From leptin to other adipokines in health and disease: Facts and expectations at the beginning of the 21st century Matthias Bl¨uher, Christos S. Mantzoros PII: DOI: Reference:

S0026-0495(14)00310-2 doi: 10.1016/j.metabol.2014.10.016 YMETA 53111

To appear in:

Metabolism

Please cite this article as: Bl¨ uher Matthias, Mantzoros Christos S., From leptin to other adipokines in health and disease: Facts and expectations at the beginning of the 21st century, Metabolism (2014), doi: 10.1016/j.metabol.2014.10.016

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

ACCEPTED MANUSCRIPT

Invited Review

T

From leptin to other adipokines in health and disease:

SC

RI P

Facts and expectations at the beginning of the 21st century

MA NU

Matthias Blüher, Christos S. Mantzoros

PT

ED

Department of Medicine, University of Leipzig, Leipzig, Germany Department of Endocrinology, Metabolism and Diabetes, VA Boston Medical Health Center, Boston, MA, USA

CE

Key words: Adipokines; obesity; type 2 diabetes, leptin, adiponectin, AdipoRon, DPP4,

AC

FGF21, TNFα, IL-1β, nampt/visfatin, BMP7

Corresponding author: Matthias Blüher, MD University of Leipzig Department of Medicine Liebigstr. 20 D-04103 Leipzig Tel. (+49) 341-9715984 Fax (+49) 341-9722439 E-mail: [email protected]

ACCEPTED MANUSCRIPT Abstract This year marks the 20th anniversary of the discovery of leptin, which has

T

tremendously stimulated translational obesity research. The discovery of leptin has led to

RI P

realizations that have established adipose tissue as an endocrine organ, secreting bioactive molecules including hormones now termed adipokines. Through adipokines, the adipose

SC

tissue influences the regulation of several important physiological functions including but not limited to appetite, satiety, energy expenditure, activity, insulin sensitivity and secretion,

MA NU

glucose and lipid metabolism, fat distribution, endothelial function, hemostasis, blood pressure, neuroendocrine regulation, and function of the immune system. Adipokines have a great potential for clinical use as potential therapeutics for obesity, obesity related metabolic, cardiovascular and other diseases. After 20 years of intense research efforts, recombinant

ED

leptin and the leptin analog metreleptin are already available for the treatment of congenital

PT

leptin deficiency and lipodystrophy. Other adipokines are also emerging as promising candidates for urgently needed novel pharmacological treatment strategies not only in obesity

CE

but also other disease states associated with and influenced by adipose tissue size and activity.

AC

In addition, prediction of reduced type 2 diabetes risk by high circulating adiponectin concentrations suggests that adipokines have the potential to be used as biomarkers for individual treatment success and disease progression, to monitor clinical responses and to identify non-responders to anti-obesity interventions. With the growing number of adipokines there is an increasing need to define their function, molecular targets and translational potential for the treatment of obesity and other diseases. In this review we present research data on adipose tissue secreted hormones, the discovery of which followed the discovery of leptin 20 years ago pointing to future research directions to unravel mechanisms of action for adipokines.

2

ACCEPTED MANUSCRIPT List of abbreviations ADA, American Diabetes Association

T

BMI, body mass index

RI P

CrP, C-reactive protein CT, computed tomography

SC

DBP, diastolic blood pressure

HbA1c, Hemoglobin A1c HDL, High Density Lipoprotein

MA NU

GIR, glucose infusion rate

HDL-C, High Density Lipoprotein-cholesterol

HOMA-IR, Homeostasis Model Assessment - Insulin-Resistance

ED

hsCrP, high sensitive C-reactive protein

IL-6, interleukin-6

PT

IGT, impaired glucose tolerance

CE

LDL, Low Density Lipoprotein

AC

LDL-C, Low Density Lipoprotein-cholesterol MCP-1, monocyte chemoattractant protein-1 mRNA, messenger ribonucleic acid NGT, normal glucose tolerance OGTT, oral glucose tolerance test SC, subcutaneous SBP, systolic blood pressure T2D, type 2 diabetes TNFα, tumor necrosis factor-α

3

ACCEPTED MANUSCRIPT 1. Introduction In 2014, the research community “celebrates” the 20th anniversary of the discovery of leptin

T

[1]. Even preceding this important discovery, adipose tissue had been identified as an

RI P

endocrine organ and adipsin/complement factor D was the first “adipokine” described [2, 3]. Adipsin circulating levels decline in animal models of diabetes and obesity [3]. Obesity and

SC

adverse (visceral) fat distribution increase the risk for metabolic (type 2 diabetes, hepatosteatosis, dyslipidemia), cardiovascular (hypertension, coronary artery disease, stroke),

MA NU

malignant, orthopaedic and psychological diseases [4, 5]. Except for bariatric surgery, current anti-obesity treatment strategies based on decreasing energy intake and increasing physical activity are frequently not successful most likely because pathogenetic factors cannot be targeted, which affect energy intake, metabolism and expenditure [6-8]. Therefore, both the

ED

treatment of obesity and the prevention of obesity related diseases may require novel

PT

pharmacotherapies targeting root causes of a sustained positive energy balance and the adverse signals from adipose tissue contributing to metabolic and cardiovascular diseases [9].

CE

Over the past two decades following the discovery of leptin which directed much needed

AC

attention to the study of adipose tissue, it has been acknowledged that several adipokines regulate important biological processes in target organs including the brain, liver, skeletal muscle, vasculature, heart, immune system and pancreatic β-cells (Figure 1) and may therefore link obesity to its metabolic and cardiovascular comorbidities [5, 9-12]. Under conditions of adipose tissue dysfunction, which is frequently found to accompany obesity, secretion of adipokines is dysregulated [5, 10]. Altered adipokine secretion may contribute to impaired regulation of appetite and satiety, fat distribution, insulin secretion and sensitivity, energy expenditure, endothelial function, inflammation, blood pressure, and hemostasis [4, 912]. Therefore, adipokines may offer exciting new opportunities for the future pharmacotherapy of obesity and obesity related diseases [12]. Translational research approaches [9, 13] are particularly important to identify novel pharmacotherapies for diseases 4

ACCEPTED MANUSCRIPT in which an incomplete understanding of the molecular mechanisms leads to a lack of etiology based treatment strategies. Obesity and other disease states associated with

RI P

T

excess/lack or dysfunction of adipose tissue certainly belongs to these diseases.

2. From leptin to more than 600 adipokines

SC

Different cell types of adipose tissue (adipocytes, immune cells, fibroblasts, endothelial cells and others) release fatty acids, other lipids and metabolites and adipokines [9, 14-16].

MA NU

Stimulated by the discovery of leptin and the notion that adipose tissue is an endocrine organ, more than six hundred adipokines have been described so far [17] and search for novel adipokines still represents a hot topic in obesity research. Adipokines have been proposed to play specific roles in immune response (e.g. adipsin/complement factor D, acylation-

ED

stimulating protein, serum amyloid A3, interleukins) and inflammation (e.g. IL-1β, IL-6, IL-8,

PT

IL-10, CrP, monocyte chemoattractant protein-1, osteopontin, progranulin, chemerin), glucose metabolism (e.g. leptin, adiponectin, dipeptidyl peptidase-4, fibroblast growth factor 21,

CE

resistin, vaspin), insulin sensitivity (e.g. leptin, adiponectin, chemerin), hypertension (e.g.

AC

angiotensinogen), cell adhesion (e.g. PAI-1), vascular growth and function (e.g. VEGF), atherosclerosis development (e.g. cathepsins, apelin), adipogenesis and bone morphogenesis (e.g. BMP-7), growth (e.g. IGF-1, TGFβ, fibronectin), lipid metabolism (e.g. CD36), regulation of appetite and satiety (e.g. leptin, vaspin), eating disorders such as anorexia nervosa (e.g. leptin, adiponectin, resistin) and other biological processes [9-11]. However, for the majority of recently identified adipokines, physiologic regulation, functions, molecular action and targets need to be defined to develop future pharmacological therapeutics [14-16]. The lack of understanding adipokine related mechanisms still represents an important road block in translational adipokine research. The path of leptin from its discovery to the clinical application for patients with congenital leptin deficiency [18] and lipodystrophy [19-21] may

5

ACCEPTED MANUSCRIPT serve as the first example for successful translational adipokine research. Other adipokines

Clinical importance of adipokines and translational adipokine research

RI P

3.

T

may follow leptin as potential novel therapeutics or treatment targets.

Translational adipokine research may lead to clinically important applications of adipokines

SC

either as biomarkers or as substrates/targets for pharmacological management of obesity and

3.1.

MA NU

prevention of obesity related disorders in the future (Figure 2).

Adipokines as predictors of mortality and obesity related diseases

Adipokines are involved in the regulation of metabolism, insulin sensitivity, inflammation and serum concentrations of distinct adipokines are associated with obesity, metabolic and

ED

cardiovascular diseases [4, 10, 14-16]. It has been therefore hypothesized that adipokine

PT

serum concentrations may serve as predictors of mortality, obesity related diseases or the individual disease outcomes [4, 10]. Indeed, in a prospective cohort study of 981 outpatients

CE

with stable coronary artery disease, low leptin serum concentrations at baseline were

AC

independently of gender, obesity and traditional cardiovascular risk factors associated with a 37% increased risk for cardiovascular events and mortality [22]. In a larger prospective study including 5,672 participants, circulating leptin was not associated with the risk of cardiovascular disease (CVD), but with type 2 diabetes risk in men [23]. In women with diabetes, circulating leptin levels were also not associated with cardiovascular morbidity and mortality [24]. A recent meta-analysis using data from 14,063 CVD patients from 15 prospective cohort and 1 nested case control studies, revealed that increased baseline plasma adiponectin levels are significantly associated with elevated risk of all-cause and cardiovascular mortality [25]. Moreover, data from the Cardiovascular Health Study have shown that higher plasma adiponectin concentrations are associated with new-onset heart failure, left ventricular systolic 6

ACCEPTED MANUSCRIPT dysfunction and left atrial enlargement in adults ≥65 years [26]. Noteworthy, in another meta-analysis, no associations between adiponectin and CV events have been observed [27]

T

and in the EPIC Norfolk Prospective Population Study, low adiponectin concentrations have

RI P

been associated with an increased CVD risk [28]. Adiponectin has also been proposed as a biomarker for stroke [29, 30]. Despite the general consideration of adiponectin as a protective

SC

molecule, increased concentrations of adiponectin in patients with type 1 diabetes were independently associated with all-cause and cardiovascular mortality [31]. The role of

MA NU

adiponectin in determining CVD health, especially in relation to heart failure [32] was recently summarized in the journal [33-35].

On the other hand, a protective role of higher adiponectin serum concentrations

ED

against the development of type 2 diabetes has been suggested [36-38]. Spranger and colleagues found in 27,548 participants of a prospective, nested case-control study within the

PT

population-based EPIC (European Prospective Investigation into Cancer and Nutrition) Potsdam cohort that high concentrations of adiponectin were associated with a substantially

CE

reduced relative risk to develop of type 2 diabetes in apparently healthy individuals [36]. This

AC

relationship was stable even after adjusting for age, gender, waist-to-hip ratio, body-mass index, smoking, exercise, alcohol consumption, education, and HbA1c [36]. In addition to type 2 diabetes and CVD, adipokine measurements maybe useful tools for the prediction of other not as commonly with obesity associated diseases including malignancies [39-41]. For instance, high adiponectin serum concentrations predispose men to a lower risk of developing high-grade prostate cancer and a lower risk of subsequently dying from it [42]. High non-HMW adiponectin as well as IL-6 and CrP serum concentrations have been found to be independently associated with increased risk for hepatocellular carcinoma [43]. We have shown, however, that although HMW adiponectin may slightly improve clinical prediction of metabolic variables [44] the added benefit is only incremental and probably not corresponding to the extra cost and burden conferred by the measurement of 7

ACCEPTED MANUSCRIPT HMW adiponectin over just measuring total adiponectin. More recent studies have raised the possibility that other measurements, such as the high serum C1q-adiponectin/total adiponectin

T

may be novel promising biomarkers of risk [45] but detailed comparative evaluations against

RI P

more traditional markers are lacking.

