circ res-2007-yang-545-59

Upload: jessica

Post on 07-Jul-2018

223 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/18/2019 Circ Res-2007-Yang-545-59

    1/16

    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 ConsequencesRonghua Yang, Lili A. Barouch

     Abstract—Leptin, among the best known hormone markers for obesity, exerts pleiotropic actions on multiple organ

    systems. In this review, we summarize major leptin signaling pathways, namely Janus-activated kinase/signal

    transducers and activators of transcription and mitogen-activated protein kinase, including possible mechanisms of 

    leptin resistance in obesity. The effects of leptin on the cardiovascular system are discussed in detail, including its

    contributions to hypertension, atherosclerosis, depressed myocardial contractile function, fatty acid metabolism,

    hypertrophic remodeling, and reduction of ischemic/reperfusion injury. The overall goal is to summarize current

    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

    Over 60% of people in the United States are overweight orobese. Extensive evidence now supports the notion thatmaladaptation of the biological system for weight mainte-

    nance 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 adipocytokinesthat 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 popu-lation 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 [email protected]

    © 2007 American Heart Association, Inc.Circulation Research   is available at http:// circres.ahajournals.org DOI: 10.1161/CIRCRESAHA.107.156596

     545

    Review

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

    http://circres.ahajournals.org/http://circres.ahajournals.org/http://circres.ahajournals.org/

  • 8/18/2019 Circ Res-2007-Yang-545-59

    2/16

    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 Emerg-

    ing 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 reproduc-

    tion, 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 pa-

    tient 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 expressionand function of peripheral organs, the relative contributions

    of central versus peripheral signal disruption are still contro-

    versial. 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 ReceptorLeptin 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 reperfu-

    sion after ischemia,24,25 and leptin concentration in cardio-

    myocyte 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 themurine 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 andActivators of TranscriptionOb-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-characterizedpathway in leptin signaling is the JAK/STAT pathway (Fig-

    ure 1).31 Ligand binding causes Ob-R to undergo homooli-

    gomerization32,33 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 ob-

    served with leptin treatment.35 However, only Ob-Rb con-

    tains 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 Phos-

    phorylation of Tyr1138 recruits STAT proteins to the Ob-Rb/ JAK2 complex. Tyrosine phosphorylated STAT3 molecules

    Figure 1.  Leptin receptor signaling. The binding of leptin to its receptor leads to formation of the Ob-R/JAK2 complex that results incross-phosphorylation. Tyr1138 on Ob-Rb is crucial for STAT3 activation, which stimulates SOCS3 expression that negatively inhibitsleptin signaling via Tyr985 and additional sites on JAK2. Protein tyrosine phosphatase 1B (PTP1B) is also capable of inhibition of leptinsignaling. 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.

     546 Circulation Research   September 14, 2007

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

    http://circres.ahajournals.org/http://circres.ahajournals.org/

  • 8/18/2019 Circ Res-2007-Yang-545-59

    3/16

  • 8/18/2019 Circ Res-2007-Yang-545-59

    4/16

    Increasing evidence suggests that leptin signaling is prefer-

    entially reduced in the arcuate nucleus of the hypothalamus

    but not in other regions, such as the ventromedial, dorsome-dial, and/or premammillary nucleus of the hypothalamus that

    also express Ob-Rs.68

    Leptin Increases Sympathetic OutflowIn addition to reducing appetite and controlling weight gain,

    leptin centrally activates the sympathetic nervous system. It

    significantly increases plasma norepinephrine and epineph-

    rine concentrations via the ventromedial hypothalamus.69

    Whereas chronic leptin overstimulation in the hypothalamus

    decreases the ability of leptin to regulate appetite, its sympa-

    thetic excitatory effects are maintained as increased arterial

    pressure and renal sympathetic nerve activity are presented in

    obesity (Figure 2).70 This observation suggests that centralleptin 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 beenconfirmed 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 increasedrenal 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 circu-

    latory 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 dis-

    ruption. Under resting conditions, it is estimated that only 5%

    to 25% of Ob-R isoforms are located at the cell surface.75 The

    ligand–receptor complex internalizes, and studies have shownthat leptin internalization was greater for the Ob-Rb iso-

    form.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 modu-lates the elastic properties of large and medium-size arteries

    Figure 2.  Systemic leptin function. Chronic hyperleptinemia impairs the centrally mediated metabolic actions of the hormone, alyhoughits activation of sympathetic outflow is preserved. Selective central leptin resistance results in obesity and adverse effects on the car-diovascular system including hypertension, atherosclerosis, and LVH. Although leptin can protect against ectopic lipid deposition innonadipose tissue, whether this effect is abolished because of (selective) peripheral leptin resistance requires further examination.

