8,331
Views
383
CrossRef citations to date
0
Altmetric
Review

Pathogenic potential of adipose tissue and metabolic consequences of adipocyte hypertrophy and increased visceral adiposity

, , , , , , & show all
Pages 343-368 | Published online: 10 Jan 2014

Abstract

When caloric intake exceeds caloric expenditure, the positive caloric balance and storage of energy in adipose tissue often causes adipocyte hypertrophy and visceral adipose tissue accumulation. These pathogenic anatomic abnormalities may incite metabolic and immune responses that promote Type 2 diabetes mellitus, hypertension and dyslipidemia. These are the most common metabolic diseases managed by clinicians and are all major cardiovascular disease risk factors. ‘Disease’ is traditionally characterized as anatomic and physiologic abnormalities of an organ or organ system that contributes to adverse health consequences. Using this definition, pathogenic adipose tissue is no less a disease than diseases of other body organs. This review describes the consequences of pathogenic fat cell hypertrophy and visceral adiposity, emphasizing the mechanistic contributions of genetic and environmental predispositions, adipogenesis, fat storage, free fatty acid metabolism, adipocyte factors and inflammation. Appreciating the full pathogenic potential of adipose tissue requires an integrated perspective, recognizing the importance of ‘cross-talk’ and interactions between adipose tissue and other body systems. Thus, the adverse metabolic consequences that accompany fat cell hypertrophy and visceral adiposity are best viewed as a pathologic partnership between the pathogenic potential adipose tissue and the inherited or acquired limitations and/or impairments of other body organs. A better understanding of the physiological and pathological interplay of pathogenic adipose tissue with other organs and organ systems may assist in developing better strategies in treating metabolic disease and reducing cardiovascular disease risk.

Adipose tissue may be pathogenic through the adverse consequences of excessive fat mass alone, and/or through deleterious endocrinologic and immunologic activity. Adipocyte hypertrophy and visceral adipose tissue accumulation are associated with many of the most common metabolic diseases found in clinical practice, including Type 2 diabetes mellitus (T2DM), hypertension, dyslipidemia and, possibly, atherosclerosis Citation[1]. For the past 20–30 years, in the USA alone, the rate of overweight or obesity has increased as follows Citation[401]:

Adults: increased from 15 to 33%

Children (2–5 years): increased from 5 to 14%

Children (6–11 years): increased from 7 to 19%

Adolescents (12–19 years): increased from 5 to 17%

Since obesity and its metabolic consequences are now epidemics within many developed nations Citation[2,3,402,403], it is important to understand the pathogenic potential of adipose tissue.

The role of adipose tissue in human health is best considered within the context of its physio-logic benefits versus its potential pathogenic contributions to ill health. A sole focus on fat mass is often inadequate in assessing the associated health risks of adipose tissue since being overweight alone is not necessarily detrimental. Modestly or moderately overweight patients may actually have decreased mortality from noncancerous, noncardiovascular disease (CVD) causes and no increased mortality due to cancer or CVD disease Citation[4]. In addition, while obesity is associated with significantly increased CVD and obesity-related cancer mortality, and while combined overweight and obesity is associated with increased mortality from T2DM and kidney disease, comparisons across surveys suggest a decrease in the association of obesity with CVD mortality over time Citation[4]. Overall, this suggests that excessive body fat is more closely associated with increased CVD risk when adipose tissue is pathogenic. Pathogenic adipose tissue may lead to major atherosclerotic coronary heart disease (CHD) risk factors, such as T2DM, hypertension and dyslipidemia, as well as to other more direct adverse effects upon the vasculature and heart Citation[1].

Current obesity guidelines are often based upon body mass index (BMI) and waist circumference, with differing therapeutic cut-off points dependent upon the presence of comorbidities Citation[404]. However, given that the increase in metabolic disease with increasing BMI is both continuous and gradual Citation[5,6], specific cut-off points may not apply to individual patients, particularly when potential ethnic and gender variances are taken into consideration Citation[7]. Another challenge is that despite its known medical, monetary and human costs Citation[8], obesity (which includes many patients with pathogenic adipose tissue) has not yet been universally recognized as a disease Citation[405]. ‘Disease’ can be defined as an impairment of body function or system, often accompanied by pathological alterations in tissues or cells, resulting in adverse clinical outcomes Citation[9]. If adipocyte hypertrophy and visceral adipose tissue accumulation occur during positive caloric balance, then the pathogenic consequences may unfavorably affect other body organs, such as liver, muscle and pancreas, resulting in adverse clinical outcomes Citation[6]. It is, therefore, through the understanding of the pathogenic potential of hypertrophied adipocytes and increased accumulation of visceral adipose tissue that helps support how an increase in body fat, in many individuals, is itself a disease. It also provides a framework for explaining why treating pathogenic adipose tissue is more rational than treating BMI alone Citation[7].

Genetic & environmental considerations

The pathogenic potential of adipose tissue is dependent upon genetic predisposition and environmental surroundings. Many Native American groups, including the Pima Indians, are genetically predisposed to insulin resistance, obesity and the development of T2DM; obesity markedly increases the risk of T2DM Citation[10]. Interestingly, the risk of CHD may not be increased Citation[11,12]. Anatomically, the prevalence of T2DM in Pima Indians is increased in the presence of hypertrophic, bloated, pathogenic adipocytes, as opposed to smaller, leaner and more functional fat cells Citation[13]. In fact, it is the presence of anatomically larger adipocytes that best predicts the onset of T2DM among Pima Indians, compared with the presence of obesity alone Citation[14]. Additionally, the prevalence of T2DM in Mexican Pima Indians is no different than other, non-Pima Mexicans. But with the excessive body fat found in US Pima Indians, this better reveals their genetic predisposition towards developing metabolic disease. Thus, US Pima Indians, who are substantially more overweight than their leaner Mexican counterparts Citation[10], have an approximately five-times greater prevalence of T2DM.

