You are on page 1of 15

Review

This Review is part of a thematic series on the Pathobiology of Obesity, which includes the following articles:
Adipose-Derived Stem Cells for Regenerative Medicine
Cardiac Energy Metabolism in Obesity

Leptin Signaling and Obesity: Cardiovascular Consequences

Lipid Disorders and the Metabolic Syndrome


Adiponectin As a Cardiovascular Protectant
Gary Lopaschuk, Guest Editor

Leptin Signaling and Obesity


Cardiovascular Consequences
Ronghua Yang, Lili A. Barouch

Abstract—Leptin, among the best known hormone markers for obesity, exerts pleiotropic actions on multiple organ
systems. In this review, we summarize major leptin signaling pathways, namely Janus-activated kinase/signal
transducers and activators of transcription and mitogen-activated protein kinase, including possible mechanisms of
leptin resistance in obesity. The effects of leptin on the cardiovascular system are discussed in detail, including its
contributions to hypertension, atherosclerosis, depressed myocardial contractile function, fatty acid metabolism,
hypertrophic remodeling, and reduction of ischemic/reperfusion injury. The overall goal is to summarize current
Downloaded from http://ahajournals.org by on April 15, 2022

understanding of how altered leptin signaling in obesity contributes to obesity-related cardiovascular disease. (Circ
Res. 2007;101:545-559.)
Key Words: leptin 䡲 obesity 䡲 cardiovascular disease

O ver 60% of people in the United States are overweight or


obese. Extensive evidence now supports the notion that
maladaptation of the biological system for weight mainte-
normal range for her age, and the same effects were observed
in her similarly affected cousin.10 Plasma leptin is generally
proportional to adipose mass.11,12 The primary physiological
nance makes it extremely difficult for people to maintain role of leptin is to communicate to the central nervous system
weight loss.1 Several genes have been identified to disclose a (CNS) the abundance of available energy stores and to
physiological system that maintains body weight within a restrain food intake and induce energy expenditure. The
range of about twenty pounds.2 A key element of this system absence of leptin therefore leads to increased appetite and
is leptin, the 16-kDa hormonal product of the obesity (ob) food intake that result in morbid obesity. Notably, only rare
gene.3 Leptin is primarily secreted by adipocytes and is a cases of severe early childhood obesity have been associated
classic member of the more than 50 identified adipocytokines with leptin deficiency.9,13 The remainder of the obese popu-
that participate in adipose tissue hormonal signaling.4 lation typically have elevated leptin levels.14 The failure of
Since its identification in 1994, leptin has attracted much leptin to induce weight loss in these cases is thought to be the
attention as one of the most important central and peripheral result of leptin resistance.
signals for the maintenance of energy homeostasis.5– 8 For Hyperleptinemia, nearly universally observed in human
example, a 9-year-old girl with extreme obesity was found to obesity and animal models, is accompanied by a disruption of
lack leptin.9 Leptin treatment reduced her weight to the the usual activities of the hormone, possibly at different

Original received May 22, 2007; revision received July 20, 2007; accepted August 6, 2007.
From the Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, Md.
This manuscript was sent to Richard A. Walsh, Consulting Editor, for review by expert referees, editorial decision, and final disposition.
Correspondence to Lili A. Barouch, MD, Johns Hopkins University, 720 Rutland Ave, Ross 1050, Baltimore, MD 21205. E-mail barouch@jhmi.edu
© 2007 American Heart Association, Inc.
Circulation Research is available at http://circres.ahajournals.org DOI: 10.1161/CIRCRESAHA.107.156596

545
546 Circulation Research September 14, 2007

Figure 1. Leptin receptor signaling. The binding of leptin to its receptor leads to formation of the Ob-R/JAK2 complex that results in
cross-phosphorylation. Tyr1138 on Ob-Rb is crucial for STAT3 activation, which stimulates SOCS3 expression that negatively inhibits
leptin signaling via Tyr985 and additional sites on JAK2. Protein tyrosine phosphatase 1B (PTP1B) is also capable of inhibition of leptin
signaling. JAK2 phosphorylation can lead to activation of MAPK and insulin receptor substrate/PI3K signaling pathways. See text.
GRB2 indicates growth factor receptor– bound protein 2.

stages in the circulatory transport and/or in the signaling thyroid hormone, and exposure to cold temperature.22,23 In the
cascade. Disruption of leptin signaling in the hypothalamus heart, increased leptin expression is seen following reperfu-
results in obesity and confirms the central role of this sion after ischemia,24,25 and leptin concentration in cardio-
hormone in the maintenance of energy balance.7,15,16 Emerg-
Downloaded from http://ahajournals.org by on April 15, 2022

myocyte culture serum is increased with endothelin (ET)-1


ing evidence suggests that leptin resistance in the CNS may and angiotensin (Ang) II treatment,26 suggesting the heart as
be selective, namely that the effects of leptin on central a site of leptin production.
metabolic processes is disrupted, whereas its other effects, Six isoforms of the Ob-R (a to f) have been identified in the
such as the sympathetic activation of blood pressure, is still murine model, and they are closely related to the class I
retained.17 In addition to its actions in the CNS, leptin cytokine receptor family. Ob-Ra and Ob-Rb represent the
receptors (Ob-Rs) are found in multiple peripheral tissue dominant isoforms in the heart, whereas the others are
types and affect many systemic processes, such as reproduc- expressed at low levels27 and are not well conserved among
tion, immunity, and cardiovascular functions.18 –20 species.28,29 Ob-Re is the secreted form that binds circulating
Obesity is also a part of the metabolic syndrome, which is leptin and regulates the concentration of free leptin.30
diagnosed by a set of criteria that include abdominal obesity,
insulin resistance, dyslipidemia, and hypertension. This pa- Janus-Activated Kinase/Signal Transducers and
tient population faces increased risk for type 2 diabetes and Activators of Transcription
cardiovascular diseases. The widely distributed Ob-Rs make Ob-Rs have been shown to activate Janus-activated kinase
the hormone an attractive candidate for a molecular link in the (JAK), signal transducers and activators of transcription
pathogenesis of obesity-related diseases. Although disruption (STAT), insulin receptor substrate, and the mitogen-activated
of leptin signaling can lead to altered phenotypic expression protein kinase (MAPK) pathways. The best-characterized
and function of peripheral organs, the relative contributions pathway in leptin signaling is the JAK/STAT pathway (Fig-
of central versus peripheral signal disruption are still contro- ure 1).31 Ligand binding causes Ob-R to undergo homooli-
versial. Increasing our understanding of how leptin and/or gomerization32,33 and to bind to JAK, primarily JAK2.34 In
leptin resistance affects the heart and vasculature will be the case of overexpression of JAKs by transient transfection,
important for gaining comprehension of obesity-related weak Ob-Rb/JAK1 and Ob-Ra/JAK2 association was ob-
threats to cardiovascular health. served with leptin treatment.35 However, only Ob-Rb con-
tains the STAT-binding site.35
Leptin Signaling Studies in vivo have demonstrated that signaling occurs
Leptin and Leptin Receptor mainly through STAT3.16 Ob-Rb binding to JAK2 leads to
Leptin is primarily secreted by adipocytes and circulates at a JAK2 autophosphorylation and the phosphorylation of
level of 5 to 15 ng/mL in lean subjects.21 Its expression is Tyr985, Tyr1077, and Tyr1138 on Ob-Rb.32,34,36 –38 Phos-
increased by overfeeding, insulin, glucocorticoids, endotoxin, phorylation of Tyr1138 recruits STAT proteins to the Ob-Rb/
and cytokines and is decreased by fasting, testosterone, JAK2 complex. Tyrosine phosphorylated STAT3 molecules
Yang and Barouch Leptin Signaling and Obesity 547

dimerize and translocate to the nucleus to activate transcrip- JNK was unaltered in cultured adult cardiomyocytes from
tion of target genes, which includes the gene for a member of C57BL/6, ob/ob, and db/db mouse strains with 15-minute
the suppressors of the cytokine signaling family leptin treatment.46 The effects of long-term leptin treatment
(SOCS3).16,36,39 SOCS3 binding to Tyr985 and other sites on JNK activity in the heart remain to be investigated.
within the Ob-Rb/JAK2 complex mediates negative feedback Nuclear factor ␬B has been proposed as an attractive down-
on leptin signaling.40,41 JAK2 phosphorylation of Tyr985 also stream target for p38 and JNK MAPK pathways because this
consequently leads to phosphorylation of the src homology 2 transcription factor is essential in the transcriptional regula-
(SH2) domain of the tyrosine phosphatase SHP-2 (src homol- tion of proinflammatory cytokines such as tumor necrosis
ogy 2-containing tyrosine phosphatase), which activates the factor (TNF)-␣ and interleukin (IL)-1␤.52 Additional leptin
extracellular signal-regulated kinase (ERK) signaling path- signaling pathways are difficult to consolidate because of
way.36 Overexpression of SHP-2 has been shown to blunt tissue specificity. For a comprehensive review of leptin
SOCS3-mediated inhibition, likely through competitive bind- signaling capabilities, see reviews by Fruhbeck52 or
ing to Tyr985.40 Sweeney.53
JAK2 autophosphorylation is independent of tyrosine
phosphorylation sites on Ob-Rb and has many subsequent Leptin Signaling in the CNS
effects. One of these effects is to phosphorylate insulin Hypothalamic Leptin Decreases Food Intake
receptor substrate proteins, which recruit the phosphatidyl- Most Ob-Rs in the CNS are located in the basomedial
inositol 3⬘-kinase (PI3K) to activate downstream signals.7,42 hypothalamus.54,55 In neurons that synthesize proopiomelano-
In the heart, the leptin-associated PI3K pathway, along with cortin and express Ob-Rb, leptin stimulates the synthesis of
ERK cascades, seems to be important in cardiomyocyte proopiomelanocortin, which is processed to produce ␣-mela-
proliferation and protecting cardiac tissue from ischemia/ nocyte–stimulating hormone and activate downstream
reperfusion injury.24,43 melanocortin-3 and -4 receptors to decrease appetite.56 Leptin
also inhibits the appetite-stimulating hormone neuropeptide
Mitogen-Activated Protein Kinase Y in the arcuate nucleus, and causes inhibition of agouti-
Phosphorylation of Tyr985 on Ob-Rb leads to SHP-2 and related peptide that restrain melanocortin-3 and -4 receptor
Grb-2 activation of ERK1/2 of the MAPK family.36 ERK1/2 signaling.55,56
activation also occurs via a pathway independent of Tyr985. Ob-Rb–stimulated JAK2/STAT3 signaling is crucial in
In this case, JAK2 associates with the SH2 domain- leptin control of feeding and energy expenditure. Although
containing SHP-2.36,44 Therefore, both Ob-Ra and Ob-Rb can short forms of the leptin receptor may be capable of activating
Downloaded from http://ahajournals.org by on April 15, 2022

