type IV collagen, crystalline, Exhibits a slow turnover rate of about ten years. It [81]
type of structure and the apo B100, accumulates with a yearly rate of about 3,7%,
origin of the substrates reaching a 30–50% increase at 80 years of age.
Low molecular AGEs Pyralline, CML, CEL, Pentosidine, Biomarker of tissue amine modification linked to the [82]
Imidazole presence of (unmeasured) toxic molecules with cross-
linking potential or the ability to activate specific AGE
receptors
Produced from a particular AGE-1, AGE-2, AGE-3, AGE-4, AGE-5, Interaction of angiogenic bone marrow cells with [83]
glycating agent AGE-6, AA-AGE AGE-modified collagen results in reduced cell
adhesion, increased susceptibility to apoptosis, fewer
progenitor cells, and reduced angiogenic potential.

Abbreviations: AGEs (advanced glycation end-products), MRX (Maillard reaction product X), GOLD (glyoxal lysine dimmer), MOLD (methylglyoxal lysine dimer), IDL
(imidazolium dilysine), AFGP (alkyl formyl glycosyl pyrrole), ALI (arginine lysine imidazole), MG-imidazolones (methylglyoxal-imidazolones), 3-DG-imidizolones (3-
deoxyglucosone-imidizolones), GA-pyridine (gallic acid-pyridine), CML (N-carboxymethyllysine), CEL (N-carboxyethyl-lysine), FL (N-fructosyl-lysine), apoB100
(apolipoprotein B100), HbA1c (Hemoglobin A1c), AGE-1 (glucose-derived AGEs), AGE-2 (glyceraldehyde-derived AGEs), AGE-3 (glycoaldehyde derived AGEs), AGE-4
(Methylglyoxal-derived AGEs), AGE-5 (glyoxal-derived AGEs), AGE-6 (3-deoxyglucosone-derived AGEs), AA-AGE (acetaldehyde-derived AGEs)

atherogenesis by maintaining the stability of atherosclerotic plaques, N-carboxyethyl-lysine, methylglyoxal lysine dimer, glyoxal lysine dim­
mainly by AGEs formation [75]. Different AGEs have inflammatory ef­ mer) [53]. These AGEs are principal cardiometabolic risk factors.
fects on DCM through either receptor or non-receptor-mediated path­ AGEs-induced lipid accumulation is correlated with autophagy; cells
ways (Fig. 4) [10]. Glycation of apoprotein B and LDL leads to functional encourage autophagy before lipid accumulation, which is how glycation
disruption and turns their momentums towards oxidation, forming induces cell death [76]. AGEs interfere with native proteins and can
glyoxal, glycol-aldehydes, and other carbonyl intermediates. The resist their expected degradation via intracellular proteasomal complex,
carbonyl compounds further interact with extracellular matrix proteins thereby elevating oxidative stress inside or outside the cardiomyocytes
and form AGEs (e.g., imidazolone, N-carboxymethyl lysine, [77]. This alters the permeability of the extracellular matrix and traps

5
M. Thakur and R.S. Tupe Prostaglandins and Other Lipid Mediators 164 (2023) 106698

Fig. 5. Biosynthesis of LXs and their epimers in inflammation settings. Glycation and disrupted lipid metabolism-induced inflammation stimulates cardiomyocyte’s
membrane phospholipids and facilitate the release of arachidonic acid by the cytosolic phospholipase 2 enzyme, producing LXs and their isomers via LO and COX-2
pathways. Abbreviations: AGEs, advanced glycation end products; RAGE, receptor for AGEs; LDL, low-density lipoprotein; CD36, cluster of differentiation 36; ROS,
reactive oxygen species; cPLA2, cytosolic phospholipase A2; LO, lipoxygenase; 15(s)-HETE, 15(s)- hydroxyeicosatetraenoic acid; 15(R)-HETE, 15(R)- hydrox­
yeicosatetraenoic acid; COX 2, cycloxygenase 2; FLAP, 5-lipoxygenase activating protein; LTA4, leukotriene 4; LTB4, LTC4, leukotriene cysteinyl 4; LX, lipoxin.

the macromolecules against the vessel wall. As a result, the extracellular including neutrophils and macrophages [90]. LXs come from non-classic
matrix releases inflammatory cytokines and growth factors, enhancing eicosanoids and are members of the specialized pro-resolving mediators
the endothelium’s antithrombotic properties and the vessel wall’s abil­ family of polyunsaturated fatty acid metabolites, arachidonic acid [91].
ity to modulate vascular tone, leading to low-grade inflammation LXs cast an anti-inflammatory and pro-resolving effect on many cells and
associated with DCM [78]. Table 1 summarizes the involvement of are supposed to be associated with the suppressed cell injuries mediated
various glycation adducts in DCM pathogenesis. by inflammatory leukocytes [92,93], leading to reduced proin­
flammatory cytokines release and production. They first came into the
light of research when Serhan et al. isolated them from human leuko­
2.1. Receptor-mediated pathways of AGEs
cytes [90]. They are described as unsaturated molecules having an
anti-inflammatory activity against various metabolic diseases, including
The most dominant AGEs receptors in DCM pathogenesis are the
atherosclerosis and CVDs. Still, little is known about their mechanism of
receptors for AGEs (RAGE) with different isoforms, namely, soluble
action in glycation and glycation-induced DCM.
RAGE and endogenous secretory RAGE [84]. Soluble RAGE circulates in
the blood system, and its elevated levels are associated with the
increased risk of DCM in diabetes mellitus patients [85]. Thus, they can 3.1. Synthesis and mode of action of LXs
be considered an indicator of the destabilization of vulnerable plaque.
High RAGE levels are commonly seen in any inflammatory lesion, LXs are synthesized by interactions with lipoxygenase (LO) products
including those found in blood vessel walls [84,86], indicating the produced from arachidonic acid metabolism. They are generated as a
importance of the AGEs-RAGE axis as a therapeutic target for DCM response to the cell-cell interactions at the site of injury and host re­
amelioration. AGEs and their receptors have inflammatory effects, sponses to inflammation or infection [94]. This review hypothesizes that
including endothelial dysfunction, smooth muscle cell proliferation and glycation-induced inflammation can act as the stimulator for releasing
migration, macrophage activation, atrial stiffness, mitochondrial and arachidonic acid leading to LXs synthesis (Fig. 5). Arachidonic acid
cytoskeletal dysfunction, enhanced ROS formation, impaired calcium further synthesizes LXs through two pathways by three major LO, viz.
homeostasis, atherosclerosis, enhanced uptake of ox-LDL, accelerated 5-LO, 15-LO, and 12-LO, (i) the first route involves a series of actions of
foam cell formation, contributing to abnormal lipid metabolism and LO in monocytes and neutrophils, where arachidonic acid is converted
regulating DCM pathogenesis [87]. Upregulated RAGE is often associ­ to 15(s)- hydroxyeicosatetraenoic acid, one of the stereoisomers of 15-
ated with granular calcification, which exposes the lipid core of hydroxyeicosatetraenoic acid, which further produces LXs, (ii) the sec­
atherosclerotic plaques and favors its rupture [88], thus, initiating in­ ond route takes place in platelets where leukotriene A4 acts with 12-LO
flammatory responses through lipid deposit-induced and forms LXs (iii) Aspirin leads an additional route of LXs synthesis
metalloproteinases. with the help of which their epimers are formed. Aspirin acetylates
cyclooxygenase-2 and forms 15(R)- hydroxyeicosatetraenoic acid,
3. Lipoxins in DCM another stereoisomer of 15- hydroxyeicosatetraenoic acid [90]. In the
end, LXs and their epimers are generated through 5(s), 6(s), 15(s)-
The immune system releases anti-inflammatory molecules and cy­ epoxytetraeneoic acid intermediate [87]. It is to be noted that various
tokines in response to inflammation [89]. Lipoxins (LXs) are important pro- and anti-inflammatory molecules are generated during
anti-inflammatory lipid mediators secreted by various immune cells, the production of LXs, giving therapeutic directions to arachidonic acid

6
M. Thakur and R.S. Tupe Prostaglandins and Other Lipid Mediators 164 (2023) 106698

Table 2 hydroxyeicosatetraenoic acid formed by the action of 5-, 12- and 15-LO
Different types of LXs and their biological roles in CVDs. has been suggested to have hypertrophic effects on human ventricular
Sr. Role of LXs in CVDs Study Model References cardiomyocytes; the hypertrophic activity was shown to be MAPK- and
No. NF-κB dependent [98]. Arachidonic acid metabolism via the cyclo­
LXA4 & LXB4
oxygenase pathway generates a family of prostanoid mediators having
1. Resolve atherosclerotic Apolipoprotein E deficient [92, pro-inflammatory activities in DCM pathogenesis. Thromboxanes are
plaques and increase mice 111–115]
plaque stability
such prostanoid mediators, which are potential vasoconstrictors and
2. Recruit non-inflammatory Primary Human Umbilical [111,116, platelet aggregates. They hamper the metabolism of extracellular matrix
monocytes to Vein Endothelial Cells 117] proteins and promote fibrosis and scarring. They also enhance the
inflammatory sites, functioning of lymphocytes and macrophages, directing immunomod­
suppress neutrophil
ulatory effects in DCM [99]. However, the actions of cycloxygenase
chemotaxis, adhesion &
transmigration, diminish metabolites are complex, and certain prostanoids (prostaglandins,
P-selectin-mediated prostacyclin) inhibit immune and inflammatory responses associated
neutrophil-endothelial with DCM. Therefore, it is hypothesized that non-steroidal anti-in­
interactions, flammatory drugs are supposed to inhibit inflammatory prostanoids like
3. Reduce oxLDL cholesterol Patients who underwent [111,118,
and foam cell formation coronary angiography for 119]
thromboxane.
chest pain
LXA4, 15-epi-LX4 3.1.2. Anti-inflammatory molecules in LXs biosynthesis
4. Reduce reperfusion- Human leukocytes [111], On the other side, a metabolite from the arachidonic acid pathway
associated vascular [120]
formed from cytochrome P450 monooxygenases and epoxygenases is
inflammation
5. Reduce pro-inflammatory TNF-α-initiated neutrophil [111,116, epoxyeicosatrienoic acid, which has the potential to restore cardiac
cytokine secretion and (polymorphonuclear 121] remodeling and pro-inflammatory cytokine secretion [100]. A review
ROS production leukocyte) responses in vitro from Romashko and colleagues has postulated that epoxyeicosatrienoic
and in vivo using acid promotes vasodilation, anti-inflammatory, and anti-apoptotic
metabolically stable LX
analogs
properties by reducing blood pressure, adiposity, and myocardial
6. Inhibit angiogenic Primary Human Umbilical [111,117, fibrosis [101]. In addition, it can also limit atherosclerosis and DCM,
pathways, including Vein Endothelial Cells 122–124] lowering heart failure incidences [102]. Prostaglandin E receptor sub­
endothelial cells type 4 acts via FOXO1/CD36 signaling and mediates cardiac uptake of
proliferation and
fatty acids and ATP generation, thus, protecting against DCM [103].
activation
LXA4 Evidence shows that disturbed prostaglandin D2 signaling enhances
7. Decrease monocyte IL-1β 15 patients with severe [111,125] atherosclerosis, hypertension, myocardial ischemia, and DCM patho­
& calcium efflux preeclampsia and 20 normal genesis [104]. Cycloxygenase-2 mediated increased prostaglandin E
pregnant women (control receptor subtype two syntheses in cardiac inflammation milieu suggests
group)
8. Modulate K+ channel and Rat cardiomyocytes [111,125,
the cardioprotective role of prostaglandin E2 in DCM [105–107]. Pros­
activate p38 MAPK 126] tacyclin is yet another inflammation mediator produced during arach­
pathways and nuclear idonic acid metabolism through cycloxygenase-2. A study showed a
translocation of NRF2; critical role of prostacyclin in cardiac inflammation by increased
induces the release of the
atherosclerosis and thrombosis with enhanced proliferative response to
protective heme
oxygenase-1 carotid vascular injury on knockout of prostacyclin receptor in mice
9. Decrease proliferation, Human and mouse B cells [111,127] [108].
IgG & IgM production of
human memory B-cells, 3.2. LXs in DCM and glycation
inhibiting NF-kB p65
nuclear translocation
10. A vasodilator promoting Wistar rats [128] Increased extracellular cholesterol levels cause the shifting of 15-lip­
the endothelial release of oxygenase to LDL oxidation and induce inflammation while negatively
prostacyclin and nitric regulating blood pressure. LXs and other pro-resolvins in human serum
oxide while inhibiting
can reverse this shift back to cholesterol [109]. LXs are shown to have
ROS production
anti-inflammatory actions, such as inhibited foam cell formation,
Abbreviations: oxLDL (oxidized low-density lipoprotein), TNFα (Tumor necrosis ox-LDL-induced inflammation, and apoptotic signaling in macrophages.
factor alpha), ROS (reactive oxygen species), IL-1β (interleukin − 1β), MAPK Table 2 enlists some such major bioactions of LXs in CVDs. Literature
(mitogen-activated protein kinase), Nrf2 (nuclear erythroid 2-related factor 2),
supports the hypothesis that LXs have therapeutic potential in
IgG (immunoglobulin G), IgM (immunoglobulin M), NF-kB (nuclear factor
diabetes-associated vascular complications [110].
Kappa-light-chain-enhancer of activated B cells)
LXs help combat atherogenesis by reducing fibroblast maturation,
collagen production, and myofibroblast differentiation; hence, they can
metabolic signaling.
be the target molecules as their actions may have therapeutic potential
in the treatment of diabetes-induced atherosclerosis, suggested the in-
3.1.1. Pro-inflammatory molecules in LXs biosynthesis
vivo study performed by Brennan and group [110]. This review points
LO activities can be associated with DCM pathogenesis as they
out that LXA4 has been extensively studied in various metabolic disor­
oxidize LDLs and triggers atherosclerosis [95]. 5-LO predominantly
ders such as atherosclerosis and CVDs. At the same time, LXA4 and
forms 5- hydroxyeicosatetraenoic acid and leukotrienes, which are
respective epimers are not well explored in glycation-associated meta­
involved in leukocyte recruitment, inflammatory cytokine production,
bolic diseases. This indicates the research potential of LXs and their
hampered vascular permeability, and endothelial dysfunction [95,96].
epimers in glycation-induced DCM.
Synthesis of leukotrienes requires a 5-LO activating protein, which as­
sembles other enzymes needed for leukotriene synthesis in leukocytes
3.3. LX-ALX/FPR2 receptor-mediated action
[97]. In this sense, inhibitors for 5-LO activating protein can have a
strong therapeutic potential in alleviating DCM. 15-
Lipid mediators LXs act via G-protein coupled receptors, namely