Asians are another population illustrating the importance of genetics in predisposing patients to the adverse clinical consequences of pathogenic adipose tissue Citation[15–19]. Asians have a high prevalence of T2DM, the metabolic syndrome and CHD. Particularly well described are individuals from the South Asian subcontinent who have increased adipocyte size Citation[20], increased visceral adipose tissue Citation[16–18], increased circulating free fatty acids Citation[15], increased leptin levels Citation[15,17], increased pro-inflammatory factors (e.g., increased Creactive protein levels) Citation[19], decreased anti-inflammatory factors (e.g., adiponectin) Citation[15,17], increased insulin resistance Citation[15] and increased CHD risk Citation[21]. Anatomically, Asians have the typical findings of pathogenic adipose tissue, which includes an increase in the relative amount of visceral fat, and a lower number of adipocytes Citation[20]. The reduced number of adipocytes may be due to a limited ability to undergo adipogenesis, which is a process that involves the recruitment and proliferation of more functional adipocytes Citation[22]. If adipogenesis is impaired during positive caloric balance, then existing adipocytes must undergo hypertrophy to store excessive energy. If adipocyte hypertrophy results in metabolic dysfunction, then this may help account for the increase in metabolic disease in Asians compared with Caucasians at the same level of BMI Citation[20]. This, again, supports the theory that within populations (and individuals) who are genetically predisposed, it is pathogenic adipose tissue that often ‘triggers’ the expression of metabolic disease. In fact, it is because the pathogenic potential of adipocyte and adipose tissue varies among those with differing genetic predispositions that international organizations have proposed that Asians should have different cut-off points for the determination of the terms ‘overweight’ and ‘obesity’ Citation[23].

Acquired or environmental factors may also affect the pathogenic potential of adipose tissue. Hypercortisolemia, such as occurs with Cushing’s syndrome or exogenous corticosteroid use, is an example of a pathologic environment whose effects upon multiple body organs contribute to T2DM and other findings associated with the metabolic syndrome (increased waist circumference, hypertension and dyslipidemia). Glucocorticoids are normally anabolic in the liver, causing increased gluconeogenesis and increased glucose release. Hypercortisolism may also increase the appetite of patients, often resulting in positive caloric balance. Conversely, glucocorticoids are catabolic to muscle, causing muscle wasting and insulin resistance, and catabolic to adipose tissue, where lipolysis is modestly increased Citation[1,24]. In peripheral, subcutaneous adipose tissue, exogenous corticosteroids may promote smaller adipocyte size, presumably due to catabolism and modest lipolytic activity Citation[25]. However, visceral adipose tissue may undergo relative and absolute accumulation, due to a fourfold increase in glucocorticoid receptors in visceral adipose tissue when compared with peripheral, subcutaneous adipose tissue Citation[26]. An increase in visceral adipocyte hypertrophy may be attributable to a glucocorticoid-induced increase in appetite and a glucocorticoidinduced increase in adipocyte differentiation and decrease in adipocyte proliferation – both of which promote adipocyte hypertrophy Citation[27]. Additionally, the hyperinsulinemia that accompanies hypercortisol-induced insulin resistance may overwhelm the relatively minor lipolytic effects and promote an increase in triglyceride storage in visceral fat cells, a process termed lipogenesis. Overall, abdominal fat cells typically become enlarged and visceral adiposity is increased Citation[25]. Finally, patients with hypercortisolism may have increased adipose tissue inflammatory responses, relative to those with less visceral adipose tissue accumulation Citation[28]. Thus, hypercortisolemia is an example of an acquired environment that helps generate pathogenic adipose tissue. When coupled with a glucocorticoid-induced increase in gluconeogenesis from the liver and a glucocorticoid-induced increase in insulin resistance in the muscle, all of this contributes to the hyperglycemia so often found with hypercortisolemia.

Adipogenesis

Typically, the cellular content of adipose tissue is approximately 50% adipocytes, with the remaining 50% being the stromal vasculature fraction of fibroblasts, endothelial cells, macrophages and preadipocytes. During positive caloric balance, increased storage of energy optimally occurs through the generation of added, functional fat cells, achieved through adipogenesis from preadipocytes Citation[29,30]. Patients with lipodystrophy have a variable lack of adipose tissue that results in impaired adipose tissue function, manifested by low adiponectin levels and high circulating free fatty acids. Without adequate adipose tissue, storage of free fatty acids is inadequate and increased circulating free fatty acids often results in ‘lipotoxicity’ Citation[31]. Lipotoxicity is characterized by ectopic fat deposition in muscle and liver (contributing to insulin resistance) and deposition in the pancreas (promoting insulinopenia), all potentially leading to T2DM Citation[7,32]. Lipoatrophic mice have virtually no white adipose tissue and express a severe form of lipoatrophic T2DM. Surgical transplantation of functional adipose tissue in lipoatrophic mice reverses hyperglycemia, dramatically lowers insulin levels and improves muscle insulin sensitivity Citation[33].

But just as too little adipose tissue can result in metabolic diseases Citation[34], so can too much adipose tissue. This especially occurs when excessive body fat results in adipocyte hypertrophy, sometimes described as ‘acquired lipodystrophy’Citation[35,36]. During positive caloric balance, adipocytes initially undergo hypertrophy, which normally triggers adipose tissue paracrine adipogenic signaling for the purpose of adding functional fat cells and towards maintaining adipose tissue physiologic functions during increased energy storage Citation[37–39]. In the past, it has been suggested that the number of human adipocytes were fixed early in life and that a ‘fixed adipocyte-number’ pre-destined individuals to be lean or obese. However, this is no longer thought to be true Citation[40]. Not only does adipocyte hyper-trophy occur in humans Citation[1,35,37,39,41–45], but the recruitment and proliferation of preadipocytes is also thought to occur in adult humans Citation[1,35,39,41–43,46]. Adipogenesis is, therefore, an important physiologic process whose function or dysfunction may prevent or promote metabolic disease Citation[1,47,48].

During persistent positive caloric balance, if adipogenesis is impaired after initial adipocyte hypertrophy, then further adipocyte hypertrophy may result in adipocyte dysfunction Citation[47,49]. Some have even suggested that an increase in fat cell size might be viewed as a failure of adipocytes to adequately proliferate Citation[35,50–52]. This may have pathological consequences. It has been known at least since the 1970s that during times of positive caloric balance, excessive fat cell enlargement results in adipocyte metabolic and immune abnormalities Citation[53,54]. Animal studies have shown that a decrease in the expression of adipogenic genes is associated with metabolic diseases, such as T2DM Citation[51]. Similarly, human studies have shown that in obese and T2DM patients, the proliferation and differentiation of adipocytes are decreased, as reflected by the decreased expression of adipogenic genes Citation[42]. In summary, during times of positive caloric balance, if energy is stored predominantly through lipogenesis and fat cell hypertrophy of existing adipocytes, as opposed to adipogenesis with recruitment and differentiation of new fat cells and fat cell hyperplasia, then this may lead to pathologic adipose tissue responses that contribute to metabolic disease Citation[1,14,35,36,42–45,49,51–60].