activate MAPK.35,36 Shc, another SH-2– containing protein JAK, insulin receptor substrate, and MAPK proteins, only
that is able to associate with Grb-2, has also been shown to Ob-Rb is capable of activating STAT3.35,57,58 A homologous
phosphorylate tyrosine after leptin treatment.45 Leptin- replacement of Ob-Rb by a receptor mutant for Tyr1138
induced phosphorylation of STAT3 and ERK1/2 have been established that STAT3 is indispensable for the regulation of
observed in isolated adult C57BL/6 mouse cardiomyocytes, expression of proopiomelanocortin and neuropeptide Y in the
with maximal activation at 15 minutes after treatment. Four- hypothalamus.16
week leptin treatment also elevated STAT3 and ERK1/2
phosphorylation and abundance in cardiac tissue from leptin- Mechanisms of Central Leptin Resistance
deficient ob/ob mice but not from Ob-Rb– deficient db/db One theory of leptin resistance involves intracellular signal-
mice.46 Because Ob-Ra is intact in db/db mice and the distal ing disruption. STAT3 and protein tyrosine phosphatase 1B
portion of Ob-Rb is not essential for MAPK signaling,35 are 2 molecules known to attenuate leptin signaling.40,59 – 63
ERK1/2 phosphorylation would be expected to increase even Hypothalamic protein tyrosine phosphatase 1B levels are not
if STAT3 phosphorylation were unaltered. Therefore, the known to be altered in obesity, although protein tyrosine
results suggest that either Ob-Rb is the predominant signaling phosphatase 1B–null animals develop increased insulin sen-
receptor in the mouse heart or that JAK2-induced ERK1/2 sitivity and have a lean phenotype.64 As previously discussed,
phosphorylation does not occurs at a significant level in leptin can induce its own negative feedback through STAT3-
cardiomyocytes. ERK1/2 activation leads to the expression of induced SOCS3 accumulation during prolonged Ob-Rb stim-
target genes, such as c-fos and egr-1, which participate in cell ulation,36,40,61 which can occur via Tyr985 or additional sites
proliferation and differentiation. in the Ob-Rb/JAK2 complex.40,41 At low concentrations of
Phosphorylation of p38 MAPK in response to leptin has leptin, incremental changes in leptin would be almost fully
also been demonstrated. The ␣ and ␤ isoforms of p38 MAPK translated into increased Ob-Rb signaling, whereas, at high
are broadly distributed, including at relatively high levels in levels, accumulated SOCS3 would counter most of the
the heart.47 Although the specific mechanism leading to increase in Ob-Rb signaling.39,65
leptin-induced p38 MAPK activation has not been elucidated, Another possible mechanism of central leptin resistance
it is associated with the onset of hypertrophy and pro- stems from the dependency of leptin on saturable transport
grammed cell death in both rat vascular smooth muscle cells across the vascular barrier and, probably to a lesser extent,
(VSMCs) and cardiomyocytes.24,48,49 across the choroid plexus to reach the arcuate nucleus.66 The
As with other cytokines, leptin has the ability to activate activity of this blood– brain barrier transport system seems to
the stress-activated protein kinase c-Jun N-terminal kinase decrease in diet-induced obese (DIO) rodents,67 resulting in
(JNK).50,51 However, we observed that phosphorylation of failure of circulating leptin to reach its targets in the brain.
548 Circulation Research September 14, 2007

Figure 2. Systemic leptin function. Chronic hyperleptinemia impairs the centrally mediated metabolic actions of the hormone, alyhough
its activation of sympathetic outflow is preserved. Selective central leptin resistance results in obesity and adverse effects on the car-
diovascular system including hypertension, atherosclerosis, and LVH. Although leptin can protect against ectopic lipid deposition in
nonadipose tissue, whether this effect is abolished because of (selective) peripheral leptin resistance requires further examination.
Downloaded from http://ahajournals.org by on April 15, 2022

Increasing evidence suggests that leptin signaling is prefer- logic model of human obesity. In C57BL/6J mice fed a
entially reduced in the arcuate nucleus of the hypothalamus 10-week high-fat diet, intraperitoneal leptin administration
but not in other regions, such as the ventromedial, dorsome- failed to decrease appetite and body weight, but increased
dial, and/or premammillary nucleus of the hypothalamus that renal sympathetic nerve activity. Sympathetic nerve activity
also express Ob-Rs.68 in brown adipose tissue and in hindlimb did not increase on
leptin administration, indicating region-specific preservation
Leptin Increases Sympathetic Outflow of leptin sympathoactivation that serves to protect its circu-
In addition to reducing appetite and controlling weight gain, latory effects.73
leptin centrally activates the sympathetic nervous system. It The sympathoactivation effect is completely abolished by
significantly increases plasma norepinephrine and epineph- selective destruction of the arcuate nucleus.74 As the arcuate
rine concentrations via the ventromedial hypothalamus.69 nucleus is required for both metabolic and sympathetic
Whereas chronic leptin overstimulation in the hypothalamus effects of leptin, these results seem to suggest that leptin
decreases the ability of leptin to regulate appetite, its sympa- resistance occurs mainly through intracellular signaling dis-
thetic excitatory effects are maintained as increased arterial ruption. Under resting conditions, it is estimated that only 5%
pressure and renal sympathetic nerve activity are presented in to 25% of Ob-R isoforms are located at the cell surface.75 The
obesity (Figure 2).70 This observation suggests that central ligand–receptor complex internalizes, and studies have shown
leptin resistance is selective. that leptin internalization was greater for the Ob-Rb iso-
The concept of selective resistance is suggested by a form.75,76 Preferential downregulation of Ob-Rb in response
comparison between ob/ob and Agouti yellow obese mice. to prolonged leptin exposure may be important in regulating
Lower arterial pressure is observed in ob/ob mice, which is different tissue sensitivity to leptin and another cause for
increased by leptin reconstitution despite the accompanying selective leptin resistance.
weight loss.71 In contrast, Agouti yellow obese mice develop Leptin stimulation of adrenergic overdrive can lead to
obesity resulting from ubiquitous overexpression of agouti numerous adverse effects on the cardiovascular system. Both
protein, which blocks MC4R rather than directly affecting in vitro and in vivo studies have demonstrated adrenergic
leptin. They have elevated arterial pressure similarly to DIO influences on the growth of cardiomyocytes.77,78 Patients with
mice, despite the fact that they have milder obesity than ob/ob the metabolic syndrome have increased sympathetic activity,
mice.71,72 This preservation of sympathoactivation effects of hypertension, and higher occurrences of left ventricular
leptin despite disruption of its weight control effects has been hypertrophy (LVH).79,80 Sympathetic influences also modu-
confirmed in DIO mice, which is considered a more physio- lates the elastic properties of large and medium-size arteries
Yang and Barouch Leptin Signaling and Obesity 549

Summary of Leptin Signaling in the Cardiovascular System


Leptin Action Pathways Involved
Development of hypertension Increased catecholamine release, preserved renal
sympathetic nerve activity increase, NO
Development of atherosclerosis Endothelial cell NO and ROS production, VSMC
proliferation and hypertrophy, platelet
aggregation, proinflammatory cytokine production
Impaired cardiomyocyte contractility JAK/STAT, NO-cGMP, G protein–coupled
receptor, adenylate cyclase, ET-1, NAPDH, ROS
Liporegulation Increased FA oxidation
Increase in size of cardiomyocyte ET-1, ROS
Deficiency leads to increased rate of p38, caspase-3, DNA damage, Ang II
cardiomyocyte apoptosis
Enhanced cardiomyocyte mitosis and ERK1/2, p38, PI3K, peroxisome
proliferation proliferator-activated receptor-␣
Protection from ischemia/reperfusion injury PI3K, ERK1/2, NO, ROS

and promote endothelial dysfunction, contributing to the arginine methyl ester did not unmask any pressor effect.95 At
development of vascular structural alterations and the occur- a concentration sufficient to increase sympathetic nerve
rence of atherosclerotic lesions.81 outflow, leptin did not change arterial pressure or blood flow
in mesenteric, lower aortic, and renal arteries.96 Blockade of
Leptin and the Cardiovascular System NO synthesis increased heart rate and renal vascular and
Hyperleptinemia is associated with obesity-related hyperten- glomerular response but did not substantially augment the
sion and chronic congestive heart failure (HF) in humans, and pressor response to leptin,97 indicating a negligible role of
vascular endothelial and myocardial dysfunction in animal leptin-stimulated NO production on blood pressure in vivo.
models.82– 85 The role of leptin and leptin resistance in the Leptin has been shown to increases the release of ET-1, a
pathogenesis of LVH and HF in obesity remains controver- vasoconstrictor secreted primarily by endothelial cells but
sial. The Table provides a summary of the leptin signaling
Downloaded from http://ahajournals.org by on April 15, 2022

also by macrophages, fibroblasts, and cardiomyocytes, pos-


pathways involved in the cardiovascular system. sibly countering the effects of NO.98

Vascular Effects of Leptin Development of Atherosclerosis


The proatherogenic action of leptin is likely attributable to a
Development of Hypertension
Both intravenous infusion and intracerebroventricular admin- combination of its effects on various cell types. In endothelial
istration of leptin lead to increased arterial pressure and heart cells, leptin induces oxidative stress, increases production of
rate in rodents.86,87 Blockade of the adrenergic system inhibits monocyte chemoattractant protein-1 and ET-1 and potentiates
the pressor response to leptin.88 In addition to increased proliferation.50,89,98,99 In VSMCs, leptin promotes migration,
sympathetic activity in hyperleptinemia, other mechanisms proliferation, and hypertrophy.100 –102 Leptin also promotes
may also contribute to the development of obesity-related calcification of cells of the vascular wall and facilitates
hypertension. Leptin has been shown to increase the genera- thrombosis by increasing platelet aggregation.103,104
tion of reactive oxygen species (ROS) in endothelial cells50,89 Hyperleptinemia has been associated with coronary athero-
and to stimulate secretion of proinflammatory cytokines such sclerosis in type 2 diabetes, and this association has been
as TNF-␣ and IL-6, both of which are promoters of hyper- shown to be independent of insulin resistance.105 In apoli-
tension and atherosclerosis.90 poprotein E– deficient (apoE⫺/⫺) mice that are atherosclerosis
While stimulating blood flow via increased sympathetic prone, recombinant leptin treatment further promoted athero-
output, leptin has also been shown to have direct vasodilatory sclerosis and thrombosis, despite its metabolic benefits.106
effects. In both intact rodents and in endothelial cells, leptin This result indicates that elevation of leptin concentration in
induces NO production.83,91 Leptin administration in anesthe- obesity increases the risk for atherosclerotic damage.
tized rats causes a dose-dependent increase in NO metabolite Paradoxically, leptin resistance is also proatherogenic.
concentrations, and inhibition of NO synthesis increases ApoE⫺/⫺ mice lacking Ob-Rb (apoE⫺/⫺ db/db) are character-
arterial pressure.91 In vitro studies have shown that leptin ized by a 5-fold higher area of spontaneous atherosclerotic
evokes an endothelium-dependent relaxation of arterial lesions in the aorta than apoE⫺/⫺ with intact Ob-R.107 Unlike
rings.92,93 In endothelial cells, leptin activates a PI3K- apoE⫺/⫺ mice, apoE⫺/⫺ db/db mice are also obese and insulin
independent Akt-endothelial NO synthase (eNOS) phosphor- resistant. Taken together, these data suggest either that a
ylation pathway to increase NO production, which can be supraphysiological level of exogenous leptin self-induces
abolished by erbstatin A, a Ca2⫹-independent tyrosine kinase resistance that is harmful or that the atherogenic effect seen in
inhibitor.94 However, the vasodilator action of leptin was not apoE⫺/⫺ db/db is an effect more relevant to lipid profile
found in conscious rats and the NOS inhibitor NG-nitro-L- change, which is absent in apoE⫺/⫺ mice.
550 Circulation Research September 14, 2007

Figure 3. Leptin and myocardial contractility. Leptin directly depresses cardiomyocyte contractility. The signaling pathways implicated
Downloaded from http://ahajournals.org by on April 15, 2022

in this process include the NO-cGMP pathway and pathways that lead to increased ROS production. Changes that occur in a chronic
leptin-deficient state are also illustrated. TG indicates triacylglycerol; iNOS, inducible NO synthase; nNOS, neuronal NO synthase; AC,
adenylate cyclase; PKA, protein kinase A; XOR, xanthine oxidoreductase; SR, sarcoplasmic reticulum.