     548 Circulation Research   September 14, 2007

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

    http://circres.ahajournals.org/http://circres.ahajournals.org/

  • 8/18/2019 Circ Res-2007-Yang-545-59

    5/16

  • 8/18/2019 Circ Res-2007-Yang-545-59

    6/16

    Leptin Attenuates Cardiomyocyte Contractility

     Possible Mechanisms Leading to Increased  NO ProductionSimilar 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 N G-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 upregula-

    tion 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 under-

    standing of its role in myocardial contractility and hypertro-

    phy responses. We have shown that spatial confinement of 

    different constitutive NOS isoforms within separate subcel-

    lular compartments of the cardiac myocyte allows NO signals

    to have independent, and even opposite, effects on cardiacphenotype and contractile response.111 Overexpression of 

    eNOS inhibits hypertrophy in the remote myocardium and

    preserved cardiac function after myocardial infarction, pos-

    sibly through attenuation of   -adrenergic–stimulated com-

    pensatory hypertrophy.112 Neuronal NO synthase and eNOS

    independently contribute to the development of cardiac hy-

    pertrophy, leading to marked age-related concentric hyper-

    trophic 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 in-creased 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 depres-

    sant effect may become important when considered in con-

     juncture with alterations occurring in obese states.

     Leptin Deficiency Leads to Decreased Responsiveness to-Adrenergic StimulationIn 10 week-old   ob/ob   isolated myocytes, attenuated sarco-

    mere shortening and calcium transients and depressed sarco-

    plasmic reticulum Ca2

    stores were seen in response toisoproterenol stimulation of the  -adrenergic receptor or to

    Figure 3.  Leptin and myocardial contractility. Leptin directly depresses cardiomyocyte contractility. The signaling pathways implicatedin this process include the NO-cGMP pathway and pathways that lead to increased ROS production. Changes that occur in a chronicleptin-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.

     550 Circulation Research   September 14, 2007

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

    http://circres.ahajournals.org/http://circres.ahajournals.org/

  • 8/18/2019 Circ Res-2007-Yang-545-59

    7/16

    post–receptor 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 phos-

    phorylated 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 catechol-

    amine-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 in-

    creased 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 inmyocytes, as Ob-R expression is seen to decrease at 18-hour

    leptin treatment.115 Taken together, leptin deficiency or resis-

    tance leads to decreased   -adrenergic response, whereas

    moderate leptin stimulation can improve the contractile

    response.

     Mechanisms Involving ROSMitochondrial formation of ROS is enhanced in obesity.

    Xanthine oxidoreductase and nicotinamide adenine dinucle-

    otide 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 cardiaccontractile function is accompanied by elevated oxidative

    stress, lipid peroxidation, protein carbonyl formation, redis-

    tribution 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 nitroso–redox 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 dys-

    function in  ob/ob  myocytes but failed to reserve SERCA2a

    oxidative modification.116 8-Bromo-cGMP, a membrane-

    permeable 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 pre-

    vents the enhanced negative cGMP effects in  ob/ob  cardio-

    myocytes.120 NAPDH-mediated reduction in NO bioavail-

    ability could explain the failure of NO donor to elicit further

    negative inotropic response in obese models.121,122 Addition-

    ally, the interaction between NO and ROS produces per-oxynitrite, which can nitrosylate proteins and exert positive

    inotropic effects, thereby offsetting the cGMP-dependent

    reduction in contractility.120 Peroxynitrite has shown both

    negative and positive inotropy in isolated cardiomyo-

    cytes,123,124 although it is generally accepted to trigger apo-

    ptosis 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, ETAand ETB. Both are known to mediate cardiomyocyte inotropic

    response, and ETA receptors also affect hypertrophy.127 Lep-

    tin administration to rat neonatal cardiomyocytes induced

    intracellular O2. generation and upregulated protein expres-

    sion of p67 phox  and p47 phox  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 con-

    tractile response.126

     Additional Mechanisms Affecting ContractilityObesity 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 ef-

    fects on myocyte shortening and intracellular Ca2 transients.