In addition to circulating adiponectin [46, 47] several papers have also indicated that

SC

adiponectin gene variants as well as variants of the adiponectin receptor are also strongly related to risk for metabolic diseases [48, 49]. Taken together, associations of adipokine

MA NU

serum concentrations with obesity related diseases (type 2 diabetes, CVD) and mortality have been frequently reported. However, it is not clear whether these sometimes contradictory associations are due to a mechanistic link between altered adipokine levels and an increased disease risk or simply reflect uncontrolled confounding. Further studies are required to

ED

identify potential mechanisms underlying the association between differences in adipokine

Adipokines – potential role as biomarkers

CE

3.2.

PT

serum concentrations and individual disease risks or outcomes.

AC

At least theoretically, adipokines have the potential to serve as biomarkers for the functional status of adipose tissue [5, 10] (Figure 2). Altered adipokine serum concentrations maybe related to hypertrophy of adipocytes, infiltration of AT with immune cells and processes within AT including hypoxia [50], autophagy [51], apoptosis [52], and different types of stresses [53]. In this context, we could recently show that using a specific circulating adipokine pattern with higher adiponectin, but lower RBP4, chemerin, progranulin, and fetuin-A serum concentrations an insulin sensitive obese subphenotype could be distinguished from an insulin resistant otherwise metabolically healthy obese subphenotype [54]. These adipokine changes were associated with an adverse (visceral) fat distribution, hypertrophic adipocytes and AT inflammation [54]. Moreover, several adipokines (e.g. omentin, RBP4, Nampt) have been suggested as biomarkers for visceral fat mass [5, 10]. In the future, 8

ACCEPTED MANUSCRIPT adipokines could become relevant biomarkers for the prediction of individual weight dynamics in response to lifestyle or bariatric surgery interventions, as markers of food

T

preferences or diet adherence (Figure 2). So far, adipokines have not been proven to predict

RI P

obesity related metabolic diseases better than classical parameters to define the metabolic syndrome [55]. Recently, we identified distinct adipokine clusters related to fat mass and

SC

inflammation (ANGPTL3, chemerin, clusterin, DLL1, GPX3, Nampt, resistin), insulin sensitivity, glucose and lipid metabolism (adiponectin, ANGPTL6, progranulin, RBP4, BMP7,

MA NU

DLK1, SFRP5, vaspin, glypican4, CTRP3 and 5, omentin) [55]. Interestingly, an adipokine panel consisting of ANGPTL6, DLK1, Nampt and progranulin was independently correlated to T2D in obese individuals, but with a lower sensitivity and specificity than established

ED

parameters of glucose metabolism [55].

PT

3.2.1 Adipokine dynamics and weight loss Weight loss interventions frequently induce a rapid decline in body weight followed by a

CE

weight stabilization and/or regain phase with significant intra-individual variation. There is

AC

still an unmet need to predict individual outcomes from weight loss interventions with the aim to define the individual who will benefit the most from such weight loss interventions. Measurement of baseline adipokine serum concentrations or changes of adipokines in the initial rapid weight loss phase could provide such information. Recently, we analyzed the dynamics of adipokines among 322 participants in the 2-year Dietary Intervention Randomized Controlled Trial (DIRECT) of low-fat, Mediterranean or low-carbohydrate diets for weight loss [56]. In the DIRECT a weight loss phase of ~ 6 months was followed by either weight stabilization or partial to full weight regain despite continued dieting [56]. In the context of this dietary intervention study, we found that adipokine serum concentrations independently of gender, diabetes diagnosis, or diet group either follow the weight loss pattern (leptin, chemerin, MCP-1, RBP4) or the continued healthy diet (adiponectin, fetuin-A, 9

ACCEPTED MANUSCRIPT progranulin, vaspin) [57]. However, we were unable to link a specific adipokine (or its early change) serum concentration to specific dietary components, food preference or diet

T

adherence [57]. On the other hand, weight regain in the DIRECT was significantly and

RI P

independently associated with genetic variations in the leptin gene (P = 0.006 for both rs4731426 and rs2071045) [58]. The inclusion of the leptin genotype to the phenotypic

SC

multivariate model increased its predictive value for weight regain by 34% [58]. Moreover, the importance of adipokines as biomarkers is further supported by data from the DIOGENES

MA NU

study, which demonstrated that high baseline RBP4 serum concentrations (together with low total testosterone and low SHBG) significantly predicts weight regain after weight loss [59].

3.2.2 Adipokines as predictors of adverse fat distribution

ED

Visceral fat distribution is clearly a stronger predictor for obesity related metabolic and

PT

cardiovascular diseases as whole body fat mass or BMI [4, 60, 61]. However, quantification of total visceral fat mass requires sophisticated and expensive imaging techniques. Both

CE

magnetic resonance imaging (MRI) and computerized tomography (CT) have been widely

AC

used for total visceral fat assessment. The exposure to ionizing radiation makes CT a largely inappropriate technique for prospective studies. MRI, on the other hand, is safer but typically involves longer acquisition times and higher costs [62]. Serum concentrations of adipokines including fetuin-A, chemerin, RBP-4, omentin, vaspin, progranulin, Nampt and others have been suggested to predict visceral fat mass [5, 10]. However, associations between circulating adipokines and visceral fat mass are not always robust across different cohorts and therefore no generally accepted adipokine marker of visceral fat mass exists. In addition, it is not clear, whether adipokines cause or reflect visceral fat accumulation. The discovery of Nampt, also named visfatin, is one example for how difficult the search for circulating markers of visceral fat mass may be. Nampt/visfatin has been defined as an adipokine exclusively secreted from visceral fat with insulin-mimetic 10

ACCEPTED MANUSCRIPT effects [63], however, subsequent human studies revealed that other tissues and adipose tissue depots may also express Nampt/visfatin [64]. Noteworthy, with aging and hypercaloric

T

feeding, impaired NAMPT-mediated NAD+ biosynthesis may contribute to T2D development

RI P

[65]. For another adipokine, RBP4, different studies found either strong positive correlations between circulating RBP4 and visceral fat distribution [54, 66, 67] or no associations [68, 69].

SC

Such contradictory findings may at least in part be explained by the fact that in addition to adipose tissue other tissues such as the liver may determine circulating levels. For the

MA NU

adipokines chemerin [70, 71], progranulin [72] and omentin [73], associations between increased visceral fat mass and higher serum concentrations may be influenced by AT immune cell infiltration and AT dysfunction [5]. Noteworthy, so far there are no genome wide association studies using visceral or subcutaneous AT distribution as traits. Such studies may

PT

ED

unravel adipokines, which are directly involved in the regulation of fat distribution.

3.2.3 Altered circulating adipokines reflect adipose tissue dysfunction

CE

Storage of excessive calories as triglycerides and release of fatty acids during fasting and

AC

prolonged food deprivation may represent the main functions of AT [74]. In addition, AT is an important organ for thermoregulation, mechanical organ protection and endocrine signalling. With increasing body weight upon excessive energy intake, AT dysfunction may develop and seems to determine the individual risk to develop metabolic and cardiovascular comorbidities [4, 10, 74]. The inability to store excess calories in subcutaneous fat depots may represent a critical node in the development of subsequent ectopic fat deposition in visceral depots, the liver and other cell types [74]. Typical symptoms of AT dysfunction are therefore: 1) visceral (ectopic) fat accumulation, 2) changes in the cellular and intracellular matrix composition of adipose tissue, 3) increased number of immune cells within AT, 4) adipocyte hypertrophy, 5) increased autophagy and apoptosis, 6) AT extracellular matrix changes (AT fibrosis), 7) alterations in AT mRNA and protein expression patterns [74]. An 11

ACCEPTED MANUSCRIPT adipokine pattern with low circulating adiponectin and high serum concentrations of chemerin, progranulin, RBP4, fetuin-A, CrP, glypican-4, DPP-4 and others may reflect AT

T

dysfunction [54, 75, 76]. Importantly, further studies are required to identify robust predictors

RI P

of AT function.

SC

3.2.4 Adipokines and neurodegenerative diseases

Obesity has been associated with changes in brain structure, cognitive deficits, dementia and

MA NU

Alzheimer‫׳‬s disease (AD) [77]. The association between obesity, structural changes in the brain and neurodegenerative diseases could be mediated by pleiotropic effects of adipokines, particularly those adipokines which may cross the blood brain barrier including leptin, adiponectin, plasminogen activator inhibitor-1 (PAI-1), IL-6, TNF-α and angiotensingen [77].

ED

Indeed, circulating leptin levels correlate with the development of AD, leptin has previously

PT

been shown to attenuate tau hyperphosphorylation in neuronal cells and to be modulated by amyloid-β [78, 79]. Recent data further suggest that leptin resistance in the hippocampus may

CE

play a role in the characteristic changes associated with AD [79]. Moreover, leptin treatment

AC

has been proposed as a potential clinical application for AD [80]. In a mouse model for AD with an overexpression of the amyloid-β protein precursor, chronic administration of leptin results in a significant improvement in the cognitive performance [80]. Noteworthy, weight loss often precedes the onset of dementia and AD and the level of circulating leptin is inversely proportional to the severity of cognitive decline [80]. However, the concept that leptin treatment may improve the natural course of AD needs to be proven in clinical studies. Another example for the association between changes in adipokine patterns and neurodegenerative disorders is adiponectin [77]. Decreased adiponectin serum concentrations are associated with mild cognitive impairment and AD, but do not predict cognitive decline in elderly individuals [81]. In contrast to that notion, in dementia-free women participating in the Framingham Heart Study, increased plasma adiponectin levels have been detected as 12

ACCEPTED MANUSCRIPT independent risk factor for the development of both all-cause dementia and AD [82]. In patients with probable AD, lower leptin and higher circulating adiponectin serum

T

concentrations are positively correlated with the severity of dementia [83]. However, other

RI P

studies did not find associations between adiponectin and neurodegenerative disorders [84-85]. In addition to associations between leptin and adiponectin with neurodegenerative diseases, it

SC

has been recently shown that resistin may protect against endogenous amyoloid β neuronal cytotoxicity in a mouse model of AD, raising the possibility of novel AD therapies using

MA NU

resistin [86]. However, further mechanistic studies are required to unravel the role of adipokines on structural changes in the brain, cognitive deficits, dementia, and AD.

3.3

Specific adipokines in health and disease: Are adipokines a novel therapeutic tool

ED

to treat obesity and obesity-related diseases?

PT

Changes in circulating adipokines and adipokine expression in AT have been linked to metabolic diseases (type 2 diabetes, fatty liver disease), cardiovascular diseases, malignancies

CE

and compromised reproductive health [12]. Therefore, translational research efforts have been

AC

put into the path from the discovery of adipokines to their clinical use in the treatment of such diseases. There are important obstacles in the successful translation of biomedical research into clinical implementation of new adipokine-based therapeutic concepts, which have been extensively discussed elsewhere [9]. Some adipokines, including leptin [18, 19], adiponectin [87] and FGF21 [88] have been systematically developed into drug applications. For other adipokines including DPP-4 [89] and BMP-7 [90], clinical application preceded their recognition as adipokines. The translational potential of adipokines research is not restricted to metabolic or cardiovascular diseases – indication fields such as infertility, lipodystrophy, fractures or musculo-skeletal diseases are either in clinical development, have been tested in clinical phase I trials or clinical pilot studies already [9].