Leptin Attenuates Cardiomyocyte Contractility eNOS inhibits hypertrophy in the remote myocardium and
Possible Mechanisms Leading to Increased preserved cardiac function after myocardial infarction, pos-
NO Production sibly through attenuation of ␤-adrenergic–stimulated com-
Similar to its effects in endothelial cells, acute leptin infusion pensatory hypertrophy.112 Neuronal NO synthase and eNOS
in isolated rat ventricular myocytes increases NO activity, independently contribute to the development of cardiac hy-
which leads to attenuated cardiac contractility (Figure 3).108 pertrophy, leading to marked age-related concentric hyper-
Intracellular Ca2⫹ transients were lowered and NO production trophic remodeling in double-knockout mice lacking both
were increased with leptin. These effects were blocked by the neuronal NO synthase and eNOS.113,114 Understanding the
NO inhibitor NG-nitro-L-arginine methyl ester108 and by JAK2 leptin crosstalk with the ␤-adrenergic signaling system in
or p38 MAPK inhibitors AG-490 and SB203580.109 cardiomyocytes would provide significant insight into the
The intermediate steps by which leptin signaling leads to understanding of myocardial dysfunction in obesity.
increase in NO production and the specific NOS isoforms that Whereas leptin-induced NO increase directly depresses
mediate the effects of leptin have not been fully elucidated. In cardiomyocyte contractility, systemic actions such as in-
rat VSMCs, leptin inhibits the contractile response induced creased sympathetic modulation may indirectly stimulate
by Ang II through increased NO production. The upregula- contractility. Just as leptin-stimulated NO production in
tion of inducible NO synthase through mechanisms involving endothelial cells may have a negligible role in blood pressure
JAK2/STAT3 and PI3K/Akt pathways is responsible for the in vivo, cardiac contractile depression may not manifest
increase of NO bioavailability in VSMCs.110 under normal physiologic conditions. However, the depres-
Elucidation of the NOS isoform(s) responsible for the sant effect may become important when considered in con-
actions of leptin in the heart would lead to a better under- juncture with alterations occurring in obese states.
standing of its role in myocardial contractility and hypertro- Leptin Deficiency Leads to Decreased Responsiveness to
phy responses. We have shown that spatial confinement of ␤-Adrenergic Stimulation
different constitutive NOS isoforms within separate subcel- In 10 week-old ob/ob isolated myocytes, attenuated sarco-
lular compartments of the cardiac myocyte allows NO signals mere shortening and calcium transients and depressed sarco-
to have independent, and even opposite, effects on cardiac plasmic reticulum Ca2⫹ stores were seen in response to
phenotype and contractile response.111 Overexpression of isoproterenol stimulation of the ␤-adrenergic receptor or to
Yang and Barouch Leptin Signaling and Obesity 551

post–receptor level stimulation with forskolin and dibutyryl– inotropic effects, thereby offsetting the cGMP-dependent
cAMP. Leptin replenishment in ob/ob mice restored each of reduction in contractility.120 Peroxynitrite has shown both
these abnormalities toward normal without affecting gross negative and positive inotropy in isolated cardiomyo-
(wall thickness) or microscopic (cell size) measures of cytes,123,124 although it is generally accepted to trigger apo-
cardiac architecture. Decreased G␣s (52 kDa), increased ptosis in cardiomyocytes in vitro and in vivo, possibly
sarcoplasmic reticulum Ca2⫹-ATPase, and depressed phos- through a pathway involving caspase-3 activation and the
phorylated phospholamban abundance were detected in ob/ob cleavage of poly(ADP-ribose) polymerase.125
mice. In addition, protein kinase A activity in ob/ob mice was Another recent study proposes that ET-1 is upstream of
depressed at baseline and corrected toward wild-type (WT) NADPH oxidase in leptin-induced myocardial contractile
level with leptin repletion.46 In the H9c2 cardiac cell line, response.126 There are at least 2 cardiac ET-1 receptors, ETA
30-minute leptin treatment increased basal and catechol- and ETB. Both are known to mediate cardiomyocyte inotropic
amine-stimulated adenylate cyclase activity, whereas 18-hour response, and ETA receptors also affect hypertrophy.127 Lep-
treatment was associated with a reduced adenylate cyclase tin administration to rat neonatal cardiomyocytes induced
activity and a different responsiveness to isoproterenol and intracellular O2. generation and upregulated protein expres-
norepinephrine stimulation, likely attributable to differential sion of p67phox and p47phox subunits of NAPDH, the effect
activation of G␣s. Adenylate cyclase, G␣s (52 kDa), G␣i, of which is attenuated by ETA and ETB receptor antagonists
p21-ras, and phosphorylated ERK1/2 expressions were in- and apocynin, suggesting that the ET-1 receptors are likely
creased with short-term leptin treatment and decreased at 18 upstream of NADPH oxidase in leptin-induced cardiac con-
hours, whereas G␣s (45 kDa) continued to increase at 18 tractile response.126
hours. Receptor level leptin resistance is conceivable in
myocytes, as Ob-R expression is seen to decrease at 18-hour Additional Mechanisms Affecting Contractility
Obesity is a lipotoxic disease featuring overtly elevated
leptin treatment.115 Taken together, leptin deficiency or resis-
ceramide levels (see section below, Leptin Shifts Myocardial
tance leads to decreased ␤-adrenergic response, whereas
Metabolism Toward Fatty Acid Utilization). The de novo
moderate leptin stimulation can improve the contractile
ceramide pathway has been postulated to be key to the
response.
lipoapoptosis of pancreatic ␤ cells and cardiomyocytes in
Mechanisms Involving ROS obese individuals.128,129 The ability of ceramide to amplify
Mitochondrial formation of ROS is enhanced in obesity. leptin-induced depression of contractility in adult rat left
Xanthine oxidoreductase and nicotinamide adenine dinucle- ventricular myocytes was recently demonstrated.130 Although
Downloaded from http://ahajournals.org by on April 15, 2022

otide phosphate (NADPH) oxidase are 2 main sources of ceramide alone did not elicit any effect on cell mechanics and
superoxide (O2. ) production in the heart. O2. is capable of intracellular Ca2⫹ transients, it sensitized leptin-induced ef-
generating a large family of ROS by interacting with other fects on myocyte shortening and intracellular Ca2⫹ transients.
molecular compounds. In ob/ob hearts, impaired cardiac In vivo obese concentrations of plasma leptin lie in the low
contractile function is accompanied by elevated oxidative nanomolar range, which is seemingly disconnected from the
stress, lipid peroxidation, protein carbonyl formation, redis- high in vitro leptin concentration (⬎10 nmol/L) needed to
tribution of myosin heavy chain isozymes from myosin heavy affect cardiac contractile function. The observation that
chain-␣ to -␤, and oxidative modification of SERCA2a.116 ceramide may augment the cardiac depressive effect of leptin
Neuronal NO synthase constrains xanthine oxidoreductase provides an additional explanation for hyperleptinemia-
activity.117,118 Reduced neuronal NO synthase expression is associated cardiac dysfunction in obesity.
observed in 2- to 6-month-old ob/ob mice, which leads to Elevated adipose mass in obesity also increases the secre-
increased xanthine oxidoreductase production of O2. , thereby tion of other proinflammatory factors, including TNF-␣, IL-6,
causing an imbalance between the production of ROS and and Ang II. TNF-␣ and leptin both depress contractility in
reactive nitrogen species.119 This nitroso–redox imbalance adult rat ventricular myocytes, although no additive response
may be partially responsible for the myocardial dysfunction by the 2 proinflammatory factors was observed. Inhibitory
seen in ob/ob mice. Activation of NADPH oxidase is also effects were abolished by NG-monomethyl-L-arginine in both
seen in the ob/ob heart.116 Treatment with apocynin, a cases and in the case of combined exposure.131 Thus, the
NADPH oxidase inhibitor, reversed cardiac contractile dys- inhibitory effect on cardiac contraction by TNF-␣ and leptin
function in ob/ob myocytes but failed to reserve SERCA2a may mask each other and share a common mechanism
oxidative modification.116 8-Bromo-cGMP, a membrane- dependent on NO.131
permeable cGMP analog, induced a greater negative effect in It is interesting, however, that hypertension seems to
ob/ob than lean C57BL/6J mice. However, the effect of attenuate leptin-induced cardiomyocyte contractile depres-
adding a NO donor was similar in the obese and lean models, sion. Isolated rat ventricular myocytes from spontaneously
indicating that some cGMP-independent effect of NO pre- hypertensive rats (SHR) displayed decreased leptin-induced
vents the enhanced negative cGMP effects in ob/ob cardio- depression of myocyte shortening and intracellular Ca2⫹
myocytes.120 NAPDH-mediated reduction in NO bioavail- transients, as well as blunted leptin-induced NOS activity
ability could explain the failure of NO donor to elicit further compared with the normotensive control mice. Additionally,
negative inotropic response in obese models.121,122 Addition- treatment of SHR myocytes with JAK or p38 inhibitor led to
ally, the interaction between NO and ROS produces per- further inhibition of myocyte shortening by leptin instead of
oxynitrite, which can nitrosylate proteins and exert positive abolishing such effects.109 The altered signal transduction of
552 Circulation Research September 14, 2007

JAK/STAT and p38 pathways to attenuate leptin-induced transcription through MAPK and nuclear factor ␬B and are
cardiomyocyte dysfunction possibly serves as a compensa- implicated in the development of cardiomyocyte hypertro-
tory mechanism to prevent further impairment of ventricular phy.139 It seems paradoxical that enhanced expression of ␤
function in sustained hypertension or hyperleptinemia.109 oxidation enzymes such as long-chain acyl-CoA dehydroge-
nase and carnitine palmitoyltransferase-1 are seen in ob/ob
Leptin Shifts Myocardial Metabolism Toward mice,140 as leptin-deficient hearts are also characterized by
Fatty Acid Utilization increased FA utilization.141 This is likely attributable to
Accumulating evidence suggests that leptin regulates energy long-term adaptation to the changes in the lipid profile in
homeostasis through direct actions on peripheral lipid and
these animals. Genes for lipoprotein lipase, which generates
glucose metabolism.132 Fatty acid (FA) oxidation produces
free FAs, and FA transport proteins are seen at an greatly
the major source of ATP to sustain contractile function of the
elevated level in ob/ob myocytes, which causes accumulation
heart. AMP-activated protein kinase (AMPK) has a key role
of free FAs that promote ceramide, inducible NO synthase,
as a fuel gauge in the heart and regulates cardiac FA oxidation
and NO production in the cardiomyocyte.140 When increased
by phosphorylation and inhibition of acetyl– coenzyme A
(CoA) carboxylase, which then lowers malonyl-CoA levels FA delivery and hyperinsulinemia are imposed in ob/ob
and stimulates carnitine palmitoyltransferase-1–induced FA hearts, myocardial function is maintained, albeit with de-
oxidation.133 AMPK activation is also known to reduce FA creased efficiency, whereas WT hearts are unable to adapt
incorporation into triacylglycerol.134 acutely.141
Examination of leptin effects on cardiac FA oxidation Increased FA uptake accompanied by decreased oxidation
confirmed that leptin infusion increases FA oxidation and leads to lipotoxicity in long-term leptin-treated cardiomyo-
triacylglycerol lipolysis in isolated working rat hearts, al- cytes. The antisteatotic effects of leptin in early obesity might
though this effect was independent of changes in the AMPK/ be lost with the progression of leptin resistance occurring in
acetyl-CoA carboxylase/malonyl/CoA axis. Neither did leptin late stage obesity, leading to lipotoxicity that promotes
affect glucose oxidation rates.135 Myocardial oxygen con- cardiomyocyte contractile dysfunction and apoptosis129 or
sumption was increased, possibly because of increased mito- acts in a maladaptive fashion because of increased ceramide
chondrial uncoupling protein activity.135 In DIO mice, plasma synthesis.
leptin concentration increase was associated with an induc-
tion of uncoupling protein 2 and an increase in phosphoryla- Leptin Effects on Cardiac Hypertrophy
tion of AMPK that led to increased FA oxidation.136 At this and Remodeling
Downloaded from http://ahajournals.org by on April 15, 2022