However, it is not the hypertrophy of individual fat cells alone that has potential adverse clinical consequences. Excessive expansion of the adipose tissue organ itself may also contribute to pathogenic processes. In order for adipose tissue growth to occur with maintenance of normal adipose tissue function, it must do so with an orderly and appropriate production of modulating and transcriptional factors Citation[30,43,46,50,61], dissolution and reformation of the adipose tissue extracellular matrix Citation[30,62], and angiogenesis Citation[30,63]. Extracellular matrix (ECM) remodeling and angiogenesis are biological processes that are intimately linked Citation[63], and disruption of either process may result in impaired adipose tissue function. For example, during positive caloric balance, an impairment of ECM formation limits adipose tissue growth, limits fat storage within adipose tissue and increases the potential for ectopic fat storage, which may contribute to metabolic diseases Citation[31]. Furthermore, while adipo-genesis may trigger ECM formation, adipocyte ECM factors may likewise affect adipogenesis Citation[64]. If the appropriate and orderly formation of ECM is impaired, then this may also impair adipogenesis and thus contribute to adverse clinical consequences of pathogenic adipose tissue Citation[35,52]. Similarly, an increase in adipose tissue growth also requires additional blood supply. If angiogenesis is impaired, then relative hypoxia may impair adipogenesis Citation[65], which exacerbates adipocyte metabolic dysfunction, and may promote a net proinflammatory response Citation[66–68], all contributing to metabolic disease.

Thus, adipogenesis is a process that helps explain the seemingly paradoxical finding wherein not all overweight patients have metabolic disease and not all patients with metabolic disease are significantly overweight Citation[1,2,5,69,70]. During positive caloric balance, the development of metabolic disease is more closely related to how the fat is stored (through adipocyte hypertrophy versus hyperplasia) than simply the amount of fat that is stored Citation[1,44].

The determination as to whether adipocytes respond to positive caloric balance with hypertrophy versus hyperplasia is based upon genetic predisposition and the actions of multiple regulatory factors (Box 1)Citation[30]. Some adipocyte and nonadipocyte factors may have different effects within the adipogenic process, such as differing effects upon proliferation (creation of new adipocytes from preadipocytes) versus differentiation (increased maturity and lipogenesis within existing adipocytes). Angiotensin II impairs proliferation in humans (although reports are inconsistent in rodents Citation[71]) and promotes differentiation Citation[72–77]. Angiotensinogen impairs proliferation in humans (although reports inconsistent in rodents Citation[71]) and promotes differentiation Citation[72–75,77]. Autotaxin/lysophosphatidic acid promotes proliferation and impairs differentiation Citation[78,79]. Catecholamines promote proliferation and impair differentiation Citation[80]. Glucocorticoids impair proliferation and promote differentiation Citation[27,81]. Finally, epidermal growth factor impairs proliferation and promotes differentiation Citation[82].

This has practical, clinical implications in that glucocorticoids increase the differentiation of existing adipocytes (especially visceral adipocytes) relative to subcutaneous, peripheral adipocytes, while decreasing adipocyte proliferation. The resulting hypertrophy of visceral adipocytes, coupled with a decrease in the recruitment of functional subcutaneous, peripheral adipocytes, is a contributing cause of the T2DM, hypertension and dyslipidemia often found with hypercortisolemia Citation[1,26,27]. Angiotensinogen/angiotensin II may unfavorably decrease adipogenesis. Conversely, enhanced adipogenesis is one of the proposed reasons why angiotensin-converting enzyme (ACE) inhibitors and angiotensin II blockers may improve metabolic disease, including T2DM Citation[72]. Additionally, ACE inhibitors and angiotensin II receptor blockers (ARBs) increase peroxisome proliferator-activated receptor (PPAR)-γ activity (which promotes adipogenesis), and have other effects that may involve adipose tissue. ACE inhibitors and ARBs may also increase adiponectin levels, increase translocation of glucose transporters, upregulate tyrosine phosphorylation of insulin receptor substrate-1 and enhance bradykinin and nitric oxide activities Citation[7,73,83–88]. Finally, weight gain is often associated with an increase in catecholamine activity, as may be mediated through increased circulatory and CNS leptin levels Citation[89]. An increase in catecholamines would be expected to increase adipocyte proliferation and lipolysis, and thus potentially attenuate progression to metabolic disease. However, a leptinmediated increase in catecholamines levels may also increase blood pressure. With this exception aside, the favorable influence of catecholamines on lipolysis and adipogenesis helps explain why impairment of these favorable metabolic activities, such as might occur through the dysfunction of the sympathetic nervous system, may contribute to obesity and T2DM Citation[90].

Fat storage

The pathogenesis of metabolic diseases is significantly influenced by not only how the fat is stored (hypertrophy versus hyperplasia), but also where the fat is stored Citation[91]. Patient populations described as metabolically healthy (no metabolic disease), but obese, often have less visceral adipose tissue distribution than obese patients with metabolic disease Citation[92,93]. Conversely, patients who are metabolically obese (those with metabolic disease), but normal weight, often have more visceral adipose tissue than individuals of similar weight and no metabolic disease Citation[92]. Such clinical findings are explained by the different intrinsic activities of fat depots Citation[74,94–100]. Visceral adipose tissue accumulation is the fat depot most characterized as being associated with an increased risk of metabolic disease Citation[97,99,101–109]. This is whether the visceral adipose tissue accumulation occurs by hypertrophy or hyperplasia Citation[1,97,101–108,110,111]. Conversely, if subcutaneous, peripheral adipose tissue undergoes hyperplasia with a generation of smaller and more functional adipocytes, then this added functionality may attenuate or reduce the risk of metabolic disease Citation[1,35,36,43,51,52,56,58,112].

