Sie sind auf Seite 1von 16

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

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 maintenance makes it extremely difficult for people to maintain
weight loss.1 Several genes have been identified to disclose a
physiological system that maintains body weight within a
range of about twenty pounds.2 A key element of this system
is leptin, the 16-kDa hormonal product of the obesity (ob)
gene.3 Leptin is primarily secreted by adipocytes and is a
classic member of the more than 50 identified adipocytokines
that participate in adipose tissue hormonal signaling.4
Since its identification in 1994, leptin has attracted much
attention as one of the most important central and peripheral
signals for the maintenance of energy homeostasis.5 8 For
example, a 9-year-old girl with extreme obesity was found to
lack leptin.9 Leptin treatment reduced her weight to the

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
role of leptin is to communicate to the central nervous system
(CNS) the abundance of available energy stores and to
restrain food intake and induce energy expenditure. The
absence of leptin therefore leads to increased appetite and
food intake that result in morbid obesity. Notably, only rare
cases of severe early childhood obesity have been associated
with leptin deficiency.9,13 The remainder of the obese population typically have elevated leptin levels.14 The failure of
leptin to induce weight loss in these cases is thought to be the
result of leptin resistance.
Hyperleptinemia, nearly universally observed in human
obesity and animal models, is accompanied by a disruption of
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

545at VA MED CTR BOISE on March 23, 2016


Downloaded from http://circres.ahajournals.org/

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


cascade. Disruption of leptin signaling in the hypothalamus
results in obesity and confirms the central role of this
hormone in the maintenance of energy balance.7,15,16 Emerging evidence suggests that leptin resistance in the CNS may
be selective, namely that the effects of leptin on central
metabolic processes is disrupted, whereas its other effects,
such as the sympathetic activation of blood pressure, is still
retained.17 In addition to its actions in the CNS, leptin
receptors (Ob-Rs) are found in multiple peripheral tissue
types and affect many systemic processes, such as reproduction, immunity, and cardiovascular functions.18 20
Obesity is also a part of the metabolic syndrome, which is
diagnosed by a set of criteria that include abdominal obesity,
insulin resistance, dyslipidemia, and hypertension. This patient population faces increased risk for type 2 diabetes and
cardiovascular diseases. The widely distributed Ob-Rs make
the hormone an attractive candidate for a molecular link in the
pathogenesis of obesity-related diseases. Although disruption
of leptin signaling can lead to altered phenotypic expression
and function of peripheral organs, the relative contributions
of central versus peripheral signal disruption are still controversial. Increasing our understanding of how leptin and/or
leptin resistance affects the heart and vasculature will be
important for gaining comprehension of obesity-related
threats to cardiovascular health.

Leptin Signaling
Leptin and Leptin Receptor
Leptin is primarily secreted by adipocytes and circulates at a
level of 5 to 15 ng/mL in lean subjects.21 Its expression is
increased by overfeeding, insulin, glucocorticoids, endotoxin,
and cytokines and is decreased by fasting, testosterone,

thyroid hormone, and exposure to cold temperature.22,23 In the


heart, increased leptin expression is seen following reperfusion after ischemia,24,25 and leptin concentration in cardiomyocyte culture serum is increased with endothelin (ET)-1
and angiotensin (Ang) II treatment,26 suggesting the heart as
a site of leptin production.
Six isoforms of the Ob-R (a to f) have been identified in the
murine model, and they are closely related to the class I
cytokine receptor family. Ob-Ra and Ob-Rb represent the
dominant isoforms in the heart, whereas the others are
expressed at low levels27 and are not well conserved among
species.28,29 Ob-Re is the secreted form that binds circulating
leptin and regulates the concentration of free leptin.30

Janus-Activated Kinase/Signal Transducers and


Activators of Transcription
Ob-Rs have been shown to activate Janus-activated kinase
(JAK), signal transducers and activators of transcription
(STAT), insulin receptor substrate, and the mitogen-activated
protein kinase (MAPK) pathways. The best-characterized
pathway in leptin signaling is the JAK/STAT pathway (Figure 1).31 Ligand binding causes Ob-R to undergo homooligomerization32,33 and to bind to JAK, primarily JAK2.34 In
the case of overexpression of JAKs by transient transfection,
weak Ob-Rb/JAK1 and Ob-Ra/JAK2 association was observed with leptin treatment.35 However, only Ob-Rb contains the STAT-binding site.35
Studies in vivo have demonstrated that signaling occurs
mainly through STAT3.16 Ob-Rb binding to JAK2 leads to
JAK2 autophosphorylation and the phosphorylation of
Tyr985, Tyr1077, and Tyr1138 on Ob-Rb.32,34,36 38 Phosphorylation of Tyr1138 recruits STAT proteins to the Ob-Rb/
JAK2 complex. Tyrosine phosphorylated STAT3 molecules

Downloaded from http://circres.ahajournals.org/ at VA MED CTR BOISE on March 23, 2016

Yang and Barouch


dimerize and translocate to the nucleus to activate transcription of target genes, which includes the gene for a member of
the suppressors of the cytokine signaling family
(SOCS3).16,36,39 SOCS3 binding to Tyr985 and other sites
within the Ob-Rb/JAK2 complex mediates negative feedback
on leptin signaling.40,41 JAK2 phosphorylation of Tyr985 also
consequently leads to phosphorylation of the src homology 2
(SH2) domain of the tyrosine phosphatase SHP-2 (src homology 2-containing tyrosine phosphatase), which activates the
extracellular signal-regulated kinase (ERK) signaling pathway.36 Overexpression of SHP-2 has been shown to blunt
SOCS3-mediated inhibition, likely through competitive binding to Tyr985.40
JAK2 autophosphorylation is independent of tyrosine
phosphorylation sites on Ob-Rb and has many subsequent
effects. One of these effects is to phosphorylate insulin
receptor substrate proteins, which recruit the phosphatidylinositol 3-kinase (PI3K) to activate downstream signals.7,42
In the heart, the leptin-associated PI3K pathway, along with
ERK cascades, seems to be important in cardiomyocyte
proliferation and protecting cardiac tissue from ischemia/
reperfusion injury.24,43

Mitogen-Activated Protein Kinase


Phosphorylation of Tyr985 on Ob-Rb leads to SHP-2 and
Grb-2 activation of ERK1/2 of the MAPK family.36 ERK1/2
activation also occurs via a pathway independent of Tyr985.
In this case, JAK2 associates with the SH2 domaincontaining SHP-2.36,44 Therefore, both Ob-Ra and Ob-Rb can
activate MAPK.35,36 Shc, another SH-2 containing protein
that is able to associate with Grb-2, has also been shown to
phosphorylate tyrosine after leptin treatment.45 Leptininduced phosphorylation of STAT3 and ERK1/2 have been
observed in isolated adult C57BL/6 mouse cardiomyocytes,
with maximal activation at 15 minutes after treatment. Fourweek leptin treatment also elevated STAT3 and ERK1/2
phosphorylation and abundance in cardiac tissue from leptindeficient ob/ob mice but not from Ob-Rb deficient db/db
mice.46 Because Ob-Ra is intact in db/db mice and the distal
portion of Ob-Rb is not essential for MAPK signaling,35
ERK1/2 phosphorylation would be expected to increase even
if STAT3 phosphorylation were unaltered. Therefore, the
results suggest that either Ob-Rb is the predominant signaling
receptor in the mouse heart or that JAK2-induced ERK1/2
phosphorylation does not occurs at a significant level in
cardiomyocytes. ERK1/2 activation leads to the expression of
target genes, such as c-fos and egr-1, which participate in cell
proliferation and differentiation.
Phosphorylation of p38 MAPK in response to leptin has
also been demonstrated. The and isoforms of p38 MAPK
are broadly distributed, including at relatively high levels in
the heart.47 Although the specific mechanism leading to
leptin-induced p38 MAPK activation has not been elucidated,
it is associated with the onset of hypertrophy and programmed cell death in both rat vascular smooth muscle cells
(VSMCs) and cardiomyocytes.24,48,49
As with other cytokines, leptin has the ability to activate
the stress-activated protein kinase c-Jun N-terminal kinase
(JNK).50,51 However, we observed that phosphorylation of

Leptin Signaling and Obesity

547

JNK was unaltered in cultured adult cardiomyocytes from


C57BL/6, ob/ob, and db/db mouse strains with 15-minute
leptin treatment.46 The effects of long-term leptin treatment
on JNK activity in the heart remain to be investigated.
Nuclear factor B has been proposed as an attractive downstream target for p38 and JNK MAPK pathways because this
transcription factor is essential in the transcriptional regulation of proinflammatory cytokines such as tumor necrosis
factor (TNF)- and interleukin (IL)-1.52 Additional leptin
signaling pathways are difficult to consolidate because of
tissue specificity. For a comprehensive review of leptin
signaling capabilities, see reviews by Fruhbeck52 or
Sweeney.53

Leptin Signaling in the CNS


Hypothalamic Leptin Decreases Food Intake
Most Ob-Rs in the CNS are located in the basomedial
hypothalamus.54,55 In neurons that synthesize proopiomelanocortin and express Ob-Rb, leptin stimulates the synthesis of
proopiomelanocortin, which is processed to produce -melanocytestimulating hormone and activate downstream
melanocortin-3 and -4 receptors to decrease appetite.56 Leptin
also inhibits the appetite-stimulating hormone neuropeptide
Y in the arcuate nucleus, and causes inhibition of agoutirelated peptide that restrain melanocortin-3 and -4 receptor
signaling.55,56
Ob-Rbstimulated JAK2/STAT3 signaling is crucial in
leptin control of feeding and energy expenditure. Although
short forms of the leptin receptor may be capable of activating
JAK, insulin receptor substrate, and MAPK proteins, only
Ob-Rb is capable of activating STAT3.35,57,58 A homologous
replacement of Ob-Rb by a receptor mutant for Tyr1138
established that STAT3 is indispensable for the regulation of
expression of proopiomelanocortin and neuropeptide Y in the
hypothalamus.16

Mechanisms of Central Leptin Resistance


One theory of leptin resistance involves intracellular signaling disruption. STAT3 and protein tyrosine phosphatase 1B
are 2 molecules known to attenuate leptin signaling.40,59 63
Hypothalamic protein tyrosine phosphatase 1B levels are not
known to be altered in obesity, although protein tyrosine
phosphatase 1Bnull animals develop increased insulin sensitivity and have a lean phenotype.64 As previously discussed,
leptin can induce its own negative feedback through STAT3induced SOCS3 accumulation during prolonged Ob-Rb stimulation,36,40,61 which can occur via Tyr985 or additional sites
in the Ob-Rb/JAK2 complex.40,41 At low concentrations of
leptin, incremental changes in leptin would be almost fully
translated into increased Ob-Rb signaling, whereas, at high
levels, accumulated SOCS3 would counter most of the
increase in Ob-Rb signaling.39,65
Another possible mechanism of central leptin resistance
stems from the dependency of leptin on saturable transport
across the vascular barrier and, probably to a lesser extent,
across the choroid plexus to reach the arcuate nucleus.66 The
activity of this blood brain barrier transport system seems to
decrease in diet-induced obese (DIO) rodents,67 resulting in
failure of circulating leptin to reach its targets in the brain.

Downloaded from http://circres.ahajournals.org/ at VA MED CTR BOISE on March 23, 2016

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 cardiovascular 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.