stage, leptin signaling in the heart as assessed by STAT3 Postmortem clinical studies of obesity-associated hypertro-
phosphorylation remained unaltered from WT, suggesting phy have demonstrated excessive heart weight but low
that the shift to FA oxidation is at least mediated, in part, by heart/body weight ratios.142 Fatty infiltration of the myocar-
leptin. Increased FA oxidation may confer antisteatosis pro- dium, presence of metabolic inclusions, excessive fibrosis, or
tection to the myocardium during hyperleptinemia in early increases in extracellular matrix do not appear to play a direct
stages of obesity. This is consistent with the observation that role in augmenting wall thickness in obesity-related hyper-
DIO rodents exhibit minimal rise in myocardial triacylglyc- trophy.143 The findings on endomyocardial biopsy in the
erol content, whereas ob/ob, db/db, and fa/fa animals show majority of obese patients with cardiomyopathy appear to be
significantly greater accumulation.137 Leptin deficiency per- primarily myocyte hypertrophy.144 Hyperleptinemia is ob-
turbs liporegulation in peripheral organs and results in lipo- served in patients with LVH.145 Leptin effects on cardiomyo-
toxicity, lipoapoptosis, and generalized steatosis, manifesting cyte hypertrophy is not entirely clear. We had previously
in clinical conditions such as nonalcoholic steatohepatitis, shown leptin repletion to be antihypertrophic in ob/ob hearts.
type 2 diabetes, and lipotoxic cardiomyopathy.128 Lipotoxic
Four- to 6-week leptin infusion reduced weight and reversed
cardiomyopathy has been identified in fa/fa rats and has been
LVH, whereas caloric restriction reduced weight but did not
transgenically induced in acyl–CoA synthase transgenic
effectively reduce wall thickness and myocyte size.113 Recon-
mice.138 Overexpression of acyl–CoA synthase, which actives
stitution to normal levels may provide the antisteatotic effect
and esterifies FAs, led to severe cardiomyopathy that was
of leptin that can reverse lipotoxic effects of FA deposition in
attenuated by injection of recombinant adenovirus containing
the leptin cDNA.138 the myocardium. On the other hand, leptin has been shown to
Increased de novo lipogenesis and decreased compensatory induce hypertrophy in a concentration-dependent manner in
oxidation of FAs are two possible mechanisms for intracel- cultured neonatal rat ventricular myocytes.48,146 Fasting
lular accumulation of lipids in nonadipose tissue. Enzymes plasma leptin levels are associated with increased myocardial
involved in FA oxidation such as acyl-CoA oxidase and wall thickness in hypertensive humans, although this may be
carnitine palmitoyltransferase-1 are observed at attenuated related to leptin resistance.147 The different types of cardio-
levels in fa/fa rats. Hydrolysis of the increased stores of myocytes used in experiments could also contribute to the
triacylglycerol to fatty acyl-CoA in fa/fa rats also increased observed paradoxical effects of leptin on cardiomyocyte
the substrate for de novo ceramide synthesis and activated the hypertrophy. Direct effects of leptin on cardiomyocyte hy-
expression of inducible NO synthase in the myocardium, pertrophy, apoptosis, proliferation, and extracellular matrix
which led to acceleration of apoptosis.129 Ceramide also remodeling could all contribute to maladaptive hypertrophy
activated protein kinase C isoforms, which can induce gene and HF in obesity.
Yang and Barouch Leptin Signaling and Obesity 553

Cardiomyocyte Hypertrophy nently involved in the upregulation of the renin–Ang system,


Leptin-induced neonatal cardiomyocyte hypertrophy occurs and Ang II–treated adult rat cardiomyocytes in culture exhibit
through a mechanism involving ET-1 and ROS generation.146 increased apoptosis. The somewhat paradoxical combination
ET-1 has been shown to increase cardiomyocyte surface area of antiapoptotic roles of leptin and proapoptotic actions of
without increasing cell proliferation.148 In this study, blocking JAK2 merits further investigation. Leptin acutely increases
the ETA receptor with the selective inhibitor ABT-627 par- phosphorylation of ERK1/2 and p38 MAPK in rat neonatal
tially but significantly reduced leptin-induced hypertrophy.146 cardiomyocytes, but leptin-induced p38 activation in rat
An interdependence of ET-1 and leptin signaling has been neonatal cardiomyocytes sustains for a longer period than
proposed in the progression of myocardial dysfunction and ERK1/2 activation, suggesting that the downstream transcrip-
hypertrophy, suggesting that leptin may cause chronic oxida- tion factors of p38 may be involved in the long-term
tive stress and inflammation in the myocardium, similar to maladaptive cardiac remodeling in obese HF patients.48
other agents such as TNF-␣, norepinephrine, and Ang II, all
of which induce hypertrophy via ROS upregulation.149,150 Mitosis and Proliferation
Leptin treatment at a level similar to plasma concentration in
Leptin may indeed have an autocrine role in mediating
obese individuals increased proliferation of both HL-1 car-
ET-1 and Ang II–induced cardiomyocyte hypertrophy. Treat-
diac muscle cells and human pediatric ventricular myocytes.43
ment of neonatal rat myocytes for 24 hours with leptin, Ang
The proliferation was accompanied by increased DNA syn-
II, or ET-1 significantly increased cell area by 37%, 36%, and
thesis associated with increased ERK1/2 phosphorylation and
35%, respectively. In contrast to the study mentioned above
increased association of the p85 regulatory subunit of PI3K
by Xu et al,146 Rajapurohitam et al26 have shown that
with phosphotyrosine immunoprecipitates.43 ERK1/2 inhibi-
blocking the ETA receptor did not prevent leptin-induced
tion significantly attenuated the leptin-induced proliferative
hypertrophy, neither did blocking the Ang receptor. Leptin
activity and DNA replication in HL-1 and pediatric human
blockade attenuated the hypertrophic responses generated by
cardiomyocytes43 but failed to decrease [3H]-leucine incorpo-
all 3 agents. Ang II and ET-1 significantly increased leptin
ration in neonatal rat cardiomyocytes treated with leptin.48
levels in the culture medium and increased the gene expres-
Other pathways likely involved in leptin-induced hypertro-
sion of both Ob-Ra and Ob-Rb. Additionally, Ang II and
phy include the activation of adenylate cyclase,115 peroxi-
ET-1 increased phosphorylation of ERK1/2, p38, JNK, and some proliferator-activated receptor-␣,157 and the JAK/STAT
nuclear factor ␬B, but the ability of leptin blockade to pathway associated with hsp56 and Ang II.109,158 Leptin
attenuate hypertrophic responses was generally dissociated signaling is capable of activating other traditional pathways
from these effects.26 The discrepancy between the studies by for the development of hypertrophy, such as PI3K and protein
Downloaded from http://ahajournals.org by on April 15, 2022

Xu et al and Rajapurohitam et al in terms of blocking kinase C.159 Whether these pathways are activated in cardio-
leptin-induced hypertrophic response with ETA receptors myocytes in response to leptin and the specific isoforms
antagonists (ABT-627 versus BQ123) cautions against the involved mandate further research.
interpretation of results using pharmacological agents, as the
precise mechanisms of action is unclear in many cases. Extracellular Matrix Remodeling
Another study demonstrates that in 9-week-old mice fed a Leptin has been shown to increase the expression of matrix
high-fat diet, serum and myocardial ET-1, myocardial leptin, metalloproteinase-2, and to increase collagen type III and IV
and Ob-R mRNA are all elevated, whereas in ob/ob mice, mRNA and decrease collagen type I mRNA without affecting
both serum and myocardial ET-1 levels are not higher than total collagen synthesis in human pediatric cardiomyo-
WT mice, confirming a direct role of leptin in mediating cytes.160 This suggests that leptin selectively regulates differ-
increased myocardial ET-1 signaling.151 Simvastatin, a cho- ent forms of collagen although further studies are required to
lesterol-lowering drug decreases leptin-induced ROS- validate the regulation of collagen synthesis by leptin and to
mediated hypertrophy in rat neonatal cardiomyocytes.152 confirm these effects on cardiac remodeling in obesity.

Apoptosis Protection in Ischemia/Reperfusion Injury


One of the causes of HF is cardiomyocyte apoptosis and Timely reperfusion is necessary to salvage myocardium from
necrosis.153 We recently found that leptin deficiency or acute infarction, but reperfusion usually induces additional
resistance results in increased cardiomyocyte apoptosis, as injury. A recent report shows that exogenous leptin given at
assessed by TUNEL staining and caspase-3 levels.154 Aged early reperfusion in an isolated mouse heart model reduces
ob/ob and db/db mice showed increased DNA damage infarct size.24 This cardioprotective action of leptin is asso-
compared with old WT mice. Leptin reconstitution ob/ob ciated with activation of the reperfusion injury salvage kinase
animals reduced the rate of apoptosis, although not to WT pathway that includes PI3K/Akt and ERK1/2, ultimately
levels.154 This is consistent with earlier work demonstrating leading to the inhibition of mitochondrial permeability tran-
increased apoptosis in islet cells and cardiomyocytes in fa/fa sition pore opening.161 Infarct size in C57BL/6J mouse hearts
rats.129,155 These results suggest that leptin signaling is perfused with leptin was about half that of hearts perfused
necessary to maintain normal low levels of cell death without leptin. In a rat model, leptin and Ob-Ra expressions
and that leptin provides protection against lipotoxicity- were locally upregulated in scarred tissue following reperfu-
induced apoptosis. sion, whereas Ob-Rb expression was downregulated.24 PI3K
On the other hand, JAK2 has been suggested as a mediator or ERK1/2 inhibition diminished the cardioprotective ef-
of the apoptotic response in cardiomyocytes.156 It is promi- fect.24 Interestingly, leptin did not increase phosphorylation
554 Circulation Research September 14, 2007

of Akt or its downstream targets such as eNOS. Additionally, coronary dilation, increased endothelium-derived hyperpolar-
there was increased phosphorylation of p38 MAPK and izing factor, nor changes in coronary Ob-R mRNA levels.166
reduced abundance and phosphorylation of STAT3 and A recent hypothesis relevant to both central and peripheral
AMPK.24 Leptin-stimulated ROS production and NO synthe- leptin resistance involves leptin interaction with circulating
sis has been shown also to protect against ischemia reperfu- factors in the blood.167 Five serum leptin–interacting proteins
sion injury in the gut and kidney.162,163 Clinically, it is have been isolated, one of which is C-reactive protein. It
interesting to note that patients with a higher body mass index directly inhibits the binding of leptin to Ob-Rs and blocks its
have better outcomes following an acute coronary syndrome ability to signal in cultured cells. Infusion of human
or percutaneous coronary intervention.164,165 C-reactive protein into ob/ob mice blocked leptin treatment
effects on satiety and weight reduction. Physiological con-
Leptin Resistance in the centrations of leptin stimulate expression of C-reactive pro-
Cardiovascular System? tein in human primary hepatocytes,167 and human C-reactive
The relative contributions of central and peripheral leptin protein has been correlated with increased adiposity and
effects to disease pathogenesis are difficult to decipher. plasma leptin,168 suggesting an systemic self-induced nega-
Central leptin resistance disrupts hypothalamic control of tive feedback that may cause leptin resistance in the obese
energy homeostasis, which results in obesity and increased state.167
lipid production. This in turn may lead to ectopic lipid
deposition and lipotoxicity in peripheral organs. The attempt Leptin Antagonists
to separate the effects of this pathological process from the Leptin antagonists used in animal models have been shown to
physiological effects of leptin in the cardiovascular system block central leptin effects and increase appetite and food
has proven to be challenging, complicated by different intake.169,170 Three approaches have been employed to antag-
isoform signaling capabilities and possible resistance in the onize leptin activity: (1) binding free leptin in the circulation,
periphery. The question remains whether peripheral leptin (2) competitive Ob-R binding by mutants that do not cause
signaling activation, and (3) specific anti–Ob-R monoclonal
resistance occurs in the myocardium itself in obesity. Even
antibodies. An example of the first approach is a recombinant
though chronic leptin stimulation has been seen to decrease
human and mouse Ob-R/Fc chimeric glycoprotein.171,172 Only
Ob-R expression in various studies,75,115 DIO mice show
the latter 2 approaches have been employed in cardiac-related
increased Ob-R mRNA expression.151 On the other hand,
research. In neonatal rat ventricular cardiomyocytes, rat
Ob-Rb expression in ob/ob left ventricular homogenate is
L39A/D40A/F41A leptin mutein blocked hypertrophic ef-
lower than WT.126 However, mRNA expression does not
Downloaded from http://ahajournals.org by on April 15, 2022