    In vivo obese concentrations of plasma leptin lie in the lownanomolar 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 hyperleptinemia-

    associated cardiac dysfunction in obesity.

    Elevated adipose mass in obesity also increases the secre-

    tion 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  N G-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 depres-

    sion. 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 

    Yang and Barouch Leptin Signaling and Obesity   551

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

    http://circres.ahajournals.org/http://circres.ahajournals.org/

  • 8/18/2019 Circ Res-2007-Yang-545-59

    8/16

  • 8/18/2019 Circ Res-2007-Yang-545-59

    9/16

    Cardiomyocyte HypertrophyLeptin-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 par-

    tially 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 oxida-

    tive 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 II–induced cardiomyocyte hypertrophy. Treat-

    ment 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-inducedhypertrophy, 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 expres-

    sion 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 cho-

    lesterol-lowering drug decreases leptin-induced ROS-

    mediated hypertrophy in rat neonatal cardiomyocytes.152

     ApoptosisOne 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 lipotoxicity-

    induced apoptosis.

    On the other hand, JAK2 has been suggested as a mediatorof the apoptotic response in cardiomyocytes.156 It is promi-

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

    and Ang II–treated 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 transcrip-

    tion factors of p38 may be involved in the long-term

    maladaptive cardiac remodeling in obese HF patients.48

     Mitosis and ProliferationLeptin treatment at a level similar to plasma concentration in

    obese individuals increased proliferation of both HL-1 car-

    diac muscle cells and human pediatric ventricular myocytes.43

    The proliferation was accompanied by increased DNA syn-

    thesis associated with increased ERK1/2 phosphorylation and

    increased association of the p85 regulatory subunit of PI3K

    with phosphotyrosine immunoprecipitates.43 ERK1/2 inhibi-

    tion significantly attenuated the leptin-induced proliferativeactivity and DNA replication in HL-1 and pediatric human

    cardiomyocytes43 but failed to decrease [3H]-leucine incorpo-

    ration in neonatal rat cardiomyocytes treated with leptin.48

    Other pathways likely involved in leptin-induced hypertro-

    phy include the activation of adenylate cyclase,115 peroxi-

    some 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 cardio-

    myocytes in response to leptin and the specific isoforms

    involved mandate further research.

     Extracellular Matrix RemodelingLeptin 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 cardiomyo-

    cytes.160 This suggests that leptin selectively regulates differ-

    ent 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 InjuryTimely 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 asso-

    ciated with activation of the reperfusion injury salvage kinase

    pathway that includes PI3K/Akt and ERK1/2, ultimately

    leading to the inhibition of mitochondrial permeability tran-

    sition 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 reperfu-

    sion, whereas Ob-Rb expression was downregulated.24 PI3K

    or ERK1/2 inhibition diminished the cardioprotective ef-fect.24 Interestingly, leptin did not increase phosphorylation

    Yang and Barouch Leptin Signaling and Obesity   553

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

    http://circres.ahajournals.org/http://circres.ahajournals.org/

  • 8/18/2019 Circ Res-2007-Yang-545-59

    10/16

    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 synthe-

    sis has been shown also to protect against ischemia reperfu-

    sion 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 theCardiovascular 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 systemhas 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 receptorlevel. 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 attenu-

    ated 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 leptin-

    related effects on the myocardium; that is, excessive exoge-

    nous leptin and leptin deficiency often lead to the same end.