7
M. Thakur and R.S. Tupe Prostaglandins and Other Lipid Mediators 164 (2023) 106698

5-lipoxygenase, which is also involved in the PPARγ accelerating


CD36 expression, is a precursor of LXs generation [144]. PPARγ and
5-lipoxygenase clear apoptotic neutrophils and limit inflammatory tis­
sue injury [145]. LXs upregulate the expression of CD36 independent of
PPARγ [144]. It also can upregulate PPARγ expression. All three entities,
glycation, LXs, and SREBPs, are well established with their respective
positive or negative effects on lipid metabolism, either leading to or
inhibiting DCM pathogenesis. However, glycation-induced inflamma­
tion can trigger LXs biosynthesis by immune cells; the LXs are not
studied in the glycation pathway to date, which can be a significant
therapeutic breakthrough against deteriorated lipid metabolism in
DCM. LXs and SREBP signaling are associated with lipid metabolism
and, when studied together, can be a vital tool for treating disrupted
immunometabolism in many metabolic disorders, including DCM.
Perhaps the literature lacks the interaction of LXs and SREBP signaling;
they both regulate PPARγ receptors in one way or another, showing the
potential of LXs in SREBP signaling in glycation-induced disrupted lipid
metabolism in DCM through these nuclear receptors (Fig. 6).
Fig. 6. Scheme illustrating the connection of glycation and LXs in SREBP sig­ This review has already described the mechanism of action of gly­
nalling. Glycation, LXs, and PPARγ expression regulate SREBP-1c signaling in cation and LXs in DCM. To understand their roles in SREBP signaling, it
DCM through their specific mechanism of action. Regulation of SREBP-1c is essential to know how the signaling works in cardiac energy
expression by keeping a check on glycation-LXs crosstalk in glycation-induced
metabolism.
DCM can be a novel therapeutic approach. Abbreviations: AGEs, advanced
glycation end products; SREBP-1c, sterol regulatory element binding proteins-
1c, PPARγ, peroxisome proliferator-activated receptor gamma.
5. SREBP signaling in lipid metabolism

Transcription factors SREBPs are extensively involved in regulating


ALX/FPR2 [129,130]. It limits leukocyte trafficking and activation,
gene expressions in synthesizing lipids and function in global biological
stimulating efferocytosis, granulocyte apoptosis, and leukocyte egress
signaling pathways related to various physiological and pathophysio­
[130]. It also suppresses calcium-sensing kinase and
logical processes [146]. They are of three types with three different
calcium-calmodulin-dependent protein kinase and inhibits p38
established functions in lipid metabolism: (i) SREBP1a in global lipid
mitogen-activated protein kinase phosphorylation [131]. Failed
synthesis, (ii) SREBP1c in fatty acid synthesis and energy storage; (iii)
inflammation resolution in terms of increased pro-inflammatory ago­
SREBP2 involved in cholesterol regulation [147]. Pathways related to
nists and decreased levels of pro-resolving agonists in atherosclerosis
cholesterol synthesis and uptake are controlled via end-product feed­
was observed by a disrupted balance between ALX/FPR2 ligands [132].
back inhibition of 3-hydroxy-3- methylglutaryl CoA reductase at the
Hence, restoration of the pro-resolving ALX/FPR2 signaling with the
transcriptional level. 3-hydroxy-3- methylglutaryl CoA reductase is a
help of lipid or peptide agonists can be a novel therapeutic approach in
rate-limiting enzyme of cholesterol synthesis through LDLR, facilitating
atherosclerosis. Accelerating ALX/FPR2 signaling is essential in limiting
endocytosis of cholesterol-rich LDLs [148,149]. These genes have a
neutrophil-mediated ischemia, reperfusion injury, smooth muscle
sterol regulatory element (SRE) on their promoter region, regulating
cell-mediated intimal hyperplasia, and restenosis after different revas­
sterol-dependent expression regulation [150]. In their native form,
cularization procedures [133]. Also, there is an urgent need to study
SREBPs are manufactured and stored at the endoplasmic reticulum
ALX/FPR2 signaling in DCM and glycation-induced complications.
membrane. The active N-terminal of SREBPs needs to undergo proteo­
lytic cleavage to facilitate their transcriptional activities and nuclear
4. LXs and glycation in sterol regulating element binding
translocation. Nuclear cleavage depends on cellular cholesterol avail­
protein (SREBP) signaling
ability [146,150,151]. When cholesterol is low, SCAP binds to and es­
corts SREBP in COP II vesicles to the Golgi complex, where the SREBPs
Regulation of cardiac lipid metabolism is dominated by transcrip­
are cleaved by a serine protease, site 1, and zinc metalloprotease, site 2.
tional factors SREBPs [134,135]. A statin group of SREBP is responsible
Once the cholesterol level is restored, Insig, an endoplasmic reticulum
for blocking lipid generation by inhibiting the 3-hydroxy-3- methyl­
membrane protein regulator, traps and brings the SREBP/SCAP complex
glutaryl CoA pathway [136]. AGEs produced in glycation reactions can
to the endoplasmic reticulum. It controls SREBP cleavage in the Golgi,
increase lipid generation through activated SREBP cleavage-activating
thus, downregulating the LDL receptor biosynthesis [152]. They are
protein (SCAP)/SREBP signaling; SCAP is a functional cholesterol
strongly implicated in various pathological consequences, including
sensor [137].
inflammation, autophagy and apoptosis, and endoplasmic reticulum
Additionally, glycation has been shown to have an inversely pro­
stress, thereby contributing to obesity, dyslipidemia, diabetes, fatty liver
portional expression with peroxisome proliferator-activated receptors
disease, chronic kidney disease, neurodegenerative diseases, and can­
(PPARγ), nuclear receptors with active participation in lipid and glucose
cers [146]. As atherosclerosis, dyslipidemia, and cardiac metabolism are
homeostasis as transcriptional factors [138]. Upregulated PPARγ ex­
closely related and intertwined, it is necessary to study the SREBP
pressions showed AGEs-damaged chondrocyte repair in a study carried
signaling in glycation-induced DCM.
out in 2015 [139]. In another study, AGEs enhance the IL-1 and TNF-α
expressions and inhibit PPARγ in a time and dose-dependent manner
5.1. SREBPs and cardiac metabolism: Clinical prospective
[140]. Short-chain fatty acids and their derivatives help activate PPARγ
and create a lipid signaling network between the nucleus and cell sur­
Pathogenesis of DCM is multifactorial and involves cardiac immu­
face. Less PPARγ hampers adipose tissue’s ability to store fat, thereby
nometabolism [153]. During DCM, augmented cardiac lipid levels in­
increasing lipotoxicity and causing DCM [141,142]. PPARγ also en­
crease mitochondrial and endoplasmic reticulum stress [154]. The
hances the expression of CD36 and resolves inflammation [17].
functions of SREBPs are closely associated with the endoplasmic retic­
Furthermore, PPARγ positively regulates the expression of SREBP-1c; it
ulum. Chronic endoplasmic reticulum stress dysregulates SREBPs
enhances lipid oxidation and synthesis [143].
expression and enhances intracellular lipid levels [155]. A study

8
M. Thakur and R.S. Tupe Prostaglandins and Other Lipid Mediators 164 (2023) 106698

Table 3
Pharmacological interventions used in the treatment of CVDs.
Drug Class Associated drugs Mechanism of action Physiological actions Adverse effects References

Calcium Channel Drug classification


Blockers or calcium Dihydropyridine derivative: High affinity for Ca+ channels in The strong hypotensive effect, Excessive hypotension, edema,
agonists[166–168] Nifedipine, Nicardipine, vascular smooth muscle cells, strong coronary vasodilation, tachycardia reflex
Nilvadipine, low membrane potential, strong peripheral vasodilation,
Phenylalkylamine derivative: increased activity in vessels than in high cardiac contraction,
Verapamil cardiac muscle cells. increased heart rate,
Benzodiazepine derivative: moderate atrioventricular
Diltiazem conduction
Diarylaminopropylamine Act on the impulse conduction system Strong coronary vasodilation, Vasoconstriction, hemostasis,
derivative: with high membrane potential and Ca+ strong peripheral vasodilation, thrombosis, ischemia, seizures,
Bepridil channels in ventricular muscle, decreased cardiac contraction, hyperthermia, and rhabdomyolysis
adverse inotropic, chronotropic, and moderate heart rate, Impaired muscle coordination
dromotropic effects. decreased atrioventricular
conduction
The intermediate effect between the Extreme coronary vasodilation,
dihydropyridine and phenylalkylamine weak peripheral vasodilation,
derivatives, decreased cardiac contraction,
act on the myocardium and impulse decreased heart rate,
conduction system, particularly in decreased atrioventricular
atrioventricular node conduction conduction
A long-acting, non-selective, significant Significant coronary Urticaria, gastrointestinal
anti-anginal, antihypertensive, vasodilation, modest peripheral disturbances
selective anti-arrhythmia activity acts effects
as a calmodulin antagonist.
Sodium-glucose Drug classification
Cotransporter SGLT1[169] High-affinity, low-capacity glucose The putative intestinal target for Genital infections, diabetic
inhibitors Sotagliflozin, Zynquista transporter is predominantly found in antidiabetic therapy; ketoacidosis
enterocytes of the small intestine, physiological regulation is
where it transports glucose and incompletely understood,
galactose from the gut lumen across the regulated by dietary composition
gut wall over days
SGLT2[170] Reduces reabsorption of filtered glucose Effective in improving glycaemic urinary and genital tract infections in
Canagliflozin, Ertugliflozin, from the tubular lumen and lowers the control, with added benefits of a small proportion of users
Empagliflozin RTG some reductions in BP and
weight
GLP-1 receptor Drug classification
agonists[171] Short-acting drugs[172]: Decreases glucagon release during The pancreas produces insulin Hypoglycemia in combination with
exenatide, lixisenatide, hyperglycemic periods, which reduces more efficiently; insulin or sulphonylurea
semaglutide hepatic glucose output and decreases helps control blood sugar levels. Risk of hypoglycemia
insulin demand. Delayed gastric
emptying decreases the rate at which
glucose arrives in the bloodstream.
Long-acting drugs[172]: Delayed atherosclerosis progression by Improved gastrointestinal
Liraglutide[173] regulating inflammation through LDL tolerability, moderate plasma
Semaglutide[173,174] receptor-deficient and apolipoprotein drug concentration fluctuations
Exenatide[175] E-deficient mice
Decreases systolic BP, body weight, and
blood glucose levels.
Improves insulin sensitivity and
intramyocellular lipid. Activates AMP-
activated protein kinase signaling
pathway without BW reduction
PCSK9 inhibitors Alirocuma,Evolocumab, Human IgG1 monoclonal antibody that Increase the clearance of LDL Injection-site reactions, influenza-like
Bococizumab[176] binds and inhibits PCSK9 cholesterol from the circulation illness, myalgia
Anti-Advanced Aminoguanidine[177] Binds to sugars, thus preventing them Increased vascular elasticity, Gastrointestinal disturbance,
Glycation End from binding to the lysine group of improved left-ventricular- abnormalities in liver function tests,
Products proteins. Prevent oxidative arterial coupling, and decreased flu-like symptoms, development of
Compounds modification of LDL and inhibit the vascular permeability in diabetic antinuclear antibody, pernicious-like
formation of atherosclerotic plaques rats. anemia, rare vasculitis
Benfotiamine[178] Post-Amadori inhibitor by trapping Activates transketolase, an Angioedema, low blood pressure
dicarbonyl intermediates, inhibition of enzyme that helps apparent
NADPH oxidase AGEs,
Inhibit significant pathways
implicated in oxidative stress and
vascular dysfunction
ACEIs (ex. captopril, enalapril, Pre & post-Amadori inhibitor, It improves heart failure, Hypotension, disturbed kidney
lisinopril, ramipril) transition metal ion chelator decreases afterload, preload, and functions
systolic wall stress, and increases
cardiac output without
increasing heart rate
Statins Stimulates RAGE shedding and lowers Stimulate the production of Adverse effects on cognition
(Atorvastatin, fluvastatin, cholesterol in LDL, non-HDL, and RLP soluble RAGE
lovastatin, pravastatin, and in the fasting and fed states
simvastatin)[179,180]
(continued on next page)