Increasing evidence suggests that leptin signaling is preferentially reduced in the arcuate nucleus of the hypothalamus
but not in other regions, such as the ventromedial, dorsomedial, and/or premammillary nucleus of the hypothalamus that
also express Ob-Rs.68

Leptin Increases Sympathetic Outflow


In addition to reducing appetite and controlling weight gain,
leptin centrally activates the sympathetic nervous system. It
significantly increases plasma norepinephrine and epinephrine concentrations via the ventromedial hypothalamus.69
Whereas chronic leptin overstimulation in the hypothalamus
decreases the ability of leptin to regulate appetite, its sympathetic excitatory effects are maintained as increased arterial
pressure and renal sympathetic nerve activity are presented in
obesity (Figure 2).70 This observation suggests that central
leptin resistance is selective.
The concept of selective resistance is suggested by a
comparison between ob/ob and Agouti yellow obese mice.
Lower arterial pressure is observed in ob/ob mice, which is
increased by leptin reconstitution despite the accompanying
weight loss.71 In contrast, Agouti yellow obese mice develop
obesity resulting from ubiquitous overexpression of agouti
protein, which blocks MC4R rather than directly affecting
leptin. They have elevated arterial pressure similarly to DIO
mice, despite the fact that they have milder obesity than ob/ob
mice.71,72 This preservation of sympathoactivation effects of
leptin despite disruption of its weight control effects has been
confirmed in DIO mice, which is considered a more physio-

logic model of human obesity. In C57BL/6J mice fed a


10-week high-fat diet, intraperitoneal leptin administration
failed to decrease appetite and body weight, but increased
renal sympathetic nerve activity. Sympathetic nerve activity
in brown adipose tissue and in hindlimb did not increase on
leptin administration, indicating region-specific preservation
of leptin sympathoactivation that serves to protect its circulatory effects.73
The sympathoactivation effect is completely abolished by
selective destruction of the arcuate nucleus.74 As the arcuate
nucleus is required for both metabolic and sympathetic
effects of leptin, these results seem to suggest that leptin
resistance occurs mainly through intracellular signaling disruption. Under resting conditions, it is estimated that only 5%
to 25% of Ob-R isoforms are located at the cell surface.75 The
ligandreceptor complex internalizes, and studies have shown
that leptin internalization was greater for the Ob-Rb isoform.75,76 Preferential downregulation of Ob-Rb in response
to prolonged leptin exposure may be important in regulating
different tissue sensitivity to leptin and another cause for
selective leptin resistance.
Leptin stimulation of adrenergic overdrive can lead to
numerous adverse effects on the cardiovascular system. Both
in vitro and in vivo studies have demonstrated adrenergic
influences on the growth of cardiomyocytes.77,78 Patients with
the metabolic syndrome have increased sympathetic activity,
hypertension, and higher occurrences of left ventricular
hypertrophy (LVH).79,80 Sympathetic influences also modulates the elastic properties of large and medium-size arteries

Downloaded from http://circres.ahajournals.org/ at VA MED CTR BOISE on March 23, 2016

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


Liporegulation
Increase in size of cardiomyocyte

JAK/STAT, NO-cGMP, G proteincoupled


receptor, adenylate cyclase, ET-1, NAPDH, ROS
Increased FA oxidation
ET-1, ROS

Deficiency leads to increased rate of


cardiomyocyte apoptosis

p38, caspase-3, DNA damage, Ang II

Enhanced cardiomyocyte mitosis and


proliferation

ERK1/2, p38, PI3K, peroxisome


proliferator-activated receptor-

Protection from ischemia/reperfusion injury

and promote endothelial dysfunction, contributing to the


development of vascular structural alterations and the occurrence of atherosclerotic lesions.81

Leptin and the Cardiovascular System


Hyperleptinemia is associated with obesity-related hypertension and chronic congestive heart failure (HF) in humans, and
vascular endothelial and myocardial dysfunction in animal
models.82 85 The role of leptin and leptin resistance in the
pathogenesis of LVH and HF in obesity remains controversial. The Table provides a summary of the leptin signaling
pathways involved in the cardiovascular system.

Vascular Effects of Leptin


Development of Hypertension
Both intravenous infusion and intracerebroventricular administration of leptin lead to increased arterial pressure and heart
rate in rodents.86,87 Blockade of the adrenergic system inhibits
the pressor response to leptin.88 In addition to increased
sympathetic activity in hyperleptinemia, other mechanisms
may also contribute to the development of obesity-related
hypertension. Leptin has been shown to increase the generation of reactive oxygen species (ROS) in endothelial cells50,89
and to stimulate secretion of proinflammatory cytokines such
as TNF- and IL-6, both of which are promoters of hypertension and atherosclerosis.90
While stimulating blood flow via increased sympathetic
output, leptin has also been shown to have direct vasodilatory
effects. In both intact rodents and in endothelial cells, leptin
induces NO production.83,91 Leptin administration in anesthetized rats causes a dose-dependent increase in NO metabolite
concentrations, and inhibition of NO synthesis increases
arterial pressure.91 In vitro studies have shown that leptin
evokes an endothelium-dependent relaxation of arterial
rings.92,93 In endothelial cells, leptin activates a PI3Kindependent Akt-endothelial NO synthase (eNOS) phosphorylation pathway to increase NO production, which can be
abolished by erbstatin A, a Ca2-independent tyrosine kinase
inhibitor.94 However, the vasodilator action of leptin was not
found in conscious rats and the NOS inhibitor NG-nitro-L-

PI3K, ERK1/2, NO, ROS

arginine methyl ester did not unmask any pressor effect.95 At


a concentration sufficient to increase sympathetic nerve
outflow, leptin did not change arterial pressure or blood flow
in mesenteric, lower aortic, and renal arteries.96 Blockade of
NO synthesis increased heart rate and renal vascular and
glomerular response but did not substantially augment the
pressor response to leptin,97 indicating a negligible role of
leptin-stimulated NO production on blood pressure in vivo.
Leptin has been shown to increases the release of ET-1, a
vasoconstrictor secreted primarily by endothelial cells but
also by macrophages, fibroblasts, and cardiomyocytes, possibly countering the effects of NO.98
Development of Atherosclerosis
The proatherogenic action of leptin is likely attributable to a
combination of its effects on various cell types. In endothelial
cells, leptin induces oxidative stress, increases production of
monocyte chemoattractant protein-1 and ET-1 and potentiates
proliferation.50,89,98,99 In VSMCs, leptin promotes migration,
proliferation, and hypertrophy.100 102 Leptin also promotes
calcification of cells of the vascular wall and facilitates
thrombosis by increasing platelet aggregation.103,104
Hyperleptinemia has been associated with coronary atherosclerosis in type 2 diabetes, and this association has been
shown to be independent of insulin resistance.105 In apolipoprotein E deficient (apoE/) mice that are atherosclerosis
prone, recombinant leptin treatment further promoted atherosclerosis and thrombosis, despite its metabolic benefits.106
This result indicates that elevation of leptin concentration in
obesity increases the risk for atherosclerotic damage.
Paradoxically, leptin resistance is also proatherogenic.
ApoE/ mice lacking Ob-Rb (apoE/ db/db) are characterized by a 5-fold higher area of spontaneous atherosclerotic
lesions in the aorta than apoE/ with intact Ob-R.107 Unlike
apoE/ mice, apoE/ db/db mice are also obese and insulin
resistant. Taken together, these data suggest either that a
supraphysiological level of exogenous leptin self-induces
resistance that is harmful or that the atherogenic effect seen in
apoE/ db/db is an effect more relevant to lipid profile
change, which is absent in apoE/ mice.

Downloaded from http://circres.ahajournals.org/ at VA MED CTR BOISE on March 23, 2016

550

Circulation Research

September 14, 2007

Figure 3. Leptin and myocardial contractility. Leptin directly depresses cardiomyocyte contractility. The signaling pathways implicated
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


Possible Mechanisms Leading to Increased
NO Production
Similar to its effects in endothelial cells, acute leptin infusion
in isolated rat ventricular myocytes increases NO activity,
which leads to attenuated cardiac contractility (Figure 3).108
Intracellular Ca2 transients were lowered and NO production
were increased with leptin. These effects were blocked by the
NO inhibitor NG-nitro-L-arginine methyl ester108 and by JAK2
or p38 MAPK inhibitors AG-490 and SB203580.109
The intermediate steps by which leptin signaling leads to
increase in NO production and the specific NOS isoforms that
mediate the effects of leptin have not been fully elucidated. In
rat VSMCs, leptin inhibits the contractile response induced
by Ang II through increased NO production. The upregulation of inducible NO synthase through mechanisms involving
JAK2/STAT3 and PI3K/Akt pathways is responsible for the
increase of NO bioavailability in VSMCs.110
Elucidation of the NOS isoform(s) responsible for the
actions of leptin in the heart would lead to a better understanding of its role in myocardial contractility and hypertrophy responses. We have shown that spatial confinement of
different constitutive NOS isoforms within separate subcellular compartments of the cardiac myocyte allows NO signals
to have independent, and even opposite, effects on cardiac
phenotype and contractile response.111 Overexpression of

eNOS inhibits hypertrophy in the remote myocardium and


preserved cardiac function after myocardial infarction, possibly through attenuation of -adrenergicstimulated compensatory hypertrophy.112 Neuronal NO synthase and eNOS
independently contribute to the development of cardiac hypertrophy, leading to marked age-related concentric hypertrophic remodeling in double-knockout mice lacking both
neuronal NO synthase and eNOS.113,114 Understanding the
leptin crosstalk with the -adrenergic signaling system in
cardiomyocytes would provide significant insight into the
understanding of myocardial dysfunction in obesity.
Whereas leptin-induced NO increase directly depresses
cardiomyocyte contractility, systemic actions such as increased sympathetic modulation may indirectly stimulate
contractility. Just as leptin-stimulated NO production in
endothelial cells may have a negligible role in blood pressure
in vivo, cardiac contractile depression may not manifest
under normal physiologic conditions. However, the depressant effect may become important when considered in conjuncture with alterations occurring in obese states.
Leptin Deficiency Leads to Decreased Responsiveness to
-Adrenergic Stimulation
In 10 week-old ob/ob isolated myocytes, attenuated sarcomere shortening and calcium transients and depressed sarcoplasmic reticulum Ca2 stores were seen in response to
isoproterenol stimulation of the -adrenergic receptor or to

Downloaded from http://circres.ahajournals.org/ at VA MED CTR BOISE on March 23, 2016