13

ACCEPTED MANUSCRIPT To fully develop an adipokine until its clinical applications are proven may take ~15 years. Even if we celebrate this year the 20th anniversary of the discovery of leptin, the long

T

development process makes it obvious that for more recently identified adipokines clinical

RI P

applications can not be expected in the next few years. For many adipokines, we still have an incomplete knowledge about the mechanism of action and the major interaction partners (e.g.

SC

receptors).

In addition, since many adipokines are upregulated in disease states, it may be useful to

MA NU

develop an inhibitor for such adipokines. For TNFα [91, 92] or IL-1β [93] – which could also be considered as adipokines - inhibition of action have been already tested for different indications. Moreover, a chemerin (ChemR23) antagonist CCX832 has been shown to protect against chemerin-related arterial contraction, thus linking higher chemerin concentrations in

ED

obesity to impaired vascular function [94]. There are several additional adipokines in the

CE

3.3.1 Leptin

PT

“pipeline” from drug discovery to clinical testing [9].

AC

Leptin has been considered as the prototype for all adipocyte secreted hormones [12]. Leptin levels are pulsatile [95] and are associated with fat mass/BMI as well as circulating inflammatory markers [95-98]. Leptin in low leptin states plays an important role in satiety, appetite [99, 100], food intake, reproductive function and puberty [101-106] activity and energy expenditure regulation [107]. Originally, leptin was viewed as a potential anti-obesity therapeutic agent and was originally developed mainly with the aim to reduce body fat mass in obesity [18, 108-111]. In mice [108] and humans [109] with genetically inherited leptin deficiency, leptin has been proven as an effective weight loss drug. In contrast to leptin substitution treatment, in human obese patients [110, 111] with obesity as well as in high caloric diet induced obesity in mice [112], leptin had only little effects on weight loss [113, 114]. Even high doses of leptin and once weekly treatment with pegylated leptin did not 14

ACCEPTED MANUSCRIPT improve weight loss effects, but increased the risk for side effects of recombinant leptin therapy of obesity [111]. Despite these hurdles in the human application of leptin as an anti-

T

obesity drug, there are still research efforts to improve leptin analogues for their clinical use.

RI P

For example several site-specifically enhanced leptin analogues have been designed to increase the potency and lead to a more sustained action leptin, which provided enhanced

SC

weight loss in diet induced obesity mice [115].Administration of leptin to human obese individuals does not significantly affect appetite and body weight [21]. It has been suggested

MA NU

that the lack of leptin efficacy in human obesity is due to the development of central leptin resistance or tolerance in parallel with increased circulating leptin levels upon and before obesity development [21]. Leptin signaling pathways, the abnormality of which may eventually lead to the development of leptin resistance, have only recently started to be

ED

studied in humans [116-121]. In the search for agents that may restore leptin sensitivity,

PT

amylin (pramlintide) has been shown to induce weight loss in obese rodent models and humans when given together with leptin [122]. The leptin analogue metreleptin combined

CE

with pramlintide has been advanced to a clinical proof of concept study, which demonstrated

AC

a significant weight-lowering effect in a 24-week study in human obesity [123]. Unfortunately, a recent randomized clinical trial using pramlintide/metreleptin as anti-obesity therapeutic had to be stopped due to adverse events such as antibody generation and skin reactions [9, 118].

3.3.2 Clinical applications of leptin and other adipokines Recombinant leptin is now available for compassionate use for patients with congenital leptin deficiency [19]. Metreleptin also has been already approved for the treatment of lipodystrophy in Japan and in the US and is under consideration at European regulatory agencies for approval [20]. The FDA Advisory Committee voted in favor of metreleptin for the treatment of diabetes and/or hypertriglyceridemia, in patients with rare forms of 15

ACCEPTED MANUSCRIPT congenital lipodystrophy [20]. An additional potential future indication for metreleptin treatment has been recently emerged from clinical studies in five patients with Rabson-

T

Mendenhall syndrome (RMS), which is caused by mutations in the insulin receptor [124]. In

RI P

these patients, metreleptin treatment for 12 months was associated with a 1.7% reduction in HbA1c and reduced body weight suggesting metreleptin is a promising treatment option for

SC

rare RMS patients [124]. We have recently reviewed the role of leptin in lipodystrophy elsewhere [125] and herein [126]. Moreover, the effects of metreleptin administration in

MA NU

replacement doses were investigated in a randomized, double-blinded, placebo-controlled trial in women with hypothalamic amenorrhea and acquired chronic hypoleptinemia induced by negative energy balance [101, 121, 127-131]. This study could demonstrate that metreleptin restores both CD4(+) T-cell counts and their in vitro proliferative responses in these women

ED

suggesting metreleptin as an effective therapy for selective CD4(+) T-cell immune

PT

reconstitution in hypoleptinemic states such as tuberculosis and HIV infection in which CD4(+) T cells are reduced [121]. In contrast, patients with HIV-associated dyslipidemic

CE

lipodystrophy did not significantly improve their leptin kinetics in response to metreleptin

AC

replacement treatment over 2-4 months [132] although metreleptin was successful in both open label and randomized placebo controlled trials in patients with HIV-associated lipodystrophy [133-135]. Proof of concept studies demonstrated that a safe leptin analogue has a potential clinical application in the treatment of hypothalamic amenorrhea and related infertility [101, 121, 127-131]. Moreover, serum leptin concentrations in women with anorexia nervosa are lower and show reduced diurnal variation than those of normal-weight controls as a result of decreased body weight and fat mass [reviewed in 127]. In addition to these leptin alterations, anorexia nervosa is characterized by significant changes of hormones (e.g. IGF-1, cortisol, ghrelin, PYY) including adipokines (e.g. adiponectin, resistin). Importantly, dietary therapy of anorexia nervosa leads to increased circulating leptin which correlates with increasing gonadotropins, indicating that increasing leptin in response to 16

ACCEPTED MANUSCRIPT weight gain could activate the hypothalamic-pituitary-gonadal axis [127]. Because the nutritional therapy for women with anorexia nervosa takes time, maintaining recovered

T

weight maybe difficult and long-term effectiveness of this method remains to be proven,

RI P

treatment of eating disorders such as anorexia nervosa with safe leptin analogues and careful dose titration could be an additional clinical application of leptin in the future [127-129].

SC

However, so far recombinant leptin has not yet been successfully introduced for the treatment of this eating disorder.

MA NU

To date, leptin is the best example for a successful translational adipokine research, which already successfully advanced to clinical application. In the future, more clinical applications of adipokine-based treatment strategies are under consideration or tested in randomized clinical proof of concept trials for the treatment of impaired glucose metabolism (e.g.

ED

adiponectin, FGF21, BMP7), insulin resistance (e.g. adiponectin, FGF21), dyslipidemia (e.g.

PT

leptin, FGF21), chronic systemic inflammation (e.g. anti IL-1 antibodies), eating disorders

CE

such as anorexia nervosa (e.g. leptin), bone fracture healing (e.g. BMP7) and others (Figure 2).

AC

3.3.3 Adiponectin

Adiponectin belongs to the highest expressed proteins in adipocytes and represents ~0.01% of total plasma protein [136]. Discovered shortly after leptin in 1995 [137-139], adiponectin gained wide interest due to its insulin sensitizing, anti-inflammatory and anti-apoptotic actions on a number of different cell types [14, 140]. In the past almost 20 years and in more than 11,000 references in the international literature, several aspects of adiponectin´s actions have been discovered, which make adiponectin-based therapies a promising approach for both treatment of obesity itself as well as for obesity-related disorders [14, 136]. Importantly, the basic principles of adiponectin physiology have been maintained remarkably well across the different species [140]. Adiponectin emerged as a suitable biomarker, because it is easily detectable in blood, stable upon collection and relatively inert to the method of collection and 17

ACCEPTED MANUSCRIPT diurnal changes [14, 137]. Circulating adiponectin levels inversely correlate with multiple metabolic disorders including obesity and related diseases [136, 137, 140]. Moreover, in large

T

scale longitudinal studies circulating adiponectin has been identified as a marker for all-cause

RI P

mortality, heart failure, coronary artery disease and type 2 diabetes [26-28, 31, 36]. In addition to beneficial peripheral effects on insulin sensitivity, adiponectin acts in the brain to

SC

increase energy expenditure and may thereby promote weight loss [140]. Administration of recombinant adiponectin results in improved insulin sensitivity (predominantly in the liver),

MA NU

increased insulin secretion [141], reduction of body weight and improved glycemia [reviewed in [136, 140]. In some studies, which can not confirm these adiponectin treatment data, ineffective recombinant adiponectin preparations may underlie the lack of efficacy [142]. Last year, a potential breakthrough in the adiponectin field has been reported by Okada-Iwabu and

ED

coworkers, who synthesized an orally active, synthetic small-molecule adiponectin receptor

PT

agonist [87, 143]. The molecule which has been given the name AdipoRon significantly improves insulin sensitivity and glucose tolerance in mice, thereby extending lifespan of

CE

db/db mice on a high-fat diet [87, 143]. The observation that agonists of the adiponectin

AC

receptor may have beneficial effects on increasing longevity in a mouse model of obesity provides strong evidence for the hypothesis that higher circulating adiponectin levels are associated with prolonged life expectancy under conditions of shortened life span due to obesity-related diseases [143-145]. In this context, increased adiponectin levels are associated with extended longevity in mice [145-148]. Transgenic mice expressing high levels of human adiponectin have increased longevity [146] and various mouse models of extended longevity including fat-specific insulin receptor knockout mice [147], mice with defects in growth hormone production [148], and calorie-restricted mice [149] have increased adiponectin. Moreover, female centenarians have significantly higher adiponectin plasma concentrations compared to BMI-matched women at other ages [150]. Together with evidence from longitudinal human studies that lower circulating adiponectin is related to increased all-cause 18

ACCEPTED MANUSCRIPT mortality [31] these data support the concept that adiponectin has a potential beneficial role in extending life span [145].

T

These promising recent advances in the adiponectin further establish adiponectin itself or

RI P

adiponectin receptor agonists as candidates for the further development as pharmacotherapy for diseases associated with obesity and insulin resistance, including not only diabetes and

SC

cardiovascular disease but also possibly obesity associated malignancies [14, 39, 49, 151-180].

MA NU

3.3.4 FGF21

Fibroblast growth factor 21 (FGF21) is a hepato-adipokine that has been shown to stimulate glucose uptake into adipocytes in an additive manner to insulin [88, 181]. The initial breakthrough in uncovering the function of FGF21 came as a result of a phenotypic screen

ED

carried out at Lilly Research Laboratories in early 2000 [182]. In patients with obesity and

PT

T2D, circulating FGF21 levels are significantly increased [183]. The obesity and diabetes research community considers FGF21 as promising molecule for the treatment of metabolic

CE

diseases, because this molecule may increase thermogenesis, energy expenditure, fat

AC

utilization and has significant glucose and lipid lowering effects [88]. In animal studies, recombinant FGF21 reduces hyperglycemia and produces additional beneficial metabolic effects [88]. In a truly translational research effort, many of the animal data have been reproduced in a randomized, placebo-controlled, double-blind trial on the effects of a FGF21 variant (LY2405319) in patients with obesity and type 2 diabetes [88]. LY2405319 administration caused body weight reduction, as well as significant improvements in fasting insulin, LDL-cholesterol, HDL-cholesterol, triglycerides and apolipoprotein profile [88]. In contrast to FGF21 overexpression studies in transgenic mice in which FGF21 not only protected animals from diet-induced obesity, but also lowered blood glucose [181], LY2405319 did not significantly improve glucose concentrations [88].