    Whether these effects occur through entirely different mech-

    anisms, are mediated through differential regulation of Ob-R

    isoforms, or are attributable to peripheral resistance requires

    further investigation. If peripheral myocardial resistance doesoccur, 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 inthe vasculature. This resistance is not attributable to altered

    coronary dilation, increased endothelium-derived hyperpolar-

    izing 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 leptin–interacting 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 con-

    centrations of leptin stimulate expression of C-reactive pro-

    tein 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 nega-

    tive feedback that may cause leptin resistance in the obese

    state.167

    Leptin AntagonistsLeptin antagonists used in animal models have been shown to

    block central leptin effects and increase appetite and foodintake.169,170 Three approaches have been employed to antag-

    onize 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 anti–Ob-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 ef-

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

    ited by leptin, Ang II, or ET-1.26 The hypertrophic effects of 

    leptin are also prevented by antibodies to Ob-Ra and Ob-

    Rb.26 Cardiac dysfunction did not develop in rats treated withOb-R antibodies after coronary artery ligation compared with

    sham, indicating that blocking Ob-R can improve postinfarc-

    tion 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.

    SummaryObesity 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 re-

    sponse. 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. Interac-

    tion with enhanced ROS production pathways in obesity, onthe other hand, can cause lipotoxicity and deleterious myo-

     554 Circulation Research   September 14, 2007

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

    http://circres.ahajournals.org/http://circres.ahajournals.org/

  • 8/18/2019 Circ Res-2007-Yang-545-59

    11/16

    cardial effects such as cell death and maladaptive hypertro-

    phy. Perturbations of leptin signaling and other signal trans-

    duction 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 car-

    diomyocytes via SOCS3, PI3K/Akt, protein kinase C, and

    other signaling pathways could provide a more comprehen-

    sive 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 exper-

    imental 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 FundingThis work was supported in part by the Donald W. ReynoldsFoundation, NIH grant K08-HL076220, the W.W. Smith CharitableTrust, and the Irvin Talles Endowed Fund for CardiomyopathyResearch.

    DisclosuresNone.

    References1. Friedman JM. A war on obesity, not the obese.   Science. 2003;299:

    856–858.

    2. O’Rahilly S, Farooqi IS, Yeo GS, Challis BG. Minireview: Human

    obesity-lessons from monogenic disorders.  Endocrinology. 2003;144:

    3757–3764.

    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;19:525–546.

    5. Friedman JM, Halaas JL. Leptin and the regulation of body weight in

    mammals. Nature. 1998;395:763–770.

    6. Elmquist JK, Elias CF, Saper CB. From lesions to leptin: hypothalamic

    control of food intake and body weight.  Neuron. 1999;22:221–232.

    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;71:345–372.

    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, O’Rahilly S. Congenital leptin defi-

    ciency is associated with severe early-onset obesity in humans.  Nature.

    1997;387:903–908.

    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,

    O’Rahilly S. Beneficial effects of leptin on obesity, T cell hyporespon-

    siveness, and neuroendocrine/metabolic dysfunction of human con-

    genital leptin deficiency.  J Clin Invest . 2002;110:1093–1103.

    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-reducedsubjects. Nat Med . 1995;1:1155–1161.

    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;334:292–295.

    13. Strobel A, Issad T, Camoin L, Ozata M, Strosberg AD. A leptin

    missense mutation associated with hypogonadism and morbid obesity.

     Nat Genet . 1998;18:213–215.

    14. Frederich RC, Hamann A, Anderson S, Lollmann B, Lowell BB, Flier

    JS. Leptin levels reflect body lipid content in mice: evidence for diet-induced resistance to leptin action.  Nat Med . 1995;1:1311–1314.

    15. Cohen P, Zhao C, Cai X, Montez JM, Rohani SC, Feinstein P, Mom-

    baerts P, Friedman JM. Selective deletion of leptin receptor in neurons

    leads to obesity. J Clin Invest . 2001;108:1113–1121.

    16. Bates SH, Myers MG. The role of leptin-STAT3 signaling in neu-

    roendocrine function: an integrative perspective.  J Mol Med . 2004;82:

    12–20.

    17. Mark AL, Correia ML, Rahmouni K, Haynes WG. Selective leptin

    resistance: a new concept in leptin physiology with cardiovascular

    implications.  J Hypertens. 2002;20:1245–1250.