fects and abolished increases in Ob-R gene expression elic-


necessarily correlate with receptor density at the membrane;
ited by leptin, Ang II, or ET-1.26 The hypertrophic effects of
therefore, these results are not conclusive in determining
leptin are also prevented by antibodies to Ob-Ra and Ob-
whether leptin resistance can occur at the myocyte receptor
Rb.26 Cardiac dysfunction did not develop in rats treated with
level. One recent study suggests that leptin resistance does
Ob-R antibodies after coronary artery ligation compared with
not occur in the myocardium in a model of early central
sham, indicating that blocking Ob-R can improve postinfarc-
resistance. Eight-week DIO C57BL/6 mice showed attenu- tion HF in rats.173 The recent proposal of nanobodies (a
ated leptin phosphorylation of STAT3 in hypothalamic tissue, unique form of antibodies that is characterized by a single
whereas no such attenuation was shown in whole-heart antigen-binding domain and generally does not cross the
homogenate.136 blood– brain barrier) may lead to an antagonist that could
Paradoxical results have been reported in almost all leptin- selectively inhibit peripheral activities of leptin.174 This form
related effects on the myocardium; that is, excessive exoge- of leptin antagonist might be clinically useful, as they can
nous leptin and leptin deficiency often lead to the same end. target peripheral adverse effect of leptin without inducing
Whether these effects occur through entirely different mech- central weight gain.
anisms, are mediated through differential regulation of Ob-R
isoforms, or are attributable to peripheral resistance requires Summary
further investigation. If peripheral myocardial resistance does Obesity leads to cardiac hypertrophy, ventricular dysfunction,
occur, these differences could be resolved if we consider reduced diastolic compliance, and hypertension, as well as
leptin deficiency and leptin resistance both to be states of type 2 diabetes and hyperlipidemia.175 The high risk of
dysfunctional downstream signaling. developing cardiovascular diseases in obesity has drawn
Interestingly, obesity-induced leptin resistance, although much effort to study the neurohormone effects of leptin on
not reported in the myocardium, has been shown to extend to cardiac function and remodeling. Hyperleptinemia, central
affect platelets and the vascular wall.166 Obese concentrations leptin resistance, and leptin deficiency are all associated with
of leptin significantly attenuate coronary vasodilation to impaired postreceptor leptin signaling and contractile re-
intracoronarily administered acetylcholine and significantly sponse. Short-term administration of leptin seems to have
attenuate relaxation in left circumflex coronary rings in beneficial effects on the myocardium, including antisteatotic
control dogs. These effects were not seen when the same actions, protection against ischemia/reperfusion injury, and
concentrations of leptin were administered to dogs fed a participation in compensatory myocyte hypertrophy. Interac-
high-fat diet, suggesting that leptin resistance does occur in tion with enhanced ROS production pathways in obesity, on
the vasculature. This resistance is not attributable to altered the other hand, can cause lipotoxicity and deleterious myo-
Yang and Barouch Leptin Signaling and Obesity 555

cardial effects such as cell death and maladaptive hypertro- 12. Considine RV, Sinha MK, Heiman ML, Kriauciunas A, Stephens TW,
phy. Perturbations of leptin signaling and other signal trans- Nyce MR, Ohannesian JP, Marco CC, McKee LJ, Bauer TL. Serum
immunoreactive-leptin concentrations in normal-weight and obese
duction pathways regulated by leptin in cardiomyocytes humans. N Engl J Med. 1996;334:292–295.
likely underlie the pathology of cardiomyocyte hypertrophy 13. Strobel A, Issad T, Camoin L, Ozata M, Strosberg AD. A leptin
in obesity. In particular, alterations in JAK/STAT, MAPK, missense mutation associated with hypogonadism and morbid obesity.
NO, and ␤-adrenergic pathways have been implicated in the Nat Genet. 1998;18:213–215.
14. Frederich RC, Hamann A, Anderson S, Lollmann B, Lowell BB, Flier
negative inotropic and hypertrophic responses. Additional JS. Leptin levels reflect body lipid content in mice: evidence for diet-
studies investigating the integrated effects of leptin on car- induced resistance to leptin action. Nat Med. 1995;1:1311–1314.
diomyocytes via SOCS3, PI3K/Akt, protein kinase C, and 15. Cohen P, Zhao C, Cai X, Montez JM, Rohani SC, Feinstein P, Mom-
baerts P, Friedman JM. Selective deletion of leptin receptor in neurons
other signaling pathways could provide a more comprehen-
leads to obesity. J Clin Invest. 2001;108:1113–1121.
sive understanding of leptin action on the cardiovascular 16. Bates SH, Myers MG. The role of leptin-⬎STAT3 signaling in neu-
system. roendocrine function: an integrative perspective. J Mol Med. 2004;82:
The unresolved debate about selective preservation of 12–20.
17. Mark AL, Correia ML, Rahmouni K, Haynes WG. Selective leptin
peripheral leptin signaling in the setting of hyperleptinemia resistance: a new concept in leptin physiology with cardiovascular
and central resistance complicates the interpretation of exper- implications. J Hypertens. 2002;20:1245–1250.
imental results involving the myocardium. Despite such 18. Lord GM, Matarese G, Howard JK, Baker RJ, Bloom SR, Lechler RI.
challenges, a picture is emerging in which the risk of obesity Leptin modulates the T-cell immune response and reverses starvation-
induced immunosuppression. Nature. 1998;394:897–901.
is not merely attributable to the physical burden of extra 19. Margetic S, Gazzola C, Pegg GG, Hill RA. Leptin: a review of its
weight but is, rather, a complex condition of hormonal peripheral actions and interactions. Int J Obes Relat Metab Disord.
dysregulation. Improved understanding of the actions of 2002;26:1407–1433.
leptin within the cardiovascular system will greatly improve 20. Rahmouni K, Haynes WG. Leptin and the cardiovascular system. Recent
Prog Horm Res. 2004;59:225–244.
our understanding of obesity-associated heart disease. 21. Sinha MK, Opentanova I, Ohannesian JP, Kolaczynski JW, Heiman ML,
Hale J, Becker GW, Bowsher RR, Stephens TW, Caro JF. Evidence of
Sources of Funding free and bound leptin in human circulation. Studies in lean and obese
This work was supported in part by the Donald W. Reynolds subjects and during short-term fasting. J Clin Invest. 1996;98:
Foundation, NIH grant K08-HL076220, the W.W. Smith Charitable 1277–1282.
Trust, and the Irvin Talles Endowed Fund for Cardiomyopathy 22. Coleman RA, Herrmann TS. Nutritional regulation of leptin in humans.
Research. Diabetologia. 1999;42:639 – 646.
23. Fried SK, Ricci MR, Russell CD, Laferrere B. Regulation of leptin
production in humans. J Nutr. 2000;130:3127S–3131S.
Disclosures
Downloaded from http://ahajournals.org by on April 15, 2022

24. Smith CC, Mocanu MM, Davidson SM, Wynne AM, Simpkin JC,
None. Yellon DM. Leptin, the obesity-associated hormone, exhibits direct
cardioprotective effects. Br J Pharmacol. 2006;149:5–13.
References 25. Matsui H, Motooka M, Koike H, Inoue M, Iwasaki T, Suzuki T,
1. Friedman JM. A war on obesity, not the obese. Science. 2003;299: Kurabayashi M, Yokoyama T. Ischemia/reperfusion in rat heart induces
856 – 858. leptin and leptin receptor gene expression. Life Sci. 2007;80:672– 680.
2. O’Rahilly S, Farooqi IS, Yeo GS, Challis BG. Minireview: Human 26. Rajapurohitam V, Javadov S, Purdham DM, Kirshenbaum LA,
obesity-lessons from monogenic disorders. Endocrinology. 2003;144: Karmazyn M. An autocrine role for leptin in mediating the cardiomyo-
3757–3764. cyte hypertrophic effects of angiotensin II and endothelin-1. J Mol Cell
3. Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Cardiol. 2006;41:265–274.
Positional cloning of the mouse obese gene and its human homologue. 27. Fei H, Okano HJ, Li C, Lee GH, Zhao C, Darnell R, Friedman JM.
Nature. 1994;372:425– 432. Anatomic localization of alternatively spliced leptin receptors (ob-R) in
4. Koerner A, Kratzsch J, Kiess W. Adipocytokines: leptin-the classical, mouse brain and other tissues. Proc Natl Acad Sci U S A. 1997;94:
resistin-the controversical, adiponectin-the promising, and more to 7001–7005.
come. Best Pract Res Clin Endocrinol Metab. 2005;19:525–546. 28. Tartaglia LA. The leptin receptor. J Biol Chem. 1997;272:6093– 6096.
5. Friedman JM, Halaas JL. Leptin and the regulation of body weight in 29. Chua SC Jr, Koutras IK, Han L, Liu SM, Kay J, Young SJ, Chung WK,
mammals. Nature. 1998;395:763–770. Leibel RL. Fine structure of the murine leptin receptor gene: splice site
6. Elmquist JK, Elias CF, Saper CB. From lesions to leptin: hypothalamic suppression is required to form two alternatively spliced transcripts.
control of food intake and body weight. Neuron. 1999;22:221–232. Genomics. 1997;45:264 –270.
7. Bates SH, Myers MG Jr. The role of leptin receptor signaling in feeding 30. Ge H, Huang L, Pourbahrami T, Li C. Generation of soluble leptin
and neuroendocrine function. Trends Endocrinol Metab. 2003;14: receptor by ectodomain shedding of membrane-spanning receptors in
447– 452. vitro and in vivo. J Biol Chem. 2002;277:45898 – 45903.
8. Zhang F, Chen Y, Heiman M, Dimarchi R. Leptin: structure, function 31. Vaisse C, Halaas JL, Horvath CM, Darnell JE Jr, Stoffel M, Friedman
and biology. Vitam Horm. 2005;71:345–372. JM. Leptin activation of Stat3 in the hypothalamus of wild-type and
9. Montague CT, Farooqi IS, Whitehead JP, Soos MA, Rau H, Wareham ob/ob mice but not db/db mice. Nat Genet. 1996;14:95–97.
NJ, Sewter CP, Digby JE, Mohammed SN, Hurst JA, Cheetham CH, 32. White DW, Kuropatwinski KK, Devos R, Baumann H, Tartaglia LA.
Earley AR, Barnett AH, Prins JB, O’Rahilly S. Congenital leptin defi- Leptin receptor (OB-R) signaling. Cytoplasmic domain mutational anal-
ciency is associated with severe early-onset obesity in humans. Nature. ysis and evidence for receptor homo-oligomerization. J Biol Chem.
1997;387:903–908. 1997;272:4065– 4071.
10. Farooqi IS, Matarese G, Lord GM, Keogh JM, Lawrence E, Agwu C, 33. Nakashima K, Narazaki M, Taga T. Leptin receptor (OB-R) oli-
Sanna V, Jebb SA, Perna F, Fontana S, Lechler RI, DePaoli AM, gomerizes with itself but not with its closely related cytokine signal
O’Rahilly S. Beneficial effects of leptin on obesity, T cell hyporespon- transducer gp130. FEBS Lett. 1997;403:79 – 82.
siveness, and neuroendocrine/metabolic dysfunction of human con- 34. Kloek C, Haq AK, Dunn SL, Lavery HJ, Banks AS, Myers MG Jr.
genital leptin deficiency. J Clin Invest. 2002;110:1093–1103. Regulation of jak kinases by intracellular leptin receptor sequences.
11. Maffei M, Halaas J, Ravussin E, Pratley RE, Lee GH, Zhang Y, Fei H, J Biol Chem. 2002;277:41547– 41555.
Kim S, Lallone R, Ranganathan S. Leptin levels in human and rodent: 35. Bjorbaek C, Uotani S, da Silva B, Flier JS. Divergent signaling
measurement of plasma leptin and ob RNA in obese and weight-reduced capacities of the long and short isoforms of the leptin receptor. J Biol
subjects. Nat Med. 1995;1:1155–1161. Chem. 1997;272:32686 –32695.
556 Circulation Research September 14, 2007