9
M. Thakur and R.S. Tupe Prostaglandins and Other Lipid Mediators 164 (2023) 106698

Table 3 (continued )
Drug Class Associated drugs Mechanism of action Physiological actions Adverse effects References

TZDs/glitazones[181] Reduces RAGE expression Improved insulin sensitivity fluid retention, edema, and
congestive heart failure[182]
ALT-946b[183] Post-Amadori inhibitor by trapping Indicators of obesity, Abnormal ALT on a long-term
dicarbonyl intermediates, NOS cardiovascular disease, non- outcome such as end-stage liver
inhibitor alcoholic fatty liver disease disease or premature mortality
Tenilsetamb[184] Post-Amadori inhibitor by trapping Inhibits glucose-induced Adverse effects on patients with
dicarbonyl intermediates, NOS lysozyme polymerization severe liver and kidney diseases and
inhibitor, transition metal ion chelator alcohol consumption.
PPARγ agonists ARBs[185] Prevent the hypertensive effects of Vasoconstriction, simulation and Upper respiratory infection
[(ex. Olmesartan, Losartan, angiotensin II by selective blockade of synthesis of aldosterone and
Valsartan, Irbesartan, the angiotensin II type 1 receptor ADH, cardiac stimulation, and
Eprosartan)] renal reabsorption of sodium
TZDs/glitazones[186,187] Antidiabetic effects through a Improve metabolic control in Fluid retention, which can result in,
mechanism that involves activation of patients with type 2 diabetes or exacerbate, edema and congestive
the gamma isoform of the PPARγ through the improvement of heart failure
insulin sensitivity
Pharmacological Aspirin[188–190] Affects the biosynthesis of lipid Reduces platelet aggregation, Gastrointestinal bleeding, gastric
agents/drugs for mediators involved in pro- manages thrombotic disorders, irritation,
LXs inflammatory pathways. Inhibits cyclooxygenase and
prostaglandin formation
Midazolam[191,192] Anticonvulsant activity is related to the Anticonvulsant, anxiolytic, sleep- Intravenous administration causes
excess GABA action on motor circuits in inducing, muscle relaxant, and respiratory and cardiovascular
the brain. It also acts on glycine "sedative" effects depression.
receptors and produces a muscle-
relaxing effect.
Barbiturate[193,194] Potentiate GABAergic inhibition by Anticonvulsant, sedative, Reduces neuronal activity, depress
increasing the lifetime of Cl‾ channel hypnotic cerebral metabolism
opening induced by GABA
BML-111 (5(S)− 6(R)− 7- Equivalent to LXA4 in the inhibition of Upregulates the mRNA and Negligible or no side effects, however,
trihydroxy-heptanoic acid inflammatory processes protein expressions of PPARα, transgenic mice unable to produce LX
methyl ester)[195,196] inhibits neutrophil recruitment, controlled the M. tuberculosis bacteria
reduces the inflammatory better with reduced mortality in
response comparison to wild-type
SREBP-1c inhibitors Betulin[197,198] Anti-inflammatory action through Suppresses cellular glucose Halitosis, orofacial pain, reduced
glucocorticoid receptor-mediated metabolism salivary flow, oral lesions associated
pathway with CVDs, diabetes mellitus
Clenbuterol[199] Upregulates CREB1 expression. Decreases body fat Can induce cardiac hypertrophy
PF-429242 (Aminopyrrolidine Site 1 protease inhibitor Inhibits cholesterol and fatty acid Hypolipidemia
amide)[200,201] synthesis

Abbreviations: GLP-1, glucagon-like peptide-1; IgG1, immunoglobulin G1; BP, blood pressure; PCSK9, proprotein convertase subtilisin/kexin type 9; LDL, low-density
lipoprotein; DM, diabetes mellitus; AGE, advanced glycation end products; ACEIs, Angiotensin-converting enzyme inhibitors; ARBs, Angiotensin receptor blockers;
PPARγ, Peroxisome proliferator-activated receptor gamma; RAGE, the receptor for AGEs; HDL, high-density lipoprotein; RLP, remnant lipoprotein; TZDs, Thiazoli­
dinediones; NOS, nitric oxide synthase; ATP, adenosine triphosphate; GABA, Gamma Amino Butyric Acid; LPS, lipopolysaccharides; BMDM, Bone marrow-derived
macrophages; MSU, monosodium urate; PBMC, Peripheral Blood Mononuclear Cells; CREBP-1, Cyclic adenosine monophosphate response element binding protein
1; NADH; LXA4, lipoxin A4; mRNA, messenger ribonucleic acid, PPARα, Peroxisome proliferator-activated receptor alpha

performed on Trypanosoma cruzi, an intracellular parasite causing inflammation by excess ROS formation, inflammatory cytokines release,
devastating chronic Chagas cardiomyopathy, conveyed that inhibition leukocyte activation, and their accumulation on the extracellular ma­
of SREBP activation improves cardiac mitochondrial stress and sup­ trix. Together, these help glycations promote lipotoxicity, and systolic
presses cardiac endoplasmic reticulum stress, and gradually suppresses and diastolic dysfunctions in the heart through cardiac hypertrophy and
inflammation [156]. A study of genome scanning has associated the fibrosis [160], promoting DCM (Fig. 2). Glycation upregulates lipid
17p11 region, consisting of the human SREBP-1 gene locus, with plasma biogenesis by inhibiting SCAP/SREBP interaction, thereby inducing
leptin concentrations and type 2 diabetes [157]. The glycoprotein78 cardiac output pressure. It becomes enlarged due to persistent hyper­
knockout (L-gp782/2) mice are shielded from diet-/ age-induced tension and undergoes pathological hypertrophy causing contractile
obesity, improving hyperlipidemia and insulin resistance. In 2012, Liu dysfunctions in cardiac muscles.
and co-researchers generated liver-specific L-gp782/2 mice. These mice A study showed that metabolites generated in the arachidonic acid
have increased Insig-1 and Insig-2, causing SREBP inhibition [158]. pathway could mediate cholesterol homeostasis—aspirin-induced
Thus, gp782/2 can be inhibited to provide therapeutic direction to biosynthesis of LXs-epimers reverse cholesterol transport, which is the
alleviate metabolic diseases by inhibiting SREBP signaling. primary function of high-density lipoproteins. LXs mimetics also lower
plasma LDL-cholesterol, maintaining cholesterol-derived cardiac
5.2. Glycation, LXs, and SREBP signaling in contractile dysfunctions inflammation [161]. Reverse cholesterol transport moves cholesterol
out of foam and atherosclerotic plaques to circulation, from where they
Diabetes-induced systolic and diastolic dysfunctions affect heart are transported to the liver and excreted in the feces [162]. This in­
muscles, enhance muscle fibrosis, enlargement, and swelling, and design dicates that LXs and mimetics relax the contractile dysfunctions in DCM
an atherosclerotic condition and cardiac hypertrophy leading to DCM by washing off LDLs, cholesterol, inflammatory leukocytes, and platelets
[159]. LDL, free fatty acids, cholesterol crystals, and calcium phosphate from the injured site (Table 2). It is hypothesized that LXs interaction in
crystals are influx in the cells at a higher rate than their uptake in the SREBP signaling through PPARγ receptors may help deal with cardiac
glycation milieu. These entities either form AGEs and activate inflam­ muscle systolic and diastolic dysfunctions. Glycation may enhance
matory signaling pathways NF-κB through RAGE interactions or accu­ contractile dysfunctions, but if, as per the hypothesis of this review,
mulate on the target cells’ extracellular matrix, thereby inducing glycation triggers biosynthesis of LXs, it may also help in the contractile