Location is one of the more important reasons why different fat depots have different pathogenic potential. Visceral adipose tissue, which represents approximately 20% of total body fat, secretes various adipocyte factors (such as free fatty acids) into the portal vein, which supplies 80% of the hepatic blood supply Citation[107]. Also, visceral adipose tissue is genetically predetermined Citation[113] to have different functions than subcutaneous, peripheral adipose tissue (which represents approximately 80% of total body fat). For example, these two fat depots differ in the production of bioactive molecules, the activity of various receptors and the enzymatic processes involved with fat metabolism Citation[47,114–116]. An important clinical implications of these differences is that when corrected for the same age, men are at higher CHD risk than women. Comparatively, men often store more fat in the visceral region, representing a so called ‘android’ adipose tissue distribution Citation[117,118]. An increase in visceral adiposity promotes metabolic diseases that are important CVD and CHD risk factors (T2DM, hypertension and dyslipidemia). Conversely, women often have increased adipose tissue accumulation and increased adipocyte size within the peripheral, subcutaneous region, representing a so called ‘gynoid’ adipose tissue distribution Citation[119–122]. These gender differences in adipose tissue distribution, described since the 1940s Citation[123], can be at least partially explained by the influences of sex hormones Citation[1,47,74]. Analogous to the effects glucocorticoids may have on visceral adipose tissue distribution Citation[1,26,74,124], sex hormones can also affect adipose tissue distribution. Androgens are associated with increased visceral adipose tissue distribution, while estrogens are associated with increased peripheral, subcutaneous adipose tissue distribution Citation[1,26,74,125,126].

Adipose tissue depots not only differ in their metabolic activity, but also in the degree of their metabolic activity. Visceral adipose tissue, such as intraperitoneal (omental, mesenteric and umbilical), extraperitoneal (peripancreatic and perirenal), and intrapelvic (gonadal/epidydimal and urogenital) adipose tissue have a higher degree of metabolic activity compared with sub-cutaneous, peripheral adipose tissue (truncal, gluteofemoral, mammary and inguinal) Citation[47,101,127]. Some have suggested that subcutaneous adipose tissue in the abdominal region has metabolic activity, such as lipolysis and the release of inflammatory factors, between that of visceral and subcutaneous, peripheral adipose tissue Citation[101,111,128–130]. Accordingly, subcutaneous, abdominal adipose tissue may contribute to worsening of CHD risk factors Citation[131].

Similarly, periorgan adipose tissue (pericardial, perimuscular, perivascular, orbital and paraosseal) may also have pathogenic potential through metabolic activities including lipolysis and the release of inflammatory factors, with an intrinsic activity between that of visceral adipose tissue and subcutaneous, peripheral adipose tissue Citation[47,128,133]. Pericardial and perivascular adipose tissue accumulation may directly promote CHD and peripheral vascular disease Citation[133–137]. It has traditionally been assumed that atherosclerosis is exclusively the result of pathologic interactions of intralumenal processes within arterial subendothelia. However, pathogenic pericardial and perivascular adipose tissue may directly contribute to atherosclerosis through an ‘outside to inside’ (from the outside of the vessel to the endothelium) vascular atherogenic model Citation[1,133–135].

Although subcutaneous adipose tissue is sometimes thought of as ‘protective’, even excessive subcutaneous adipose tissue may become pathogenic Citation[138,139]. If during positive caloric balance, the recruitment and proliferation of smaller and more functional subcutaneous adipocytes occurs, then the risk of developing metabolic disease may be decreased. This may be characterized as ‘protective’ through providing additional adipose tissue functionality, including improved energy (free fatty acid) storage capacity. However, if adipogenesis is impaired and subcutaneous adipocytes become sufficiently enlarged to become dysfunctional and pathogenic, then this may contribute to metabolic diseases, such as T2DM Citation[14].

One of the more notable ways in which subcutaneous adipose tissue may be pathogenic involves the production of leptin . In obesity due to leptin deficiency, administration of leptin dramatically reduces body fat, and corrects almost all of the associated metabolic abnormalities Citation[140]. However, in humans without leptin deficiency, leptin functions mainly to signal energy adequacy, and is best viewed as a hormone whose reduced levels (such as through negative caloric balance) promote increased feeding and decreased energy expenditure Citation[1,47,141]. In the absence of leptin deficiency, the weight loss effects of increasing leptin levels appears to be near maximal at physiologic levels Citation[142]. This may be, at least in part, because the secretion of leptin by enlarging adipocytes may be simultaneously accompanied by adipose tissue physiological responses that block leptin activity (a proposed form of ‘leptin resistance’) Citation[143]. Nonetheless, while further increases in leptin levels may potentially reduce abnormalities of glucose and lipid metabolism Citation[1], hyperleptinemia may increase blood pressure Citation[1,89,144–149]. Subcutaneous adipose tissue is the major source of circulating leptin because subcutaneous adipose tissue is quantitatively the largest fat depot; subcutaneous adipocytes are larger than visceral or omental adipocytes; and leptin gene expression may be increased within this fat depot Citation[150]. Biopsy studies of femoral subcutaneous adipose tissue have shown that hyperleptinemia is more closely associated with adipose cell hypertrophy than with adipose tissue hyperplasia Citation[151]. Thus, to the extent that hyperleptinemia contributes to hypertension and to the degree that hyperleptinemia is more related to hypertrophied subcutaneous adipose tissue than visceral adipose tissue, then hypertrophy of subcutaneous adipocytes may lead to hyperleptinemia-induced hypertension. This would be supported by the findings that hypertension is directly and continuously related to BMI, Citation[5] and that arterial compliance decreases and blood pressure increases in overweight and obese individuals compared with normal weight individuals, even when their hip circumference and sum of skin folds (measures of subcutaneous adipose tissue) are increased Citation[152]. However, when adjusted for body surface area, hip circumference and sum of skin folds may be inversely related to arterial compliance, even while waist circumference may or may not have a significant correlation. This suggests that while both subcutaneous and visceral adipose tissue may contribute to hypertension, particularly when accompanied by adipocyte hypertrophy, a relative increase in visceral adipose tissue often has a greater blood pressure-raising effect. This may possibly be due to the deleterious effects of visceral adipose tissue upon various adipocyte factors , such as IL-6, C-reactive protein and TNF-α, directly leading to endothelial dysfunction Citation[152]. Obesity-induced insulin resistance may also impair the release of otherwise vasodilatory, endothelia nitric oxide, which is another mechanism that might increase blood pressure Citation[152].

Finally, while visceral adipose tissue is the major contributor to portal free fatty acids, subcutaneous adipose tissue accounts for the majority of systemic circulating free fatty acids. Specifically, the majority of postabsorptive systemic free fatty acids, which may most adversely affect muscle, pancreas and vasculature Citation[153], are derived from upper body subcutaneous adipose tissue, with only approximately 15% being derived from visceral adipose tissue Citation[153,154]. Thus, while mostly described with central adiposity, the lipolytic activity of both visceral and subcutaneous adipose tissue have the potential to be pathogenic.