36. Banks AS, Davis SM, Bates SH, Myers MG Jr. Activation of down- capabilities of interleukin 6-type cytokine receptors. Proc Natl Acad Sci
stream signals by the long form of the leptin receptor. J Biol Chem. U S A. 1996;93:8374 – 8378.
2000;275:14563–14572. 59. Zabolotny JM, Bence-Hanulec KK, Stricker-Krongrad A, Haj F, Wang
37. Eyckerman S, Broekaert D, Verhee A, Vandekerckhove J, Tavernier J. Y, Minokoshi Y, Kim YB, Elmquist JK, Tartaglia LA, Kahn BB, Neel
Identification of the Y985 and Y1077 motifs as SOCS3 recruitment sites BG. PTP1B regulates leptin signal transduction in vivo. Dev Cell.
in the murine leptin receptor. FEBS Lett. 2000;486:33–37. 2002;2:489 – 495.
38. Hekerman P, Zeidler J, Bamberg-Lemper S, Knobelspies H, Lavens D, 60. Cheng A, Uetani N, Simoncic PD, Chaubey VP, Lee-Loy A, McGlade
Tavernier J, Joost HG, Becker W. Pleiotropy of leptin receptor sig- CJ, Kennedy BP, Tremblay ML. Attenuation of leptin action and regu-
nalling is defined by distinct roles of the intracellular tyrosines. FEBS J. lation of obesity by protein tyrosine phosphatase 1B. Dev Cell. 2002;2:
2005;272:109 –119. 497–503.
39. Munzberg H, Bjornholm M, Bates SH, Myers MG Jr. Leptin receptor 61. Bjorbaek C, Elmquist JK, Frantz JD, Shoelson SE, Flier JS. Identifi-
action and mechanisms of leptin resistance. Cell Mol Life Sci. 2005;62: cation of SOCS-3 as a potential mediator of central leptin resistance.
642– 652. Mol Cell. 1998;1:619 – 625.
40. Bjorbaek C, Buchholz RM, Davis SM, Bates SH, Pierroz DD, Gu H, 62. Howard JK, Cave BJ, Oksanen LJ, Tzameli I, Bjorbaek C, Flier JS.
Neel BG, Myers MG Jr, Flier JS. Divergent roles of SHP-2 in ERK Enhanced leptin sensitivity and attenuation of diet-induced obesity in
activation by leptin receptors. J Biol Chem. 2001;276:4747– 4755. mice with haploinsufficiency of Socs3. Nat Med. 2004;10:734 –738.
41. Dunn SL, Bjornholm M, Bates SH, Chen Z, Seifert M, Myers MG Jr. 63. Mori H, Hanada R, Hanada T, Aki D, Mashima R, Nishinakamura H,
Feedback inhibition of leptin receptor/Jak2 signaling via Tyr1138 of the Torisu T, Chien KR, Yasukawa H, Yoshimura A. Socs3 deficiency in
leptin receptor and suppressor of cytokine signaling 3. Mol Endocrinol. the brain elevates leptin sensitivity and confers resistance to diet-
2005;19:925–938. induced obesity. Nat Med. 2004;10:739 –743.
42. Niswender KD, Gallis B, Blevins JE, Corson MA, Schwartz MW, 64. Klaman LD, Boss O, Peroni OD, Kim JK, Martino JL, Zabolotny JM,
Baskin DG. Immunocytochemical detection of phosphatidylinositol Moghal N, Lubkin M, Kim YB, Sharpe AH, Stricker-Krongrad A,
3-kinase activation by insulin and leptin. J Histochem Cytochem. 2003; Shulman GI, Neel BG, Kahn BB. Increased energy expenditure,
51:275–283. decreased adiposity, and tissue-specific insulin sensitivity in protein-
43. Tajmir P, Ceddia RB, Li RK, Coe IR, Sweeney G. Leptin increases tyrosine phosphatase 1B-deficient mice. Mol Cell Biol. 2000;20:
cardiomyocyte hyperplasia via extracellular signal-regulated kinase- 5479 –5489.
and phosphatidylinositol 3-kinase-dependent signaling pathways. 65. Munzberg H, Myers MG Jr. Molecular and anatomical determinants of
Endocrinology. 2004;145:1550 –1555. central leptin resistance. Nat Neurosci. 2005;8:566 –570.
44. Stofega MR, Herrington J, Billestrup N, Carter-Su C. Mutation of the 66. Banks WA. Blood-brain barrier and energy balance. Obesity (Silver
SHP-2 binding site in growth hormone (GH) receptor prolongs Spring). 2006;14(suppl 5):234S–237S.
GH-promoted tyrosyl phosphorylation of GH receptor, JAK2, and 67. Levin BE, Dunn-Meynell AA, Banks WA. Obesity-prone rats have
normal blood-brain barrier transport but defective central leptin sig-
STAT5B. Mol Endocrinol. 2000;14:1338 –1350.
naling before obesity onset. Am J Physiol Regul Integr Comp Physiol.
45. Gualillo O, Eiras S, White DW, Dieguez C, Casanueva FF. Leptin
2004;286:R143–R150.
promotes the tyrosine phosphorylation of SHC proteins and SHC asso-
68. Munzberg H, Flier JS, Bjorbaek C. Region-specific leptin resistance
ciation with GRB2. Mol Cell Endocrinol. 2002;190:83– 89.
within the hypothalamus of diet-induced obese mice. Endocrinology.
46. Raju SV, Zheng M, Schuleri KH, Phan AC, Bedja D, Saraiva RM,
2004;145:4880 – 4889.
Downloaded from http://ahajournals.org by on April 15, 2022

Yiginer O, Vandegaer K, Gabrielson KL, O’donnell CP, Berkowitz DE,


69. Satoh N, Ogawa Y, Katsuura G, Numata Y, Tsuji T, Hayase M, Ebihara
Barouch LA, Hare JM. Activation of the cardiac ciliary neurotrophic
K, Masuzaki H, Hosoda K, Yoshimasa Y, Nakao K. Sympathetic acti-
factor receptor reverses left ventricular hypertrophy in leptin-deficient
vation of leptin via the ventromedial hypothalamus: leptin-induced
and leptin-resistant obesity. Proc Natl Acad Sci U S A. 2006;103:
increase in catecholamine secretion. Diabetes. 1999;48:1787–1793.
4222– 4227.
70. Mark AL, Correia ML, Rahmouni K, Haynes WG. Loss of leptin actions
47. Wang Y, Huang S, Sah VP, Ross J Jr, Brown JH, Han J, Chien KR.
in obesity: two concepts with cardiovascular implications. Clin Exp
Cardiac muscle cell hypertrophy and apoptosis induced by distinct
Hypertens. 2004;26:629 – 636.
members of the p38 mitogen-activated protein kinase family. J Biol 71. Mark AL, Shaffer RA, Correia ML, Morgan DA, Sigmund CD, Haynes
Chem. 1998;273:2161–2168. WG. Contrasting blood pressure effects of obesity in leptin-deficient
48. Rajapurohitam V, Gan XT, Kirshenbaum LA, Karmazyn M. The ob/ob mice and agouti yellow obese mice. J Hypertens. 1999;17:
obesity-associated peptide leptin induces hypertrophy in neonatal rat 1949 –1953.
ventricular myocytes. Circ Res. 2003;93:277–279. 72. Correia ML, Haynes WG, Rahmouni K, Morgan DA, Sivitz WI, Mark
49. Shin HJ, Oh J, Kang SM, Lee JH, Shin MJ, Hwang KC, Jang Y, Chung AL. The concept of selective leptin resistance: evidence from agouti
JH. Leptin induces hypertrophy via p38 mitogen–activated protein yellow obese mice. Diabetes. 2002;51:439 – 442.
kinase in rat vascular smooth muscle cells. Biochem Biophys Res 73. Rahmouni K, Morgan DA, Morgan GM, Mark AL, Haynes WG. Role of
Commun. 2005;329:18 –24. selective leptin resistance in diet-induced obesity hypertension.
50. Bouloumie A, Marumo T, Lafontan M, Busse R. Leptin induces oxi- Diabetes. 2005;54:2012–2018.
dative stress in human endothelial cells. FASEB J. 1999;13:1231–1238. 74. Haynes WG. Interaction between leptin and sympathetic nervous system
51. Shen J, Sakaida I, Uchida K, Terai S, Okita K. Leptin enhances in hypertension. Curr Hypertens Rep. 2000;2:311–318.
TNF-alpha production via p38 and JNK MAPK in LPS-stimulated 75. Barr VA, Lane K, Taylor SI. Subcellular localization and internalization
kupffer cells. Life Sci. 2005;77:1502–1515. of the four human leptin receptor isoforms. J Biol Chem. 1999;274:
52. Fruhbeck G. Intracellular signalling pathways activated by leptin. 21416 –21424.
Biochem J. 2006;393:7–20. 76. Uotani S, Bjorbaek C, Tornoe J, Flier JS. Functional properties of leptin
53. Sweeney G. Leptin signalling. Cell Signal. 2002;14:655– 663. receptor isoforms: internalization and degradation of leptin and ligand-
54. Elmquist JK, Maratos-Flier E, Saper CB, Flier JS. Unraveling the central induced receptor downregulation. Diabetes. 1999;48:279 –286.
nervous system pathways underlying responses to leptin. Nat Neurosci. 77. Sen S, Tarazi RC, Khairallah PA, Bumpus FM. Cardiac hypertrophy in
1998;1:445– 450. spontaneously hypertensive rats. Circ Res. 1974;35:775–781.
55. Schwartz MW, Woods SC, Porte D Jr, Seeley RJ, Baskin DG. Central 78. Patel MB, Stewart JM, Loud AV, Anversa P, Wang J, Fiegel L, Hintze
nervous system control of food intake. Nature. 2000;404:661– 671. TH. Altered function and structure of the heart in dogs with chronic
56. Cowley MA, Smart JL, Rubinstein M, Cerdan MG, Diano S, Horvath elevation in plasma norepinephrine. Circulation. 1991;84:2091–2100.
TL, Cone RD, Low MJ. Leptin activates anorexigenic POMC neurons 79. Navarro J, Redon J, Cea-Calvo L, Lozano JV, Fernandez-Perez C, Bonet
through a neural network in the arcuate nucleus. Nature. 2001;411: A, Gonzalez-Esteban J. Metabolic syndrome, organ damage and cardio-
480 – 484. vascular disease in treated hypertensive patients. the ERIC-HTA study.
57. Ghilardi N, Ziegler S, Wiestner A, Stoffel R, Heim MH, Skoda RC. Blood Press. 2007;16:20 –27.
Defective STAT signaling by the leptin receptor in diabetic mice. Proc 80. Cuspidi C, Meani S, Fusi V, Severgnini B, Valerio C, Catini E, Leonetti
Natl Acad Sci U S A. 1996;93:6231– 6235. G, Magrini F, Zanchetti A. Metabolic syndrome and target organ
58. Baumann H, Morella KK, White DW, Dembski M, Bailon PS, Kim H, damage in untreated essential hypertensives. J Hypertens. 2004;22:
Lai CF, Tartaglia LA. The full-length leptin receptor has signaling 1991–1998.
Yang and Barouch Leptin Signaling and Obesity 557