10
M. Thakur and R.S. Tupe Prostaglandins and Other Lipid Mediators 164 (2023) 106698

dysfunctions, provided this pathway is studied systematically. [2] D.M. Greenfield, J.A. Snowden, Cardiovascular Diseases and Metabolic
Syndrome, The EBMT Handbook, Springer International Publishing, Cham, 2019,
pp. 415–420, https://doi.org/10.1007/978-3-030-02278-5_55.
6. Therapeutics in DCM [3] R.H. Ritchie, E.D. Abel, Basic mechanisms of diabetic heart disease, Circ. Res 126
(2020) 1501–1525, https://doi.org/10.1161/CIRCRESAHA.120.315913.
[4] G.S. Gulsin, L. Athithan, G.P. McCann, Diabetic cardiomyopathy: prevalence,
Although no particular pharmacological therapy is designed for determinants and potential treatments, 204201881983486, Ther. Adv.
DCM, regulating chronic hyperglycemia, hypertension, and hyperlipid­ Endocrinol. Metab. 10 (2019), https://doi.org/10.1177/2042018819834869.
emia can help alleviate DCM. Most anti-diabetic drugs are proven [5] T.R. Einarson, A. Acs, C. Ludwig, U.H. Panton, Prevalence of cardiovascular
disease in type 2 diabetes: a systematic literature review of scientific evidence
effective in reducing cardiovascular mortality and morbidity associated from across the world in 2007–2017, Cardiovasc Diabetol. 17 (2018) 83, https://
with DCM [163]. Most cardiovascular events are treated following the doi.org/10.1186/s12933-018-0728-6.
associated clinical phenotypes established in cardiology guidelines [6] M.S. Shah, M. Brownlee, Molecular and cellular mechanisms of cardiovascular
disorders in diabetes, Circ. Res 118 (2016) 1808–1829, https://doi.org/10.1161/
[164,165] (Table 3). This review shows a link between glycation, LXs,
CIRCRESAHA.116.306923.
and SREBP-1c signaling via PPARγ nuclear receptors. Current treatment [7] S. Yamagishi, N. Nakamura, T. Matsui, Glycation and cardiovascular disease in
strategies have multiple adverse factors associated; further studies can diabetes: a perspective on the concept of metabolic memory, J. Diabetes 9 (2017)
target lipid metabolism therapies for DCM therapeutics in terms of 141–148, https://doi.org/10.1111/1753-0407.12475.
[8] S. Tsalamandris, A.S. Antonopoulos, E. Oikonomou, G.-A. Papamikroulis,
anti-diabetic drugs, AGEs inhibitors, LXs mimics, SREBP-1c inhibitors, G. Vogiatzi, S. Papaioannou, S. Deftereos, D. Tousoulis, The role of inflammation
and PPARγ agonists. in diabetes: current concepts and future perspectives, Eur. Cardiol. Rev. 14
(2019) 50–59, https://doi.org/10.15420/ecr.2018.33.1.
[9] T. Miyazawa, K. Nakagawa, S. Shimasaki, R. Nagai, Lipid glycation and protein
7. Conclusion glycation in diabetes and atherosclerosis, Amino Acids 42 (2012) 1163–1170,
https://doi.org/10.1007/s00726-010-0772-3.
[10] R.V. Giglio, A.P. Stoian, M. Haluzik, K. Pafili, A.M. Patti, A.A. Rizvi, M. Ciaccio,
In DCM, increased oxidative stress facilitates LDL oxidation and in­
N. Papanas, M. Rizzo, Novel molecular markers of cardiovascular disease risk in
duces lipotoxicity. Disrupted fatty acid metabolism in DCM is associated type 2 diabetes mellitus, Biochim. Et. Biophys. Acta (BBA) - Mol. Basis Dis. 1867
with high SREBP-1c expression, positively regulated by PPARγ nuclear (2021), 166148, https://doi.org/10.1016/j.bbadis.2021.166148.
receptors. Glycation increases lipotoxicity, while LXs upregulate PPARγ [11] G.D. Lopaschuk, Q.G. Karwi, R. Tian, A.R. Wende, E.D. Abel, Cardiac energy
metabolism in heart failure, Circ. Res 128 (2021) 1487–1513, https://doi.org/
expressions and resolve inflammation. Glycation and LXs have been 10.1161/CIRCRESAHA.121.318241.
studied independently in various metabolic disorders. Furthermore, [12] T. Doenst, T.D. Nguyen, E.D. Abel, Cardiac metabolism in heart failure, Circ. Res
SREBP signaling, especially SREBP-1c, is distinct in disrupting lipid 113 (2013) 709–724, https://doi.org/10.1161/CIRCRESAHA.113.300376.
[13] C. Bartolacci, C. Andreani, Y. El-Gammal, P.P. Scaglioni, Lipid metabolism
metabolism during DCM. This review suggests that the interplay of LXs regulates oxidative stress and ferroptosis in ras-driven cancers: a perspective on
can be studied in glycation and SRBP-1c signaling pathways in cancer progression and therapy, Front Mol. Biosci. 8 (2021), https://doi.org/
glycation-induced DCM (Fig. 6). 10.3389/fmolb.2021.706650.
[14] A.K. Hauck, D.A. Bernlohr, Oxidative stress and lipotoxicity, J. Lipid Res 57
Different treatment comorbidities are available for DCM, as (2016) 1976–1986, https://doi.org/10.1194/jlr.R066597.
described above (Table 3), yet, glycation-induced DCM and associated [15] J. Ritterhoff, R. Tian, Metabolism in cardiomyopathy: every substrate matters,
lipotoxicity lack a specific and clear treatment line. Applying LXs in Cardiovasc Res 113 (2017) 411–421, https://doi.org/10.1093/cvr/cvx017.
[16] I. Ballesteros, M.I. Cuartero, J.M. Pradillo, J. de la Parra, A. Perez-Ruiz, A. Corbi,
glycation and SREBP-1c signaling can lead to a novel therapeutic M. Ricote, J.A. Hamilton, M. Sobrado, J. Vivancos, F. Nombela, I. Lizasoain, M.
approach to treating glycation-induced DCM and lipotoxicity. As PPARγ A. Moro, Rosiglitazone-induced CD36 up-regulation resolves inflammation by
agonists have various adverse effects, treatment strategies can focus on PPAR and 5-LO-dependent pathways, J. Leukoc. Biol. 95 (2014) 587–598,
https://doi.org/10.1189/jlb.0613326.
regulating its expression through AGEs/RAGE-LXs crosstalk in SREBP-1c
[17] M. Kawai, C.J. Rosen, PPARγ: a circadian transcription factor in adipogenesis and
signaling. In summary, inhibition of the AGEs/RAGE axis, enhancement osteogenesis, Nat. Rev. Endocrinol. 6 (2010) 629–636, https://doi.org/10.1038/
in LXs and PPARγ activity, and adequate SREBP-1c signaling can nrendo.2010.155.
contribute to developing novel clinical therapies for diminishing [18] L.H. Opie, J. Knuuti, The adrenergic-fatty acid load in heart failure, J. Am. Coll.
Cardiol. 54 (2009) 1637–1646, https://doi.org/10.1016/j.jacc.2009.07.024.
glycation-induced pathogenesis DCM. [19] N. Fillmore, J. Mori, G.D. Lopaschuk, Mitochondrial fatty acid oxidation
alterations in heart failure, ischaemic heart disease and diabetic cardiomyopathy,
Data Availability Br. J. Pharm. 171 (2014) 2080–2090, https://doi.org/10.1111/bph.12475.
[20] J. Glatz, J. Luiken, M. Nabben, Metabolic modulation to treat cardiac diseases:
role for membrane substrate transporters, J. Cardiol. Cardiovasc Sci. 4 (2020)
No data was used for the research described in the article. 24–29, https://doi.org/10.29245/2578-3025/2020/1.1187.
[21] J.F.C. Glatz, F. Wang, M. Nabben, J.J.F.P. Luiken, CD36 as a target for metabolic
modulation therapy in cardiac disease, Expert Opin. Ther. Targets 25 (2021)
Acknowledgments 393–400, https://doi.org/10.1080/14728222.2021.1941865.
[22] M. Tong, J. Sadoshima, Nuclear miR-320 controls lipotoxicity, Circ. Res 125
We thank Symbiosis International (Deemed University), Pune, for (2019) 1121–1123, https://doi.org/10.1161/CIRCRESAHA.119.316199.
[23] H. Li, J. Fan, Y. Zhao, X. Zhang, B. Dai, J. Zhan, Z. Yin, X. Nie, X.-D. Fu, C. Chen,
providing the Junior Research Fellowship to Ms. Muskan Thakur (Ref: D.W. Wang, Nuclear miR-320 mediates diabetes-induced cardiac dysfunction by
SIU/SCRI/2021/6.4/S-06/3921). activating transcription of fatty acid metabolic genes to cause lipotoxicity in the
heart, Circ. Res 125 (2019) 1106–1120, https://doi.org/10.1161/
CIRCRESAHA.119.314898.
[24] N. Ghosh, S. Fenton, I. van Hout, G.T. Jones, S. Coffey, M.J.A. Williams,
Declaration of Competing Interest
R. Sugunesegran, D. Parry, P. Davis, D.O. Schwenke, A. Chatterjee, R. Katare,
Therapeutic knockdown of miR-320 improves deteriorated cardiac function in a
The authors declare no conflict of interest. pre-clinical model of non-ischemic diabetic heart disease, Mol. Ther. Nucleic
Acids 29 (2022) 330–342, https://doi.org/10.1016/j.omtn.2022.07.007.
[25] H. Li, D. Song, Q. Liu, L. Li, X. Sun, J. Guo, D. Li, P. Li, miR-351 promotes
atherosclerosis in diabetes by inhibiting the ITGB3/PIK3R1/Akt pathway and
Grant support
induces endothelial cell injury and lipid accumulation, Mol. Med. 28 (2022) 120,
https://doi.org/10.1186/s10020-022-00547-9.
This work received no specific grant from the public, commercial, or [26] G. Caputa, J.E. Schaffer, RNA regulation of lipotoxicity and metabolic stress,
Diabetes 65 (2016) 1816–1823, https://doi.org/10.2337/db16-0147.
not-for-profit funding agencies.
[27] S. Li, B. Culver, J. Ren, Benefit and risk of exercise on myocardial function in
diabetes, Pharm. Res 48 (2003) 127–132, https://doi.org/10.1016/S1043-6618
References (03)00099-9.
[28] J.B. Halter, N. Musi, F. McFarland Horne, J.P. Crandall, A. Goldberg, L. Harkless,
W.R. Hazzard, E.S. Huang, M.S. Kirkman, J. Plutzky, K.E. Schmader, S. Zieman, K.
[1] C.J. McAloon, F. Osman, P. Glennon, P.B. Lim, S.A. Hayat, Global epidemiology
P. High, Diabetes and cardiovascular disease in older adults: current status and
and incidence of cardiovascular disease, in: Cardiovasc Dis, Elsevier, 2016,
pp. 57–96, https://doi.org/10.1016/B978-0-12-803312-8.00004-5.