19

ACCEPTED MANUSCRIPT Despite several lines of evidence about the positive metabolic actions of FGF21, safety concerns associated with targeting the FGF21 pathway have also emerged [182]. The

T

translational adipokine research related to FGF21 may serve as an example how basic

RI P

research may unravel data to guide drug discovery, but may also lead to unexpected findings. FGF21 was initially discovered using in vitro assays as a potential insulin mimetic, emerged

SC

as a potential agent in animals which improved more than only hyperglycemia, and ended up providing several metabolic benefits yet failed to deliver sustained glucose lowering in

MA NU

humans [182]. Taken together, FGF21 serves as a prime example that novel and potentially better medicines also derived from translational adipokine research are within the reach [182].

3.3.5 DPP-4

ED

Compared to leptin, adiponectin and FGF21, the drug discovery path went a totally different

PT

way for the clinical importance of the molecule dipeptidyl peptidase-4 (DPP-4). In an attempt to develop incretin-based therapies for type 2 diabetes, DPP-4 emerged as the most prominent

CE

indirect target, because DPP-4 rapidly degrades incretins including GLP-1 [89]. DPP-4

AC

inhibitors lower increased plasma glucose by increasing the concentration, half life time and action of incretins thereby stimulating glucose-induced insulin secretion and suppressing glucagon production [89]. Although research has focused on the role of DPP-4 in the degradation of GLP-1, recent data suggest that DPP-4 also exerts direct effects, as it is able to induce insulin resistance in adipocytes and skeletal muscle cells in concentrations that can be found in the circulation of overweight and obese subjects [184]. Only very recently, it has been appreciated that adipose tissue maybe an important source of circulating DPP-4 and DPP-4 may be considered an adipokine [184]. Importantly, obese individuals particularly those with impaired insulin sensitivity [54] have a significantly increased adipose tissue DPP-4 secretion compared to lean controls [76]. From such studies, it has been concluded that increased DPP-4 production in AT could contribute to obesity and 20

ACCEPTED MANUSCRIPT insulin resistance [9]. However, there are open questions about the net contribution of AT derived DPP-4 to changes in endogenous GLP-1 levels and whether DPP-4 inhibitors may

T

disproportionately affect AT produced DPP-4 from that of other sources.

RI P

It has been shown that DPP-4 serum concentration and adipose tissue DPP-4 expression correlate with hypertrophy of adipocytes and AT dysfunction [76, 184]. Therefore,

SC

DPP-4 might not only reflect adipose tissue function, but it may also have local effects within adipose tissue and systemic effects. Increased DPP-4 activity and serum concentrations in

MA NU

obesity may serve as a model how altered adipokine secretion may be successfully used as therapeutic target in the treatment of obesity related diseases. However, further work is needed to elucidate the functional role of DPP-4 within adipose tissue and to define whether higher DPP-4 expression and serum concentration may contribute to higher efficacy of DPP-4

PT

ED

inhibitors in patients with type 2 diabetes [9].

3.3.6 TNFα and IL-1β

CE

Both TNFα and IL-1β are highly expressed in and secreted from adipose [185, 186] and may

AC

therefore qualify for the term adipokine. Both cytokines are attractive targets for antiinflammatory therapies with beneficial effects on insulin sensitivity and glucose metabolism [9]. In a rat model of obesity, neutralization of TNFα leads to an increase in peripheral glucose uptake in response to insulin [187]. Genetic ablation of the TNFα gene in mice results in improved insulin sensitivity in different obesity models suggesting that blocking TNFα action may improve insulin sensitivity and its related traits [188]. Treatment with TNFα antibodies resulted in inhibited inflammatory activity, improved fatty liver disease [189], protection against diet induced obesity and insulin resistance [190] in several animal models. However, these beneficial metabolic effects using anti-TNF treatment could not be proven to be successful in obese Zucker rats [191] or recent clinical studies [192]. On the other hand, there are studies reporting improvements in insulin sensitivity or hyperglycemia in insulin 21

ACCEPTED MANUSCRIPT resistant individuals during prolonged treatment with TNFα targeting strategies infliximab [91] or etanercept [92].

T

IL-1β is another potential drug target, because it may contribute to β-cell destruction and

RI P

apoptosis associated with the pathogenesis of both type 1 and type 2 diabetes [93]. In the first clinical double-blind, parallel-group proof of concept study, Larsen and coworkers

SC

demonstrated that blocking IL-1 using the recombinant human IL-1-receptor antagonist anakinra significantly lowers plasma glucose, improves β-cell function and reduced markers

MA NU

of systemic inflammation [93]. These data support the concept that targeting proinflammatory adipokines/cytokines may be an approach to improve metabolic diseases.

3.3.7 BMP-7

ED

Bone morphogenetic proteins (BMPs) are developmental factors that belong to the

PT

transforming growth factor (TGF) β superfamily and regulate several processes of organogenesis and patterning during embryonic development [193-196]. Moreover, BMPs

CE

may act as a molecular switch in regulating white versus brown adipogenesis [195, 196].

AC

Recently, several members of the BMP family of ligands and receptors have been found to associate with obesity-related traits in humans [61, 197, 198]. Therefore, targeting BMPs has emerged as a novel therapeutic concept to treat obesity and its related diseases by inducing increased energy expenditure and browning of white adipose tissue [193, 194]. As an example, BMP7 has gained attention after the notion that BMP-7 stimulates brown adipogenesis, reduces food intake, increases energy expenditure resulting in reduced weight gain [195, 196]. For BMP7 translational adipokine research is facilitated by the fact that human recombinant BMP7 is already available [90]. Under the brand name OP1, recombinant BMP7 is locally applied to aid in the fusion of vertebral bodies in the treatment of non-union after tibia fractures [90]. From the clinical use, it is known that local recombinant BMP7 application does not have local or systemic toxic effects or other 22

ACCEPTED MANUSCRIPT adverse events [90]. So far, BMP7 has not been tested in the treatment of obesity or metabolic

Conclusions

RI P

4.

T

diseases.

Several important advances in our understanding of the function of the adipose tissue and

SC

important changes in the way we approach and treat several disease states have occurred over the twenty years since the discovery of leptin. Adipokines contribute to the regulation of

MA NU

appetite, satiety, energy expenditure and physical activity. Targeting or using adipokine-based mechanisms to treat obesity and diseases which are caused by a positive energy balance is therefore a promising strategy, and has driven translational adipokine research during the past 20 years. Moreover, we have also realized during the same period of time that several

ED

adipokines, including but not limited to adiponectin, leptin, etc. serve as messages linking

PT

adipose tissue with specific organs and conveying information that may alter human

AC

issue [14].

CE

physiology and pathophysiology as outlined in part in this renew and elsewhere in this special

Thus, all these newly discovered adipokines are not only promising new potential biomarkers and therapeutic agents; we have already started realizing tangible benefits. In the energy deficiency part of the energy homeostasis spectrum, congenital leptin deficiency and generalized lipodystrophy are now indications for therapy with leptin. Proof of concept studies have demonstrated that a safe leptin analogue could be a reasonable, physiology based, approach for the treatment of hypothalamic amenorrhea and related infertility and possibly, in well controlled doses, for some cases of anorexia nervosa. In the treatment of obesity, lifestyle changes, psychological interventions or pharmaco-therapies have provided only very limited sustained beneficial effects. In the long-term, weight loss surgery are much more effective compared to the non-surgical intervention, but the bare an increased perioperative 23

ACCEPTED MANUSCRIPT mortality risk. In particular, pharmacological anti-obesity therapies rarely lead to leanness or a complete remission of obesity-associated diseases.

T

At the 20th anniversary of the discovery of leptin, translational research has already led

RI P

to clinical application of the leptin analogue metreleptin as a pharmacotherapy in individuals with congenital leptin deficiency and lipodystrophies [199]. More adipokine-based treatment

SC

strategies using for example adiponectin, FGF21 or BMP7 may follow leptin in the path of drug discovery. In summary, adipokines may open exciting new diagnostic and treatment

MA NU

opportunities for diseases with unmet clinical needs.

Conflict of interest

ED

Dr. Mantzoros is a consultant for Astra Zeneca. Dr. Blüher does not have any conflict of

AC

CE

PT

interest related to this manuscript.

24

ACCEPTED MANUSCRIPT Figure legends Figure 1. Adipokines regulate important physiologic processes. Secreted factors from

T

adipose tissue (representative histologic slide of human subcutaneous adipose tissue,

RI P

hematoxylin and eosin staining; magnification: 20X) play an important role in the regulation of appetite and satiety, energy expenditure, insulin sensitivity and insulin secretion,

SC

inflammation, blood pressure, hemostasis, endothelial function and others.

MA NU

Figure 2. Translational adipokine research. Adipokines have the potential to be used in clinical practice as markers (predictors) for obesity related diseases, mortality, individual disease outcomes or as substrates/targets for pharmacological management of obesity and prevention of obesity related disorders. For several adipokines there is evidence for the

ED

prediction of obesity complications, but also a need for future development and validation.

PT

Some adipokines have been already used in clinical studies for different indications, whereas

AC

CE

for more recently discovered adipokines only data from animal experiments are available.

25

ACCEPTED MANUSCRIPT Table 1. Examples of adipokines and main function(s).

Adipokine

Target action Regulation of satiety, appetite, food intake,

T

Leptin

RI P

activity and energy expenditure, fertility, atherogenesis and growth induction Antidiabetic, anti-atherogenic and anti-

SC

Adiponectin

inflammatory

Chemoattractant protein, regulation of

Chemerin

MA NU

adipogenesis

Pro-inflammatory

TNFα

Pro-inflammatory

IL-1β

Stimulates glucose uptake into adipocytes,

FGF21

ED

increases thermogenesis, energy expenditure,

PT

Retinol binding protein-4 (RBP4)

CE

Vaspin

AC

Visfatin/PBEF/Nampt

fat utilization, lowers glucose and triglycerides Related to insulin resistance, visceral fat distribution Decreases food intake, improves hyperglycemia Nampt-mediated systemic NAD biosynthesis is critical for β cell function

Monocyte chemotactic protein-1

Chemoattractant protein, promotes adipose

(MCP-1)

tissue inflammation

Progranulin

Promotes adipose tissue inflammation

DPP-4

Degrades GIP and GLP-1 Inhibitors in clinical use for type 2 diabetes

BMP-7

Stimulates brown adipogenesis, reduces food intake, increases energy expenditure

26

ACCEPTED MANUSCRIPT References

T

[1] Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning

RI P

of the mouse obese gene and its human homologue. Nature 1994;372:425-32. [2] Cook KS, Min HY, Johnson D, Chaplinsky RJ, Flier JS, Hunt CR, et al. Adipsin: a

SC

circulating serine protease homolog secreted by adipose tissue and sciatic nerve. Science 1987;237:402-5.

MA NU

[3] Flier JS, Cook KS, Usher P, Spiegelman BM. Severely impaired adipsin expression in genetic and acquired obesity. Science 1987;237:405-8.

[4] Van Gaal LF, Mertens IL, De Block CE. Mechanisms linking obesity with cardiovascular disease. Nature 2006;444:875-80.

ED

[5] Blüher M. Adipose tissue dysfunction in obesity. Exp Clin Endocrinol Diabetes

PT

2009;117:241-50.

[6] Sharma AM, Padwal R. Obesity is a sign - over-eating is a symptom: an aetiological

CE

framework for the assessment and management of obesity. Obes Rev 2010;11:362-70.

AC

[7] Wadden TA. Treatment of obesity by moderate and severe caloric restriction. Results of clinical research trials. Ann Intern Med 1993;119:688-93. [8] Sjostrom L, Narbro K, Sjostrom CD, Karason K, Larsson B, Wedel H, et al. Effects of bariatric surgery on mortality in Swedish obese subjects. N Engl J Med 2007;357:741-52. [9] Blüher M. Adipokines - removing road blocks to obesity and diabetes therapy. Mol Metab 2014;3:230-40. [10] Blüher M. Clinical relevance of adipokines. Diabetes Metab J 2012;36:317-27. [11] Catalan V, Gomez-Ambrosi J, Rodriguez A, Salvador J, Fruhbeck G. Adipokines in the treatment of diabetes mellitus and obesity. Expert Opin Pharmacother 2009;10:239-54. [12] Sahin-Efe A, Katsikeris F, Mantzoros CS. Advances in adipokines. Metabolism 2012;61:1659-65. 27

ACCEPTED MANUSCRIPT [13] Tangpricha V. Translational Research in Endocrinology. Journal of Clinical & Translational Endocrinology 2014;1:e1.