It has also been proposed that the pathogenic effects of visceral adipose tissue may contribute to the pathogenic responses of both abdominal and gluteal subcutaneous adipose tissue. Insulin inhibits lipolysis through trapping circulating free fatty acid within adipocytes Citation[1]. Impaired insulin activity from visceral adipose tissue-induced insulin resistance Citation[154] might increase adipocyte lipolysis, increase circulating free fatty acids, and further worsen insulin resistance. Thus, an increase in visceral adiposity may cause or exacerbate the pathogenic potential of subcutaneous adipose tissue.

Recognizing their physiologic and potential pathologic importance, various fat depots are frequently measured in clinical trials and sometimes assessed clinically Citation[155]. From a research standpoint, while ultrasound is sometimes used for evaluation of intra-abdominal adipose tissue, computed tomography (CT) and magnetic resonance imaging (MRI) are the imaging procedures most widely used to assess visceral fat depots Citation[156], with CT often considered to be the gold-standard imaging technique for assessing various fat depots Citation[132,136,157–159]. MRI may also provide good imaging for visceral adipose tissue and intramuscular fat, but perhaps less so for subcutaneous or inter-muscular adipose tissue Citation[132,160]. From a clinician standpoint, measurement of height, weight and waist circumference may be diagnostically helpful. An increase in abdominal girth may be more specifically associated with an increased amount of visceral fat and, thus, more predictive of an increased risk of metabolic disease when compared with BMI alone Citation[18,106,166–163]. Alternatively, since an increase in BMI is generally associated with an increased risk of metabolic disease Citation[5,6], some have suggested that BMI performs at least as well as waist circumference in identifying the potential for insulin sensitivity abnormalities and CHD risk factors Citation[162].

Free fatty acid metabolism

Adipose tissue is the major energy storage organ of the body. Approximately 80% of adipose tissue weight is lipid, and over 90% of lipids are stored triglycerides Citation[155]. The major secretory product from adipose tissue is free fatty acids Citation[29], which are derived from the lipolysis of stored triglycerides, and regulated by adipocyte and nonadipocyte factors (Box 2). If intra-cellular adipocyte hydrolysis of triglycerides (lipolysis) exceeds intracellular adipocyte esterification of free fatty acids (lipogenesis), then free fatty acids undergo a net release into the circulation. A sustained, excessive net increase in circulating free fatty acids contributes to metabolic disease Citation[31]. Chronic increases in circulating free fatty acids worsen glucose metabolism due to ‘lipotoxic’ effects upon muscle and liver (contributing to insulin resistance) Citation[153] and pancreas (contributing to insulinopenia) Citation[31,97,164,165]. This is an important reason why David Savage suggests Citation[166]:

‘The last decade has seen a shift from the traditional ‘glucocentric’ view of diabetes to an increasingly acknowledged ‘lipocentric’ viewpoint.’

Increases in circulating free fatty acids are also an independent risk factor for hypertension, possibly due to free fatty acid-induced insulin resistance, which itself contributes to high blood pressure Citation[167,168]; impairment of endothelium-dependent vasodilation Citation[167,168]; and other microvascular dysfunctions leading to hypertension Citation[167,168]. Finally, increases in circulating free fatty acids contribute to the typical dyslipidemia found with the ‘metabolic syndrome,’ which includes hypertriglyceridemia, reduced high-density lipoprotein-cholesterol levels, and abnormalities of lipoprotein particle size and subclass distribution, such as an increased proportion of small, dense low-density lipoprotein particles Citation[1,169,170].

Adipocyte factors

While its function has sometimes been characterized as little more than storing energy, adipose tissue is clearly an active endocrine organ (Box 3)Citation[48,74,171–173]. Adipocytes and adipose tissue are actively involved in metabolic processes such as angio-genesis, adipogenesis, ECM dissolution and reformation, lipo-genesis, growth factor production, glucose metabolism, production of factors associated with the renin–angiotensin system, lipid metabolism, enzyme production, hormone production, steroid metabolism, immune response, hemostasis and element binding (Box 3). The disruption of these adipose tissue processes, as occurs with adipocyte hypertrophy and visceral adipose tissue accumulation, results in adipocyte factor abnormalities that are not only associated with, but may be important contributors to metabolic diseases Citation[1,47,169].

Inflammation

Adipose tissue is not only an active endocrine organ, but it is also an active immune organ Citation[174–180]. An increase in body fat is directly related to the number of macrophages found in adipose tissue Citation[178,179,181,182]. A net proinflammatory response of adipose tissue may result from: adipose tissue secretion of proinflammatory factors; adipose tissue secretion of factors that stimulate other tissues to produce inflammatory factors; and decreased production of anti-inflammatory factors Citation[1,29]. The net proinflammatory response associated with pathogenic adipose tissue is an important contributor to metabolic disease Citation[1,166,180,183–187].

Adipose tissue inflammatory factors are produced by both adipocytes and associated inflammatory cells, such as adipose tissue-related macrophages. Adipose tissue macrophages may be responsible for almost all adipose tissue TNF-α expression and significant amounts of other inflammatory factors, including interleukins, cathepsin S, macrophage-inhibitory factor, nerve growth factor and inducible nitric oxide synthase (iNOS) Citation[178,181,188,189]. Furthermore, the origin of increases in inflammatory factors found in patients with excessive body weight are often derived from nonadipose tissue Citation[172,190], with pathogenic adipose tissue promoting the inflammatory responses from other body organs. An example would be hepatic C-reactive protein production in response to adipocyte/adipose tissue IL-6 release, as may occur with obesity Citation[191].

As previously described, adipocytes are metabolically active and have the capacity to secrete nonproteins factors such as prostaglandins, fatty acids, monobutyrin, and steroid hormones. From an immune standpoint, adipocytes and adipose tissue also produce bioactive proteins, termed adipokines, which are secretory factors that include classic cytokines, complement factors, enzymes, growth factors, hormones and matrix proteins. Increased secretion of proinflammatory adipokines with cytokine activity may contribute to metabolic disease, including atherosclerosis Citation[1,47,180,192]. Adipose tissue-derived inflammatory factors that are potentially pathogenic include acute phase reactants, such as plasminogen activator inhibitor-1 Citation[193] and possibly C-reactive protein Citation[194,195], proteins of the alternative complement system Citation[1,47,172], chemotactic/chemoattractant adipokines Citation[1,47], eicosanoids/prostaglandins Citation[1,47], and reduced secretion of anti-inflammatory factors (Box 4)Citation[1,47,192].