Yang and Barouch


postreceptor level stimulation with forskolin and dibutyryl
cAMP. Leptin replenishment in ob/ob mice restored each of
these abnormalities toward normal without affecting gross
(wall thickness) or microscopic (cell size) measures of
cardiac architecture. Decreased Gs (52 kDa), increased
sarcoplasmic reticulum Ca2-ATPase, and depressed phosphorylated phospholamban abundance were detected in ob/ob
mice. In addition, protein kinase A activity in ob/ob mice was
depressed at baseline and corrected toward wild-type (WT)
level with leptin repletion.46 In the H9c2 cardiac cell line,
30-minute leptin treatment increased basal and catecholamine-stimulated adenylate cyclase activity, whereas 18-hour
treatment was associated with a reduced adenylate cyclase
activity and a different responsiveness to isoproterenol and
norepinephrine stimulation, likely attributable to differential
activation of Gs. Adenylate cyclase, Gs (52 kDa), Gi,
p21-ras, and phosphorylated ERK1/2 expressions were increased with short-term leptin treatment and decreased at 18
hours, whereas Gs (45 kDa) continued to increase at 18
hours. Receptor level leptin resistance is conceivable in
myocytes, as Ob-R expression is seen to decrease at 18-hour
leptin treatment.115 Taken together, leptin deficiency or resistance leads to decreased -adrenergic response, whereas
moderate leptin stimulation can improve the contractile
response.
Mechanisms Involving ROS
Mitochondrial formation of ROS is enhanced in obesity.
Xanthine oxidoreductase and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase are 2 main sources of
superoxide (O2. ) production in the heart. O2. is capable of
generating a large family of ROS by interacting with other
molecular compounds. In ob/ob hearts, impaired cardiac
contractile function is accompanied by elevated oxidative
stress, lipid peroxidation, protein carbonyl formation, redistribution of myosin heavy chain isozymes from myosin heavy
chain- to -, and oxidative modification of SERCA2a.116
Neuronal NO synthase constrains xanthine oxidoreductase
activity.117,118 Reduced neuronal NO synthase expression is
observed in 2- to 6-month-old ob/ob mice, which leads to
increased xanthine oxidoreductase production of O2. , thereby
causing an imbalance between the production of ROS and
reactive nitrogen species.119 This nitrosoredox imbalance
may be partially responsible for the myocardial dysfunction
seen in ob/ob mice. Activation of NADPH oxidase is also
seen in the ob/ob heart.116 Treatment with apocynin, a
NADPH oxidase inhibitor, reversed cardiac contractile dysfunction in ob/ob myocytes but failed to reserve SERCA2a
oxidative modification.116 8-Bromo-cGMP, a membranepermeable cGMP analog, induced a greater negative effect in
ob/ob than lean C57BL/6J mice. However, the effect of
adding a NO donor was similar in the obese and lean models,
indicating that some cGMP-independent effect of NO prevents the enhanced negative cGMP effects in ob/ob cardiomyocytes.120 NAPDH-mediated reduction in NO bioavailability could explain the failure of NO donor to elicit further
negative inotropic response in obese models.121,122 Additionally, the interaction between NO and ROS produces peroxynitrite, which can nitrosylate proteins and exert positive

Leptin Signaling and Obesity

551

inotropic effects, thereby offsetting the cGMP-dependent


reduction in contractility.120 Peroxynitrite has shown both
negative and positive inotropy in isolated cardiomyocytes,123,124 although it is generally accepted to trigger apoptosis in cardiomyocytes in vitro and in vivo, possibly
through a pathway involving caspase-3 activation and the
cleavage of poly(ADP-ribose) polymerase.125
Another recent study proposes that ET-1 is upstream of
NADPH oxidase in leptin-induced myocardial contractile
response.126 There are at least 2 cardiac ET-1 receptors, ETA
and ETB. Both are known to mediate cardiomyocyte inotropic
response, and ETA receptors also affect hypertrophy.127 Leptin administration to rat neonatal cardiomyocytes induced
intracellular O2. generation and upregulated protein expression of p67phox and p47phox subunits of NAPDH, the effect
of which is attenuated by ETA and ETB receptor antagonists
and apocynin, suggesting that the ET-1 receptors are likely
upstream of NADPH oxidase in leptin-induced cardiac contractile response.126
Additional Mechanisms Affecting Contractility
Obesity is a lipotoxic disease featuring overtly elevated
ceramide levels (see section below, Leptin Shifts Myocardial
Metabolism Toward Fatty Acid Utilization). The de novo
ceramide pathway has been postulated to be key to the
lipoapoptosis of pancreatic cells and cardiomyocytes in
obese individuals.128,129 The ability of ceramide to amplify
leptin-induced depression of contractility in adult rat left
ventricular myocytes was recently demonstrated.130 Although
ceramide alone did not elicit any effect on cell mechanics and
intracellular Ca2 transients, it sensitized leptin-induced effects on myocyte shortening and intracellular Ca2 transients.
In vivo obese concentrations of plasma leptin lie in the low
nanomolar range, which is seemingly disconnected from the
high in vitro leptin concentration (10 nmol/L) needed to
affect cardiac contractile function. The observation that
ceramide may augment the cardiac depressive effect of leptin
provides an additional explanation for hyperleptinemiaassociated cardiac dysfunction in obesity.
Elevated adipose mass in obesity also increases the secretion of other proinflammatory factors, including TNF-, IL-6,
and Ang II. TNF- and leptin both depress contractility in
adult rat ventricular myocytes, although no additive response
by the 2 proinflammatory factors was observed. Inhibitory
effects were abolished by NG-monomethyl-L-arginine in both
cases and in the case of combined exposure.131 Thus, the
inhibitory effect on cardiac contraction by TNF- and leptin
may mask each other and share a common mechanism
dependent on NO.131
It is interesting, however, that hypertension seems to
attenuate leptin-induced cardiomyocyte contractile depression. Isolated rat ventricular myocytes from spontaneously
hypertensive rats (SHR) displayed decreased leptin-induced
depression of myocyte shortening and intracellular Ca2
transients, as well as blunted leptin-induced NOS activity
compared with the normotensive control mice. Additionally,
treatment of SHR myocytes with JAK or p38 inhibitor led to
further inhibition of myocyte shortening by leptin instead of
abolishing such effects.109 The altered signal transduction of

Downloaded from http://circres.ahajournals.org/ at VA MED CTR BOISE on March 23, 2016

552

Circulation Research

September 14, 2007

JAK/STAT and p38 pathways to attenuate leptin-induced


cardiomyocyte dysfunction possibly serves as a compensatory mechanism to prevent further impairment of ventricular
function in sustained hypertension or hyperleptinemia.109

Leptin Shifts Myocardial Metabolism Toward


Fatty Acid Utilization
Accumulating evidence suggests that leptin regulates energy
homeostasis through direct actions on peripheral lipid and
glucose metabolism.132 Fatty acid (FA) oxidation produces
the major source of ATP to sustain contractile function of the
heart. AMP-activated protein kinase (AMPK) has a key role
as a fuel gauge in the heart and regulates cardiac FA oxidation
by phosphorylation and inhibition of acetyl coenzyme A
(CoA) carboxylase, which then lowers malonyl-CoA levels
and stimulates carnitine palmitoyltransferase-1induced FA
oxidation.133 AMPK activation is also known to reduce FA
incorporation into triacylglycerol.134
Examination of leptin effects on cardiac FA oxidation
confirmed that leptin infusion increases FA oxidation and
triacylglycerol lipolysis in isolated working rat hearts, although this effect was independent of changes in the AMPK/
acetyl-CoA carboxylase/malonyl/CoA axis. Neither did leptin
affect glucose oxidation rates.135 Myocardial oxygen consumption was increased, possibly because of increased mitochondrial uncoupling protein activity.135 In DIO mice, plasma
leptin concentration increase was associated with an induction of uncoupling protein 2 and an increase in phosphorylation of AMPK that led to increased FA oxidation.136 At this
stage, leptin signaling in the heart as assessed by STAT3
phosphorylation remained unaltered from WT, suggesting
that the shift to FA oxidation is at least mediated, in part, by
leptin. Increased FA oxidation may confer antisteatosis protection to the myocardium during hyperleptinemia in early
stages of obesity. This is consistent with the observation that
DIO rodents exhibit minimal rise in myocardial triacylglycerol content, whereas ob/ob, db/db, and fa/fa animals show
significantly greater accumulation.137 Leptin deficiency perturbs liporegulation in peripheral organs and results in lipotoxicity, lipoapoptosis, and generalized steatosis, manifesting
in clinical conditions such as nonalcoholic steatohepatitis,
type 2 diabetes, and lipotoxic cardiomyopathy.128 Lipotoxic
cardiomyopathy has been identified in fa/fa rats and has been
transgenically induced in acylCoA synthase transgenic
mice.138 Overexpression of acylCoA synthase, which actives
and esterifies FAs, led to severe cardiomyopathy that was
attenuated by injection of recombinant adenovirus containing
the leptin cDNA.138
Increased de novo lipogenesis and decreased compensatory
oxidation of FAs are two possible mechanisms for intracellular accumulation of lipids in nonadipose tissue. Enzymes
involved in FA oxidation such as acyl-CoA oxidase and
carnitine palmitoyltransferase-1 are observed at attenuated
levels in fa/fa rats. Hydrolysis of the increased stores of
triacylglycerol to fatty acyl-CoA in fa/fa rats also increased
the substrate for de novo ceramide synthesis and activated the
expression of inducible NO synthase in the myocardium,
which led to acceleration of apoptosis.129 Ceramide also
activated protein kinase C isoforms, which can induce gene

transcription through MAPK and nuclear factor B and are


implicated in the development of cardiomyocyte hypertrophy.139 It seems paradoxical that enhanced expression of
oxidation enzymes such as long-chain acyl-CoA dehydrogenase and carnitine palmitoyltransferase-1 are seen in ob/ob
mice,140 as leptin-deficient hearts are also characterized by
increased FA utilization.141 This is likely attributable to
long-term adaptation to the changes in the lipid profile in
these animals. Genes for lipoprotein lipase, which generates
free FAs, and FA transport proteins are seen at an greatly
elevated level in ob/ob myocytes, which causes accumulation
of free FAs that promote ceramide, inducible NO synthase,
and NO production in the cardiomyocyte.140 When increased
FA delivery and hyperinsulinemia are imposed in ob/ob
hearts, myocardial function is maintained, albeit with decreased efficiency, whereas WT hearts are unable to adapt
acutely.141
Increased FA uptake accompanied by decreased oxidation
leads to lipotoxicity in long-term leptin-treated cardiomyocytes. The antisteatotic effects of leptin in early obesity might
be lost with the progression of leptin resistance occurring in
late stage obesity, leading to lipotoxicity that promotes
cardiomyocyte contractile dysfunction and apoptosis129 or
acts in a maladaptive fashion because of increased ceramide
synthesis.

Leptin Effects on Cardiac Hypertrophy


and Remodeling
Postmortem clinical studies of obesity-associated hypertrophy have demonstrated excessive heart weight but low
heart/body weight ratios.142 Fatty infiltration of the myocardium, presence of metabolic inclusions, excessive fibrosis, or
increases in extracellular matrix do not appear to play a direct
role in augmenting wall thickness in obesity-related hypertrophy.143 The findings on endomyocardial biopsy in the
majority of obese patients with cardiomyopathy appear to be
primarily myocyte hypertrophy.144 Hyperleptinemia is observed in patients with LVH.145 Leptin effects on cardiomyocyte hypertrophy is not entirely clear. We had previously
shown leptin repletion to be antihypertrophic in ob/ob hearts.
Four- to 6-week leptin infusion reduced weight and reversed
LVH, whereas caloric restriction reduced weight but did not
effectively reduce wall thickness and myocyte size.113 Reconstitution to normal levels may provide the antisteatotic effect
of leptin that can reverse lipotoxic effects of FA deposition in
the myocardium. On the other hand, leptin has been shown to
induce hypertrophy in a concentration-dependent manner in
cultured neonatal rat ventricular myocytes.48,146 Fasting
plasma leptin levels are associated with increased myocardial
wall thickness in hypertensive humans, although this may be
related to leptin resistance.147 The different types of cardiomyocytes used in experiments could also contribute to the
observed paradoxical effects of leptin on cardiomyocyte
hypertrophy. Direct effects of leptin on cardiomyocyte hypertrophy, apoptosis, proliferation, and extracellular matrix
remodeling could all contribute to maladaptive hypertrophy
and HF in obesity.