T

[14] Dalamaga M, Diakopoulos KN, Mantzoros CS. The role of adiponectin in cancer: a

RI P

review of current evidence. Endocr Rev 2012;33:547-94.

[15] Kelesidis T, Kelesidis I, Chou S, Mantzoros CS. Narrative review: the role of leptin in

SC

human physiology: emerging clinical applications. Ann Intern Med 2010;152:93-100. [16] Kelesidis T, Mantzoros CS. The emerging role of leptin in humans. Pediatr Endocrinol

MA NU

Rev 2006;3:239-48.

[17] Lehr S, Hartwig S, Sell H. Adipokines: a treasure trove for the discovery of biomarkers for metabolic disorders. Proteomics Clin Appl 2012;6:91-101. [18] Farooqi IS, Jebb SA, Langmack G, Lawrence E, Cheetham CH, Prentice AM, et al.

ED

Effects of recombinant leptin therapy in a child with congenital leptin deficiency. N Engl J

PT

Med 1999;341:879-84.

[19] Foo JP, Mantzoros CS. Leptin in congenital or HIV-associated lipodystrophy and

CE

metabolic syndrome: a need for more mechanistic studies and large, randomized, placebo-

AC

controlled trials. Metabolism 2012;61:1331-6. [20] Chou K, Perry CM. Metreleptin: first global approval. Drugs 2013;73:989-97. [21] Savage DB, O'Rahilly S. Leptin: a novel therapeutic role in lipodystrophy. J Clin Invest 2002;109:1285-6. [22] Ku IA, Farzaneh-Far R, Vittinghoff E, Zhang MH, Na B, Whooley MA. Association of low leptin with cardiovascular events and mortality in patients with stable coronary artery disease: the Heart and Soul Study. Atherosclerosis 2011;217:503-8. [23] Welsh P, Murray HM, Buckley BM, de Craen AJ, Ford I, Jukema JW, et al. Leptin predicts diabetes but not cardiovascular disease: results from a large prospective study in an elderly population. Diabetes Care 2009;32:308-10.

28

ACCEPTED MANUSCRIPT [24] Brennan AM, Li TY, Kelesidis I, Gavrila A, Hu FB, Mantzoros CS. Circulating leptin levels are not associated with cardiovascular morbidity and mortality in women with diabetes:

T

a prospective cohort study. Diabetologia 2007;50:1178-85.

RI P

[25] Wu ZJ, Cheng YJ, Gu WJ, Aung LH. Adiponectin is associated with increased mortality in patients with already established cardiovascular disease: a systematic review and meta-

SC

analysis. Metabolism 2014;63:1157-66.

[26] Karas MG, Benkeser D, Arnold AM, Bartz TM, Djousse L, Mukamal KJ, et al. Relations

MA NU

of plasma total and high-molecular-weight adiponectin to new-onset heart failure in adults >/=65 years of age (from the Cardiovascular Health study). Am J Cardiol 2014;113:328-34. [27] Sook Lee E, Park SS, Kim E, Sook Yoon Y, Ahn HY, Park CY, et al. Association between adiponectin levels and coronary heart disease and mortality: a systematic review and

ED

meta-analysis. Int J Epidemiol 2013;42:1029-39.

PT

[28] Cote M, Cartier A, Reuwer AQ, Arsenault BJ, Lemieux I, Despres JP, et al. Adiponectin and risk of coronary heart disease in apparently healthy men and women (from the EPIC-

CE

Norfolk Prospective Population Study). Am J Cardiol 2011;108:367-73.

AC

[29] Brault M, Ray J, Gomez YH, Mantzoros CS, Daskalopoulou SS. Statin treatment and new-onset diabetes: a review of proposed mechanisms. Metabolism 2014;63:735-45. [30] Kuwashiro T, Ago T, Kamouchi M, Matsuo R, Hata J, Kuroda J, et al. Significance of plasma adiponectin for diagnosis, neurological severity and functional outcome in ischemic stroke - Research for Biomarkers in Ischemic Stroke (REBIOS). Metabolism 2014;63:1093103. [31] Forsblom C, Thomas MC, Moran J, Saraheimo M, Thorn L, Waden J, et al. Serum adiponectin concentration is a positive predictor of all-cause and cardiovascular mortality in type 1 diabetes. J Intern Med 2011;270:346-55.

29

ACCEPTED MANUSCRIPT [32] Christensen HM, Kistorp C, Schou M, Keller N, Zerahn B, Frystyk J, et al. Cross-talk between the heart and adipose tissue in cachectic heart failure patients with respect to

T

alterations in body composition: a prospective study. Metabolism 2014;63:141-9.

RI P

[33] Kizer JR. Adiponectin, cardiovascular disease, and mortality: Parsing the dual prognostic implications of a complex adipokine. Metabolism 2014;63:1079-83.

SC

[34] Sahebkar A. Head-to-head comparison of fibrates versus statins for elevation of circulating adiponectin concentrations: a systematic review and meta-analysis. Metabolism

MA NU

2013;62:1876-85.

[35] Meyvis K, Verrijken A, Wouters K, Van Gaal L. Plasma adiponectin level is inversely correlated with albuminuria in overweight and obese nondiabetic individuals. Metabolism 2013;62:1570-6.

ED

[36] Spranger J, Kroke A, Mohlig M, Bergmann MM, Ristow M, Boeing H, et al.

PT

Adiponectin and protection against type 2 diabetes mellitus. Lancet 2003;361:226-8. [37] Heidemann C, Sun Q, van Dam RM, Meigs JB, Zhang C, Tworoger SS, et al. Total and

CE

high-molecular-weight adiponectin and resistin in relation to the risk for type 2 diabetes in

AC

women. Ann Intern Med 2008;149:307-16. [38] Chan JL, Matarese G, Shetty GK, Raciti P, Kelesidis I, Aufiero D, et al. Differential regulation of metabolic, neuroendocrine, and immune function by leptin in humans. Proc Natl Acad Sci U S A 2006;103:8481-6. [39] Spyridopoulos TN, Petridou ET, Skalkidou A, Dessypris N, Chrousos GP, Mantzoros CS, et al. Low adiponectin levels are associated with renal cell carcinoma: a case-control study. Int J Cancer 2007;120:1573-8. [40] Vetvik KK, Sonerud T, Lindeberg M, Luders T, Storkson RH, Jonsdottir K, et al. Globular adiponectin and its downstream target genes are up-regulated locally in human colorectal tumors: ex vivo and in vitro studies. Metabolism 2014;63:672-81.

30

ACCEPTED MANUSCRIPT [41] Michalakis K, Williams CJ, Mitsiades N, Blakeman J, Balafouta-Tselenis S, Giannopoulos A, et al. Serum adiponectin concentrations and tissue expression of adiponectin

T

receptors are reduced in patients with prostate cancer: a case control study. Cancer Epidemiol

RI P

Biomarkers Prev 2007;16:308-13.

[42] Li H, Stampfer MJ, Mucci L, Rifai N, Qiu W, Kurth T, et al. A 25-year prospective study

SC

of plasma adiponectin and leptin concentrations and prostate cancer risk and survival. Clin Chem 2010;56:34-43.

MA NU

[43] Aleksandrova K, Boeing H, Nothlings U, Jenab M, Fedirko V, Kaaks R, et al. Inflammatory and metabolic biomarkers and risk of liver and biliary tract cancer. Hepatology 2014;60:858-71.

[44] Blüher M, Brennan AM, Kelesidis T, Kratzsch J, Fasshauer M, Kralisch S, et al. Total

ED

and high-molecular weight adiponectin in relation to metabolic variables at baseline and in

Care 2007;30:280-5.

PT

response to an exercise treatment program: comparative evaluation of three assays. Diabetes

CE

[45] Hirata A, Kishida K, Kobayashi H, Nakatsuji H, Funahashi T, Shimomura I. Correlation

AC

between serum C1q-adiponectin/total adiponectin ratio and polyvascular lesions detected by vascular ultrasonography in Japanese type 2 diabetics. Metabolism 2013;62:376-85. [46] Wolfe BE, Jimerson DC, Orlova C, Mantzoros CS. Effect of dieting on plasma leptin, soluble leptin receptor, adiponectin and resistin levels in healthy volunteers. Clin Endocrinol (Oxf) 2004;61:332-8. [47] Shea SA, Hilton MF, Orlova C, Ayers RT, Mantzoros CS. Independent circadian and sleep/wake regulation of adipokines and glucose in humans. J Clin Endocrinol Metab 2005;90:2537-44. [48] Qi L, Doria A, Manson JE, Meigs JB, Hunter D, Mantzoros CS, et al. Adiponectin genetic variability, plasma adiponectin, and cardiovascular risk in patients with type 2 diabetes. Diabetes 2006;55:1512-6. 31

ACCEPTED MANUSCRIPT [49] Kaklamani VG, Wisinski KB, Sadim M, Gulden C, Do A, Offit K, et al. Variants of the adiponectin (ADIPOQ) and adiponectin receptor 1 (ADIPOR1) genes and colorectal cancer

T

risk. JAMA 2008;300:1523-31.

dysfunction in obesity. Annu Rev Nutr 2014;34:207-36.

RI P

[50] Trayhurn P. Hypoxia and adipocyte physiology: implications for adipose tissue

SC

[51] Kovsan J, Blüher M, Tarnovscki T, Klöting N, Kirshtein B, Madar L, et al. Altered autophagy in human adipose tissues in obesity. J Clin Endocrinol Metab 2011;96:E268-77.

MA NU

[52] Blüher M, Klöting N, Wueest S, Schoenle EJ, Schon MR, Dietrich A, et al. Fas and FasL expression in human adipose tissue is related to obesity, insulin resistance, and type 2 diabetes. J Clin Endocrinol Metab 2014;99:E36-44.

[53] Rudich A, Kanety H, Bashan N. Adipose stress-sensing kinases: linking obesity to

ED

malfunction. Trends Endocrinol Metab 2007;18:291-9.

PT

[54] Klöting N, Fasshauer M, Dietrich A, Kovacs P, Schon MR, Kern M, et al. Insulinsensitive obesity. Am J Physiol Endocrinol Metab 2010;299:E506-15.

CE

[55] Flehmig G, Scholz M, Klöting N, Fasshauer M, Tonjes A, Stumvoll M, et al.

AC

Identification of adipokine clusters related to parameters of fat mass, insulin sensitivity and inflammation. PloS One 2014;9:e99785. [56] Shai I, Schwarzfuchs D, Henkin Y, Shahar DR, Witkow S, Greenberg I, et al. Weight loss with a low-carbohydrate, Mediterranean, or low-fat diet. N Engl J Med 2008;359:229-41. [57] Blüher M, Rudich A, Klöting N, Golan R, Henkin Y, Rubin E, et al. Two patterns of adipokine and other biomarker dynamics in a long-term weight loss intervention. Diabetes Care 2012;35:342-9. [58] Erez G, Tirosh A, Rudich A, Meiner V, Schwarzfuchs D, Sharon N, et al. Phenotypic and genetic variation in leptin as determinants of weight regain. Int J Obes (Lond) 2011;35:785-92.

32

ACCEPTED MANUSCRIPT [59] Wang P, Menheere PP, Astrup A, Andersen MR, van Baak MA, Larsen TM, et al. Metabolic syndrome, circulating RBP4, testosterone, and SHBG predict weight regain at 6

T

months after weight loss in men. Obesity 2013;21:1997-2006.