81. Grassi G, Giannattasio C. Obesity and vascular stiffness: when body fat 103. Nakata M, Yada T, Soejima N, Maruyama I. Leptin promotes aggre-
has an adverse impact on arterial dynamics. J Hypertens. 2005;23: gation of human platelets via the long form of its receptor. Diabetes.
1789 –1791. 1999;48:426 – 429.
82. Aizawa-Abe M, Ogawa Y, Masuzaki H, Ebihara K, Satoh N, Iwai H, 104. Maruyama I, Nakata M, Yamaji K. Effect of leptin in platelet and
Matsuoka N, Hayashi T, Hosoda K, Inoue G, Yoshimasa Y, Nakao K. endothelial cells. Obesity and arterial thrombosis. Ann N Y Acad Sci.
Pathophysiological role of leptin in obesity-related hypertension. J Clin 2000;902:315–319.
Invest. 2000;105:1243–1252. 105. Reilly MP, Iqbal N, Schutta M, Wolfe ML, Scally M, Localio AR, Rader
83. Winters B, Mo Z, Brooks-Asplund E, Kim S, Shoukas A, Li D, Nyhan DJ, Kimmel SE. Plasma leptin levels are associated with coronary
D, Berkowitz DE. Reduction of obesity, as induced by leptin, reverses atherosclerosis in type 2 diabetes. J Clin Endocrinol Metab. 2004;89:
endothelial dysfunction in obese (lep(ob)) mice. J Appl Physiol. 2000; 3872–3878.
89:2382–2390. 106. Bodary PF, Gu S, Shen Y, Hasty AH, Buckler JM, Eitzman DT.
84. Leyva F, Godsland IF, Ghatei M, Proudler AJ, Aldis S, Walton C, Recombinant leptin promotes atherosclerosis and thrombosis in apoli-
Bloom S, Stevenson JC. Hyperleptinemia as a component of a metabolic poprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2005;25:
syndrome of cardiovascular risk. Arterioscler Thromb Vasc Biol. 1998; e119 – e122.
18:928 –933. 107. Wu KK, Wu TJ, Chin J, Mitnaul LJ, Hernandez M, Cai TQ, Ren N,
85. Morawietz H, Bornstein SR. Leptin, endothelin, NADPH oxidase, and Waters MG, Wright SD, Cheng K. Increased hypercholesterolemia
heart failure. Hypertension. 2006;47:e20; author reply e20 – e21. and atherosclerosis in mice lacking both ApoE and leptin receptor.
86. Shek EW, Brands MW, Hall JE. Chronic leptin infusion increases Atherosclerosis. 2005;181:251–259.
arterial pressure. Hypertension. 1998;31:409 – 414. 108. Nickola MW, Wold LE, Colligan PB, Wang GJ, Samson WK, Ren J.
87. Correia ML, Morgan DA, Sivitz WI, Mark AL, Haynes WG. Leptin acts Leptin attenuates cardiac contraction in rat ventricular myocytes. Role of
in the central nervous system to produce dose– dependent changes in NO. Hypertension. 2000;36:501–505.
arterial pressure. Hypertension. 2001;37:936 –942. 109. Wold LE, Relling DP, Duan J, Norby FL, Ren J. Abrogated leptin-
88. Carlyle M, Jones OB, Kuo JJ, Hall JE. Chronic cardiovascular and renal induced cardiac contractile response in ventricular myocytes under
actions of leptin: role of adrenergic activity. Hypertension. 2002;39: spontaneous hypertension: role of Jak/STAT pathway. Hypertension.
496 –501. 2002;39:69 –74.
89. Yamagishi SI, Edelstein D, Du XL, Kaneda Y, Guzman M, Brownlee 110. Rodriguez A, Fruhbeck G, Gomez-Ambrosi J, Catalan V, Sainz N, Diez
M. Leptin induces mitochondrial superoxide production and monocyte J, Zalba G, Fortuno A. The inhibitory effect of leptin on angiotensin
chemoattractant protein-1 expression in aortic endothelial cells by II-induced vasoconstriction is blunted in spontaneously hypertensive
increasing fatty acid oxidation via protein kinase A. J Biol Chem. rats. J Hypertens. 2006;24:1589 –1597.
2001;276:25096 –25100. 111. Barouch LA, Harrison RW, Skaf MW, Rosas GO, Cappola TP, Kobeissi
90. Loffreda S, Yang SQ, Lin HZ, Karp CL, Brengman ML, Wang DJ, ZA, Hobai IA, Lemmon CA, Burnett AL, O’Rourke B, Rodriguez ER,
Klein AS, Bulkley GB, Bao C, Noble PW, Lane MD, Diehl AM. Leptin
Huang PL, Lima JA, Berkowitz DE, Hare JM. Nitric oxide regulates the
regulates proinflammatory immune responses. FASEB J. 1998;12:
heart by spatial confinement of nitric oxide synthase isoforms. Nature.
57– 65.
2002;416:337–339.
91. Fruhbeck G. Pivotal role of nitric oxide in the control of blood pressure
112. Bloch KD, Janssens S. Cardiomyocyte-specific overexpression of nitric
after leptin administration. Diabetes. 1999;48:903–908.
oxide synthase 3: impact on left ventricular function and myocardial
92. Kimura K, Tsuda K, Baba A, Kawabe T, Boh-oka S, Ibata M, Moriwaki
Downloaded from http://ahajournals.org by on April 15, 2022

infarction. Trends Cardiovasc Med. 2005;15:249 –253.


C, Hano T, Nishio I. Involvement of nitric oxide in endothelium-
113. Barouch LA, Cappola TP, Harrison RW, Crone JK, Rodriguez ER,
dependent arterial relaxation by leptin. Biochem Biophys Res Commun.
Burnett AL, Hare JM. Combined loss of neuronal and endothelial nitric
2000;273:745–749.
oxide synthase causes premature mortality and age-related hypertrophic
93. Lembo G, Vecchione C, Fratta L, Marino G, Trimarco V, d’Amati G,
cardiac remodeling in mice. J Mol Cell Cardiol. 2003;35:637– 644.
Trimarco B. Leptin induces direct vasodilation through distinct endo-
114. Cappola TP, Cope L, Cernetich A, Barouch LA, Minhas K, Irizarry RA,
thelial mechanisms. Diabetes. 2000;49:293–297.
Parmigiani G, Durrani S, Lavoie T, Hoffman EP, Ye SQ, Garcia JG,
94. Vecchione C, Maffei A, Colella S, Aretini A, Poulet R, Frati G, Gentile
MT, Fratta L, Trimarco V, Trimarco B, Lembo G. Leptin effect on Hare JM. Deficiency of different nitric oxide synthase isoforms activates
endothelial nitric oxide is mediated through akt-endothelial nitric oxide divergent transcriptional programs in cardiac hypertrophy. Physiol
synthase phosphorylation pathway. Diabetes. 2002;51:168 –173. Genomics. 2003;14:25–34.
95. Gardiner SM, Kemp PA, March JE, Bennett T. Regional haemodynamic 115. Illiano G, Naviglio S, Pagano M, Spina A, Chiosi E, Barbieri M,
effects of recombinant murine or human leptin in conscious rats. Br J Paolisso G. Leptin affects adenylate cyclase activity in H9c2 cardiac cell
Pharmacol. 2000;130:805– 810. line: effects of short- and long-term exposure. Am J Hypertens. 2002;
96. Mitchell JL, Morgan DA, Correia ML, Mark AL, Sivitz WI, Haynes 15:638 – 643.
WG. Does leptin stimulate nitric oxide to oppose the effects of sympa- 116. Li SY, Yang X, Ceylan-Isik AF, Du M, Sreejayan N, Ren J. Cardiac
thetic activation? Hypertension. 2001;38:1081–1086. contractile dysfunction in Lep/Lep obesity is accompanied by NADPH
97. Kuo JJ, Jones OB, Hall JE. Inhibition of NO synthesis enhances chronic oxidase activation, oxidative modification of sarco(endo)plasmic
cardiovascular and renal actions of leptin. Hypertension. 2001;37: reticulum Ca2⫹-ATPase and myosin heavy chain isozyme switch.
670 – 676. Diabetologia. 2006;49:1434 –1446.
98. Quehenberger P, Exner M, Sunder-Plassmann R, Ruzicka K, 117. Khan SA, Lee K, Minhas KM, Gonzalez DR, Raju SV, Tejani AD, Li
Bieglmayer C, Endler G, Muellner C, Speiser W, Wagner O. Leptin D, Berkowitz DE, Hare JM. Neuronal nitric oxide synthase nega-
induces endothelin-1 in endothelial cells in vitro. Circ Res. 2002;90: tively regulates xanthine oxidoreductase inhibition of cardiac
711–718. excitation-contraction coupling. Proc Natl Acad Sci U S A. 2004;101:
99. Park HY, Kwon HM, Lim HJ, Hong BK, Lee JY, Park BE, Jang Y, Cho 15944 –15948.
SY, Kim HS. Potential role of leptin in angiogenesis: leptin induces 118. Kinugawa S, Huang H, Wang Z, Kaminski PM, Wolin MS, Hintze TH.
endothelial cell proliferation and expression of matrix metallopro- A defect of neuronal nitric oxide synthase increases xanthine oxidase-
teinases in vivo and in vitro. Exp Mol Med. 2001;33:95–102. derived superoxide anion and attenuates the control of myocardial
100. Kellerer M, Koch M, Metzinger E, Mushack J, Capp E, Haring HU. oxygen consumption by nitric oxide derived from endothelial nitric
Leptin activates PI-3 kinase in C2C12 myotubes via janus kinase-2 oxide synthase. Circ Res. 2005;96:355–362.
(JAK-2) and insulin receptor substrate-2 (IRS-2) dependent pathways. 119. Saraiva RM, Minhas KM, Zheng M, Pitz E, Treuer A, Gonzalez D,
Diabetologia. 1997;40:1358 –1362. Schuleri KH, Vandegaer KM, Barouch LA, Hare JM. Reduced neuronal
101. Li L, Mamputu JC, Wiernsperger N, Renier G. Signaling pathways nitric oxide synthase expression contributes to cardiac oxidative stress
involved in human vascular smooth muscle cell proliferation and matrix and nitroso-redox imbalance in ob/ob mice. Nitric Oxide. 2007;16:
metalloproteinase-2 expression induced by leptin: inhibitory effect of 331–338.
metformin. Diabetes. 2005;54:2227–2234. 120. Su J, Zhang S, Tse J, Scholz PM, Weiss HR. Alterations in nitric
102. Oda A, Taniguchi T, Yokoyama M. Leptin stimulates rat aortic smooth oxide-cGMP pathway in ventricular myocytes from obese leptin-
muscle cell proliferation and migration. Kobe J Med Sci. 2001;47: deficient mice. Am J Physiol Heart Circ Physiol. 2003;285:
141–150. H2111–H2117.
558 Circulation Research September 14, 2007