11
M. Thakur and R.S. Tupe Prostaglandins and Other Lipid Mediators 164 (2023) 106698

future directions, diabetes 63 (2014) 2578–2589, https://doi.org/10.2337/db14- [55] T. Kita, N. Kume, M. Yokode, K. Ishii, H. Arai, H. Horiuchi, H. Moriwaki,
0020. M. Minami, H. Kataoka, Y. Wakatsuki, Oxidized-LDL and atherosclerosis: role of
[29] A. Sepe, T. Tchkonia, T. Thomou, M. Zamboni, J.L. Kirkland, Aging and regional LOX-1, Ann. N. Y Acad. Sci. 902 (2006) 95–102, https://doi.org/10.1111/j.1749-
differences in fat cell progenitors - a mini-review, Gerontology 57 (2011) 66–75, 6632.2000.tb06304.x.
https://doi.org/10.1159/000279755. [56] C. Khatana, N.K. Saini, S. Chakrabarti, V. Saini, A. Sharma, R. v Saini, A.K. Saini,
[30] E.S. Ford, W.H. Giles, W.H. Dietz, Prevalence of the metabolic syndrome among Mechanistic insights into the oxidized low-density lipoprotein-induced
US adults: findings from the third National Health and Nutrition Examination atherosclerosis, Oxid. Med Cell Longev. 2020 (2020) 1–14, https://doi.org/
Survey, JAMA 287 (2002) 356–359, https://doi.org/10.1001/jama.287.3.356. 10.1155/2020/5245308.
[31] M.J. Hughes, H.M. McGettrick, E. Sapey, Shared mechanisms of multimorbidity [57] G. Schmitz, M. Grandl, The molecular mechanisms of HDL and associated
in COPD, atherosclerosis and type-2 diabetes: the neutrophil as a potential vesicular trafficking mechanisms to mediate cellular lipid homeostasis,
inflammatory target, Eur. Respir. Rev. 29 (2020), 190102, https://doi.org/ Arterioscler. Thromb. Vasc. Biol. 29 (2009) 1718–1722, https://doi.org/
10.1183/16000617.0102-2019. 10.1161/ATVBAHA.108.179507.
[32] J. Nikolich-Žugich, T cell aging, J. Exp. Med. 201 (2005) 837–840, https://doi. [58] T. Nishikawa, D. Edelstein, X.L. Du, S. Yamagishi, T. Matsumura, Y. Kaneda, M.
org/10.1084/jem.20050341. A. Yorek, D. Beebe, P.J. Oates, H.-P. Hammes, I. Giardino, M. Brownlee,
[33] S. Giubilato, G. Liuzzo, S. Brugaletta, D. Pitocco, F. Graziani, C. Smaldone, R. Normalizing mitochondrial superoxide production blocks three pathways of
A. Montone, V. Pazzano, D. Pedicino, L.M. Biasucci, G. Ghirlanda, F. Crea, hyperglycaemic damage, Nature 404 (2000) 787–790, https://doi.org/10.1038/
Expansion of CD4+CD28 null T-lymphocytes in diabetic patients: exploring new 35008121.
pathogenetic mechanisms of increased cardiovascular risk in diabetes mellitus, [59] S. Mittra, V.S. Bansal, P.K. Bhatnagar, From a glucocentric to a lipocentric
Eur. Heart J. 32 (2011) 1214–1226, https://doi.org/10.1093/eurheartj/ehq499. approach towards metabolic syndrome, Drug Disco Today 13 (2008) 211–218,
[34] E.Y.M. Lau, E.C. Carroll, L.A. Callender, G.A. Hood, V. Berryman, M. Pattrick, https://doi.org/10.1016/j.drudis.2008.01.006.
S. Finer, G.A. Hitman, G.L. Ackland, S.M. Henson, Type 2 diabetes is associated [60] G.L. King, The role of inflammatory cytokines in diabetes and its complications,
with the accumulation of senescent T cells, Clin. Exp. Immunol. 197 (2019) J. Periodo 79 (2008) 1527–1534, https://doi.org/10.1902/jop.2008.080246.
205–213, https://doi.org/10.1111/cei.13344. [61] C. Borza, D. Muntean, C. Dehelean, G. Savoiu, C. Serban, G. Simu, M. Andoni,
[35] Y. Lee, S.R. Kim, D.H. Han, H.T. Yu, Y.D. Han, J.H. Kim, S.H. Kim, C.J. Lee, B.- M. Butur, S. Drag, Oxidative stress and lipid peroxidation – a lipid metabolism
H. Min, D.-H. Kim, K.H. Kim, J.W. Cho, W.-W. Lee, E.-C. Shin, S. Park, Senescent T dysfunction, in: lipid metabolism, InTech, 2013, https://doi.org/10.5772/51627.
cells predict the development of hyperglycemia in humans, Diabetes 68 (2019) [62] L.A. Sena, N.S. Chandel, Physiological roles of mitochondrial reactive oxygen
156–162, https://doi.org/10.2337/db17-1218. species, Mol. Cell 48 (2012) 158–167, https://doi.org/10.1016/j.
[36] S. Boudina, Cardiac aging and insulin resistance: could insulin/insulin-like molcel.2012.09.025.
growth factor (IGF) signaling be used as a therapeutic target? Curr. Pharm. Des. [63] W.I. Sivitz, M.A. Yorek, Mitochondrial dysfunction in diabetes: from molecular
19 (2013) 5684–5694, https://doi.org/10.2174/1381612811319320004. mechanisms to functional significance and therapeutic opportunities, Antioxid.
[37] B.G. Childs, M. Gluscevic, D.J. Baker, R.-M. Laberge, D. Marquess, J. Dananberg, Redox Signal 12 (2010) 537–577, https://doi.org/10.1089/ars.2009.2531.
J.M. van Deursen, Senescent cells: an emerging target for diseases of ageing, Nat. [64] H. Zhong, J. Lu, L. Xia, M. Zhu, H. Yin, Formation of electrophilic oxidation
Rev. Drug Disco 16 (2017) 718–735, https://doi.org/10.1038/nrd.2017.116. products from mitochondrial cardiolipin in vitro and in vivo in the context of
[38] F. Olivieri, R. Recchioni, F. Marcheselli, A. Marie Abbatecola, G. Santini, apoptosis and atherosclerosis, Redox Biol. 2 (2014) 878–883, https://doi.org/
G. Borghetti, R. Antonicelli, A. Domenico Procopio, Cellular senescence in 10.1016/j.redox.2014.04.003.
cardiovascular diseases: potential age-related mechanisms and implications for [65] W. Liu, N.A. Porter, C. Schneider, A.R. Brash, H. Yin, Formation of 4-hydroxy­
treatment, Curr. Pharm. Des. 19 (2013) 1710–1719, https://doi.org/10.2174/ nonenal from cardiolipin oxidation: intramolecular peroxyl radical addition and
1381612811319090018. decomposition, Free Radic. Biol. Med 50 (2011) 166–178, https://doi.org/
[39] EUGenMed Cardiovascular Clinical Study Group. Vera Regitz-Zagrosek, Gender in 10.1016/j.freeradbiomed.2010.10.709.
cardiovascular diseases: impact on clinical manifestations, management, and [66] A. Ayala, M.F. Muñoz, S. Argüelles, Lipid peroxidation: production, metabolism,
outcomes, Eur. Heart J. 37 (2016) 24–34, https://doi.org/10.1093/eurheartj/ and signaling mechanisms of malondialdehyde and 4-Hydroxy-2-Nonenal, Oxid.
ehv598. Med Cell Longev. 2014 (2014) 1–31, https://doi.org/10.1155/2014/360438.
[40] M.Y. Farhat, M.C. Lavigne, P.W. Ramwell, The vascular protective effects of [67] G. Paradies, G. Petrosillo, V. Paradies, F.M. Ruggiero, Role of cardiolipin
estrogen, FASEB J. 10 (1996) 615–624, https://doi.org/10.1096/ peroxidation and Ca2+ in mitochondrial dysfunction and disease, Cell Calcium
fasebj.10.5.8621060. 45 (2009) 643–650, https://doi.org/10.1016/j.ceca.2009.03.012.
[41] S.S. Mahmood, D. Levy, R.S. Vasan, T.J. Wang, The framingham heart study and [68] D.L. Eizirik, A.K. Cardozo, M. Cnop, The role for endoplasmic reticulum stress in
the epidemiology of cardiovascular disease: a historical perspective, Lancet 383 diabetes mellitus, Endocr. Rev. 29 (2008) 42–61, https://doi.org/10.1210/
(2014) 999–1008, https://doi.org/10.1016/S0140-6736. er.2007-0015.
[42] M.T.R. Subbiah, Estrogen replacement therapy and cardioprotection: mechanisms [69] H.P. Harding, D. Ron, Endoplasmic reticulum stress and the development of
and controversies, Braz. J. Med. Biol. Res. 35 (2002) 271–276, https://doi.org/ diabetes, Diabetes 51 (2002) S455–S461, https://doi.org/10.2337/
10.1590/S0100-879X2002000300001. diabetes.51.2007.S455.
[43] E.D. Staren, S. Omer, Hormone replacement therapy in postmenopausal women, [70] J. Xu, Q. Zhou, W. Xu, L. Cai, Endoplasmic reticulum stress and diabetic
Am. J. Surg. 188 (2004) 136–149, https://doi.org/10.1016/j. cardiomyopathy, Exp. Diabetes Res 2012 (2012), 827971, https://doi.org/
amjsurg.2003.12.063. 10.1155/2012/827971.
[44] A.A. Knowlton, A.R. Lee, Estrogen and the cardiovascular system, Pharm. Ther. [71] L. Cai, Y.J. Kang, Cell death and diabetic cardiomyopathy, Cardiovasc Toxicol. 3
135 (2012) 54–70, https://doi.org/10.1016/j.pharmthera.2012.03.007. (2003) 219–228, https://doi.org/10.1385/CT:3:3:219.
[45] A.S. Lee, W.Y. Chen, H.C. Chan, J.F. Hsu, M.Y. Shen, C.M. Chang, H. Bair, M.J. Su, [72] Z. Li, T. Zhang, H. Dai, G. Liu, H. Wang, Y. Sun, Y. Zhang, Z. Ge, Involvement of
K.C. Chang, C.H. Chen, Gender disparity in LDL-induced cardiovascular damage endoplasmic reticulum stress in myocardial apoptosis of streptozocin-induced
and the protective role of estrogens against electronegative LDL, Cardiovasc diabetic rats, J. Clin. Biochem Nutr. 41 (2007) 58–67, https://doi.org/10.3164/
Diabetol. 13 (2014) 64, https://doi.org/10.1186/1475-2840-13-64. jcbn.2007008.
[46] N. Gray, G. Picone, F. Sloan, A. Yashkin, Relation between BMI and diabetes [73] B.S. Zha, H. Zhou, ER stress and lipid metabolism in adipocytes, Biochem Res Int
mellitus and its complications among US older adults, South Med J. 108 (2015) 2012 (2012) 1–9, https://doi.org/10.1155/2012/312943.
29–36, https://doi.org/10.14423/SMJ.0000000000000214. [74] A.P. Lakshmanan, M. Harima, K. Suzuki, V. Soetikno, M. Nagata, T. Nakamura,
[47] G.S. Hotamisligil, Inflammation and metabolic disorders, Nature 444 (2006) T. Takahashi, H. Sone, H. Kawachi, K. Watanabe, The hyperglycemia stimulated
860–867, https://doi.org/10.1038/nature05485. myocardial endoplasmic reticulum (ER) stress contributes to diabetic
[48] G.S. Hotamisligil, Foundations of immunometabolism and implications for cardiomyopathy in the transgenic non-obese type 2 diabetic rats: A differential
metabolic health and disease, Immunity 47 (2017) 406–420, https://doi.org/ role of unfolded protein response (UPR) signaling proteins, Int J. Biochem Cell
10.1016/j.immuni.2017.08.009. Biol. 45 (2013) 438–447, https://doi.org/10.1016/j.biocel.2012.09.017.
[49] A. Poznyak, A. v Grechko, P. Poggio, V.A. Myasoedova, V. Alfieri, A.N. Orekhov, [75] F.R. Paolillo, V.S. Mattos, A. Borghi-Silva, V.S. Bagnato, J.C. de Castro Neto,
The diabetes mellitus–atherosclerosis connection: the role of lipid and glucose Advanced glycation endproducts as biomarkers for risk of diabetes and
metabolism and chronic inflammation, Int J. Mol. Sci. 21 (2020) 1835, https:// cardiovascular diseases by skin autofluorescence: a noninvasive optical screening,
doi.org/10.3390/ijms21051835. Photo Photo Laser Surg. 37 (2019) 168–174, https://doi.org/10.1089/
[50] T. Hirano, Pathophysiology of diabetic dyslipidemia, J. Atheroscler. Thromb. 25 photob.2018.4563.
(2018) 771–782, https://doi.org/10.5551/jat.RV17023. [76] S. Mahali, N. Raviprakash, P.B. Raghavendra, S.K. Manna, Advanced glycation
[51] M. Rafieian-Kopaei, M. Setorki, M. Doudi, A. Baradaran, H. Nasri, end products (AGEs) induce apoptosis via a novel pathway, J. Biol. Chem. 286
Atherosclerosis: process, indicators, risk factors and new hopes, Int J. Prev. Med 5 (2011) 34903–34913, https://doi.org/10.1074/jbc.M111.279190.
(2014) 927–946. 〈https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4258672/〉. [77] N.J. Byrne, N.S. Rajasekaran, E.D. Abel, H. Bugger, Therapeutic potential of
[52] H. Nording, L. Baron, H.F. Langer, Platelets as therapeutic targets to prevent targeting oxidative stress in diabetic cardiomyopathy, Free Radic. Biol. Med 169
atherosclerosis, Atherosclerosis 307 (2020) 97–108, https://doi.org/10.1016/j. (2021) 317–342, https://doi.org/10.1016/j.freeradbiomed.2021.03.046.
atherosclerosis.2020.05.018. [78] M.A. Hill, Y. Yang, Z. Sun, L. Zhang, J.R. Sowers, Mechanisms underlying
[53] G. Basta, S. del Turco, R. de Caterina, Advanced glycation endproducts: vascular stiffening in obesity, insulin resistance, and type 2 diabetes, in: The
implications for accelerated atherosclerosis in diabetes. Recent. Prog. Med 95 Science, Etiology and Mechanobiology of Diabetes and Its Complications,
(2004) 67–80. 〈https://pubmed.ncbi.nlm.nih.gov/15072391/〉. Elsevier, 2021, pp. 63–88, https://doi.org/10.1016/B978-0-12-821070-3.00021-
[54] B.B. Dokken, The pathophysiology of cardiovascular disease and diabetes: beyond 0.
blood pressure and lipids, Diabetes Spectr. 21 (2008) 160–165, https://doi.org/ [79] T.A. Collier, A. Nash, H.L. Birch, N.H. de Leeuw, Intra-molecular lysine-arginine
10.2337/diaspect.21.3.160. derived advanced glycation end-product cross-linking in type I collagen: A