RI P

[60] Despres JP, Nadeau A, Tremblay A, Ferland M, Moorjani S, Lupien PJ, et al. Role of deep abdominal fat in the association between regional adipose tissue distribution and glucose

SC

tolerance in obese women. Diabetes 1989;38:304-9.

Diabetologia 2014;57:1276-86.

MA NU

[61] Schleinitz D, Bottcher Y, Blüher M, Kovacs P. The genetics of fat distribution.

[62] Machann J, Thamer C, Schnoedt B, Haap M, Haring HU, Claussen CD, et al. Standardized assessment of whole body adipose tissue topography by MRI. J Magn Reson Imaging 2005;21:455-62.

ED

[63] Fukuhara A, Matsuda M, Nishizawa M, Segawa K, Tanaka M, Kishimoto K, et al.

PT

Visfatin: a protein secreted by visceral fat that mimics the effects of insulin. Science 2005;307:426-30.

CE

[64] Berndt J, Klöting N, Kralisch S, Kovacs P, Fasshauer M, Schon MR, et al. Plasma

AC

visfatin concentrations and fat depot-specific mRNA expression in humans. Diabetes 2005;54:2911-6.

[65] Yoshino J, Mills KF, Yoon MJ, Imai S. Nicotinamide mononucleotide, a key NAD(+) intermediate, treats the pathophysiology of diet- and age-induced diabetes in mice. Cell Metab 2011;14:528-36. [66] Klöting N, Graham TE, Berndt J, Kralisch S, Kovacs P, Wason CJ, et al. Serum retinolbinding protein is more highly expressed in visceral than in subcutaneous adipose tissue and is a marker of intra-abdominal fat mass. Cell Metab 2007;6:79-87. [67] Kelly KR, Kashyap SR, O'Leary VB, Major J, Schauer PR, Kirwan JP. Retinol-binding protein 4 (RBP4) protein expression is increased in omental adipose tissue of severely obese patients. Obesity 2010;18:663-6. 33

ACCEPTED MANUSCRIPT [68] Stefan N, Hennige AM, Staiger H, Machann J, Schick F, Schleicher E, et al. High circulating retinol-binding protein 4 is associated with elevated liver fat but not with total,

T

subcutaneous, visceral, or intramyocellular fat in humans. Diabetes Care 2007;30:1173-8.

protein 4 in human obesity. Diabetes 2006;55:2805-10.

RI P

[69] Janke J, Engeli S, Boschmann M, Adams F, Bohnke J, Luft FC, et al. Retinol-binding

SC

[70] Mussig K, Staiger H, Machicao F, Thamer C, Machann J, Schick F, et al. RARRES2, encoding the novel adipokine chemerin, is a genetic determinant of disproportionate regional

MA NU

body fat distribution: a comparative magnetic resonance imaging study. Metabolism 2009;58:519-24.

[71] Chakaroun R, Raschpichler M, Klöting N, Oberbach A, Flehmig G, Kern M, et al. Effects of weight loss and exercise on chemerin serum concentrations and adipose tissue

ED

expression in human obesity. Metabolism 2012;61:706-14.

PT

[72] Youn BS, Bang SI, Klöting N, Park JW, Lee N, Oh JE, et al. Serum progranulin concentrations may be associated with macrophage infiltration into omental adipose tissue.

CE

Diabetes 2009;58:627-36.

AC

[73] Yang RZ, Lee MJ, Hu H, Pray J, Wu HB, Hansen BC, et al. Identification of omentin as a novel depot-specific adipokine in human adipose tissue: possible role in modulating insulin action. Am J Physiol Endocrinol Metab 2006;290:E1253-61. [74] Blüher M. Adipose tissue dysfunction contributes to obesity related metabolic diseases. Best Pract Res Clin Endocrinol Metab 2013;27:163-77. [75] Ussar S, Bezy O, Blüher M, Kahn CR. Glypican-4 enhances insulin signaling via interaction with the insulin receptor and serves as a novel adipokine. Diabetes 2012;61:228998. [76] Sell H, Blüher M, Klöting N, Schlich R, Willems M, Ruppe F, et al. Adipose dipeptidyl peptidase-4 and obesity: correlation with insulin resistance and depot-specific release from adipose tissue in vivo and in vitro. Diabetes Care 2013;36:4083-90. 34

ACCEPTED MANUSCRIPT [77] Arnoldussen IA, Kiliaan AJ, Gustafson DR. Obesity and dementia: Adipokines interact with the brain. Eur Neuropsychopharmacol. 2014; pii: S0924-977X(14)00087-X.

T

[78] Bonda DJ, Stone JG, Torres SL, Siedlak SL, Perry G, Kryscio R, et al. Dysregulation of

RI P

leptin signaling in Alzheimer disease: evidence for neuronal leptin resistance. J Neurochem 2014;128:162-72.

SC

[79] Greco SJ, Sarkar S, Johnston JM, Zhu X, Su B, Casadesus G, et al. Leptin reduces Alzheimer's disease-related tau phosphorylation in neuronal cells. Biochem Biophys Res

MA NU

Commun 2008;376:536–41.

[80] Tezapsidis N, Johnston JM, Smith MA, Ashford JW, Casadesus G, Robakis NK, et al. Leptin: a novel therapeutic strategy for Alzheimer's disease. J Alzheimers Dis 2009;16:73140.

ED

[81] Teixeira AL, Diniz BS, Campos AC, Miranda AS, Rocha NP, Talib LL, et al. Decreased

PT

levels of circulating adiponectin in mild cognitive impairment and Alzheimer's disease. Neuromolecular Med 2013;15:115-21.

CE

[82] van Himbergen TM, Beiser AS, Ai M, Seshadri S, Otokozawa S, Au R, et al. Biomarkers

AC

for insulin resistance and inflammation and the risk for all-cause dementia and alzheimer disease: results from the Framingham Heart Study. Arch Neurol 2012;69:594-600. [83] Khemka VK, Bagchi D, Bandyopadhyay K, Bir A, Chattopadhyay M, Biswas A Altered serum levels of adipokines and insulin in probable Alzheimer's disease. J Alzheimers Dis 2014;41:525-33. [84] Warren MW1, Hynan LS, Weiner MF. Lipids and adipokines as risk factors for Alzheimer's disease. J Alzheimers Dis 2012;29:151-7. [85] Kiliaan AJ, Arnoldussen IA, Gustafson DR. Adipokines: a link between obesity and dementia? Lancet Neurol 2014;13:913-23.

35

ACCEPTED MANUSCRIPT [86] Liu J, Chi N, Chen H, Zhang J, Bian Y, Cui G, et al. Resistin protection against endogenous Aβ neuronal cytotoxicity from mitochondrial pathway. Brain Res 2013;1523:77-

T

84.

RI P

[87] Okada-Iwabu M, Yamauchi T, Iwabu M, Honma T, Hamagami K, Matsuda K, et al. A small-molecule AdipoR agonist for type 2 diabetes and short life in obesity. Nature

SC

2013;503:493-9.

[88] Gaich G, Chien JY, Fu H, Glass LC, Deeg MA, Holland WL, et al. The effects of

MA NU

LY2405319, an FGF21 analog, in obese human subjects with type 2 diabetes. Cell Metab 2013;18:333-40.

[89] Drucker DJ. The role of gut hormones in glucose homeostasis. J Clin Invest 2007;117:24-32.

ED

[90] Vaccaro AR, Whang PG, Patel T, Phillips FM, Anderson DG, Albert TJ, et al. The safety

PT

and efficacy of OP-1 (rhBMP-7) as a replacement for iliac crest autograft for posterolateral lumbar arthrodesis: minimum 4-year follow-up of a pilot study. Spine J 2008;8:457-65.

CE

[91] Yazdani-Biuki B, Stelzl H, Brezinschek HP, Hermann J, Mueller T, Krippl P, et al.

AC

Improvement of insulin sensitivity in insulin resistant subjects during prolonged treatment with the anti-TNF-alpha antibody infliximab. Eur J Clin Invest 2004;34:641-2. [92] Stanley TL, Zanni MV, Johnsen S, Rasheed S, Makimura H, Lee H, et al. TNF-alpha antagonism with etanercept decreases glucose and increases the proportion of high molecular weight adiponectin in obese subjects with features of the metabolic syndrome. J Clin Endocrinol Metab 2011;96:E146-50. [93] Larsen CM, Faulenbach M, Vaag A, Volund A, Ehses JA, Seifert B, et al. Interleukin-1receptor antagonist in type 2 diabetes mellitus. N Engl J Med 2007;356:1517-26. [94] Watts SW, Dorrance AM, Penfold ME, Rourke JL, Sinal CJ, Seitz B, et al. Chemerin connects fat to arterial contraction. Arterioscler Thromb Vasc Biol 2013;33:1320-8.

36

ACCEPTED MANUSCRIPT [95] Licinio J, Mantzoros C, Negrao AB, Cizza G, Wong ML, Bongiorno PB, et al. Human leptin levels are pulsatile and inversely related to pituitary-adrenal function. Nat Med

T

1997;3:575-9.

RI P

[96] Mantzoros CS, Moschos S, Avramopoulos I, Kaklamani V, Liolios A, Doulgerakis DE, et al. Leptin concentrations in relation to body mass index and the tumor necrosis factor-alpha

SC

system in humans. J Clin Endocrinol Metab 1997;82:3408-13.

[97] Mantzoros CS, Liolios AD, Tritos NA, Kaklamani VG, Doulgerakis DE, Griveas I, et al.

MA NU

Circulating insulin concentrations, smoking, and alcohol intake are important independent predictors of leptin in young healthy men. Obes Res 1998;6:179-86. [98] Mantzoros CS, Varvarigou A, Kaklamani VG, Beratis NG, Flier JS. Effect of birth weight and maternal smoking on cord blood leptin concentrations of full-term and preterm

ED

newborns. J Clin Endocrinol Metab 1997;82:2856-61.

PT

[99] Ahima RS, Prabakaran D, Mantzoros C, Qu D, Lowell B, Maratos-Flier E, et al. Role of leptin in the neuroendocrine response to fasting. Nature 1996;382:250-2.

CE

[100] Chan JL, Heist K, DePaoli AM, Veldhuis JD, Mantzoros CS. The role of falling leptin

AC

levels in the neuroendocrine and metabolic adaptation to short-term starvation in healthy men. J Clin Invest 2003;111:1409-21. [101] Welt CK, Chan JL, Bullen J, Murphy R, Smith P, DePaoli AM, et al. Recombinant human leptin in women with hypothalamic amenorrhea. N Engl J Med 2004;351:987-97. [102] Mantzoros CS, Flier JS, Rogol AD. A longitudinal assessment of hormonal and physical alterations during normal puberty in boys. V. Rising leptin levels may signal the onset of puberty. J Clin Endocrinol Metab 1997;82:1066-70. [103] Mantzoros CS, Dunaif A, Flier JS. Leptin concentrations in the polycystic ovary syndrome. J Clin Endocrinol Metab 1997;82:1687-91.

37

ACCEPTED MANUSCRIPT [104] Christou H, Connors JM, Ziotopoulou M, Hatzidakis V, Papathanassoglou E, Ringer SA, et al. Cord blood leptin and insulin-like growth factor levels are independent predictors of

T

fetal growth. J Clin Endocrinol Metab 2001;86:935-8.

RI P

[105] Roemmich JN, Clark PA, Berr SS, Mai V, Mantzoros CS, Flier JS, et al. Gender differences in leptin levels during puberty are related to the subcutaneous fat depot and sex

SC

steroids. Am J Physiol 1998;275:E543-51.