Downloaded from http://circres.ahajournals.org/ at VA MED CTR BOISE on March 23, 2016

Yang and Barouch


Cardiomyocyte Hypertrophy
Leptin-induced neonatal cardiomyocyte hypertrophy occurs
through a mechanism involving ET-1 and ROS generation.146
ET-1 has been shown to increase cardiomyocyte surface area
without increasing cell proliferation.148 In this study, blocking
the ETA receptor with the selective inhibitor ABT-627 partially but significantly reduced leptin-induced hypertrophy.146
An interdependence of ET-1 and leptin signaling has been
proposed in the progression of myocardial dysfunction and
hypertrophy, suggesting that leptin may cause chronic oxidative stress and inflammation in the myocardium, similar to
other agents such as TNF-, norepinephrine, and Ang II, all
of which induce hypertrophy via ROS upregulation.149,150
Leptin may indeed have an autocrine role in mediating
ET-1 and Ang IIinduced cardiomyocyte hypertrophy. Treatment of neonatal rat myocytes for 24 hours with leptin, Ang
II, or ET-1 significantly increased cell area by 37%, 36%, and
35%, respectively. In contrast to the study mentioned above
by Xu et al,146 Rajapurohitam et al26 have shown that
blocking the ETA receptor did not prevent leptin-induced
hypertrophy, neither did blocking the Ang receptor. Leptin
blockade attenuated the hypertrophic responses generated by
all 3 agents. Ang II and ET-1 significantly increased leptin
levels in the culture medium and increased the gene expression of both Ob-Ra and Ob-Rb. Additionally, Ang II and
ET-1 increased phosphorylation of ERK1/2, p38, JNK, and
nuclear factor B, but the ability of leptin blockade to
attenuate hypertrophic responses was generally dissociated
from these effects.26 The discrepancy between the studies by
Xu et al and Rajapurohitam et al in terms of blocking
leptin-induced hypertrophic response with ETA receptors
antagonists (ABT-627 versus BQ123) cautions against the
interpretation of results using pharmacological agents, as the
precise mechanisms of action is unclear in many cases.
Another study demonstrates that in 9-week-old mice fed a
high-fat diet, serum and myocardial ET-1, myocardial leptin,
and Ob-R mRNA are all elevated, whereas in ob/ob mice,
both serum and myocardial ET-1 levels are not higher than
WT mice, confirming a direct role of leptin in mediating
increased myocardial ET-1 signaling.151 Simvastatin, a cholesterol-lowering drug decreases leptin-induced ROSmediated hypertrophy in rat neonatal cardiomyocytes.152
Apoptosis
One of the causes of HF is cardiomyocyte apoptosis and
necrosis.153 We recently found that leptin deficiency or
resistance results in increased cardiomyocyte apoptosis, as
assessed by TUNEL staining and caspase-3 levels.154 Aged
ob/ob and db/db mice showed increased DNA damage
compared with old WT mice. Leptin reconstitution ob/ob
animals reduced the rate of apoptosis, although not to WT
levels.154 This is consistent with earlier work demonstrating
increased apoptosis in islet cells and cardiomyocytes in fa/fa
rats.129,155 These results suggest that leptin signaling is
necessary to maintain normal low levels of cell death
and that leptin provides protection against lipotoxicityinduced apoptosis.
On the other hand, JAK2 has been suggested as a mediator
of the apoptotic response in cardiomyocytes.156 It is promi-

Leptin Signaling and Obesity

553

nently involved in the upregulation of the reninAng system,


and Ang IItreated adult rat cardiomyocytes in culture exhibit
increased apoptosis. The somewhat paradoxical combination
of antiapoptotic roles of leptin and proapoptotic actions of
JAK2 merits further investigation. Leptin acutely increases
phosphorylation of ERK1/2 and p38 MAPK in rat neonatal
cardiomyocytes, but leptin-induced p38 activation in rat
neonatal cardiomyocytes sustains for a longer period than
ERK1/2 activation, suggesting that the downstream transcription factors of p38 may be involved in the long-term
maladaptive cardiac remodeling in obese HF patients.48
Mitosis and Proliferation
Leptin treatment at a level similar to plasma concentration in
obese individuals increased proliferation of both HL-1 cardiac muscle cells and human pediatric ventricular myocytes.43
The proliferation was accompanied by increased DNA synthesis associated with increased ERK1/2 phosphorylation and
increased association of the p85 regulatory subunit of PI3K
with phosphotyrosine immunoprecipitates.43 ERK1/2 inhibition significantly attenuated the leptin-induced proliferative
activity and DNA replication in HL-1 and pediatric human
cardiomyocytes43 but failed to decrease [3H]-leucine incorporation in neonatal rat cardiomyocytes treated with leptin.48
Other pathways likely involved in leptin-induced hypertrophy include the activation of adenylate cyclase,115 peroxisome proliferator-activated receptor-,157 and the JAK/STAT
pathway associated with hsp56 and Ang II.109,158 Leptin
signaling is capable of activating other traditional pathways
for the development of hypertrophy, such as PI3K and protein
kinase C.159 Whether these pathways are activated in cardiomyocytes in response to leptin and the specific isoforms
involved mandate further research.
Extracellular Matrix Remodeling
Leptin has been shown to increase the expression of matrix
metalloproteinase-2, and to increase collagen type III and IV
mRNA and decrease collagen type I mRNA without affecting
total collagen synthesis in human pediatric cardiomyocytes.160 This suggests that leptin selectively regulates different forms of collagen although further studies are required to
validate the regulation of collagen synthesis by leptin and to
confirm these effects on cardiac remodeling in obesity.

Protection in Ischemia/Reperfusion Injury


Timely reperfusion is necessary to salvage myocardium from
acute infarction, but reperfusion usually induces additional
injury. A recent report shows that exogenous leptin given at
early reperfusion in an isolated mouse heart model reduces
infarct size.24 This cardioprotective action of leptin is associated with activation of the reperfusion injury salvage kinase
pathway that includes PI3K/Akt and ERK1/2, ultimately
leading to the inhibition of mitochondrial permeability transition pore opening.161 Infarct size in C57BL/6J mouse hearts
perfused with leptin was about half that of hearts perfused
without leptin. In a rat model, leptin and Ob-Ra expressions
were locally upregulated in scarred tissue following reperfusion, whereas Ob-Rb expression was downregulated.24 PI3K
or ERK1/2 inhibition diminished the cardioprotective effect.24 Interestingly, leptin did not increase phosphorylation

Downloaded from http://circres.ahajournals.org/ at VA MED CTR BOISE on March 23, 2016

554

Circulation Research

September 14, 2007

of Akt or its downstream targets such as eNOS. Additionally,


there was increased phosphorylation of p38 MAPK and
reduced abundance and phosphorylation of STAT3 and
AMPK.24 Leptin-stimulated ROS production and NO synthesis has been shown also to protect against ischemia reperfusion injury in the gut and kidney.162,163 Clinically, it is
interesting to note that patients with a higher body mass index
have better outcomes following an acute coronary syndrome
or percutaneous coronary intervention.164,165

Leptin Resistance in the


Cardiovascular System?
The relative contributions of central and peripheral leptin
effects to disease pathogenesis are difficult to decipher.
Central leptin resistance disrupts hypothalamic control of
energy homeostasis, which results in obesity and increased
lipid production. This in turn may lead to ectopic lipid
deposition and lipotoxicity in peripheral organs. The attempt
to separate the effects of this pathological process from the
physiological effects of leptin in the cardiovascular system
has proven to be challenging, complicated by different
isoform signaling capabilities and possible resistance in the
periphery. The question remains whether peripheral leptin
resistance occurs in the myocardium itself in obesity. Even
though chronic leptin stimulation has been seen to decrease
Ob-R expression in various studies,75,115 DIO mice show
increased Ob-R mRNA expression.151 On the other hand,
Ob-Rb expression in ob/ob left ventricular homogenate is
lower than WT.126 However, mRNA expression does not
necessarily correlate with receptor density at the membrane;
therefore, these results are not conclusive in determining
whether leptin resistance can occur at the myocyte receptor
level. One recent study suggests that leptin resistance does
not occur in the myocardium in a model of early central
resistance. Eight-week DIO C57BL/6 mice showed attenuated leptin phosphorylation of STAT3 in hypothalamic tissue,
whereas no such attenuation was shown in whole-heart
homogenate.136
Paradoxical results have been reported in almost all leptinrelated effects on the myocardium; that is, excessive exogenous leptin and leptin deficiency often lead to the same end.
Whether these effects occur through entirely different mechanisms, are mediated through differential regulation of Ob-R
isoforms, or are attributable to peripheral resistance requires
further investigation. If peripheral myocardial resistance does
occur, these differences could be resolved if we consider
leptin deficiency and leptin resistance both to be states of
dysfunctional downstream signaling.
Interestingly, obesity-induced leptin resistance, although
not reported in the myocardium, has been shown to extend to
affect platelets and the vascular wall.166 Obese concentrations
of leptin significantly attenuate coronary vasodilation to
intracoronarily administered acetylcholine and significantly
attenuate relaxation in left circumflex coronary rings in
control dogs. These effects were not seen when the same
concentrations of leptin were administered to dogs fed a
high-fat diet, suggesting that leptin resistance does occur in
the vasculature. This resistance is not attributable to altered

coronary dilation, increased endothelium-derived hyperpolarizing factor, nor changes in coronary Ob-R mRNA levels.166
A recent hypothesis relevant to both central and peripheral
leptin resistance involves leptin interaction with circulating
factors in the blood.167 Five serum leptininteracting proteins
have been isolated, one of which is C-reactive protein. It
directly inhibits the binding of leptin to Ob-Rs and blocks its
ability to signal in cultured cells. Infusion of human
C-reactive protein into ob/ob mice blocked leptin treatment
effects on satiety and weight reduction. Physiological concentrations of leptin stimulate expression of C-reactive protein in human primary hepatocytes,167 and human C-reactive
protein has been correlated with increased adiposity and
plasma leptin,168 suggesting an systemic self-induced negative feedback that may cause leptin resistance in the obese
state.167

Leptin Antagonists
Leptin antagonists used in animal models have been shown to
block central leptin effects and increase appetite and food
intake.169,170 Three approaches have been employed to antagonize leptin activity: (1) binding free leptin in the circulation,
(2) competitive Ob-R binding by mutants that do not cause
signaling activation, and (3) specific antiOb-R monoclonal
antibodies. An example of the first approach is a recombinant
human and mouse Ob-R/Fc chimeric glycoprotein.171,172 Only
the latter 2 approaches have been employed in cardiac-related
research. In neonatal rat ventricular cardiomyocytes, rat
L39A/D40A/F41A leptin mutein blocked hypertrophic effects and abolished increases in Ob-R gene expression elicited by leptin, Ang II, or ET-1.26 The hypertrophic effects of
leptin are also prevented by antibodies to Ob-Ra and ObRb.26 Cardiac dysfunction did not develop in rats treated with
Ob-R antibodies after coronary artery ligation compared with
sham, indicating that blocking Ob-R can improve postinfarction HF in rats.173 The recent proposal of nanobodies (a
unique form of antibodies that is characterized by a single
antigen-binding domain and generally does not cross the
blood brain barrier) may lead to an antagonist that could
selectively inhibit peripheral activities of leptin.174 This form
of leptin antagonist might be clinically useful, as they can
target peripheral adverse effect of leptin without inducing
central weight gain.