12
M. Thakur and R.S. Tupe Prostaglandins and Other Lipid Mediators 164 (2023) 106698

molecular dynamics simulation study, Biophys. Chem. 218 (2016) 42–46, [102] R.N. Lemaitre, P.N. Jensen, M. Zeigler, A.M. Fretts, J.G. Umans, B. v Howard, C.
https://doi.org/10.1016/j.bpc.2016.09.003. M. Sitlani, B. McKnight, S.A. Gharib, I.B. King, D.S. Siscovick, B.M. Psaty,
[80] D.J. Gomes, A.P. Velosa, L.S. Okuda, F.B. Fusco, K.S. da Silva, P.R. Pinto, E. N. Sotoodehnia, R.A. Totah, Plasma epoxyeicosatrienoic acids and diabetes-
R. Nakandakare, M.L. Correa-Giannella, T. Woods, M.A. Brimble, R. Pickford, K.- related cardiovascular disease: The cardiovascular health study, EBioMedicine 83
A. Rye, W.R. Teodoro, S. Catanozi, M. Passarelli, Glycated albumin induces lipid (2022), 104189, https://doi.org/10.1016/j.ebiom.2022.104189.
infiltration in mice aorta independently of DM and RAS local modulation by [103] F. Ying, H. Liu, E.H. Ching Tang, I. Lakhani, N. Liu, Z. Xia, S. Liu, Prostaglandin E
inducing lipid peroxidation and inflammation, J. Diabetes Complicat. 30 (2016) receptor subtype 4 protects against diabetic cardiomyopathy by modulating
1614–1621, https://doi.org/10.1016/j.jdiacomp.2016.07.001. cardiac fatty acid metabolism via FOXO1/CD36 signalling, Biochem Biophys. Res
[81] R.S. Pinto, C.A. Minanni, A.L. de Araújo Lira, M. Passarelli, Advanced glycation Commun. 548 (2021) 196–203, https://doi.org/10.1016/j.bbrc.2021.01.038.
end products: a sweet flavor that embitters cardiovascular disease, Int J. Mol. Sci. [104] D. Kong, Y. Yu, Prostaglandin D2 signaling and cardiovascular homeostasis,
23 (2022) 2404, https://doi.org/10.3390/ijms23052404. J. Mol. Cell Cardiol. 167 (2022) 97–105, https://doi.org/10.1016/j.
[82] M.A. Roberts, M.C. Thomas, D. Fernando, N. Macmillan, D.A. Power, F.L. Ierino, yjmcc.2022.03.011.
Low molecular weight advanced glycation end products predict mortality in [105] R. Bolli, Discovery of a new function of cyclooxygenase (COX)-2: COX-2 is a
asymptomatic patients receiving chronic haemodialysis, Nephrol. Dial. cardioprotective protein that alleviates ischemia/reperfusion injury and mediates
Transplant. 21 (2006) 1611–1617, https://doi.org/10.1093/ndt/gfl053. the late phase of preconditioning, Cardiovasc Res 55 (2002) 506–519, https://
[83] N.J.R. Blackburn, B. Vulesevic, B. McNeill, C.E. Cimenci, A. Ahmadi, doi.org/10.1016/S0008-6363(02)00414-5.
M. Gonzalez-Gomez, A. Ostojic, Z. Zhong, M. Brownlee, P.J. Beisswenger, R. [106] D. Wang, V. v Patel, E. Ricciotti, R. Zhou, M.D. Levin, E. Gao, Z. Yu, V.A. Ferrari,
W. Milne, E.J. Suuronen, Methylglyoxal-derived advanced glycation end products M.M. Lu, J. Xu, H. Zhang, Y. Hui, Y. Cheng, N. Petrenko, Y. Yu, G.A. FitzGerald,
contribute to negative cardiac remodeling and dysfunction post-myocardial Cardiomyocyte cyclooxygenase-2 influences cardiac rhythm and function, Proc.
infarction, Basic Res Cardiol. 112 (2017) 57, https://doi.org/10.1007/s00395- Natl. Acad. Sci. 106 (2009) 7548–7552, https://doi.org/10.1073/
017-0646-x. pnas.0805806106.
[84] R. Ramasamy, S.F. Yan, A.M. Schmidt, Receptor for AGE (RAGE): signaling [107] K.J. Bosma, M. Ghosh, S.R. Andrei, L. Zhong, J.C. Dunn, V.F. Ricciardi, J.
mechanisms in the pathogenesis of diabetes and its complications, Ann. N. Y B. Burkett, A.K. Hatzopoulos, D.S. Damron, M. Gannon, Pharmacological
Acad. Sci. 1243 (2011) 88–102, https://doi.org/10.1111/j.1749- modulation of prostaglandin E 2 (PGE2) EP receptors improves cardiomyocyte
6632.2011.06320.x. function under hyperglycemic conditions, Physiol. Rep. 10 (2022), https://doi.
[85] H. Maillard-Lefebvre, E. Boulanger, M. Daroux, C. Gaxatte, B.I. Hudson, org/10.14814/phy2.15212.
M. Lambert, Soluble receptor for advanced glycation end products: a new [108] E. Arehart, S. Gleim, Z. Kasza, K.M. Fetalvero, K.A. Martin, J. Hwa, Prostacyclin,
biomarker in diagnosis and prognosis of chronic inflammatory diseases, atherothrombosis, and cardiovascular disease, Curr. Med Chem. 14 (2007)
Rheumatology 48 (2009) 1190–1196, https://doi.org/10.1093/rheumatology/ 2161–2169, https://doi.org/10.2174/092986707781389637.
kep199. [109] H. Yücel, A.T. Özdemir, Low LXA4, RvD1 and RvE1 levels may be an indicator of
[86] D. Bu, V. Rai, X. Shen, R. Rosario, Y. Lu, V. D’Agati, S.F. Yan, R.A. Friedman, the development of hypertension, Prostaglandins Leukot. Ess. Fat. Acids 174
E. Nuglozeh, A.M. Schmidt, Activation of the ROCK1 branch of the transforming (2021), 102365, https://doi.org/10.1016/j.plefa.2021.102365.
growth factor-β pathway contributes to RAGE-dependent acceleration of [110] E.P. Brennan, M. Mohan, A. McClelland, M. de Gaetano, C. Tikellis, M. Marai,
atherosclerosis in diabetic ApoE-Null mice, Circ. Res 106 (2010) 1040–1051, D. Crean, A. Dai, O. Beuscart, S. Derouiche, S.P. Gray, R. Pickering, S.M. Tan,
https://doi.org/10.1161/CIRCRESAHA.109.201103. M. Godson-Treacy, S. Sheehan, J.F. Dowdall, M. Barry, O. Belton, S.T. Ali-Shah, P.
[87] T.-W. Lee, Y.-H. Kao, Y.-J. Chen, T.-F. Chao, T.-I. Lee, Therapeutic potential of J. Guiry, K. Jandeleit-Dahm, M.E. Cooper, C. Godson, P. Kantharidis, Lipoxins
vitamin D in AGE/RAGE-related cardiovascular diseases, Cell. Mol. Life Sci. 76 protect against inflammation in diabetes-associated atherosclerosis, Diabetes 67
(2019) 4103–4115, https://doi.org/10.1007/s00018-019-03204-3. (2018) 2657–2667, https://doi.org/10.2337/db17-1317.
[88] D.A. Towler, Vascular calcification, Arterioscler. Thromb. Vasc. Biol. 31 (2011) [111] E. Börgeson, The role of lipoxins in cardiometabolic physiology and disease,
237–239, https://doi.org/10.1161/ATVBAHA.110.220038. Cardiovasc Endocrinol. 5 (2016) 4–13, https://doi.org/10.1097/
[89] M.D. Turner, B. Nedjai, T. Hurst, D.J. Pennington, Cytokines and chemokines: at XCE.0000000000000068.
the crossroads of cell signalling and inflammatory disease, Biochim. Et. Biophys. [112] I. Tabas, G. García-Cardeña, G.K. Owens, Recent insights into the cellular biology
Acta (BBA) - Mol. Cell Res. 1843 (2014) 2563–2582, https://doi.org/10.1016/j. of atherosclerosis, J. Cell Biol. 209 (2015) 13–22, https://doi.org/10.1083/
bbamcr.2014.05.014. jcb.201412052.
[90] N. Sharma-walia, J. Chandrasekharan, Lipoxins: nature’s way to resolve [113] P. Libby, I. Tabas, G. Fredman, E.A. Fisher, Inflammation and its resolution as
inflammation, J. Inflamm. Res (2015) 181, https://doi.org/10.2147/JIR.S90380. determinants of acute coronary syndromes, Circ. Res 114 (2014) 1867–1879,
[91] C.N. Serhan, Lipoxins and aspirin-triggered 15-epi-lipoxins are the first lipid https://doi.org/10.1161/CIRCRESAHA.114.302699.
mediators of endogenous anti-inflammation and resolution, Prostaglandins [114] M. Romano, E. Cianci, F. Simiele, A. Recchiuti, Lipoxins and aspirin-triggered
Leukot. Ess. Fat. Acids 73 (2005) 141–162, https://doi.org/10.1016/j. lipoxins in resolution of inflammation, Eur. J. Pharm. 760 (2015) 49–63, https://
plefa.2005.05.002. doi.org/10.1016/j.ejphar.2015.03.083.
[92] C.N. Serhan, Pro-resolving lipid mediators are leads for resolution physiology, [115] M.H. Petri, A. Laguna-Fernandez, H. Arnardottir, C.E. Wheelock, M. Perretti, G.
Nature 510 (2014) 92–101, https://doi.org/10.1038/nature13479. K. Hansson, M. Bäck, Aspirin-triggered lipoxin A4 inhibits atherosclerosis
[93] C.N. Serhan, N. Chiang, T.E. van Dyke, Resolving inflammation: dual anti- progression in apolipoprotein E − /− mice, Br. J. Pharm. 174 (2017) 4043–4054,
inflammatory and pro-resolution lipid mediators, Nat. Rev. Immunol. 8 (2008) https://doi.org/10.1111/bph.13707.
349–361, https://doi.org/10.1038/nri2294. [116] N. Chiang, C.N. Serhan, S.-E. Dahlén, J.M. Drazen, D.W.P. Hay, G.E. Rovati,
[94] C.N. Serhan, B.D. Levy, Lipid mediators | Lipoxins, in: Encyclopedia of T. Shimizu, T. Yokomizo, C. Brink, The lipoxin receptor ALX: potent ligand-
Respiratory Medicine, Elsevier, 2006, pp. 594–599, https://doi.org/10.1016/B0- specific and stereoselective actions in vivo, Pharm. Rev. 58 (2006) 463–487,
12-370879-6/00490-7. https://doi.org/10.1124/pr.58.3.4.
[95] R. Wisastra, F. Dekker, Inflammation, cancer and oxidative lipoxygenase activity [117] N. Baker, S.J. O’Meara, M. Scannell, P. Maderna, C. Godson, Lipoxin A4: anti-
are intimately linked, Cancers (Basel) 6 (2014) 1500–1521, https://doi.org/ inflammatory and anti-angiogenic impact on endothelial cells, J. Immunol. 182
10.3390/cancers6031500. (2009) 3819–3826, https://doi.org/10.4049/jimmunol.0803175.
[96] M. Sun, R. Wang, Q. Han, Inhibition of leukotriene B4 receptor 1 attenuates [118] S. Ramon, S. Bancos, C.N. Serhan, R.P. Phipps, Lipoxin A4 modulates adaptive
lipopolysaccharide-induced cardiac dysfunction: role of AMPK-regulated immunity by decreasing memory B-cell responses via an ALX/FPR2–dependent
mitochondrial function, Sci. Rep. 7 (2017) 44352, https://doi.org/10.1038/ mechanism, Eur. J. Immunol. 44 (2014) 357–369, https://doi.org/10.1002/
srep44352. eji.201343316.
[97] A.P. Sampson, FLAP inhibitors for the treatment of inflammatory diseases, Curr. [119] N. Tułowiecka, D. Kotlęga, A. Bohatyrewicz, M. Szczuko, Could lipoxins represent
Opin. Invest. Drugs 10 (2009) 1163–1172. 〈https://pubmed.ncbi.nlm.nih.gov/1 a new standard in ischemic stroke treatment? Int J. Mol. Sci. 22 (2021) 4207,
9876784〉. https://doi.org/10.3390/ijms22084207.
[98] Z.H. Maayah, A.O.S. El-Kadi, 5-, 12- and 15-Hydroxyeicosatetraenoic acids [120] V. Brancaleone, T. Gobbetti, N. Cenac, P. le Faouder, B. Colom, R.J. Flower,
induce cellular hypertrophy in the human ventricular cardiomyocyte, RL-14 cell N. Vergnolle, S. Nourshargh, M. Perretti, A vasculo-protective circuit centered on
line, through MAPK- and NF-κB-dependent mechanism, Arch. Toxicol. 90 (2016) lipoxin A4 and aspirin-triggered 15-epi-lipoxin A4 operative in murine
359–373, https://doi.org/10.1007/s00204-014-1419-z. microcirculation, Blood 122 (2013) 608–617, https://doi.org/10.1182/blood-
[99] D.W. Thomas, P.N. Rocha, C. Nataraj, L.A. Robinson, R.F. Spurney, B.H. Koller, T. 2013-04-496661.
M. Coffman, Proinflammatory actions of thromboxane receptors to enhance [121] L. József, C. Zouki, N.A. Petasis, C.N. Serhan, J.G. Filep, Lipoxin A4 and aspirin-
cellular immune responses, J. Immunol. 171 (2003) 6389–6395, https://doi.org/ triggered 15-epi-lipoxin A4 inhibit peroxynitrite formation, NF-κB and AP-1
10.4049/jimmunol.171.12.6389. activation, and IL-8 gene expression in human leukocytes, Proc. Natl. Acad. Sci.
[100] S. PS, M. JA, B. L, C. J, W. M, S. J, A. M, H. E, F. JR, W. JA, P. SJ, A. NG, CYP-450 99 (2002) 13266–13271, https://doi.org/10.1073/pnas.202296999.
epoxygenase derived epoxyeicosatrienoic acid contribute to reversal of heart [122] N. Baker, S.J. O’Meara, M. Scannell, P. Maderna, C. Godson, Lipoxin A4: anti-
failure in obesity-induced diabetic cardiomyopathy via PGC-1α activation, inflammatory and anti-angiogenic impact on endothelial cells, J. Immunol. 182
Cardiovasc Pharm. Open Access 07 (2018), https://doi.org/10.4172/2329- (2009) 3819–3826, https://doi.org/10.4049/jimmunol.0803175.
6607.1000233. [123] Y. Jin, M. Arita, Q. Zhang, D.R. Saban, S.K. Chauhan, N. Chiang, C.N. Serhan,
[101] M. Romashko, J. Schragenheim, N.G. Abraham, J.A. McClung, R. Dana, Anti-angiogenesis effect of the novel anti-inflammatory and pro-
Epoxyeicosatrienoic acid as therapy for diabetic and ischemic cardiomyopathy, resolving lipid mediators, Invest. Opthalmology Vis. Sci. 50 (2009) 4743, https://
Trends Pharm. Sci. 37 (2016) 945–962, https://doi.org/10.1016/j. doi.org/10.1167/iovs.08-2462.
tips.2016.08.001. [124] P.F.T. Cezar-de-Mello, A.M. Vieira, V. Nascimento-Silva, C.G. Villela, C. Barja-
Fidalgo, I.M. Fierro, ATL-1, an analogue of aspirin-triggered lipoxin A4, is a potent