It is not known which, if any, proinflammatory factors are best to measure in relation to adipose tissue’s promotion of metabolic disease. Among the more commonly described adipose tissue inflammatory factors associated with T2DM are TNF-α, IL-6 Citation[196] and C-reactive protein, which are all positively correlated to adipocyte size Citation[197]. Adiponectin is the adipocyte/adipose tissue anti-inflammatory factor whose decreased levels are best described to be associated with metabolic disease. Adiponectin levels are negatively correlated with adipocyte size Citation[197].

Cross-talk & interactions with other body tissues

The onset or worsening of many metabolic diseases might best be considered the net result of a pathologic partnership between adipose tissue and limitations and/or dysfunction of other body organs. Impaired cross-talk with adjacent adipocytes, such as paracrine signaling Citation[198], may account for impaired adipogenesis and promotion of metabolic disease Citation[199]. Cross-talk is defined as biological signaling exchanges between body organs. This cross-talk is important for the integration of metabolic functions of adipocytes/adipose tissue with other adipocytes, other adipose tissue depots and other body organs. Disrupted or adverse cross-talk between adipose tissue and other body organs may contribute to metabolic disease Citation[200,201]. Organs systems affected by signaling from adipose tissue include the nervous system Citation[202], immune system Citation[176], skeletal muscle Citation[203–206], cardiovascular system Citation[133,167,187,207–213] liver Citation[214,215], gastrointestinal system Citation[216], adrenal cortex Citation[217] and thyroid Citation[218]. The best example might be the CNS Citation[202,219–226], which would include the CNS activity of leptin/insulin Citation[143,224,227–235], pro-opiomelanocortin (POMC)/cocaine amphetamine-regulated transcript (CART) Citation[223,229,236,237], neuropeptide Y (NPY)/agoutirelated peptide (AgRP) Citation[223,229,238–242], melano-cortin system, Citation[223,236,239,243–248] and CNS neuroendocrine activity (e.g., thyroid-releasing hormone and corticotropin-releasing hormone) Citation[223,249,250]. More recently, the endocannabinoid system is becoming more recognized as playing an important role in adipocyte function, and the pathogenic potential of adipose tissue Citation[7,223,251–259]. Abnormalities of other organ systems may also affect the pathogenic potential of adipose tissue Citation[260–262].

In addition to impaired cross-talk signaling, nonadipose body organs may have inherent or acquired dysfunction or limitations that increase the risk of developing metabolic disease. Some patients may have an inability to metabolize intra-muscular fat due to genetic or acquired ‘inflexibility’ in their oxidation of free fatty acids Citation[263]. If fat cell hypertrophy and/or increase in visceral adipose tissue results in increased circulating free fatty acids, this may cause excessive ectopic free fatty acid storage in muscle. Such ‘lipotoxicity’ is pathogenic in that the accumulation of intramyocellular lipids, such as diacyl-glycerol, fatty acyl CoA and ceramides, promote insulin resistance Citation[263–267]. Fat weight loss through hypocaloric nutritional intervention may not necessarily improve ‘inflexible’ muscle’s inherent ability to metabolize free fatty acids. But such intervention is nonetheless therapeutic in that through fat weight loss, the triglyceride content of skeletal muscle is reduced, improving insulin sensitivity Citation[266]. Thus, in the presence of limitations or dysfunction of target organs, positive caloric balance may promote metabolic disease, while negative caloric balance may improve metabolic disease. Conversely, if skeletal muscle were to become ‘hyperflexible’ (i.e., develop an increased capacity to metabolize free fatty acids), either through genetic predisposition or through the use of drug therapies (such as PPAR-γ agents), then metabolic disease may be theoretically prevented or improved Citation[268], even in the presence of pathogenic adipose tissue.

The liver is also an important organ in oxidizing and metabolizing free fatty acids. With positive caloric balance, adipocyte hypertrophy and visceral adipose tissue accumulation may increase the flow of free fatty acids to the liver, increasing hepatic lipid content, and resulting in the common clinical finding of hepatosteatosis (‘fatty liver’). Patients with ‘inflexibility’ in hepatic free fatty acid oxidization may be more susceptible to lipid accumulation in the liver and, thus, more prone to developing insulin resistance and dyslipidemia Citation[34]. Finally, in patients with an inherent or acquired insulinopenia, the chronic increase in circulating free fatty acid from pathogenic adipose tissue and ectopic free fatty acid deposition in the pancreas may decrease insulin secretion and also contribute to T2DM Citation[269].

In summary, adipose tissue does not act alone in its potential to promote metabolic disease. In patients who are predisposed to metabolic disease due to genetic background, age Citation[270], gender, nutritional intake, physical activity level, comorbid conditions, concurrent drug treatments and other predispositions, it is the limitation or dysfunction of other body organs that determines the degree by which the pathogenic potential of adipose tissue will promote metabolic disease. Thus, the variability in fat weight gain which results in metabolic disease is not only due to how fat is stored (adipogenesis), where the fat is stored (visceral versus other fat depots), but is also dependent upon the signaling and interactions with other body organs.

Adverse clinical consequences of excessive fat mass

Excessive fat mass alone may contribute to other clinical disorders, such as cardiovascular Citation[138,139,207,271–273], neurologic Citation[138,139], pulmonary Citation[138,139,271], musculoskeletal Citation[138,139], dermatologic Citation[138,139], gastrointestinal, Citation[138,139] genitourinary Citation[138,139], renal Citation[138,139,272] and psychological diseases Citation[138,139].