Summary
Obesity leads to cardiac hypertrophy, ventricular dysfunction,
reduced diastolic compliance, and hypertension, as well as
type 2 diabetes and hyperlipidemia.175 The high risk of
developing cardiovascular diseases in obesity has drawn
much effort to study the neurohormone effects of leptin on
cardiac function and remodeling. Hyperleptinemia, central
leptin resistance, and leptin deficiency are all associated with
impaired postreceptor leptin signaling and contractile response. Short-term administration of leptin seems to have
beneficial effects on the myocardium, including antisteatotic
actions, protection against ischemia/reperfusion injury, and
participation in compensatory myocyte hypertrophy. Interaction with enhanced ROS production pathways in obesity, on
the other hand, can cause lipotoxicity and deleterious myo-

Downloaded from http://circres.ahajournals.org/ at VA MED CTR BOISE on March 23, 2016

Yang and Barouch


cardial effects such as cell death and maladaptive hypertrophy. Perturbations of leptin signaling and other signal transduction pathways regulated by leptin in cardiomyocytes
likely underlie the pathology of cardiomyocyte hypertrophy
in obesity. In particular, alterations in JAK/STAT, MAPK,
NO, and -adrenergic pathways have been implicated in the
negative inotropic and hypertrophic responses. Additional
studies investigating the integrated effects of leptin on cardiomyocytes via SOCS3, PI3K/Akt, protein kinase C, and
other signaling pathways could provide a more comprehensive understanding of leptin action on the cardiovascular
system.
The unresolved debate about selective preservation of
peripheral leptin signaling in the setting of hyperleptinemia
and central resistance complicates the interpretation of experimental results involving the myocardium. Despite such
challenges, a picture is emerging in which the risk of obesity
is not merely attributable to the physical burden of extra
weight but is, rather, a complex condition of hormonal
dysregulation. Improved understanding of the actions of
leptin within the cardiovascular system will greatly improve
our understanding of obesity-associated heart disease.

Sources of Funding
This work was supported in part by the Donald W. Reynolds
Foundation, NIH grant K08-HL076220, the W.W. Smith Charitable
Trust, and the Irvin Talles Endowed Fund for Cardiomyopathy
Research.

Disclosures
None.

References
1. Friedman JM. A war on obesity, not the obese. Science. 2003;299:
856 858.
2. ORahilly S, Farooqi IS, Yeo GS, Challis BG. Minireview: Human
obesity-lessons from monogenic disorders. Endocrinology. 2003;144:
37573764.
3. Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM.
Positional cloning of the mouse obese gene and its human homologue.
Nature. 1994;372:425 432.
4. Koerner A, Kratzsch J, Kiess W. Adipocytokines: leptin-the classical,
resistin-the controversical, adiponectin-the promising, and more to
come. Best Pract Res Clin Endocrinol Metab. 2005;19:525546.
5. Friedman JM, Halaas JL. Leptin and the regulation of body weight in
mammals. Nature. 1998;395:763770.
6. Elmquist JK, Elias CF, Saper CB. From lesions to leptin: hypothalamic
control of food intake and body weight. Neuron. 1999;22:221232.
7. Bates SH, Myers MG Jr. The role of leptin receptor signaling in feeding
and neuroendocrine function. Trends Endocrinol Metab. 2003;14:
447 452.
8. Zhang F, Chen Y, Heiman M, Dimarchi R. Leptin: structure, function
and biology. Vitam Horm. 2005;71:345372.
9. Montague CT, Farooqi IS, Whitehead JP, Soos MA, Rau H, Wareham
NJ, Sewter CP, Digby JE, Mohammed SN, Hurst JA, Cheetham CH,
Earley AR, Barnett AH, Prins JB, ORahilly S. Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature.
1997;387:903908.
10. Farooqi IS, Matarese G, Lord GM, Keogh JM, Lawrence E, Agwu C,
Sanna V, Jebb SA, Perna F, Fontana S, Lechler RI, DePaoli AM,
ORahilly S. Beneficial effects of leptin on obesity, T cell hyporesponsiveness, and neuroendocrine/metabolic dysfunction of human congenital leptin deficiency. J Clin Invest. 2002;110:10931103.
11. Maffei M, Halaas J, Ravussin E, Pratley RE, Lee GH, Zhang Y, Fei H,
Kim S, Lallone R, Ranganathan S. Leptin levels in human and rodent:
measurement of plasma leptin and ob RNA in obese and weight-reduced
subjects. Nat Med. 1995;1:11551161.

Leptin Signaling and Obesity

555

12. Considine RV, Sinha MK, Heiman ML, Kriauciunas A, Stephens TW,
Nyce MR, Ohannesian JP, Marco CC, McKee LJ, Bauer TL. Serum
immunoreactive-leptin concentrations in normal-weight and obese
humans. N Engl J Med. 1996;334:292295.
13. Strobel A, Issad T, Camoin L, Ozata M, Strosberg AD. A leptin
missense mutation associated with hypogonadism and morbid obesity.
Nat Genet. 1998;18:213215.
14. Frederich RC, Hamann A, Anderson S, Lollmann B, Lowell BB, Flier
JS. Leptin levels reflect body lipid content in mice: evidence for dietinduced resistance to leptin action. Nat Med. 1995;1:13111314.
15. Cohen P, Zhao C, Cai X, Montez JM, Rohani SC, Feinstein P, Mombaerts P, Friedman JM. Selective deletion of leptin receptor in neurons
leads to obesity. J Clin Invest. 2001;108:11131121.
16. Bates SH, Myers MG. The role of leptin-STAT3 signaling in neuroendocrine function: an integrative perspective. J Mol Med. 2004;82:
1220.
17. Mark AL, Correia ML, Rahmouni K, Haynes WG. Selective leptin
resistance: a new concept in leptin physiology with cardiovascular
implications. J Hypertens. 2002;20:12451250.
18. Lord GM, Matarese G, Howard JK, Baker RJ, Bloom SR, Lechler RI.
Leptin modulates the T-cell immune response and reverses starvationinduced immunosuppression. Nature. 1998;394:897901.
19. Margetic S, Gazzola C, Pegg GG, Hill RA. Leptin: a review of its
peripheral actions and interactions. Int J Obes Relat Metab Disord.
2002;26:14071433.
20. Rahmouni K, Haynes WG. Leptin and the cardiovascular system. Recent
Prog Horm Res. 2004;59:225244.
21. Sinha MK, Opentanova I, Ohannesian JP, Kolaczynski JW, Heiman ML,
Hale J, Becker GW, Bowsher RR, Stephens TW, Caro JF. Evidence of
free and bound leptin in human circulation. Studies in lean and obese
subjects and during short-term fasting. J Clin Invest. 1996;98:
12771282.
22. Coleman RA, Herrmann TS. Nutritional regulation of leptin in humans.
Diabetologia. 1999;42:639 646.
23. Fried SK, Ricci MR, Russell CD, Laferrere B. Regulation of leptin
production in humans. J Nutr. 2000;130:3127S3131S.
24. Smith CC, Mocanu MM, Davidson SM, Wynne AM, Simpkin JC,
Yellon DM. Leptin, the obesity-associated hormone, exhibits direct
cardioprotective effects. Br J Pharmacol. 2006;149:513.
25. Matsui H, Motooka M, Koike H, Inoue M, Iwasaki T, Suzuki T,
Kurabayashi M, Yokoyama T. Ischemia/reperfusion in rat heart induces
leptin and leptin receptor gene expression. Life Sci. 2007;80:672 680.
26. Rajapurohitam V, Javadov S, Purdham DM, Kirshenbaum LA,
Karmazyn M. An autocrine role for leptin in mediating the cardiomyocyte hypertrophic effects of angiotensin II and endothelin-1. J Mol Cell
Cardiol. 2006;41:265274.
27. Fei H, Okano HJ, Li C, Lee GH, Zhao C, Darnell R, Friedman JM.
Anatomic localization of alternatively spliced leptin receptors (ob-R) in
mouse brain and other tissues. Proc Natl Acad Sci U S A. 1997;94:
70017005.
28. Tartaglia LA. The leptin receptor. J Biol Chem. 1997;272:6093 6096.
29. Chua SC Jr, Koutras IK, Han L, Liu SM, Kay J, Young SJ, Chung WK,
Leibel RL. Fine structure of the murine leptin receptor gene: splice site
suppression is required to form two alternatively spliced transcripts.
Genomics. 1997;45:264 270.
30. Ge H, Huang L, Pourbahrami T, Li C. Generation of soluble leptin
receptor by ectodomain shedding of membrane-spanning receptors in
vitro and in vivo. J Biol Chem. 2002;277:45898 45903.
31. Vaisse C, Halaas JL, Horvath CM, Darnell JE Jr, Stoffel M, Friedman
JM. Leptin activation of Stat3 in the hypothalamus of wild-type and
ob/ob mice but not db/db mice. Nat Genet. 1996;14:9597.
32. White DW, Kuropatwinski KK, Devos R, Baumann H, Tartaglia LA.
Leptin receptor (OB-R) signaling. Cytoplasmic domain mutational analysis and evidence for receptor homo-oligomerization. J Biol Chem.
1997;272:4065 4071.
33. Nakashima K, Narazaki M, Taga T. Leptin receptor (OB-R) oligomerizes with itself but not with its closely related cytokine signal
transducer gp130. FEBS Lett. 1997;403:79 82.
34. Kloek C, Haq AK, Dunn SL, Lavery HJ, Banks AS, Myers MG Jr.
Regulation of jak kinases by intracellular leptin receptor sequences.
J Biol Chem. 2002;277:41547 41555.
35. Bjorbaek C, Uotani S, da Silva B, Flier JS. Divergent signaling
capacities of the long and short isoforms of the leptin receptor. J Biol
Chem. 1997;272:32686 32695.

Downloaded from http://circres.ahajournals.org/ at VA MED CTR BOISE on March 23, 2016