13
M. Thakur and R.S. Tupe Prostaglandins and Other Lipid Mediators 164 (2023) 106698

inhibitor of several steps in angiogenesis induced by vascular endothelial growth [147] H. Shimano, R. Sato, SREBP-regulated lipid metabolism: convergent physiology
factor, Br. J. Pharm. 153 (2008) 956–965, https://doi.org/10.1038/sj. — divergent pathophysiology, Nat. Rev. Endocrinol. 13 (2017) 710–730, https://
bjp.0707650. doi.org/10.1038/nrendo.2017.91.
[125] X.-Q. Chen, S.-H. Wu, Y. Zhou, Y.-R. Tang, Involvement of K+ channel-dependant [148] S. Nammi, M.S. Kim, N.S. Gavande, G.Q. Li, B.D. Roufogalis, Regulation of low-
pathways in lipoxin A4-induced protective effects on hypoxia/reoxygenation density lipoprotein receptor and 3-Hydroxy-3-Methylglutaryl Coenzyme A
injury of cardiomyocytes, Prostaglandins Leukot. Ess. Fat. Acids 88 (2013) reductase expression by Zingiber officinale in the liver of high-fat diet-fed rats,
391–397, https://doi.org/10.1016/j.plefa.2013.03.007. Basic Clin. Pharm. Toxicol. 106 (2009) 389–395, https://doi.org/10.1111/
[126] X.-Q. Chen, S.-H. Wu, Y. Zhou, Y.-R. Tang, Lipoxin A4-induced heme oxygenase-1 j.1742-7843.2009.00497.x.
protects cardiomyocytes against hypoxia/reoxygenation injury via p38 MAPK [149] D. Eberlé, B. Hegarty, P. Bossard, P. Ferré, F. Foufelle, SREBP transcription
activation and Nrf2/ARE complex, PLoS One 8 (2013), e67120, https://doi.org/ factors: master regulators of lipid homeostasis, Biochimie 86 (2004) 839–848,
10.1371/journal.pone.0067120. https://doi.org/10.1016/j.biochi.2004.09.018.
[127] S. Ramon, S. Bancos, C.N. Serhan, R.P. Phipps, Lipoxin A4 modulates adaptive [150] J.N. Athanikar, T.F. Osborne, Specificity in cholesterol regulation of gene
immunity by decreasing memory B-cell responses via an ALX/FPR2–dependent expression by coevolution of sterol regulatory DNA element and its binding
mechanism, Eur. J. Immunol. 44 (2014) 357–369, https://doi.org/10.1002/ protein, Proc. Natl. Acad. Sci. 95 (1998) 4935–4940, https://doi.org/10.1073/
eji.201343316. pnas.95.9.4935.
[128] M.J. Paul-Clark, T. van Cao, N. Moradi-Bidhendi, D. Cooper, D.W. Gilroy, 15-epi- [151] H. Shimano, Sterol regulatory element-binding proteins (SREBPs): transcriptional
lipoxin A4–mediated induction of nitric oxide explains how aspirin inhibits acute regulators of lipid synthetic genes, Prog. Lipid Res 40 (2001) 439–452, https://
inflammation, J. Exp. Med. 200 (2004) 69–78, https://doi.org/10.1084/ doi.org/10.1016/S0163-7827(01)00010-8.
jem.20040566. [152] A.J. Brown, H.W. Coates, L.J. Sharpe, Cholesterol synthesis, in: biochemistry of
[129] C.-S. Wang, O.J. Baker, The G-protein–coupled receptor ALX/FPR2 regulates lipids, lipoproteins and membranes, Elsevier, 2021, pp. 317–355, https://doi.org/
adaptive immune responses in mouse submandibular glands, Am. J. Pathol. 188 10.1016/B978-0-12-824048-9.00005-5.
(2018) 1555–1562, https://doi.org/10.1016/j.ajpath.2018.04.003. [153] N. Alonso, P. Moliner, D. Mauricio, Pathogenesis, clinical features and treatment
[130] M.H. Petri, S. Thul, T. Andonova, M. Lindquist-Liljeqvist, H. Jin, N.-T. Skenteris, of diabetic cardiomyopathy, in: (2017) 197–217. https://doi.org/10.1007/5584
H. Arnardottir, L. Maegdefessel, K. Caidahl, M. Perretti, J. Roy, M. Bäck, _2017_105.
Resolution of inflammation through the lipoxin and ALX/FPR2 receptor pathway [154] X. Chang, Y. Li, C. Cai, F. Wu, J. He, Y. Zhang, J. Zhong, Y. Tan, R. Liu, H. Zhu,
protects against abdominal aortic aneurysms, JACC Basic Transl. Sci. 3 (2018) H. Zhou, Mitochondrial quality control mechanisms as molecular targets in
719–727, https://doi.org/10.1016/j.jacbts.2018.08.005. diabetic heart, Metabolism (2022), 155313, https://doi.org/10.1016/j.
[131] M.H. Petri, S. Thul, O. Ovchinnikova, M. Bäck, Differential regulation of metabol.2022.155313.
monocytic expression of leukotriene and lipoxin receptors, Prostaglandins Other [155] S.M. Colgan, A.A. Al-Hashimi, R.C. Austin, Endoplasmic reticulum stress and lipid
Lipid Mediat 121 (2015) 138–143, https://doi.org/10.1016/j. dysregulation, Expert Rev. Mol. Med 13 (2011), e4, https://doi.org/10.1017/
prostaglandins.2015.07.005. S1462399410001742.
[132] J. Pirault, M. Bäck, Lipoxin and resolvin receptors transducing the resolution of [156] J.P. Ayyappan, K. Lizardo, S. Wang, E. Yurkow, J.F. Nagajyothi, Inhibition of
inflammation in cardiovascular disease, Front Pharm. 9 (2018), https://doi.org/ SREBP improves cardiac lipidopathy, improves endoplasmic reticulum stress, and
10.3389/fphar.2018.01273. modulates chronic chagas cardiomyopathy, J. Am. Heart Assoc. 9 (2020), https://
[133] M.H. Petri, A. Laguna-Fernández, M. Gonzalez-Diez, G. Paulsson-Berne, G. doi.org/10.1161/JAHA.119.014255.
K. Hansson, M. Bäck, The role of the FPR2/ALX receptor in atherosclerosis [157] P. Ferré, F. Foufelle, SREBP-1c transcription factor and lipid homeostasis: clinical
development and plaque stability, Cardiovasc Res 105 (2015) 65–74, https://doi. perspective, Horm. Res Paediatr. 68 (2007) 72–82, https://doi.org/10.1159/
org/10.1093/cvr/cvu224. 000100426.
[134] P. Ferré, F. Foufelle, SREBP-1c transcription factor and lipid homeostasis: clinical [158] T.-F. Liu, J.-J. Tang, P.-S. Li, Y. Shen, J.-G. Li, H.-H. Miao, B.-L. Li, B.-L. Song,
perspective, Horm. Res Paediatr. 68 (2007) 72–82, https://doi.org/10.1159/ Ablation of gp78 in Liver Improves Hyperlipidemia and Insulin Resistance by
000100426. Inhibiting SREBP to Decrease Lipid Biosynthesis, Cell Metab. 16 (2012) 213–225,
[135] J. Kotzka, S. Lehr, G. Roth, H. Avci, B. Knebel, D. Muller-Wieland, Insulin- https://doi.org/10.1016/j.cmet.2012.06.014.
activated Erk-mitogen-activated protein kinases phosphorylate sterol regulatory [159] I. Falcão-Pires, G. Palladini, N. Gonçalves, J. van der Velden, D. Moreira-
element-binding protein-2 at serine residues 432 and 455 in vivo, J. Biol. Chem. Gonçalves, D. Miranda-Silva, F. Salinaro, W.J. Paulus, H.W.M. Niessen, S. Perlini,
279 (2004) 22404–22411, https://doi.org/10.1074/jbc.M401198200. A.F. Leite-Moreira, Distinct mechanisms for diastolic dysfunction in diabetes
[136] A. Cachefo, P. Boucher, E. Dusserre, P. Bouletreau, M. Beylot, C. Chambrier, mellitus and chronic pressure-overload, Basic Res Cardiol. 106 (2011) 801–814,
Stimulation of cholesterol synthesis and hepatic lipogenesis in patients with https://doi.org/10.1007/s00395-011-0184-x.
severe malabsorption, J. Lipid Res 44 (2003) 1349–1354, https://doi.org/ [160] P. Daneii, S. Neshat, M.S. Mirnasiry, Z. Moghimi, F. Dehghan Niri, A. Farid,
10.1194/jlr.M300030-JLR200. M. Shekarchizadeh, K. Heshmat-Ghahdarijani, Lipids and diastolic dysfunction:
[137] R. Mastrocola, M. Collino, D. Nigro, F. Chiazza, G. D’Antona, M. Aragno, M. Recent evidence and findings, Nutr., Metab. Cardiovasc. Dis. 32 (2022)
A. Minetto, Accumulation of advanced glycation end-products and activation of 1343–1352, https://doi.org/10.1016/j.numecd.2022.03.003.
the SCAP/SREBP lipogenetic pathway occur in diet-induced obese mouse skeletal [161] E. Demetz, A. Schroll, K. Auer, C. Heim, J.R. Patsch, P. Eller, M. Theurl, I. Theurl,
muscle, PLoS One 10 (2015), e0119587, https://doi.org/10.1371/journal. M. Theurl, M. Seifert, D. Lener, U. Stanzl, D. Haschka, M. Asshoff, S. Dichtl,
pone.0119587. M. Nairz, E. Huber, M. Stadlinger, A.R. Moschen, X. Li, P. Pallweber,
[138] J.H. Lee, H.S. Kang, H.Y. Park, Y.A. Moon, Y.N. Kang, B.C. Oh, D.K. Song, J. H. Scharnagl, T. Stojakovic, W. März, M.E. Kleber, K. Garlaschelli, P. Uboldi, A.
H. Bae, S.S. Im, PPARα-dependent Insig2a overexpression inhibits SREBP-1c L. Catapano, F. Stellaard, M. Rudling, K. Kuba, Y. Imai, M. Arita, J.D. Schuetz, P.
processing during fasting, Sci. Rep. 7 (2017) 9958, https://doi.org/10.1038/ P. Pramstaller, U.J.F. Tietge, M. Trauner, G.D. Norata, T. Claudel, A.A. Hicks,
s41598-017-10523-7. G. Weiss, I. Tancevski, The arachidonic acid metabolome serves as a conserved
[139] C. Ma, Y. Zhang, Y. Li, C. Chen, W. Cai, Y. Zeng, The role of PPARγ in advanced regulator of cholesterol metabolism, Cell Metab. 20 (2014) 787–798, https://doi.
glycation end products-induced inflammatory response in human chondrocytes, org/10.1016/j.cmet.2014.09.004.
PLoS One 10 (2015), e0125776, https://doi.org/10.1371/journal.pone.0125776. [162] M. Ouimet, T.J. Barrett, E.A. Fisher, HDL and reverse cholesterol transport, Circ.
[140] C. Ma, Y. Zhang, Y. Li, C. Chen, W. Cai, Y. Zeng, The role of PPARγ in advanced Res 124 (2019) 1505–1518, https://doi.org/10.1161/CIRCRESAHA.119.312617.
glycation end products-induced inflammatory response in human chondrocytes, [163] M. Arad, M. Waldman, N.G. Abraham, E. Hochhauser, Therapeutic approaches to
PLoS One 10 (2015), e0125776, https://doi.org/10.1371/journal.pone.0125776. diabetic cardiomyopathy: Targeting the antioxidant pathway, Prostaglandins
[141] A.R. Tovar, N. Torres, The role of dietary protein on lipotoxicity, Biochim. Et. Other Lipid Mediat 150 (2020), 106454, https://doi.org/10.1016/j.
Biophys. Acta (BBA) - Mol. Cell Biol. Lipids 1801 (2010) 367–371, https://doi. prostaglandins.2020.106454.
org/10.1016/j.bbalip.2009.09.007. [164] S. Murarka, M.R. Movahed, Diabetic cardiomyopathy, J. Card. Fail 16 (2010)
[142] W. Wahli, L. Michalik, PPARs at the crossroads of lipid signaling and 971–979, https://doi.org/10.1016/j.cardfail.2010.07.249.
inflammation, Trends Endocrinol. Metab. 23 (2012) 351–363, https://doi.org/ [165] B. Maisch, P. Alter, S. Pankuweit, Diabetic cardiomyopathy—fact or fiction? Herz
10.1016/j.tem.2012.05.001. 36 (2011) 102–115, https://doi.org/10.1007/s00059-011-3429-4.
[143] C.C. Chyau, H.F. Wang, W.J. Zhang, C.C. Chen, S.H. Huang, C.C. Chang, R. [166] D. Sueta, N. Tabata, S. Hokimoto, Clinical roles of calcium channel blockers in
Y. Peng, Antrodan alleviates high-fat and high-fructose diet-induced fatty liver ischemic heart diseases, Hypertens. Res. 40 (2017) 423–428, https://doi.org/
disease in C57BL/6 mice model via AMPK/Sirt1/SREBP-1c/PPARγ pathway, Int 10.1038/hr.2016.183.
J. Mol. Sci. 21 (2020) 360, https://doi.org/10.3390/ijms21010360. [167] R.P. Russell, Side effects of calcium channel blockers, Hypertension 11 (1988),
[144] I. Ballesteros, M.I. Cuartero, J.M. Pradillo, J. de la Parra, A. Perez-Ruiz, A. Corbi, https://doi.org/10.1161/01.HYP.11.3_Pt_2.II42.
M. Ricote, J.A. Hamilton, M. Sobrado, J. Vivancos, F. Nombela, I. Lizasoain, M. [168] W.J. Elliott, C.V.S. Ram, Calcium channel blockers, J. Clin. Hypertens. 13 (2011)
A. Moro, Rosiglitazone-induced CD36 up-regulation resolves inflammation by 687–689, https://doi.org/10.1111/j.1751-7176.2011.00513.x.
PPAR and 5-LO-dependent pathways, J. Leukoc. Biol. 95 (2014) 587–598, [169] A.T. Stearns, A. Balakrishnan, D.B. Rhoads, A. Tavakkolizadeh, Rapid
https://doi.org/10.1189/jlb.0613326. upregulation of sodium-glucose transporter SGLT1 in response to intestinal sweet
[145] A. von Knethen, L.K. Sha, L. Kuchler, A.K. Heeg, D. Fuhrmann, H. Heide, I. Wittig, taste stimulation, Ann. Surg. 251 (2010) 865–871, https://doi.org/10.1097/
T.J. Maier, D. Steinhilber, B. Brüne, 5-Lipoxygenase contributes to PPARγ SLA.0b013e3181d96e1f.
activation in macrophages in response to apoptotic cells, Cell Signal 25 (2013) [170] R. Johnston, O. Uthman, E. Cummins, C. Clar, P. Royle, J. Colquitt, B.K. Tan,
2762–2768, https://doi.org/10.1016/j.cellsig.2013.08.045. A. Clegg, S. Shantikumar, R. Court, J.P. O’Hare, D. McGrane, T. Holt, N. Waugh,
[146] H. Shimano, R. Sato, SREBP-regulated lipid metabolism: convergent physiology Canagliflozin, dapagliflozin and empagliflozin monotherapy for treating type 2
— divergent pathophysiology, Nat. Rev. Endocrinol. 13 (2017) 710–730, https:// diabetes: systematic review and economic evaluation, Health Technol. Assess.
doi.org/10.1038/nrendo.2017.91. (Rockv. ) 21 (2017) 1–218, https://doi.org/10.3310/hta21020.