Expert commentary: defining pathogenic adipose tissue & its metabolic complications

Currently, the most common term defining the clustering of metabolic abnormalities that increase atherogenic risk is the ‘metabolic syndrome.’ The diagnostic parameters for metabolic syndrome include increased waist circumference, elevated fasting glucose levels, elevated blood pressure, elevated triglyceride levels and low HDL-C levels Citation[169,272–277]. However, this term does not reflect a description of the unified, pathophysiologic process leading to these clustering of metabolic disorders. Nor is there uniform agreement as to its definition Citation[274,169]. Finally, the diagnosis of the metabolic syndrome may not be a better predictor of future metabolic disease than assessment of its individual components Citation[278–280]. Many (but not all) clinicians find the term metabolic syndrome useful, Citation[281–276,284–286] with the hope that corralling key physical examination and laboratory measures into a group may help better focus attention to parameters that increase CHD risk. However, from a patient perspective, survey studies have suggested that the self-reported diagnosis of metabolic syndrome is often inaccurate and misunderstood Citation[287]. The lack of universally accepted terminology to describe the interrelationship between excessive body weight and metabolic abnormalities is unsatisfying. This is especially so given analyses supporting the theory that the components of the metabolic syndrome are likely due to a unified pathophysiologic process Citation[288], sometimes described as a ‘common soil’ hypothesis Citation[289,290]. Pathogenic adipose tissue represents such a unified pathophysiologic process leading to metabolic diseases, which are often major CVD and CHD risk factors, and possibly leading directly to atherosclerosis itself Citation[1,7,47,169,272,406].

Other terms have been proposed to better define the relationship of pathogenic adipose tissue to metabolic disease. One such term is adiposopathy (‘adipose-opathy’) Citation[1,7,9,47,169,213,223,274,291–294,406,407], which is defined as pathogenic adipose tissue that is promoted by positive caloric balance and sedentary lifestyle in genetically and environmentally susceptible patients. Adiposopathy is anatomically manifested by adipocyte hypertrophy, visceral adipose tissue accumulation and ectopic fat deposition. Physiologically, adiposopathy results in adverse metabolic and immune consequences resulting in clinical metabolic disease Citation[169]. The suffix ‘-pathy’ is often used to describe anatomical abnormalities of body organs that result in clinical disease, such as cardiomyopathy, myopathy, encephalopathy, ophthalmopathy, retinopathy, enteropathy, nephropathy, neuropathy and dermopathy Citation[9]. Cardiomyopathy is a term that describes pathologic enlargement of cardiac cells and heart leading to clinical disease. Enlargement of adipocytes and adipose tissue also leads to clinical disease. Myopathy is a term that describes the pathologic dysfunction of muscle. Muscle cell hypertrophy is found in some types of muscular dystrophies. Muscle is an organ located in widespread locations in the body, with differing types of muscle having differing physiology and differing pathogenic potentials, depending upon the muscle type (skeletal, smooth and cardiac). Adipocyte hypertrophy and visceral fat accumulation are also pathogenic. Furthermore, similar to muscle, adipose tissue is located in widespread locations in the body, with differing depots having different physiology and differing pathogenic potentials depending upon the depot (visceral, subcutaneous or perivascular). Adiposopathy describes adipocyte and adipose tissue anatomical abnormalities accompanied by pathophysiologic metabolic and immune responses that lead to metabolic illnesses. It is the ‘pathos’ of adipose tissue that helps to explain why the increasing epidemic of obesity is associated with an increased prevalence of T2DM, hypertension and dyslipidemia. The term adiposopathy highlights that adipose tissue has no less pathogenic potential than the pathos or pathologic dysfunction of other body organs and clinically represents no less of a ‘disease’ Citation[9].

Another term that attempts to define the relationship between excessive fat mass and metabolic disease is ‘diabesity’, which represents an interpretation of a relationship between obesity and T2DM Citation[295,296]. ‘Acquired lipodystrophy’ describes how the pathogenic potential of adipose tissue may be expressed by positive caloric balance through adipocyte hypertrophy-induced impairments of adipocyte functions. This is, paradoxically, not unlike the physiologic processes responsible for the adverse metabolic consequences associated with too little adipose tissue, as found with genetic lipoatrophy Citation[35]. Finally, ‘Cushing’s disease of the omentum’ is a term describing the relationship between visceral fat and metabolic disease Citation[9,27,81,102,297–302].

Five-year view: treatment of pathogenic adipose tissue to reduce CHD & CVD risk

No currently approved treatment indications exist for treatment of the non-mass-related consequences of pathogenic adipose tissue. However, therapies that improve pathogenic adipose tissue function may also improve clinical disease Citation[47]. One of the most important reasons to consider adipocyte hypertrophy and visceral adipose tissue accumulation as a viable treatment target is because their pathogenic potential is modifiable. Treatment modalities that favorably modify pathogenic adipose tissue include nutritional interventions, increased physical activity Citation[47,303,304] and drug therapy Citation[293,47]. Acute caloric deprivation, such as through starvation or use of very low calorie meals, promptly improves many metabolic parameters associated with metabolic diseases Citation[47]. These benefits may occur even before significant changes in overall fat mass, and are likely due to acute gene-expression responses of adipose tissue and other body organs Citation[305,306]. The more clinically relevant gradual reduction in fat mass Citation[47] through nutritional inter-ventions Citation[307–309] may favorably modify adipocyte size and gene expression Citation[310]. Clinical observations support that it is not nutrition or physical activity, but rather weight (fat) loss that has the strongest effect upon reducing the risk of T2DM and dyslipidemia Citation[311,312], especially when accompanied by a reduction in visceral adipose tissue Citation[313]. Overall, nonpharmacological interventions that reduce adipocyte hypertrophy and reduce visceral adiposity improve glucose metabolism Citation[314–317], hypertension Citation[318,319] and dyslipidemia Citation[316,320,321]. Not only are these therapeutic interventions effective for improving established disease, but regular physical activity Citation[322] and weight loss in overweight and obese patients can also help delay and/or prevent the onset of T2DM, hypertension, and dyslipidemia Citation[323,324].

Pharmacologically, therapeutic agents such as PPAR-γ agonists are effective in treating metabolic diseases such as T2DM, with some PPAR agonists also improving some atherogenic lipid parameters Citation[325]. The metabolic benefits of PPAR agonists are significantly due to their effects in promoting the recruitment of additional adipocytes and in improving the function of existing fat cells Citation[31,47,326]. This may help to explain why administration of PPAR-γ agonists to patients with impaired glucose tolerance or impaired fasting glucose reduces the progression to T2DM Citation[327]. Thus, PPAR-γ agonists promote the recruitment and proliferation of adipocytes, and decrease the ratio of visceral to subcutaneous adipose tissue. This helps resolve the apparent paradox wherein adding more (functional) adipose tissue is employed as a therapeutic strategy to improve metabolic disease, which in turn, is significantly due to too much (dysfunctional and pathogenic) adipose tissue Citation[47,169].