    18. Lord GM, Matarese G, Howard JK, Baker RJ, Bloom SR, Lechler RI.

    Leptin modulates the T-cell immune response and reverses starvation-

    induced immunosuppression. Nature. 1998;394:897–901.

    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;26:1407–1433.20. Rahmouni K, Haynes WG. Leptin and the cardiovascular system. Recent 

    Prog Horm Res. 2004;59:225–244.

    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:

    1277–1282.

    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;130:3127S–3131S.

    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;149:5–13.

    25. Matsui H, Motooka M, Koike H, Inoue M, Iwasaki T, Suzuki T,

    Kurabayashi M, Yokoyama T. Ischemia/reperfusion in rat heart inducesleptin 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 cardiomyo-

    cyte hypertrophic effects of angiotensin II and endothelin-1.  J Mol Cell

    Cardiol. 2006;41:265–274.

    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:

    7001–7005.

    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 invitro 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;14:95–97.

    32. White DW, Kuropatwinski KK, Devos R, Baumann H, Tartaglia LA.

    Leptin receptor (OB-R) signaling. Cytoplasmic domain mutational anal-

    ysis and evidence for receptor homo-oligomerization.   J Biol Chem.

    1997;272:4065–4071.

    33. Nakashima K, Narazaki M, Taga T. Leptin receptor (OB-R) oli-

    gomerizes 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 BiolChem. 1997;272:32686–32695.

    Yang and Barouch Leptin Signaling and Obesity   555

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

    http://circres.ahajournals.org/http://circres.ahajournals.org/

  • 8/18/2019 Circ Res-2007-Yang-545-59

    12/16

    36. Banks AS, Davis SM, Bates SH, Myers MG Jr. Activation of down-

    stream signals by the long form of the leptin receptor.   J Biol Chem.

    2000;275:14563–14572.

    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;486:33–37.

    38. Hekerman P, Zeidler J, Bamberg-Lemper S, Knobelspies H, Lavens D,

    Tavernier J, Joost HG, Becker W. Pleiotropy of leptin receptor sig-

    nalling 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;19:925–938.

    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;

    51:275–283.

    43. Tajmir P, Ceddia RB, Li RK, Coe IR, Sweeney G. Leptin increasescardiomyocyte hyperplasia via extracellular signal-regulated kinase-

    and 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 asso-

    ciation 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, O’donnell 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;273:2161–2168.

    48. Rajapurohitam V, Gan XT, Kirshenbaum LA, Karmazyn M. The

    obesity-associated peptide leptin induces hypertrophy in neonatal rat

    ventricular myocytes.  Circ Res. 2003;93:277–279.

    49. Shin HJ, Oh J, Kang SM, Lee JH, Shin MJ, Hwang KC, Jang Y, Chung

    JH. Leptin induces hypertrophy via p38 mitogen–activated 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 oxi-

    dative stress in human endothelial cells.  FASEB J . 1999;13:1231–1238.

    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;77:1502–1515.52. Fruhbeck G. Intracellular signalling pathways activated by leptin.

     Biochem J . 2006;393:7–20.

    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

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

    U S A. 1996;93:8374–8378.

    59. 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.

    60. Cheng A, Uetani N, Simoncic PD, Chaubey VP, Lee-Loy A, McGlade

    CJ, Kennedy BP, Tremblay ML. Attenuation of leptin action and regu-

    lation of obesity by protein tyrosine phosphatase 1B.  Dev Cell. 2002;2:

    497–503.

    61. Bjorbaek C, Elmquist JK, Frantz JD, Shoelson SE, Flier JS. Identifi-

    cation of SOCS-3 as a potential mediator of central leptin resistance.

     Mol Cell. 1998;1:619–625.

    62. 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.

    63. 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 diet-

    induced obesity.  Nat Med . 2004;10:739–743.

    64. 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 protein-

    tyrosine phosphatase 1B-deficient mice.   Mol Cell Biol. 2000;20:

    5479–5489.

    65. Munzberg H, Myers MG Jr. Molecular and anatomical determinants of 

    central leptin resistance.  Nat Neurosci.   2005;8:566–570.

    66. Banks WA. Blood-brain barrier and energy balance.   Obesity (Silver 

    Spring).  2006;14(suppl 5):234S–237S.