556

Circulation Research

September 14, 2007

36. Banks AS, Davis SM, Bates SH, Myers MG Jr. Activation of downstream signals by the long form of the leptin receptor. J Biol Chem.
2000;275:1456314572.
37. Eyckerman S, Broekaert D, Verhee A, Vandekerckhove J, Tavernier J.
Identification of the Y985 and Y1077 motifs as SOCS3 recruitment sites
in the murine leptin receptor. FEBS Lett. 2000;486:3337.
38. Hekerman P, Zeidler J, Bamberg-Lemper S, Knobelspies H, Lavens D,
Tavernier J, Joost HG, Becker W. Pleiotropy of leptin receptor signalling is defined by distinct roles of the intracellular tyrosines. FEBS J.
2005;272:109 119.
39. Munzberg H, Bjornholm M, Bates SH, Myers MG Jr. Leptin receptor
action and mechanisms of leptin resistance. Cell Mol Life Sci. 2005;62:
642 652.
40. Bjorbaek C, Buchholz RM, Davis SM, Bates SH, Pierroz DD, Gu H,
Neel BG, Myers MG Jr, Flier JS. Divergent roles of SHP-2 in ERK
activation by leptin receptors. J Biol Chem. 2001;276:4747 4755.
41. Dunn SL, Bjornholm M, Bates SH, Chen Z, Seifert M, Myers MG Jr.
Feedback inhibition of leptin receptor/Jak2 signaling via Tyr1138 of the
leptin receptor and suppressor of cytokine signaling 3. Mol Endocrinol.
2005;19:925938.
42. Niswender KD, Gallis B, Blevins JE, Corson MA, Schwartz MW,
Baskin DG. Immunocytochemical detection of phosphatidylinositol
3-kinase activation by insulin and leptin. J Histochem Cytochem. 2003;
51:275283.
43. Tajmir P, Ceddia RB, Li RK, Coe IR, Sweeney G. Leptin increases
cardiomyocyte hyperplasia via extracellular signal-regulated kinaseand phosphatidylinositol 3-kinase-dependent signaling pathways.
Endocrinology. 2004;145:1550 1555.
44. Stofega MR, Herrington J, Billestrup N, Carter-Su C. Mutation of the
SHP-2 binding site in growth hormone (GH) receptor prolongs
GH-promoted tyrosyl phosphorylation of GH receptor, JAK2, and
STAT5B. Mol Endocrinol. 2000;14:1338 1350.
45. Gualillo O, Eiras S, White DW, Dieguez C, Casanueva FF. Leptin
promotes the tyrosine phosphorylation of SHC proteins and SHC association with GRB2. Mol Cell Endocrinol. 2002;190:83 89.
46. Raju SV, Zheng M, Schuleri KH, Phan AC, Bedja D, Saraiva RM,
Yiginer O, Vandegaer K, Gabrielson KL, Odonnell CP, Berkowitz DE,
Barouch LA, Hare JM. Activation of the cardiac ciliary neurotrophic
factor receptor reverses left ventricular hypertrophy in leptin-deficient
and leptin-resistant obesity. Proc Natl Acad Sci U S A. 2006;103:
4222 4227.
47. Wang Y, Huang S, Sah VP, Ross J Jr, Brown JH, Han J, Chien KR.
Cardiac muscle cell hypertrophy and apoptosis induced by distinct
members of the p38 mitogen-activated protein kinase family. J Biol
Chem. 1998;273:21612168.
48. Rajapurohitam V, Gan XT, Kirshenbaum LA, Karmazyn M. The
obesity-associated peptide leptin induces hypertrophy in neonatal rat
ventricular myocytes. Circ Res. 2003;93:277279.
49. Shin HJ, Oh J, Kang SM, Lee JH, Shin MJ, Hwang KC, Jang Y, Chung
JH. Leptin induces hypertrophy via p38 mitogenactivated protein
kinase in rat vascular smooth muscle cells. Biochem Biophys Res
Commun. 2005;329:18 24.
50. Bouloumie A, Marumo T, Lafontan M, Busse R. Leptin induces oxidative stress in human endothelial cells. FASEB J. 1999;13:12311238.
51. Shen J, Sakaida I, Uchida K, Terai S, Okita K. Leptin enhances
TNF-alpha production via p38 and JNK MAPK in LPS-stimulated
kupffer cells. Life Sci. 2005;77:15021515.
52. Fruhbeck G. Intracellular signalling pathways activated by leptin.
Biochem J. 2006;393:720.
53. Sweeney G. Leptin signalling. Cell Signal. 2002;14:655 663.
54. Elmquist JK, Maratos-Flier E, Saper CB, Flier JS. Unraveling the central
nervous system pathways underlying responses to leptin. Nat Neurosci.
1998;1:445 450.
55. Schwartz MW, Woods SC, Porte D Jr, Seeley RJ, Baskin DG. Central
nervous system control of food intake. Nature. 2000;404:661 671.
56. Cowley MA, Smart JL, Rubinstein M, Cerdan MG, Diano S, Horvath
TL, Cone RD, Low MJ. Leptin activates anorexigenic POMC neurons
through a neural network in the arcuate nucleus. Nature. 2001;411:
480 484.
57. Ghilardi N, Ziegler S, Wiestner A, Stoffel R, Heim MH, Skoda RC.
Defective STAT signaling by the leptin receptor in diabetic mice. Proc
Natl Acad Sci U S A. 1996;93:6231 6235.
58. Baumann H, Morella KK, White DW, Dembski M, Bailon PS, Kim H,
Lai CF, Tartaglia LA. The full-length leptin receptor has signaling

59.

60.

61.

62.

63.

64.

65.
66.
67.

68.

69.

70.

71.

72.

73.

74.
75.

76.

77.
78.

79.

80.

capabilities of interleukin 6-type cytokine receptors. Proc Natl Acad Sci


U S A. 1996;93:8374 8378.
Zabolotny JM, Bence-Hanulec KK, Stricker-Krongrad A, Haj F, Wang
Y, Minokoshi Y, Kim YB, Elmquist JK, Tartaglia LA, Kahn BB, Neel
BG. PTP1B regulates leptin signal transduction in vivo. Dev Cell.
2002;2:489 495.
Cheng A, Uetani N, Simoncic PD, Chaubey VP, Lee-Loy A, McGlade
CJ, Kennedy BP, Tremblay ML. Attenuation of leptin action and regulation of obesity by protein tyrosine phosphatase 1B. Dev Cell. 2002;2:
497503.
Bjorbaek C, Elmquist JK, Frantz JD, Shoelson SE, Flier JS. Identification of SOCS-3 as a potential mediator of central leptin resistance.
Mol Cell. 1998;1:619 625.
Howard JK, Cave BJ, Oksanen LJ, Tzameli I, Bjorbaek C, Flier JS.
Enhanced leptin sensitivity and attenuation of diet-induced obesity in
mice with haploinsufficiency of Socs3. Nat Med. 2004;10:734 738.
Mori H, Hanada R, Hanada T, Aki D, Mashima R, Nishinakamura H,
Torisu T, Chien KR, Yasukawa H, Yoshimura A. Socs3 deficiency in
the brain elevates leptin sensitivity and confers resistance to dietinduced obesity. Nat Med. 2004;10:739 743.
Klaman LD, Boss O, Peroni OD, Kim JK, Martino JL, Zabolotny JM,
Moghal N, Lubkin M, Kim YB, Sharpe AH, Stricker-Krongrad A,
Shulman GI, Neel BG, Kahn BB. Increased energy expenditure,
decreased adiposity, and tissue-specific insulin sensitivity in proteintyrosine phosphatase 1B-deficient mice. Mol Cell Biol. 2000;20:
5479 5489.
Munzberg H, Myers MG Jr. Molecular and anatomical determinants of
central leptin resistance. Nat Neurosci. 2005;8:566 570.
Banks WA. Blood-brain barrier and energy balance. Obesity (Silver
Spring). 2006;14(suppl 5):234S237S.
Levin BE, Dunn-Meynell AA, Banks WA. Obesity-prone rats have
normal blood-brain barrier transport but defective central leptin signaling before obesity onset. Am J Physiol Regul Integr Comp Physiol.
2004;286:R143R150.
Munzberg H, Flier JS, Bjorbaek C. Region-specific leptin resistance
within the hypothalamus of diet-induced obese mice. Endocrinology.
2004;145:4880 4889.
Satoh N, Ogawa Y, Katsuura G, Numata Y, Tsuji T, Hayase M, Ebihara
K, Masuzaki H, Hosoda K, Yoshimasa Y, Nakao K. Sympathetic activation of leptin via the ventromedial hypothalamus: leptin-induced
increase in catecholamine secretion. Diabetes. 1999;48:17871793.
Mark AL, Correia ML, Rahmouni K, Haynes WG. Loss of leptin actions
in obesity: two concepts with cardiovascular implications. Clin Exp
Hypertens. 2004;26:629 636.
Mark AL, Shaffer RA, Correia ML, Morgan DA, Sigmund CD, Haynes
WG. Contrasting blood pressure effects of obesity in leptin-deficient
ob/ob mice and agouti yellow obese mice. J Hypertens. 1999;17:
1949 1953.
Correia ML, Haynes WG, Rahmouni K, Morgan DA, Sivitz WI, Mark
AL. The concept of selective leptin resistance: evidence from agouti
yellow obese mice. Diabetes. 2002;51:439 442.
Rahmouni K, Morgan DA, Morgan GM, Mark AL, Haynes WG. Role of
selective leptin resistance in diet-induced obesity hypertension.
Diabetes. 2005;54:20122018.
Haynes WG. Interaction between leptin and sympathetic nervous system
in hypertension. Curr Hypertens Rep. 2000;2:311318.
Barr VA, Lane K, Taylor SI. Subcellular localization and internalization
of the four human leptin receptor isoforms. J Biol Chem. 1999;274:
21416 21424.
Uotani S, Bjorbaek C, Tornoe J, Flier JS. Functional properties of leptin
receptor isoforms: internalization and degradation of leptin and ligandinduced receptor downregulation. Diabetes. 1999;48:279 286.
Sen S, Tarazi RC, Khairallah PA, Bumpus FM. Cardiac hypertrophy in
spontaneously hypertensive rats. Circ Res. 1974;35:775781.
Patel MB, Stewart JM, Loud AV, Anversa P, Wang J, Fiegel L, Hintze
TH. Altered function and structure of the heart in dogs with chronic
elevation in plasma norepinephrine. Circulation. 1991;84:20912100.
Navarro J, Redon J, Cea-Calvo L, Lozano JV, Fernandez-Perez C, Bonet
A, Gonzalez-Esteban J. Metabolic syndrome, organ damage and cardiovascular disease in treated hypertensive patients. the ERIC-HTA study.
Blood Press. 2007;16:20 27.
Cuspidi C, Meani S, Fusi V, Severgnini B, Valerio C, Catini E, Leonetti
G, Magrini F, Zanchetti A. Metabolic syndrome and target organ
damage in untreated essential hypertensives. J Hypertens. 2004;22:
19911998.

Downloaded from http://circres.ahajournals.org/ at VA MED CTR BOISE on March 23, 2016

Yang and Barouch


81. Grassi G, Giannattasio C. Obesity and vascular stiffness: when body fat
has an adverse impact on arterial dynamics. J Hypertens. 2005;23:
1789 1791.
82. Aizawa-Abe M, Ogawa Y, Masuzaki H, Ebihara K, Satoh N, Iwai H,
Matsuoka N, Hayashi T, Hosoda K, Inoue G, Yoshimasa Y, Nakao K.
Pathophysiological role of leptin in obesity-related hypertension. J Clin
Invest. 2000;105:12431252.
83. Winters B, Mo Z, Brooks-Asplund E, Kim S, Shoukas A, Li D, Nyhan
D, Berkowitz DE. Reduction of obesity, as induced by leptin, reverses
endothelial dysfunction in obese (lep(ob)) mice. J Appl Physiol. 2000;
89:23822390.
84. Leyva F, Godsland IF, Ghatei M, Proudler AJ, Aldis S, Walton C,
Bloom S, Stevenson JC. Hyperleptinemia as a component of a metabolic
syndrome of cardiovascular risk. Arterioscler Thromb Vasc Biol. 1998;
18:928 933.
85. Morawietz H, Bornstein SR. Leptin, endothelin, NADPH oxidase, and
heart failure. Hypertension. 2006;47:e20; author reply e20 e21.
86. Shek EW, Brands MW, Hall JE. Chronic leptin infusion increases
arterial pressure. Hypertension. 1998;31:409 414.
87. Correia ML, Morgan DA, Sivitz WI, Mark AL, Haynes WG. Leptin acts
in the central nervous system to produce dose dependent changes in
arterial pressure. Hypertension. 2001;37:936 942.
88. Carlyle M, Jones OB, Kuo JJ, Hall JE. Chronic cardiovascular and renal
actions of leptin: role of adrenergic activity. Hypertension. 2002;39:
496 501.
89. Yamagishi SI, Edelstein D, Du XL, Kaneda Y, Guzman M, Brownlee
M. Leptin induces mitochondrial superoxide production and monocyte
chemoattractant protein-1 expression in aortic endothelial cells by
increasing fatty acid oxidation via protein kinase A. J Biol Chem.
2001;276:25096 25100.
90. Loffreda S, Yang SQ, Lin HZ, Karp CL, Brengman ML, Wang DJ,
Klein AS, Bulkley GB, Bao C, Noble PW, Lane MD, Diehl AM. Leptin
regulates proinflammatory immune responses. FASEB J. 1998;12:
57 65.
91. Fruhbeck G. Pivotal role of nitric oxide in the control of blood pressure
after leptin administration. Diabetes. 1999;48:903908.
92. Kimura K, Tsuda K, Baba A, Kawabe T, Boh-oka S, Ibata M, Moriwaki
C, Hano T, Nishio I. Involvement of nitric oxide in endotheliumdependent arterial relaxation by leptin. Biochem Biophys Res Commun.
2000;273:745749.
93. Lembo G, Vecchione C, Fratta L, Marino G, Trimarco V, dAmati G,
Trimarco B. Leptin induces direct vasodilation through distinct endothelial mechanisms. Diabetes. 2000;49:293297.
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
endothelial nitric oxide is mediated through akt-endothelial nitric oxide
synthase phosphorylation pathway. Diabetes. 2002;51:168 173.
95. Gardiner SM, Kemp PA, March JE, Bennett T. Regional haemodynamic
effects of recombinant murine or human leptin in conscious rats. Br J
Pharmacol. 2000;130:805 810.
96. Mitchell JL, Morgan DA, Correia ML, Mark AL, Sivitz WI, Haynes
WG. Does leptin stimulate nitric oxide to oppose the effects of sympathetic activation? Hypertension. 2001;38:10811086.
97. Kuo JJ, Jones OB, Hall JE. Inhibition of NO synthesis enhances chronic
cardiovascular and renal actions of leptin. Hypertension. 2001;37:
670 676.
98. Quehenberger P, Exner M, Sunder-Plassmann R, Ruzicka K,
Bieglmayer C, Endler G, Muellner C, Speiser W, Wagner O. Leptin
induces endothelin-1 in endothelial cells in vitro. Circ Res. 2002;90:
711718.
99. Park HY, Kwon HM, Lim HJ, Hong BK, Lee JY, Park BE, Jang Y, Cho
SY, Kim HS. Potential role of leptin in angiogenesis: leptin induces
endothelial cell proliferation and expression of matrix metalloproteinases in vivo and in vitro. Exp Mol Med. 2001;33:95102.
100. Kellerer M, Koch M, Metzinger E, Mushack J, Capp E, Haring HU.
Leptin activates PI-3 kinase in C2C12 myotubes via janus kinase-2
(JAK-2) and insulin receptor substrate-2 (IRS-2) dependent pathways.
Diabetologia. 1997;40:1358 1362.
101. Li L, Mamputu JC, Wiernsperger N, Renier G. Signaling pathways
involved in human vascular smooth muscle cell proliferation and matrix
metalloproteinase-2 expression induced by leptin: inhibitory effect of
metformin. Diabetes. 2005;54:22272234.
102. Oda A, Taniguchi T, Yokoyama M. Leptin stimulates rat aortic smooth
muscle cell proliferation and migration. Kobe J Med Sci. 2001;47:
141150.