[106] Mantzoros CS, Cramer DW, Liberman RF, Barbieri RL. Predictive value of serum and

MA NU

follicular fluid leptin concentrations during assisted reproductive cycles in normal women and in women with the polycystic ovarian syndrome. Human reproduction. 2000;15:539-44. [107] Ahima RS, Flier JS. Leptin. Annu Rev Physiol 2000;62:413-37. [108] Halaas JL, Gajiwala KS, Maffei M, Cohen SL, Chait BT, Rabinowitz D, et al. Weight-

ED

reducing effects of the plasma protein encoded by the obese gene. Science 1995;269:543-6.

PT

[109] Farooqi IS, Matarese G, Lord GM, Keogh JM, Lawrence E, Agwu C, et al. Beneficial effects of leptin on obesity, T cell hyporesponsiveness, and neuroendocrine/metabolic

CE

dysfunction of human congenital leptin deficiency. J Clin Invest 2002;110:1093-103.

AC

[110] Heymsfield SB, Greenberg AS, Fujioka K, Dixon RM, Kushner R, Hunt T, et al. Recombinant leptin for weight loss in obese and lean adults: a randomized, controlled, doseescalation trial. JAMA 1999;282:1568-75. [111] Hukshorn CJ, Saris WH, Westerterp-Plantenga MS, Farid AR, Smith FJ, Campfield LA. Weekly subcutaneous pegylated recombinant native human leptin (PEG-OB) administration in obese men. J Clin Endocrinol Metab 2000;85:4003-9. [112] Van Heek M, Compton DS, France CF, Tedesco RP, Fawzi AB, Graziano MP, et al. Diet-induced obese mice develop peripheral, but not central, resistance to leptin. J Clin Invest 1997;99:385-90.

38

ACCEPTED MANUSCRIPT [113] Mantzoros CS, Magkos F, Brinkoetter M, Sienkiewicz E, Dardeno TA, Kim SY, et al. Leptin in human physiology and pathophysiology. Am J Physiol Endocrinol Metab

T

2011;301:E567-84.

RI P

[114] Dardeno TA, Chou SH, Moon HS, Chamberland JP, Fiorenza CG, Mantzoros CS. Leptin in human physiology and therapeutics. Front Neuroendocrinol 2010;31:377-93.

SC

[115] Muller TD, Sullivan LM, Habegger K, Yi CX, Kabra D, Grant E, et al. Restoration of leptin responsiveness in diet-induced obese mice using an optimized leptin analog in

MA NU

combination with exendin-4 or FGF21. J Pept Sci 2012;18:383-93. [116] Moon HS, Chamberland JP, Diakopoulos KN, Fiorenza CG, Ziemke F, Schneider B, et al. Leptin and amylin act in an additive manner to activate overlapping signaling pathways in peripheral tissues: in vitro and ex vivo studies in humans. Diabetes Care 2011;34:132-8.

ED

[117] Moon HS, Chamberland JP, Mantzoros CS. Amylin and leptin activate overlapping

PT

signalling pathways in an additive manner in mouse GT1-7 hypothalamic, C(2)C(1)(2) muscle and AML12 liver cell lines. Diabetologia 2012;55:215-25.

CE

[118] Moon HS, Dalamaga M, Kim SY, Polyzos SA, Hamnvik OP, Magkos F, et al. Leptin's

AC

role in lipodystrophic and nonlipodystrophic insulin-resistant and diabetic individuals. Endocr Rev 2013;34:377-412. [119] Moon HS, Matarese G, Brennan AM, Chamberland JP, Liu X, Fiorenza CG, et al. Efficacy of metreleptin in obese patients with type 2 diabetes: cellular and molecular pathways underlying leptin tolerance. Diabetes 2011;60:1647-56. [120] Moon HS DF, Huh JY, Schneider BE, Mantzoros CS. Identification, and saturable nature, of signaling pathways induced by metreleptin in humans: comparative evaluation of in vivo, ex vivo and in vitro administration. Diabetes 2014. [121] Matarese G, La Rocca C, Moon HS, Huh JY, Brinkoetter MT, Chou S, et al. Selective capacity of metreleptin administration to reconstitute CD4+ T-cell number in females with acquired hypoleptinemia. Proc Natl Acad Sci U S A 2013;110:E818-27. 39

ACCEPTED MANUSCRIPT [122] Roth JD, Roland BL, Cole RL, Trevaskis JL, Weyer C, Koda JE, et al. Leptin responsiveness restored by amylin agonism in diet-induced obesity: evidence from nonclinical

T

and clinical studies. Proc Natl Acad Sci U S A 2008;105:7257-62.

RI P

[123] Ravussin E, Smith SR, Mitchell JA, Shringarpure R, Shan K, Maier H, et al. Enhanced weight loss with pramlintide/metreleptin: an integrated neurohormonal approach to obesity

SC

pharmacotherapy. Obesity 2009;17:1736-43.

[124] Brown RJ, Cochran E, Gorden P. Metreleptin improves blood glucose in patients with

MA NU

insulin receptor mutations. J Clin Endocrinol Metab 2013;98:E1749-56. [125] Paruthi J, Gill N, Mantzoros CS. Adipokines in the HIV/HAART-associated lipodystrophy syndrome. Metabolism: clinical and experimental. 2013;62:1199-205. [126] Tsoukas MA, Farr OM, Mantzoros CS. Advances in metabolism. Metabolism

ED

2013;62:1700-13.

PT

[127] Chan JL, Mantzoros CS. Role of leptin in energy-deprivation states: normal human physiology and clinical implications for hypothalamic amenorrhoea and anorexia nervosa.

CE

Lancet 2005;366:74-85.

AC

[128] Chou SH, Chamberland JP, Liu X, Matarese G, Gao C, Stefanakis R, et al. Leptin is an effective treatment for hypothalamic amenorrhea. Proc Natl Acad Sci U S A 2011;108:658590.

[129] Audi L, Mantzoros CS, Vidal-Puig A, Vargas D, Gussinye M, Carrascosa A. Leptin in relation to resumption of menses in women with anorexia nervosa. Mol Psychiatry 1998;3:544-7. [130] Sienkiewicz E, Magkos F, Aronis K, Brinkoetter MT, Chamberland JP, Chou SH, et al. Long-term metreleptin treatment increases bone mineral density and content at the lumbar spine of lean hypoleptinemic women. Metabolism 2011;60:1211-21. [131] Chan JL, Moschos SJ, Bullen J, Heist K, Li X, Kim YB, et al. Recombinant methionyl human leptin administration activates signal transducer and activator of transcription 3 40

ACCEPTED MANUSCRIPT signaling in peripheral blood mononuclear cells in vivo and regulates soluble tumor necrosis factor-alpha receptor levels in humans with relative leptin deficiency. J Clin Endocrinol

T

Metab 2005;90:1625-31.

RI P

[132] Sekhar RV, Jahoor F, Iyer D, Guthikonda A, Paranilam J, Elhaj F, et al. Leptin replacement therapy does not improve the abnormal lipid kinetics of hypoleptinemic patients

SC

with HIV-associated lipodystrophy syndrome. Metabolism 2012;61:1395-403. [133] Lee JH, Chan JL, Sourlas E, Raptopoulos V, Mantzoros CS. Recombinant methionyl

MA NU

human leptin therapy in replacement doses improves insulin resistance and metabolic profile in patients with lipoatrophy and metabolic syndrome induced by the highly active antiretroviral therapy. J Clin Endocrinol Metab 2006;91:2605-11. [134] Mantzoros CS. Whither recombinant human leptin treatment for HIV-associated

ED

lipoatrophy and the metabolic syndrome? J Clin Endocrinol Metab 2009;94:1089-91.

PT

[135] Mulligan K, Khatami H, Schwarz JM, Sakkas GK, DePaoli AM, Tai VW, et al. The effects of recombinant human leptin on visceral fat, dyslipidemia, and insulin resistance in

CE

patients with human immunodeficiency virus-associated lipoatrophy and hypoleptinemia. J

AC

Clin Endocrinol Metab 2009;94:1137-44. [136] Ye R, Scherer PE. Adiponectin, driver or passenger on the road to insulin sensitivity? Mol Metab 2013;2:133-41. [137] Scherer PE, Williams S, Fogliano M, Baldini G, Lodish HF. A novel serum protein similar to C1q, produced exclusively in adipocytes. J Biol Chem 1995;270:26746-9. [138] Mantzoros CS, Rifas-Shiman SL, Williams CJ, Fargnoli JL, Kelesidis T, Gillman MW. Cord blood leptin and adiponectin as predictors of adiposity in children at 3 years of age: a prospective cohort study. Pediatrics 2009;123:682-9. [139] Ziemke F, Mantzoros CS. Adiponectin in insulin resistance: lessons from translational research. Am J Clin Nutr 2010;91:258S-61S.

41

ACCEPTED MANUSCRIPT [140] Turer AT, Scherer PE. Adiponectin: mechanistic insights and clinical implications. Diabetologia 2012;55:2319-26.

T

[141] Okamoto M, Ohara-Imaizumi M, Kubota N, Hashimoto S, Eto K, Kanno T, et al.

RI P

Adiponectin induces insulin secretion in vitro and in vivo at a low glucose concentration. Diabetologia 2008;51:827-35.

SC

[142] Tullin S, Sams A, Brandt J, Dahl K, Gong W, Jeppesen CB, et al. Recombinant adiponectin does not lower plasma glucose in animal models of type 2 diabetes. PloS One

MA NU

2012;7:e44270.

[143] Holland WL, Scherer PE. Cell Biology. Ronning after the adiponectin receptors. Science 2013;342:1460-1.

[144] Lubbers ER, List EO, Jara A, Sackman-Sala L, Cordoba-Chacon J, Gahete MD, et al.

PT

Endocrinol 2013;216:363-74.

ED

Adiponectin in mice with altered GH action: links to insulin sensitivity and longevity? J

[145] Kadowaki T, Yamauchi T, Okada-Iwabu M, Iwabu M. Adiponectin and its receptors:

AC

2014;2:8-9.

CE

implications for obesity-associated diseases and longevity. Lancet Diabetes Endocrinol

[146] Otabe S, Yuan X, Fukutani T, Wada N, Hashinaga T, Nakayama H, et al. Overexpression of human adiponectin in transgenic mice results in suppression of fat accumulation and prevention of premature death by high-calorie diet. Am J Physiol Endocrinol Metab 2007;293:E210–8. [147] Blüher M, Kahn BB, Kahn CR. Extended longevity in mice lacking the insulin receptor in adipose tissue. Science 2003;299:572-4. [148] Flurkey K, Papaconstantinou J, Miller RA, Harrison DE. Lifespan extension and delayed immune and collagen aging in mutant mice with defects in growth hormone production. Proc Natl Acad Sci U S A 2001;98:6736–41.

42

ACCEPTED MANUSCRIPT [149] McKee Alderman J, DePetrillo MA, Gluesenkamp AM, Hartley AC, Verhoff SV, Zavodni KL, et al.,Calorie restriction and dwarf mice in gerontological research. Gerontology

T

2010;56:404–9.

RI P

[150] Arai Y, Nakazawa S, Kojima T, Takayama M, Ebihara Y, Shimizu K, et al. High adiponectin concentration and its role for longevity in female centenarians. J Gerontol A Biol

SC

Sci Med Sci 2006;6:32–9.

[151] Moon HS, Mantzoros CS. Adiponectin and metformin additively attenuate IL1beta-

MA NU

induced malignant potential of colon cancer. Endocr Relat Cancer 2013;20:849-59. [152] Reddy SM, Sadim M, Li J, Yi N, Agarwal S, Mantzoros CS, et al. Clinical and genetic predictors of weight gain in patients diagnosed with breast cancer. Br J Cancer 2013;109:87281.

ED

[153] Kaklamani VG, Hoffmann TJ, Thornton TA, Hayes G, Chlebowski R, Van Horn L, et

PT

al. Adiponectin pathway polymorphisms and risk of breast cancer in African Americans and Hispanics in the Women's Health Initiative. Breast Cancer Res Treat 2013;139:461-8.