    67. Levin BE, Dunn-Meynell AA, Banks WA. Obesity-prone rats have

    normal blood-brain barrier transport but defective central leptin sig-

    naling before obesity onset. Am J Physiol Regul Integr Comp Physiol .

    2004;286:R143–R150.

    68. Munzberg H, Flier JS, Bjorbaek C. Region-specific leptin resistance

    within the hypothalamus of diet-induced obese mice.   Endocrinology.

    2004;145:4880–4889.

    69. Satoh N, Ogawa Y, Katsuura G, Numata Y, Tsuji T, Hayase M, Ebihara

    K, Masuzaki H, Hosoda K, Yoshimasa Y, Nakao K. Sympathetic acti-

    vation of leptin via the ventromedial hypothalamus: leptin-induced

    increase in catecholamine secretion.  Diabetes. 1999;48:1787–1793.

    70. Mark AL, Correia ML, Rahmouni K, Haynes WG. Loss of leptin actionsin obesity: two concepts with cardiovascular implications.   Clin Exp

     Hypertens. 2004;26:629–636.

    71. 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.

    72. 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.

    73. Rahmouni K, Morgan DA, Morgan GM, Mark AL, Haynes WG. Role of 

    selective leptin resistance in diet-induced obesity hypertension.

     Diabetes. 2005;54:2012–2018.

    74. Haynes WG. Interaction between leptin and sympathetic nervous system

    in hypertension.   Curr Hypertens Rep. 2000;2:311–318.

    75. Barr VA, Lane K, Taylor SI. Subcellular localization and internalization

    of the four human leptin receptor isoforms.   J Biol Chem. 1999;274:21416–21424.

    76. Uotani S, Bjorbaek C, Tornoe J, Flier JS. Functional properties of leptin

    receptor isoforms: internalization and degradation of leptin and ligand-

    induced receptor downregulation.  Diabetes. 1999;48:279–286.

    77. Sen S, Tarazi RC, Khairallah PA, Bumpus FM. Cardiac hypertrophy in

    spontaneously hypertensive rats.  Circ Res. 1974;35:775–781.

    78. 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;84:2091–2100.

    79. Navarro J, Redon J, Cea-Calvo L, Lozano JV, Fernandez-Perez C, Bonet

    A, Gonzalez-Esteban J. Metabolic syndrome, organ damage and cardio-

    vascular disease in treated hypertensive patients. the ERIC-HTA study.

     Blood Press. 2007;16:20–27.

    80. 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;22:1991–1998.

     556 Circulation Research   September 14, 2007

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

    http://circres.ahajournals.org/http://circres.ahajournals.org/

  • 8/18/2019 Circ Res-2007-Yang-545-59

    13/16

  • 8/18/2019 Circ Res-2007-Yang-545-59

    14/16

    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;114:1752–1761.

    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, Georgako-

    poulos D, Harrison RW, Kass DA, Hare JM. cGMP-independent ino-

    tropic effects of nitric oxide and peroxynitrite donors: potential role

    for nitrosylation.   Am J Physiol Heart Circ Physiol. 2000;279:

    H1982–H1988.

    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;47:222–229.

    127. Pieske B, Beyermann B, Breu V, Loffler BM, Schlotthauer K, Maier LS,

    Schmidt-Schweda S, Just H, Hasenfuss G. Functional effects of endo-

    thelin and regulation of endothelin receptors in isolated human non-

    failing and failing myocardium. Circulation. 1999;99:1802–1809.

    128. Unger RH, Orci L. Lipoapoptosis: its mechanism and its diseases.

     Biochim Biophys Acta. 2002;1585:202–212.

    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 con-

    traction is amplified by ceramide. Peptides. 2006;27:1415–1419.

    131. Ren J, Relling DP. Interaction between tumor necrosis factor-alpha and

    leptin-induced inhibition of cardiac contractile function in isolated ven-

    tricular myocytes.  Cytokine. 2005;32:213–218.

    132. Hynes GR, Jones PJ. Leptin and its role in lipid metabolism. Curr Opin

     Lipidol. 2001;12:321–327.