Leptin Signaling and Obesity

557

103. Nakata M, Yada T, Soejima N, Maruyama I. Leptin promotes aggregation of human platelets via the long form of its receptor. Diabetes.
1999;48:426 429.
104. Maruyama I, Nakata M, Yamaji K. Effect of leptin in platelet and
endothelial cells. Obesity and arterial thrombosis. Ann N Y Acad Sci.
2000;902:315319.
105. Reilly MP, Iqbal N, Schutta M, Wolfe ML, Scally M, Localio AR, Rader
DJ, Kimmel SE. Plasma leptin levels are associated with coronary
atherosclerosis in type 2 diabetes. J Clin Endocrinol Metab. 2004;89:
38723878.
106. Bodary PF, Gu S, Shen Y, Hasty AH, Buckler JM, Eitzman DT.
Recombinant leptin promotes atherosclerosis and thrombosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2005;25:
e119 e122.
107. Wu KK, Wu TJ, Chin J, Mitnaul LJ, Hernandez M, Cai TQ, Ren N,
Waters MG, Wright SD, Cheng K. Increased hypercholesterolemia
and atherosclerosis in mice lacking both ApoE and leptin receptor.
Atherosclerosis. 2005;181:251259.
108. Nickola MW, Wold LE, Colligan PB, Wang GJ, Samson WK, Ren J.
Leptin attenuates cardiac contraction in rat ventricular myocytes. Role of
NO. Hypertension. 2000;36:501505.
109. Wold LE, Relling DP, Duan J, Norby FL, Ren J. Abrogated leptininduced cardiac contractile response in ventricular myocytes under
spontaneous hypertension: role of Jak/STAT pathway. Hypertension.
2002;39:69 74.
110. Rodriguez A, Fruhbeck G, Gomez-Ambrosi J, Catalan V, Sainz N, Diez
J, Zalba G, Fortuno A. The inhibitory effect of leptin on angiotensin
II-induced vasoconstriction is blunted in spontaneously hypertensive
rats. J Hypertens. 2006;24:1589 1597.
111. Barouch LA, Harrison RW, Skaf MW, Rosas GO, Cappola TP, Kobeissi
ZA, Hobai IA, Lemmon CA, Burnett AL, ORourke B, Rodriguez ER,
Huang PL, Lima JA, Berkowitz DE, Hare JM. Nitric oxide regulates the
heart by spatial confinement of nitric oxide synthase isoforms. Nature.
2002;416:337339.
112. Bloch KD, Janssens S. Cardiomyocyte-specific overexpression of nitric
oxide synthase 3: impact on left ventricular function and myocardial
infarction. Trends Cardiovasc Med. 2005;15:249 253.
113. Barouch LA, Cappola TP, Harrison RW, Crone JK, Rodriguez ER,
Burnett AL, Hare JM. Combined loss of neuronal and endothelial nitric
oxide synthase causes premature mortality and age-related hypertrophic
cardiac remodeling in mice. J Mol Cell Cardiol. 2003;35:637 644.
114. Cappola TP, Cope L, Cernetich A, Barouch LA, Minhas K, Irizarry RA,
Parmigiani G, Durrani S, Lavoie T, Hoffman EP, Ye SQ, Garcia JG,
Hare JM. Deficiency of different nitric oxide synthase isoforms activates
divergent transcriptional programs in cardiac hypertrophy. Physiol
Genomics. 2003;14:2534.
115. Illiano G, Naviglio S, Pagano M, Spina A, Chiosi E, Barbieri M,
Paolisso G. Leptin affects adenylate cyclase activity in H9c2 cardiac cell
line: effects of short- and long-term exposure. Am J Hypertens. 2002;
15:638 643.
116. Li SY, Yang X, Ceylan-Isik AF, Du M, Sreejayan N, Ren J. Cardiac
contractile dysfunction in Lep/Lep obesity is accompanied by NADPH
oxidase activation, oxidative modification of sarco(endo)plasmic
reticulum Ca2-ATPase and myosin heavy chain isozyme switch.
Diabetologia. 2006;49:1434 1446.
117. Khan SA, Lee K, Minhas KM, Gonzalez DR, Raju SV, Tejani AD, Li
D, Berkowitz DE, Hare JM. Neuronal nitric oxide synthase negatively regulates xanthine oxidoreductase inhibition of cardiac
excitation-contraction coupling. Proc Natl Acad Sci U S A. 2004;101:
15944 15948.
118. Kinugawa S, Huang H, Wang Z, Kaminski PM, Wolin MS, Hintze TH.
A defect of neuronal nitric oxide synthase increases xanthine oxidasederived superoxide anion and attenuates the control of myocardial
oxygen consumption by nitric oxide derived from endothelial nitric
oxide synthase. Circ Res. 2005;96:355362.
119. Saraiva RM, Minhas KM, Zheng M, Pitz E, Treuer A, Gonzalez D,
Schuleri KH, Vandegaer KM, Barouch LA, Hare JM. Reduced neuronal
nitric oxide synthase expression contributes to cardiac oxidative stress
and nitroso-redox imbalance in ob/ob mice. Nitric Oxide. 2007;16:
331338.
120. Su J, Zhang S, Tse J, Scholz PM, Weiss HR. Alterations in nitric
oxide-cGMP pathway in ventricular myocytes from obese leptindeficient mice. Am J Physiol Heart Circ Physiol. 2003;285:
H2111H2117.

Downloaded from http://circres.ahajournals.org/ at VA MED CTR BOISE on March 23, 2016

558

Circulation Research

September 14, 2007

121. Furukawa S, Fujita T, Shimabukuro M, Iwaki M, Yamada Y, Nakajima


Y, Nakayama O, Makishima M, Matsuda M, Shimomura I. Increased
oxidative stress in obesity and its impact on metabolic syndrome. J Clin
Invest. 2004;114:17521761.
122. Katakam PV, Tulbert CD, Snipes JA, Erdos B, Miller AW, Busija DW.
Impaired insulin-induced vasodilation in small coronary arteries of
zucker obese rats is mediated by reactive oxygen species. Am J Physiol
Heart Circ Physiol. 2005;288:H854 H860.
123. Paolocci N, Ekelund UE, Isoda T, Ozaki M, Vandegaer K, Georgakopoulos D, Harrison RW, Kass DA, Hare JM. cGMP-independent inotropic effects of nitric oxide and peroxynitrite donors: potential role
for nitrosylation. Am J Physiol Heart Circ Physiol. 2000;279:
H1982H1988.
124. Katori T, Donzelli S, Tocchetti CG, Miranda KM, Cormaci G, Thomas
DD, Ketner EA, Lee MJ, Mancardi D, Wink DA, Kass DA, Paolocci N.
Peroxynitrite and myocardial contractility: in vivo versus in vitro effects.
Free Radic Biol Med. 2006;41:1606 1618.
125. Levrand S, Vannay-Bouchiche C, Pesse B, Pacher P, Feihl F, Waeber B,
Liaudet L. Peroxynitrite is a major trigger of cardiomyocyte apoptosis in
vitro and in vivo. Free Radic Biol Med. 2006;41:886 895.
126. Dong F, Zhang X, Ren J. Leptin regulates cardiomyocyte contractile
function through endothelin-1 receptor-NADPH oxidase pathway.
Hypertension. 2006;47:222229.
127. Pieske B, Beyermann B, Breu V, Loffler BM, Schlotthauer K, Maier LS,
Schmidt-Schweda S, Just H, Hasenfuss G. Functional effects of endothelin and regulation of endothelin receptors in isolated human nonfailing and failing myocardium. Circulation. 1999;99:18021809.
128. Unger RH, Orci L. Lipoapoptosis: its mechanism and its diseases.
Biochim Biophys Acta. 2002;1585:202212.
129. Zhou YT, Grayburn P, Karim A, Shimabukuro M, Higa M, Baetens D,
Orci L, Unger RH. Lipotoxic heart disease in obese rats: implications for
human obesity. Proc Natl Acad Sci U S A. 2000;97:1784 1789.
130. Ren J, Relling DP. Leptin-induced suppression of cardiomyocyte contraction is amplified by ceramide. Peptides. 2006;27:14151419.
131. Ren J, Relling DP. Interaction between tumor necrosis factor-alpha and
leptin-induced inhibition of cardiac contractile function in isolated ventricular myocytes. Cytokine. 2005;32:213218.
132. Hynes GR, Jones PJ. Leptin and its role in lipid metabolism. Curr Opin
Lipidol. 2001;12:321327.
133. Kudo N, Barr AJ, Barr RL, Desai S, Lopaschuk GD. High rates of fatty
acid oxidation during reperfusion of ischemic hearts are associated with
a decrease in malonyl-CoA levels due to an increase in 5-AMP-activated protein kinase inhibition of acetyl-CoA carboxylase. J Biol Chem.
1995;270:1751317520.
134. Muoio DM, Seefeld K, Witters LA, Coleman RA. AMP-activated kinase
reciprocally regulates triacylglycerol synthesis and fatty acid oxidation
in liver and muscle: evidence that sn-glycerol-3-phosphate acyltransferase is a novel target. Biochem J. 1999;338(pt 3):783791.
135. Atkinson LL, Fischer MA, Lopaschuk GD. Leptin activates cardiac fatty
acid oxidation independent of changes in the AMP-activated protein
kinase-acetyl-CoA carboxylase-malonyl-CoA axis. J Biol Chem. 2002;
277:29424 29430.
136. Somoza B, Guzman R, Cano V, Merino B, Ramos P, Diez-Fernandez C,
Fernandez-Alfonso MS, Ruiz-Gayo M. Induction of cardiac uncoupling
protein-2 expression and adenosine 5-monophosphate-activated protein
kinase phosphorylation during early states of diet-induced obesity in
mice. Endocrinology. 2007;148:924 931.
137. Lee Y, Wang MY, Kakuma T, Wang ZW, Babcock E, McCorkle K,
Higa M, Zhou YT, Unger RH. Liporegulation in diet-induced obesity.
The antisteatotic role of hyperleptinemia. J Biol Chem. 2001;276:
5629 5635.
138. Lee Y, Naseem RH, Duplomb L, Park BH, Garry DJ, Richardson JA,
Schaffer JE, Unger RH. Hyperleptinemia prevents lipotoxic cardiomyopathy in acyl CoA synthase transgenic mice. Proc Natl Acad Sci U S A.
2004;101:13624 13629.
139. Murphy S, Frishman WH. Protein kinase C in cardiac disease and as a
potential therapeutic target. Cardiol Rev. 2005;13:312.
140. Christoffersen C, Bollano E, Lindegaard ML, Bartels ED, Goetze JP,
Andersen CB, Nielsen LB. Cardiac lipid accumulation associated with
diastolic dysfunction in obese mice. Endocrinology. 2003;144:
34833490.
141. Mazumder PK, ONeill BT, Roberts MW, Buchanan J, Yun UJ,
Cooksey RC, Boudina S, Abel ED. Impaired cardiac efficiency and
increased fatty acid oxidation in insulin-resistant ob/ob mouse hearts.
Diabetes. 2004;53:2366 2374.