121. Furukawa S, Fujita T, Shimabukuro M, Iwaki M, Yamada Y, Nakajima 142. Warnes CA, Roberts WC. Sudden coronary death: relation of amount
Y, Nakayama O, Makishima M, Matsuda M, Shimomura I. Increased and distribution of coronary narrowing at necropsy to previous
oxidative stress in obesity and its impact on metabolic syndrome. J Clin symptoms of myocardial ischemia, left ventricular scarring and heart
Invest. 2004;114:1752–1761. weight. Am J Cardiol. 1984;54:65–73.
122. Katakam PV, Tulbert CD, Snipes JA, Erdos B, Miller AW, Busija DW. 143. Alpert MA. Obesity cardiomyopathy: pathophysiology and evolution of
Impaired insulin-induced vasodilation in small coronary arteries of the clinical syndrome. Am J Med Sci. 2001;321:225–236.
zucker obese rats is mediated by reactive oxygen species. Am J Physiol 144. Kasper EK, Hruban RH, Baughman KL. Cardiomyopathy of obesity: a
Heart Circ Physiol. 2005;288:H854 –H860. clinicopathologic evaluation of 43 obese patients with heart failure. Am J
123. Paolocci N, Ekelund UE, Isoda T, Ozaki M, Vandegaer K, Georgako- Cardiol. 1992;70:921–924.
poulos D, Harrison RW, Kass DA, Hare JM. cGMP-independent ino- 145. Sader S, Nian M, Liu P. Leptin: a novel link between obesity, diabetes,
tropic effects of nitric oxide and peroxynitrite donors: potential role cardiovascular risk, and ventricular hypertrophy. Circulation. 2003;108:
for nitrosylation. Am J Physiol Heart Circ Physiol. 2000;279: 644 – 646.
H1982–H1988. 146. Xu FP, Chen MS, Wang YZ, Yi Q, Lin SB, Chen AF, Luo JD. Leptin
124. Katori T, Donzelli S, Tocchetti CG, Miranda KM, Cormaci G, Thomas induces hypertrophy via endothelin-1-reactive oxygen species pathway
DD, Ketner EA, Lee MJ, Mancardi D, Wink DA, Kass DA, Paolocci N. in cultured neonatal rat cardiomyocytes. Circulation. 2004;110:
Peroxynitrite and myocardial contractility: in vivo versus in vitro effects. 1269 –1275.
Free Radic Biol Med. 2006;41:1606 –1618. 147. Paolisso G, Tagliamonte MR, Galderisi M, Zito GA, Petrocelli A,
125. Levrand S, Vannay-Bouchiche C, Pesse B, Pacher P, Feihl F, Waeber B, Carella C, de Divitiis O, Varricchio M. Plasma leptin level is associated
Liaudet L. Peroxynitrite is a major trigger of cardiomyocyte apoptosis in with myocardial wall thickness in hypertensive insulin–resistant men.
vitro and in vivo. Free Radic Biol Med. 2006;41:886 – 895. Hypertension. 1999;34:1047–1052.
126. Dong F, Zhang X, Ren J. Leptin regulates cardiomyocyte contractile 148. Yamashita T, Murakami T, Iida M, Kuwajima M, Shima K. Leptin
function through endothelin-1 receptor-NADPH oxidase pathway. receptor of zucker fatty rat performs reduced signal transduction.
Hypertension. 2006;47:222–229. Diabetes. 1997;46:1077–1080.
127. Pieske B, Beyermann B, Breu V, Loffler BM, Schlotthauer K, Maier LS, 149. Nakamura K, Fushimi K, Kouchi H, Mihara K, Miyazaki M, Ohe T,
Schmidt-Schweda S, Just H, Hasenfuss G. Functional effects of endo- Namba M. Inhibitory effects of antioxidants on neonatal rat cardiac
thelin and regulation of endothelin receptors in isolated human non- myocyte hypertrophy induced by tumor necrosis factor-alpha and an-
failing and failing myocardium. Circulation. 1999;99:1802–1809. giotensin II. Circulation. 1998;98:794 –799.
128. Unger RH, Orci L. Lipoapoptosis: its mechanism and its diseases. 150. Luo JD, Xie F, Zhang WW, Ma XD, Guan JX, Chen X. Simvastatin
Biochim Biophys Acta. 2002;1585:202–212. inhibits noradrenaline-induced hypertrophy of cultured neonatal rat car-
129. Zhou YT, Grayburn P, Karim A, Shimabukuro M, Higa M, Baetens D, diomyocytes. Br J Pharmacol. 2001;132:159 –164.
Orci L, Unger RH. Lipotoxic heart disease in obese rats: implications for 151. Adiarto S, Emoto N, Iwasa N, Yokoyama M. Obesity-induced upregu-
human obesity. Proc Natl Acad Sci U S A. 2000;97:1784 –1789. lation of myocardial endothelin-1 expression is mediated by leptin.
130. Ren J, Relling DP. Leptin-induced suppression of cardiomyocyte con- Biochem Biophys Res Commun. 2007;353:623– 627.
traction is amplified by ceramide. Peptides. 2006;27:1415–1419. 152. Hu TP, Xu FP, Li YJ, Luo JD. Simvastatin inhibits leptin-induced
131. Ren J, Relling DP. Interaction between tumor necrosis factor-alpha and hypertrophy in cultured neonatal rat cardiomyocytes. Acta Pharmacol
leptin-induced inhibition of cardiac contractile function in isolated ven- Sin. 2006;27:419 – 422.
tricular myocytes. Cytokine. 2005;32:213–218. 153. Williams SD, Zhu H, Zhang L, Bernstein HS. Adenoviral delivery of
Downloaded from http://ahajournals.org by on April 15, 2022

132. Hynes GR, Jones PJ. Leptin and its role in lipid metabolism. Curr Opin human CDC5 promotes G2/M progression and cell division in neonatal
Lipidol. 2001;12:321–327. ventricular cardiomyocytes. Gene Ther. 2006;13:837– 843.
133. Kudo N, Barr AJ, Barr RL, Desai S, Lopaschuk GD. High rates of fatty 154. Barouch LA, Gao D, Chen L, Miller KL, Xu W, Phan AC, Kittleson
acid oxidation during reperfusion of ischemic hearts are associated with MM, Minhas KM, Berkowitz DE, Wei C, Hare JM. Cardiac myocyte
a decrease in malonyl-CoA levels due to an increase in 5⬘-AMP-acti- apoptosis is associated with increased DNA damage and decreased
vated protein kinase inhibition of acetyl-CoA carboxylase. J Biol Chem. survival in murine models of obesity. Circ Res. 2006;98:119 –124.
1995;270:17513–17520. 155. Unger RH, Zhou YT, Orci L. Regulation of fatty acid homeostasis in
134. Muoio DM, Seefeld K, Witters LA, Coleman RA. AMP-activated kinase cells: novel role of leptin. Proc Natl Acad Sci U S A. 1999;96:
reciprocally regulates triacylglycerol synthesis and fatty acid oxidation 2327–2332.
in liver and muscle: evidence that sn-glycerol-3-phosphate acyltrans- 156. Mascareno E, Beckles DL, Siddiqui MA. Janus kinase-2 signaling
ferase is a novel target. Biochem J. 1999;338(pt 3):783–791. mediates apoptosis in rat cardiomyocytes. Vascul Pharmacol. 2005;43:
135. Atkinson LL, Fischer MA, Lopaschuk GD. Leptin activates cardiac fatty 327–335.
acid oxidation independent of changes in the AMP-activated protein 157. Finck BN, Lehman JJ, Leone TC, Welch MJ, Bennett MJ, Kovacs A,
kinase-acetyl-CoA carboxylase-malonyl-CoA axis. J Biol Chem. 2002; Han X, Gross RW, Kozak R, Lopaschuk GD, Kelly DP. The cardiac
277:29424 –29430. phenotype induced by PPARalpha overexpression mimics that caused
136. Somoza B, Guzman R, Cano V, Merino B, Ramos P, Diez-Fernandez C, by diabetes mellitus. J Clin Invest. 2002;109:121–130.
Fernandez-Alfonso MS, Ruiz-Gayo M. Induction of cardiac uncoupling 158. Kodama H, Fukuda K, Pan J, Makino S, Sano M, Takahashi T, Hori S,
protein-2 expression and adenosine 5⬘-monophosphate-activated protein Ogawa S. Biphasic activation of the JAK/STAT pathway by angiotensin
kinase phosphorylation during early states of diet-induced obesity in II in rat cardiomyocytes. Circ Res. 1998;82:244 –250.
mice. Endocrinology. 2007;148:924 –931. 159. Maroni P, Bendinelli P, Piccoletti R. Intracellular signal transduction
137. Lee Y, Wang MY, Kakuma T, Wang ZW, Babcock E, McCorkle K, pathways induced by leptin in C2C12 cells. Cell Biol Int. 2005;29:
Higa M, Zhou YT, Unger RH. Liporegulation in diet-induced obesity. 542–550.
The antisteatotic role of hyperleptinemia. J Biol Chem. 2001;276: 160. Madani S, De Girolamo S, Munoz DM, Li RK, Sweeney G. Direct
5629 –5635. effects of leptin on size and extracellular matrix components of human
138. Lee Y, Naseem RH, Duplomb L, Park BH, Garry DJ, Richardson JA, pediatric ventricular myocytes. Cardiovasc Res. 2006;69:716 –725.
Schaffer JE, Unger RH. Hyperleptinemia prevents lipotoxic cardiomy- 161. Hausenloy DJ, Tsang A, Yellon DM. The reperfusion injury salvage
opathy in acyl CoA synthase transgenic mice. Proc Natl Acad Sci U S A. kinase pathway: a common target for both ischemic preconditioning and
2004;101:13624 –13629. postconditioning. Trends Cardiovasc Med. 2005;15:69 –75.
139. Murphy S, Frishman WH. Protein kinase C in cardiac disease and as a 162. Brzozowski T, Konturek PC, Pajdo R, Kwiecien S, Ptak A, Sliwowski
potential therapeutic target. Cardiol Rev. 2005;13:3–12. Z, Drozdowicz D, Pawlik M, Konturek SJ, Hahn EG. Brain-gut axis in
140. Christoffersen C, Bollano E, Lindegaard ML, Bartels ED, Goetze JP, gastroprotection by leptin and cholecystokinin against ischemia-
Andersen CB, Nielsen LB. Cardiac lipid accumulation associated with reperfusion induced gastric lesions. J Physiol Pharmacol. 2001;52:
diastolic dysfunction in obese mice. Endocrinology. 2003;144: 583– 602.
3483–3490. 163. Erkasap S, Erkasap N, Koken T, Kahraman A, Uzuner K, Yazihan N,
141. Mazumder PK, O’Neill BT, Roberts MW, Buchanan J, Yun UJ, Ates E. Effect of leptin on renal ischemia-reperfusion damage in rats.
Cooksey RC, Boudina S, Abel ED. Impaired cardiac efficiency and J Physiol Biochem. 2004;60:79 – 84.
increased fatty acid oxidation in insulin-resistant ob/ob mouse hearts. 164. Kennedy LM, Dickstein K, Anker SD, Kristianson K, Willenheimer R,
Diabetes. 2004;53:2366 –2374. OPTIMAAL Study Group. The prognostic importance of body mass
Yang and Barouch Leptin Signaling and Obesity 559

index after complicated myocardial infarction. J Am Coll Cardiol. 2005; 170. Zhang J, Matheny MK, Tumer N, Mitchell MK, Scarpace PJ. Leptin
45:156 –158. antagonist reveals that the normalization of caloric intake and the
165. Nikolsky E, Stone GW, Grines CL, Cox DA, Garcia E, Tcheng JE, Griffin thermic effect of food after high-fat feeding are leptin dependent. Am J
JJ, Guagliumi G, Stuckey T, Turco M, Negoita M, Lansky AJ, Mehran R. Physiol Regul Integr Comp Physiol. 2007;292:R868 –R874.
Impact of body mass index on outcomes after primary angioplasty in acute 171. Matarese G, Carrieri PB, La Cava A, Perna F, Sanna V, De Rosa V,
myocardial infarction. Am Heart J. 2006;151:168–175. Aufiero D, Fontana S, Zappacosta S. Leptin increase in multiple
166. Knudson JD, Dincer UD, Dick GM, Shibata H, Akahane R, Saito M, sclerosis associates with reduced number of CD4(⫹)CD25⫹ regulatory
Tune JD. Leptin resistance extends to the coronary vasculature in T cells. Proc Natl Acad Sci U S A. 2005;102:5150 –5155.
prediabetic dogs and provides a protective adaptation against endo- 172. De Rosa V, Procaccini C, La Cava A, Chieffi P, Nicoletti GF, Fontana
thelial dysfunction. Am J Physiol Heart Circ Physiol. 2005;289: S, Zappacosta S, Matarese G. Leptin neutralization interferes with
H1038 –H1046.
pathogenic T cell autoreactivity in autoimmune encephalomyelitis.
167. Chen K, Li F, Li J, Cai H, Strom S, Bisello A, Kelley DE,
J Clin Invest. 2006;116:447– 455.
Friedman-Einat M, Skibinski GA, McCrory MA, Szalai AJ, Zhao AZ.
173. Purdham DM, Rajapurohitam V, Huang C, Karmazyn M. Attenuation of
Induction of leptin resistance through direct interaction of C-reactive
protein with leptin. Nat Med. 2006;12:425– 432. cardiac hypertrophy and heart failure by leptin receptor blockade.
168. Kazumi T, Kawaguchi A, Hirano T, Yoshino G. C-reactive protein in Circulation. 2005;112(suppl II):II-279 Abstract.
young, apparently healthy men: associations with serum leptin, QTc 174. Gertler A. Development of leptin antagonists and their potential use in
interval, and high-density lipoprotein-cholesterol. Metabolism. 2003;52: experimental biology and medicine. Trends Endocrinol Metab. 2006;
1113–1116. 17:372–378.
169. Scarpace PJ, Matheny M, Zhang Y, Cheng KY, Tumer N. Leptin 175. Eckel RH, Barouch WW, Ershow AG. Report of the national heart,
antagonist reveals an uncoupling between leptin receptor signal lung, and blood institute-national institute of diabetes and digestive
transducer and activator of transcription 3 signaling and metabolic and kidney diseases working group on the pathophysiology of
responses with central leptin resistance. J Pharmacol Exp Ther. 2007; obesity-associated cardiovascular disease. Circulation. 2002;105:
320:706 –712. 2923–2928.
Downloaded from http://ahajournals.org by on April 15, 2022

You might also like