    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-acti-vated protein kinase inhibition of acetyl-CoA carboxylase.  J Biol Chem.

    1995;270:17513–17520.

    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 acyltrans-

    ferase is a novel target.   Biochem J . 1999;338(pt 3):783–791.

    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 cardiomy-

    opathy 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;13:3–12.

    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:

    3483–3490.

    141. Mazumder PK, O’Neill 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;54:65–73.

    143. Alpert MA. Obesity cardiomyopathy: pathophysiology and evolution of 

    the clinical syndrome.  Am J Med Sci. 2001;321:225–236.

    144. Kasper EK, Hruban RH, Baughman KL. Cardiomyopathy of obesity: a

    clinicopathologic evaluation of 43 obese patients with heart failure. Am J 

    Cardiol. 1992;70:921–924.

    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 insulin–resistant men.

     Hypertension. 1999;34:1047–1052.

    148. Yamashita T, Murakami T, Iida M, Kuwajima M, Shima K. Leptin

    receptor of zucker fatty rat performs reduced signal transduction.

     Diabetes. 1997;46:1077–1080.

    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 an-

    giotensin 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 car-

    diomyocytes. Br J Pharmacol. 2001;132:159 –164.

    151. Adiarto S, Emoto N, Iwasa N, Yokoyama M. Obesity-induced upregu-

    lation 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 decreasedsurvival 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:

    2327–2332.

    156. Mascareno E, Beckles DL, Siddiqui MA. Janus kinase-2 signaling

    mediates apoptosis in rat cardiomyocytes.  Vascul Pharmacol. 2005;43:

    327–335.

    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;109:121–130.

    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;29:542–550.

    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 ischemia-

    reperfusion 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

     558 Circulation Research   September 14, 2007

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

    http://circres.ahajournals.org/http://circres.ahajournals.org/

  • 8/18/2019 Circ Res-2007-Yang-545-59

    15/16

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

    45:156–158.

    165. 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;151:168–175.

    166. 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 endo-

    thelial dysfunction.   Am J Physiol Heart Circ Physiol. 2005;289:H1038–H1046.

    167. Chen K, L i F , L i J , Cai H, S trom 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.

    168. 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:

    1113–1116.

    169. 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.

    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, FontanaS, 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;

    17:372–378.

    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:

    2923–2928.

    Yang and Barouch Leptin Signaling and Obesity   559

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

    http://circres.ahajournals.org/http://circres.ahajournals.org/

  • 8/18/2019 Circ Res-2007-Yang-545-59

    16/16

    Ronghua Yang and Lili A. BarouchLeptin Signaling and Obesity: Cardiovascular Consequences

    Print ISSN: 0009-7330. Online ISSN: 1524-4571Copyright © 2007 American Heart Association, Inc. All rights reserved.is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231Circulation Research

    doi: 10.1161/CIRCRESAHA.107.1565962007;101:545-559Circ Res.

    http://circres.ahajournals.org/content/101/6/545

    World Wide Web at:The online version of this article, along with updated information and services, is located on the

     

    http://circres.ahajournals.org//subscriptions/ is online at:Circulation ResearchInformation about subscribing toSubscriptions:

     http://www.lww.com/reprints

     Information about reprints can be found online at:Reprints: 

    document.Permissions and Rights Question and Answerabout this process is available in thelocated, click Request Permissions in the middle column of the Web page under Services. Further informationEditorial Office. Once the online version of the published article for which permission is being requested is

    can be obtained via RightsLink, a service of the Copyright Clearance Center, not theCirculation Researchin Requests for permissions to reproduce figures, tables, or portions of articles originally publishedPermissions:

    http://circres.ahajournals.org/content/101/6/545http://circres.ahajournals.org//subscriptions/http://circres.ahajournals.org//subscriptions/http://circres.ahajournals.org//subscriptions/http://www.lww.com/reprintshttp://www.lww.com/reprintshttp://www.lww.com/reprintshttp://www.ahajournals.org/site/rights/http://www.ahajournals.org/site/rights/http://circres.ahajournals.org//subscriptions/http://www.lww.com/reprintshttp://www.ahajournals.org/site/rights/http://circres.ahajournals.org/content/101/6/545