142. Warnes CA, Roberts WC. Sudden coronary death: relation of amount
and distribution of coronary narrowing at necropsy to previous
symptoms of myocardial ischemia, left ventricular scarring and heart
weight. Am J Cardiol. 1984;54:6573.
143. Alpert MA. Obesity cardiomyopathy: pathophysiology and evolution of
the clinical syndrome. Am J Med Sci. 2001;321:225236.
144. Kasper EK, Hruban RH, Baughman KL. Cardiomyopathy of obesity: a
clinicopathologic evaluation of 43 obese patients with heart failure. Am J
Cardiol. 1992;70:921924.
145. Sader S, Nian M, Liu P. Leptin: a novel link between obesity, diabetes,
cardiovascular risk, and ventricular hypertrophy. Circulation. 2003;108:
644 646.
146. Xu FP, Chen MS, Wang YZ, Yi Q, Lin SB, Chen AF, Luo JD. Leptin
induces hypertrophy via endothelin-1-reactive oxygen species pathway
in cultured neonatal rat cardiomyocytes. Circulation. 2004;110:
1269 1275.
147. Paolisso G, Tagliamonte MR, Galderisi M, Zito GA, Petrocelli A,
Carella C, de Divitiis O, Varricchio M. Plasma leptin level is associated
with myocardial wall thickness in hypertensive insulinresistant men.
Hypertension. 1999;34:10471052.
148. Yamashita T, Murakami T, Iida M, Kuwajima M, Shima K. Leptin
receptor of zucker fatty rat performs reduced signal transduction.
Diabetes. 1997;46:10771080.
149. Nakamura K, Fushimi K, Kouchi H, Mihara K, Miyazaki M, Ohe T,
Namba M. Inhibitory effects of antioxidants on neonatal rat cardiac
myocyte hypertrophy induced by tumor necrosis factor-alpha and angiotensin II. Circulation. 1998;98:794 799.
150. Luo JD, Xie F, Zhang WW, Ma XD, Guan JX, Chen X. Simvastatin
inhibits noradrenaline-induced hypertrophy of cultured neonatal rat cardiomyocytes. Br J Pharmacol. 2001;132:159 164.
151. Adiarto S, Emoto N, Iwasa N, Yokoyama M. Obesity-induced upregulation of myocardial endothelin-1 expression is mediated by leptin.
Biochem Biophys Res Commun. 2007;353:623 627.
152. Hu TP, Xu FP, Li YJ, Luo JD. Simvastatin inhibits leptin-induced
hypertrophy in cultured neonatal rat cardiomyocytes. Acta Pharmacol
Sin. 2006;27:419 422.
153. Williams SD, Zhu H, Zhang L, Bernstein HS. Adenoviral delivery of
human CDC5 promotes G2/M progression and cell division in neonatal
ventricular cardiomyocytes. Gene Ther. 2006;13:837 843.
154. Barouch LA, Gao D, Chen L, Miller KL, Xu W, Phan AC, Kittleson
MM, Minhas KM, Berkowitz DE, Wei C, Hare JM. Cardiac myocyte
apoptosis is associated with increased DNA damage and decreased
survival in murine models of obesity. Circ Res. 2006;98:119 124.
155. Unger RH, Zhou YT, Orci L. Regulation of fatty acid homeostasis in
cells: novel role of leptin. Proc Natl Acad Sci U S A. 1999;96:
23272332.
156. Mascareno E, Beckles DL, Siddiqui MA. Janus kinase-2 signaling
mediates apoptosis in rat cardiomyocytes. Vascul Pharmacol. 2005;43:
327335.
157. Finck BN, Lehman JJ, Leone TC, Welch MJ, Bennett MJ, Kovacs A,
Han X, Gross RW, Kozak R, Lopaschuk GD, Kelly DP. The cardiac
phenotype induced by PPARalpha overexpression mimics that caused
by diabetes mellitus. J Clin Invest. 2002;109:121130.
158. Kodama H, Fukuda K, Pan J, Makino S, Sano M, Takahashi T, Hori S,
Ogawa S. Biphasic activation of the JAK/STAT pathway by angiotensin
II in rat cardiomyocytes. Circ Res. 1998;82:244 250.
159. Maroni P, Bendinelli P, Piccoletti R. Intracellular signal transduction
pathways induced by leptin in C2C12 cells. Cell Biol Int. 2005;29:
542550.
160. Madani S, De Girolamo S, Munoz DM, Li RK, Sweeney G. Direct
effects of leptin on size and extracellular matrix components of human
pediatric ventricular myocytes. Cardiovasc Res. 2006;69:716 725.
161. Hausenloy DJ, Tsang A, Yellon DM. The reperfusion injury salvage
kinase pathway: a common target for both ischemic preconditioning and
postconditioning. Trends Cardiovasc Med. 2005;15:69 75.
162. Brzozowski T, Konturek PC, Pajdo R, Kwiecien S, Ptak A, Sliwowski
Z, Drozdowicz D, Pawlik M, Konturek SJ, Hahn EG. Brain-gut axis in
gastroprotection by leptin and cholecystokinin against ischemiareperfusion induced gastric lesions. J Physiol Pharmacol. 2001;52:
583 602.
163. Erkasap S, Erkasap N, Koken T, Kahraman A, Uzuner K, Yazihan N,
Ates E. Effect of leptin on renal ischemia-reperfusion damage in rats.
J Physiol Biochem. 2004;60:79 84.
164. Kennedy LM, Dickstein K, Anker SD, Kristianson K, Willenheimer R,
OPTIMAAL Study Group. The prognostic importance of body mass

Downloaded from http://circres.ahajournals.org/ at VA MED CTR BOISE on March 23, 2016

Yang and Barouch

165.

166.

167.

168.

169.

index after complicated myocardial infarction. J Am Coll Cardiol. 2005;


45:156 158.
Nikolsky E, Stone GW, Grines CL, Cox DA, Garcia E, Tcheng JE, Griffin
JJ, Guagliumi G, Stuckey T, Turco M, Negoita M, Lansky AJ, Mehran R.
Impact of body mass index on outcomes after primary angioplasty in acute
myocardial infarction. Am Heart J. 2006;151:168175.
Knudson JD, Dincer UD, Dick GM, Shibata H, Akahane R, Saito M,
Tune JD. Leptin resistance extends to the coronary vasculature in
prediabetic dogs and provides a protective adaptation against endothelial dysfunction. Am J Physiol Heart Circ Physiol. 2005;289:
H1038 H1046.
Chen K, Li F, Li J, Cai H, Strom S, Bisello A, Kelley DE,
Friedman-Einat M, Skibinski GA, McCrory MA, Szalai AJ, Zhao AZ.
Induction of leptin resistance through direct interaction of C-reactive
protein with leptin. Nat Med. 2006;12:425 432.
Kazumi T, Kawaguchi A, Hirano T, Yoshino G. C-reactive protein in
young, apparently healthy men: associations with serum leptin, QTc
interval, and high-density lipoprotein-cholesterol. Metabolism. 2003;52:
11131116.
Scarpace PJ, Matheny M, Zhang Y, Cheng KY, Tumer N. Leptin
antagonist reveals an uncoupling between leptin receptor signal
transducer and activator of transcription 3 signaling and metabolic
responses with central leptin resistance. J Pharmacol Exp Ther. 2007;
320:706 712.

Leptin Signaling and Obesity

559

170. Zhang J, Matheny MK, Tumer N, Mitchell MK, Scarpace PJ. Leptin
antagonist reveals that the normalization of caloric intake and the
thermic effect of food after high-fat feeding are leptin dependent. Am J
Physiol Regul Integr Comp Physiol. 2007;292:R868 R874.
171. Matarese G, Carrieri PB, La Cava A, Perna F, Sanna V, De Rosa V,
Aufiero D, Fontana S, Zappacosta S. Leptin increase in multiple
sclerosis associates with reduced number of CD4()CD25 regulatory
T cells. Proc Natl Acad Sci U S A. 2005;102:5150 5155.
172. De Rosa V, Procaccini C, La Cava A, Chieffi P, Nicoletti GF, Fontana
S, Zappacosta S, Matarese G. Leptin neutralization interferes with
pathogenic T cell autoreactivity in autoimmune encephalomyelitis.
J Clin Invest. 2006;116:447 455.
173. Purdham DM, Rajapurohitam V, Huang C, Karmazyn M. Attenuation of
cardiac hypertrophy and heart failure by leptin receptor blockade.
Circulation. 2005;112(suppl II):II-279 Abstract.
174. Gertler A. Development of leptin antagonists and their potential use in
experimental biology and medicine. Trends Endocrinol Metab. 2006;
17:372378.
175. Eckel RH, Barouch WW, Ershow AG. Report of the national heart,
lung, and blood institute-national institute of diabetes and digestive
and kidney diseases working group on the pathophysiology of
obesity-associated cardiovascular disease. Circulation. 2002;105:
29232928.

Downloaded from http://circres.ahajournals.org/ at VA MED CTR BOISE on March 23, 2016

Leptin Signaling and Obesity: Cardiovascular Consequences


Ronghua Yang and Lili A. Barouch
Circ Res. 2007;101:545-559
doi: 10.1161/CIRCRESAHA.107.156596
Circulation Research is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231
Copyright 2007 American Heart Association, Inc. All rights reserved.
Print ISSN: 0009-7330. Online ISSN: 1524-4571

The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://circres.ahajournals.org/content/101/6/545

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published
in Circulation Research can be obtained via RightsLink, a service of the Copyright Clearance Center, not the
Editorial Office. Once the online version of the published article for which permission is being requested is
located, click Request Permissions in the middle column of the Web page under Services. Further information
about this process is available in the Permissions and Rights Question and Answer document.
Reprints: Information about reprints can be found online at:
http://www.lww.com/reprints
Subscriptions: Information about subscribing to Circulation Research is online at:
http://circres.ahajournals.org//subscriptions/

Downloaded from http://circres.ahajournals.org/ at VA MED CTR BOISE on March 23, 2016

Das könnte Ihnen auch gefallen