14
M. Thakur and R.S. Tupe Prostaglandins and Other Lipid Mediators 164 (2023) 106698

[171] N.B. Dalsgaard, T. Vilsbøll, F.K. Knop, Effects of glucagon-like peptide-1 receptor [186] J.R. Greenfield, D.J. Chisholm, Experimental and clinical pharmacology:
agonists on cardiovascular risk factors: A narrative review of head-to-head Thiazolidinediones - mechanisms of action, Aust. Prescr. 27 (2004) 67–70,
comparisons, Diabetes Obes. Metab. 20 (2018) 508–519, https://doi.org/ https://doi.org/10.18773/austprescr.2004.059.
10.1111/dom.13128. [187] J. Karalliedde, R.E. Buckingham, Thiazolidinediones and their fluid-related
[172] R. Gentilella, V. Pechtner, A. Corcos, A. Consoli, Glucagon-like peptide-1 receptor adverse effects, Drug Saf. 30 (2007) 741–753, https://doi.org/10.2165/
agonists in type 2 diabetes treatment: are they all the same? Diabetes Metab. Res 00002018-200730090-00002.
Rev. 35 (2019) 3070, https://doi.org/10.1002/dmrr.3070. [188] J.R. Vane, R.M. Botting, The mechanism of action of aspirin, Thromb. Res 110
[173] G. Rakipovski, B. Rolin, J. Nøhr, I. Klewe, K.S. Frederiksen, R. Augustin, (2003) 255–258, https://doi.org/10.1016/S0049-3848(03)00379-7.
J. Hecksher-Sørensen, C. Ingvorsen, J. Polex-Wolf, L.B. Knudsen, The GLP-1 [189] J.S. Paikin, J.W. Eikelboom, Aspirin, Circulation 125 (2012), https://doi.org/
analogs liraglutide and semaglutide reduce atherosclerosis in ApoE− /− and 10.1161/CIRCULATIONAHA.111.046243.
LDLr− /− mice by a mechanism that includes inflammatory pathways, JACC Basic [190] E. v Paez Espinosa, J.P. Murad, F.T. Khasawneh, Aspirin: pharmacology and
Transl. Sci. 3 (2018) 844–857, https://doi.org/10.1016/j.jacbts.2018.09.004. clinical applications, Thrombosis 2012 (2012) 1–15, https://doi.org/10.1155/
[174] I. Avgerinos, T. Michailidis, A. Liakos, T. Karagiannis, D.R. Matthews, A. Tsapas, 2012/173124.
E. Bekiari, Oral semaglutide for type 2 diabetes: A systematic review and meta- [191] K.T. Olkkola, J. Ahonen, Midazolam and Other Benzodiazepines, in: Modern
analysis, Diabetes Obes. Metab. 22 (2020) 335–345, https://doi.org/10.1111/ Anesthetics, Springer Berlin Heidelberg, Berlin, Heidelberg, n.d. 335–360.
dom.13899. https://doi.org/10.1007/978–3-540–74806-9_16.
[175] F. Xu, H. Cao, Z. Chen, H. Gu, W. Guo, B. Lin, J. Weng, Short-term GLP-1 receptor [192] E. Gambeta, C.M. Kopruszinski, R.C. dos Reis, J.M. Zanoveli, J.G. Chichorro,
agonist exenatide ameliorates intramyocellular lipid deposition without weight Evaluation of heat hyperalgesia and anxiety like-behaviors in a rat model of
loss in ob/ob mice, Int J. Obes. 44 (2020) 937–947, https://doi.org/10.1038/ orofacial cancer, Neurosci. Lett. 619 (2016) 100–105, https://doi.org/10.1016/j.
s41366-019-0513-y. neulet.2016.03.001.
[176] M.T. Gürgöze, A.H.G. Muller-Hansma, M.M. Schreuder, A.M.H. Galema-Boers, [193] K.W. Lindsay, I. Bone, G. Fuller, Clinical presentation, anatomical concepts and
E. Boersma, J.E. Roeters, van Lennep, Adverse events associated with PCSK9 diagnostic approach, in: Neurology and Neurosurgery Illustrated, Elsevier, 2010,
inhibitors: a real-world experience, Clin. Pharm. Ther. 105 (2019) 496–504, pp. 67–216, https://doi.org/10.1016/B978-0-443-06957-4.50007-0.
https://doi.org/10.1002/cpt.1193. [194] J.W. Taylor, K.B. Simon, Possible intramuscular midazolam-associated
[177] S.-i Yamagishi, Advanced glycation end-products, in: Brenner’s Encyclopedia of cardiorespiratory arrest and death, DICP 24 (1990) 695–697, https://doi.org/
Genetics, Elsevier, 2013, pp. 36–38, https://doi.org/10.1016/B978-0-12-374984- 10.1177/106002809002400707.
0.00476-9. [195] K. Zhang, M. Hu, L. Zhang, Q. Zhang, Y. Huang, The effect of BML-111 in
[178] P. Balakumar, A. Rohilla, P. Krishan, P. Solairaj, A. Thangathirupathi, The preeclampsia rat model induced by the low dose of cadmium chloride, Am. J.
multifaceted therapeutic potential of benfotiamine, Pharm. Res 61 (2010) Perinatol. Rep. 09 (2019) 201–208, https://doi.org/10.1055/s-0039-1693016.
482–488, https://doi.org/10.1016/j.phrs.2010.02.008. [196] C.D. Russell, J. Schwarze, The role of pro-resolution lipid mediators in infectious
[179] P. Quade-Lyssy, A.M. Kanarek, M. Baiersdörfer, R. Postina, E. Kojro, Statins disease, Immunology 141 (2014) 166–173, https://doi.org/10.1111/imm.12206.
stimulate the production of a soluble form of the receptor for advanced glycation [197] F. Yin, F. Feng, L. Wang, X. Wang, Z. Li, Y. Cao, SREBP-1 inhibitor Betulin
end products, J. Lipid Res 54 (2013) 3052–3061, https://doi.org/10.1194/jlr. enhances the antitumor effect of Sorafenib on hepatocellular carcinoma via
M038968. restricting cellular glycolytic activity, Cell Death Dis. 10 (2019) 672, https://doi.
[180] C. Rojas-Fernandez, Z. Hudani, V. Bittner, Statins and cognitive side effects, org/10.1038/s41419-019-1884-7.
Cardiol. Clin. 33 (2015) 245–256, https://doi.org/10.1016/j.ccl.2015.02.008. [198] L. Ren, S. Niu, Y. Sun, Y. Liang, J. Zhao, T. Zhang, J. Zhang, Anti-inflammatory
[181] H. Hauner, The mode of action of thiazolidinediones, Diabetes Metab. Res Rev. 18 action of betulin and its potential as a dissociated glucocorticoid receptor
(2002) S10–S15, https://doi.org/10.1002/dmrr.249. modulator, Food Chem. Toxicol. 157 (2021), 112539, https://doi.org/10.1016/j.
[182] J. Karalliedde, R.E. Buckingham, Thiazolidinediones and their fluid-related fct.2021.112539.
adverse effects, Drug Saf. 30 (2007) 741–753, https://doi.org/10.2165/ [199] L. Zhou, Y. Li, T. Nie, S. Feng, J. Yuan, H. Chen, Z. Yang, Clenbuterol inhibits
00002018-200730090-00002. SREBP-1c expression by activating CREB1, BMB Rep. 40 (2007) 525–531, https://
[183] W.R. Kim, S.L. Flamm, A.M. di Bisceglie, H.C. Bodenheimer, Serum activity of doi.org/10.5483/BMBRep.2007.40.4.525.
alanine aminotransferase (ALT) as an indicator of health and disease, Hepatology [200] P.S. Gomes, M.P.D. Carneiro, P. de, A. Machado, V.V. de Andrade-Neto, A.M. da
47 (2008) 1363–1370, https://doi.org/10.1002/hep.22109. Fonseca-Martins, A. Goundry, J.V.M. Pereira da Silva, D.C.O. Gomes, A.P.C. de,
[184] H. Shoda, S. Miyata, B.-F. Liu, H. Yamada, T. Ohara, K. Suzuki, M. Oimomi, A. Lima, V. Ennes-Vidal, A.C.R. Sodero, S.G. De-Simone, H.L. de Matos Guedes,
M. Kasuga, Inhibitory effects of tenilsetam on the maillard reaction, Subtilisin of Leishmania amazonensis as potential druggable target: subcellular
Endocrinology 138 (1997) 1886–1892, https://doi.org/10.1210/ localization, in vitro Leishmanicidal activity and molecular docking of PF-
endo.138.5.5151. 429242, a subtilisin inhibitor, Curr. Issues Mol. Biol. 44 (2022) 2089–2106,
[185] S. Oparil, D. Williams, S.G. Chrysant, T.C. Marbury, J. Neutel, Comparative https://doi.org/10.3390/cimb44050141.
efficacy of olmesartan, losartan, valsartan, and irbesartan in the control of [201] L. Uchida, S. Urata, G. Ulanday, Y. Takamatsu, J. Yasuda, K. Morita, D. Hayasaka,
essential hypertension, J. Clin. Hypertens. 3 (2001) 283–318, https://doi.org/ Suppressive effects of the site 1 protease (S1P) inhibitor, PF-429242, on dengue
10.1111/j.1524-6175.2001.01136.x. virus propagation, Viruses 8 (2016) 46, https://doi.org/10.3390/v8020046.

15

You might also like