CE

[154] Huffman DM, Augenlicht LH, Zhang X, Lofrese JJ, Atzmon G, Chamberland JP, et al.

AC

Abdominal obesity, independent from caloric intake, accounts for the development of intestinal tumors in Apc(1638N/+) female mice. Cancer Prev Res (Phila) 2013;6:177-87. [155] Moon HS, Liu X, Nagel JM, Chamberland JP, Diakopoulos KN, Brinkoetter MT, et al. Salutary effects of adiponectin on colon cancer: in vivo and in vitro studies in mice. Gut 2013;62:561-70. [156] Moon HS, Chamberland JP, Aronis K, Tseleni-Balafouta S, Mantzoros CS. Direct role of adiponectin and adiponectin receptors in endometrial cancer: in vitro and ex vivo studies in humans. Mol Cancer Ther 2011;10:2234-43. [157] Mitsiades N, Pazaitou-Panayiotou K, Aronis KN, Moon HS, Chamberland JP, Liu X, et al. Circulating adiponectin is inversely associated with risk of thyroid cancer: in vivo and in vitro studies. J Clin Endocrinol Metab 2011;96:E2023-8. 43

ACCEPTED MANUSCRIPT [158] Kaklamani V, Yi N, Zhang K, Sadim M, Offit K, Oddoux C, et al. Polymorphisms of ADIPOQ and ADIPOR1 and prostate cancer risk. Metabolism 2011;60:1234-43.

T

[159] Petridou ET, Sergentanis TN, Antonopoulos CN, Dessypris N, Matsoukis IL, Aronis K,

RI P

et al. Insulin resistance: an independent risk factor for lung cancer? Metabolism 2011;60:1100-6.

SC

[160] Dalamaga M, Crotty BH, Fargnoli J, Papadavid E, Lekka A, Triantafilli M, et al. B-cell chronic lymphocytic leukemia risk in association with serum leptin and adiponectin: a case-

MA NU

control study in Greece. Cancer Causes Control 2010;21:1451-9. [161] Petridou ET, Sergentanis TN, Dessypris N, Vlachantoni IT, Tseleni-Balafouta S, Pourtsidis A, et al. Serum adiponectin as a predictor of childhood non-Hodgkin's lymphoma: a nationwide case-control study. J Clin Oncol 2009;27:5049-55.

ED

[162] Dalamaga M, Migdalis I, Fargnoli JL, Papadavid E, Bloom E, Mitsiades N, et al.

PT

Pancreatic cancer expresses adiponectin receptors and is associated with hypoleptinemia and hyperadiponectinemia: a case-control study. Cancer Causes Control 2009;20:625-33.

CE

[163] Dalamaga M, Karmaniolas K, Panagiotou A, Hsi A, Chamberland J, Dimas C, et al.

AC

Low circulating adiponectin and resistin, but not leptin, levels are associated with multiple myeloma risk: a case-control study. Cancer Causes Control 2009;20:193-9. [164] Sher DJ, Oh WK, Jacobus S, Regan MM, Lee GS, Mantzoros C. Relationship between serum adiponectin and prostate cancer grade. Prostate 2008;68:1592-8. [165] Kaklamani VG, Sadim M, Hsi A, Offit K, Oddoux C, Ostrer H, et al. Variants of the adiponectin and adiponectin receptor 1 genes and breast cancer risk. Cancer Res 2008;68:3178-84. [166] Petridou ET, Mitsiades N, Gialamas S, Angelopoulos M, Skalkidou A, Dessypris N, et al. Circulating adiponectin levels and expression of adiponectin receptors in relation to lung cancer: two case-control studies. Oncology 2007;73:261-9.

44

ACCEPTED MANUSCRIPT [167] Dalamaga M, Nikolaidou A, Karmaniolas K, Hsi A, Chamberland J, DionyssiouAsteriou A, et al. Circulating adiponectin and leptin in relation to myelodysplastic syndrome:

T

a case-control study. Oncology 2007;73:26-32.

RI P

[168] Williams CJ, Mitsiades N, Sozopoulos E, Hsi A, Wolk A, Nifli AP, et al. Adiponectin receptor expression is elevated in colorectal carcinomas but not in gastrointestinal stromal

SC

tumors. Endocr Relat Cancer 2008;15:289-99.

[169] Barb D, Williams CJ, Neuwirth AK, Mantzoros CS. Adiponectin in relation to

MA NU

malignancies: a review of existing basic research and clinical evidence. Am J Clin Nutr 2007;86:s858-66.

[170] Barb D, Neuwirth A, Mantzoros CS, Balk SP. Adiponectin signals in prostate cancer

Cancer 2007;14:995-1005.

ED

cells through Akt to activate the mammalian target of rapamycin pathway. Endocr Relat

PT

[171] Tworoger SS, Eliassen AH, Kelesidis T, Colditz GA, Willett WC, Mantzoros CS, et al.

2007;92:1510-6.

CE

Plasma adiponectin concentrations and risk of incident breast cancer. J Clin Endocrinol Metab

AC

[172] Korner A, Pazaitou-Panayiotou K, Kelesidis T, Kelesidis I, Williams CJ, Kaprara A, et al. Total and high-molecular-weight adiponectin in breast cancer: in vitro and in vivo studies. J Clin Endocrinol Metab 2007;92:1041-8. [173] Barb D, Pazaitou-Panayiotou K, Mantzoros CS. Adiponectin: a link between obesity and cancer. Expert Opin Investig Drugs 2006;15:917-31. [174] Kelesidis I, Kelesidis T, Mantzoros CS. Adiponectin and cancer: a systematic review. Br J Cancer 2006;94:1221-5. [175] Petridou E, Mantzoros CS, Dessypris N, Dikalioti SK, Trichopoulos D. Adiponectin in relation to childhood myeloblastic leukaemia. Br J Cancer 2006;94:156-60.

45

ACCEPTED MANUSCRIPT [176] Wei EK, Giovannucci E, Fuchs CS, Willett WC, Mantzoros CS. Low plasma adiponectin levels and risk of colorectal cancer in men: a prospective study. J Natl Cancer Inst

T

2005;97:1688-94.

RI P

[177] Dal Maso L, Augustin LS, Karalis A, Talamini R, Franceschi S, Trichopoulos D, et al. Circulating adiponectin and endometrial cancer risk. J Clin Endocrinol Metab 2004;89:1160-

SC

3.

[178] Mantzoros C, Petridou E, Dessypris N, Chavelas C, Dalamaga M, Alexe DM, et al.

MA NU

Adiponectin and breast cancer risk. J Clin Endocrinol Metab 2004;89:1102-7. [179] Gale SM, Castracane VD, Mantzoros CS. Energy homeostasis, obesity and eating disorders: recent advances in endocrinology. Journal Nutr 2004;134:295-8. [180] Petridou E, Mantzoros C, Dessypris N, Koukoulomatis P, Addy C, Voulgaris Z, et al.

ED

Plasma adiponectin concentrations in relation to endometrial cancer: a case-control study in

PT

Greece. J Clin Endocrinol Metab 2003;88:993-7. [181] Kharitonenkov A, Shiyanova TL, Koester A, Ford AM, Micanovic R, Galbreath EJ, et

CE

al. FGF-21 as a novel metabolic regulator. J Clin Invest 2005;115:1627-35.

AC

[182] Kharitonenkov A, Adams AC. Inventing new medicines: The FGF21 story. Mol Metab 2014;3:221-9.

[183] Cheng X, Zhu B, Jiang F, Fan H. Serum FGF-21 levels in type 2 diabetic patients. Endocr Res 2011;36:142-8. [184] Lamers D, Famulla S, Wronkowitz N, Hartwig S, Lehr S, Ouwens DM, et al. Dipeptidyl peptidase 4 is a novel adipokine potentially linking obesity to the metabolic syndrome. Diabetes 2011;60:1917-25. [185] Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM. Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. J Clin Invest 1995;95:2409-15.

46

ACCEPTED MANUSCRIPT [186] Sopasakis VR, Nagaev I, Smith U. Cytokine release from adipose tissue of nonobese individuals. Int J Obes (Lond) 2005;29:1144-7.

T

[187] Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis

RI P

factor-alpha: direct role in obesity-linked insulin resistance. Science 1993;259:87-91. [188] Uysal KT, Wiesbrock SM, Marino MW, Hotamisligil GS. Protection from obesity-

SC

induced insulin resistance in mice lacking TNF-alpha function. Nature 1997;389:610-4. [189] Li Z, Yang S, Lin H, Huang J, Watkins PA, Moser AB, et al. Probiotics and antibodies

MA NU

to TNF inhibit inflammatory activity and improve nonalcoholic fatty liver disease. Hepatology 2003;37:343-50.

[190] Liang H, Yin B, Zhang H, Zhang S, Zeng Q, Wang J, et al. Blockade of tumor necrosis factor (TNF) receptor type 1-mediated TNF-alpha signaling protected Wistar rats from diet-

ED

induced obesity and insulin resistance. Endocrinology 2008;149:2943-51.

PT

[191] Lopez-Soriano J, Lopez-Soriano FJ, Bagby GJ, Williamson DH, Argiles JM. Anti-TNF treatment does not reverse the abnormalities in lipid metabolism of the obese Zucker rat. Am

CE

J Phys 1997;272:E656-60.

AC

[192] Wascher TC, Lindeman JH, Sourij H, Kooistra T, Pacini G, Roden M. Chronic TNFalpha neutralization does not improve insulin resistance or endothelial function in "healthy" men with metabolic syndrome. Mol Med 2011;17:189-93. [193] Schulz TJ, Huang P, Huang TL, Xue R, McDougall LE, Townsend KL, et al. Brown-fat paucity due to impaired BMP signalling induces compensatory browning of white fat. Nature 2013;495:379-83. [194] Schulz TJ, Tseng YH. Emerging role of bone morphogenetic proteins in adipogenesis and energy metabolism. Cytokine Growth Factor Rev 2009;20:523-31. [195] Tseng YH, Kokkotou E, Schulz TJ, Huang TL, Winnay JN, Taniguchi CM, et al. New role of bone morphogenetic protein 7 in brown adipogenesis and energy expenditure. Nature 2008;454:1000-4. 47

ACCEPTED MANUSCRIPT [196] Townsend KL, Suzuki R, Huang TL, Jing E, Schulz TJ, Lee K, et al. Bone morphogenetic protein 7 (BMP7) reverses obesity and regulates appetite through a central

T

mTOR pathway. FASEB J 2012;26:2187-96.

RI P

[197] Böttcher Y, Unbehauen H, Klöting N, Ruschke K, Körner A, Schleinitz D, et al. Adipose tissue expression and genetic variants of the bone morphogenetic protein receptor 1A

SC

gene (BMPR1A) are associated with human obesity. Diabetes 2009;58:2119-28. [198] Schleinitz D, Klöting N, Böttcher Y, Wolf S, Dietrich K, Tönjes A, et al. Genetic and

MA NU

evolutionary analyses of the human bone morphogenetic protein receptor 2 (BMPR2) in the pathophysiology of obesity. PLoS One 2011;6:e16155.

[199] Dalamaga M, Chou SH, Shields K, Papageorgiou P, Polyzos SA, Mantzoros CS. Leptin at the intersection of neuroendocrinology and metabolism: current evidence and therapeutic

AC

CE

PT

ED

perspectives. Cell Metab 2013;18:29-42.

48

AC

CE

PT

ED

MA NU

SC

RI P

T

ACCEPTED MANUSCRIPT

49

AC

CE

PT

ED

MA NU

SC

RI P

T

ACCEPTED MANUSCRIPT

50

From leptin to other adipokines in health and disease: facts and expectations at the beginning of the 21st century.

This year marks the 20th anniversary of the discovery of leptin, which has tremendously stimulated translational obesity research. The discovery of le...
475KB Sizes 0 Downloads 5 Views