Improving adipose tissue functionality has also been suggested to contribute to the favorable clinical outcomes found with common therapeutic agents such as ACE inhibitors and ARBs Citation[7,83,84,328], as well as statins Citation[329]. Finally, favorable effects upon both adipose tissue anatomic and metabolic parameters are found with the use of antiobesity, weight-loss agents Citation[293] such as orlistat, sibutramine and cannabinoid receptor antagonists (not yet approved in the USA Citation[291]). Improvements in adipocyte and adipose tissue function help explain why these agents improve metabolic disease Citation[7,9,47].

Finally, the pathological consequences of adipocyte hyper-trophy and visceral adipose tissue accumulation go far beyond the metabolic diseases highlighted in this review. Pathogenic adipose tissue may also contribute to hepatosteatosis Citation[169,215], cancer Citation[330], thrombosis, polycystic ovarian syndrome Citation[331,332], hyperandrogenemia in women Citation[1,119,331,333], hyperestrogenemia in men Citation[1] and other metabolic abnormalities. Since adipose tissue health may affect patient health, clinicians should understand the importance of pathogenic adipose tissue in the genesis of the most common diseases encountered in medical practice, many of which are important CVD risk factors. Scientific organizations should work towards a consensus to define, diagnosis and eventually treat pathogenic adipose tissue.

Table 1. Comparison of subcutaneous, peripheral adipose tissue versus visceral adipose tissue*.

Table 2. Abnormalities in adipose tissue factors that may contribute to metabolic disease*.

Box 1. Examples of factors influencing adipogenesis.

Modulating and transcriptional factors produced by adipocytes

Adipocyte determination and differentiation factor-1/ sterol regulatory element-binding proteins

CCAAT/ enhancer-binding proteins

Peroxisome proliferator-activated γ receptors

E2F proteins

Cyclin D

Cyclin-dependent kinases

Nuclear receptors, such as farnesoid X receptor, liver X receptor and retinoid X receptors

Extracellular matrix factors produced by adipocytes

Actin

Bone morphogenic protein

Collagen-binding protein (colligin)

Collagens

Cystatin C

Cysteine protease cathepsin S (CTSS)

Fibroblast growth factor

Fibronectin

Gelsolin

Integrin

Laminin

Matrix metalloproteases

Myosin

Nidogen (entactin)

Osteonectin (secreted protein acidic and rich in cysteine [SPARC])

Procollagen

Stromal cell-derived factor 1

Tubulin

Vimentin

Angiogenesis factors produced by adipocytes

Adiponectin

Angiogenin

Angiopoietin-2

Autotaxin

Endostatin

Fibroblast growth factor

Hepatocyte growth factor

Hypoxia inducible factor-1

Leptin

Monobutyrin

Matrix metalloproteinases

Nitric oxide synthase

Osteonectin (SPARC)

Pigment epithelium-derived factor

Plasminogen activator inhibitor-1

Platelet derived growth factor

Prostaglandin E2

Prostaglandin I2 (prostacyclin)

Tissue factor

Transforming growth factor

Vascular endothelial growth factor

Other adipose tissue factors that may facilitate adipogenesis

Acylation-stimulating protein (mainly lipogenesis)

Adipogenin

Adiponectin

Agouti protein

Angiotensin II (promotes differentiation)

Angiotensinogen (promotes differentiation)

Angiotensin-converting enzyme (promotes differentiation)

Autotaxin (promotes proliferation)

cAMP-response element-binding protein

Epidermal growth factor (promotes differentiation)

Estrogens

F-Box proteins (such as S-phase kinase-associated protein [Skp]2)

Free fatty acids

Galectin 12 (promotes differentiation)

Hormone sensitive lipase

Insulin-like growth factor

Leptin (inconsistent reports in medical literature)

Leukemia inhibitory factor (inconsistent reports in medical literature)

Lipin

Lysophosphosphatidic acid (promotes differentiation)

Macrophage colony stimulating factor

Neuronatin

Nitric oxide

Phosphoinositide 3-kinase

Plasminogen activator inhibitor-1 (inconsistent reports in medical literature)

Prolactin

Resistin (inconsistent reports in medical literature)

Retinoids (inconsistent reports in medical literature)

Adipose tissue factors that inhibit adipogenesis

Androgens (testosterone)

Angiotensin II (inhibits preadipocyte recruitment)

Angiotensin-converting enzyme (inhibits preadipocyte recruitment)

Angiotensionogen (inhibits preadipocyte recruitment)

Autotaxin (inhibits differentiation)

Ceramide

Chemerin

Epidermal growth factor (inhibits proliferation)

Insulin-like growth factor-binding protein

IL-1, IL-6, IL-8, IL-11

Leptin (inconsistent reports in medical literature)

Leukemia inhibitory factor (inconsistent reports in medical literature)

Lysophosphatidic acid (inhibits differentiation)

Macrophage inflammatory protein-1α

Mitogen-activated protein kinase

Monocyte chemoattractant protein-1

Necdin

Plasminogen activator inhibitor (inconsistent reports in medical literature)

Pre-adipocyte factor-1

Prostaglandin F2

Resistin (inconsistent reports in medical literature)

Resistin-like molecules

Retinoids (inconsistent reports in medical literature)

Transforming growth factor-β

TNF-α

Other factors, not necessarily adipocyte in origin, that facilitate adipogenesis

Catecholamines (neurologic signaling promotes adipocyte hyperplasia)

Hormones

– Estrogens

– Ghrelin (inconsistent reports in the literature)

– Glucocorticoids (stimulates differentiation)

– Insulin

– Insulin growth factor-1

– Prolactin

– Thyroid hormone

Lipoproteins (very-low-density lipoproteins)

Lipids

– Endocannabinoids (anandamide)

– Dietary fats

– Free fatty acids

– Prostaglandins (PGE2, PGI2, PGJ2)

Proteins

– Plasminogen/plasmin

– Neuropeptide Y

– Protein kinase C (PKC-bI)

– Protein kinase C inhibitor

Other factors, not necessarily adipocyte in origin, that impair adipogenesis

Androgens

Catecholamines (neurologic signaling decreases adipocyte hypertrophy)

Cytokines (such as TNF-α)

Flavonoids

Ghrelin (inconsistent reports in the literature)

Glucagon-like peptide-1

Glucocorticoids (impairs proliferation)

Growth factors

– Epidermal growth factor, (which impairs proliferation and promotes differentiation)

– Fibroblast growth factor

– Platelet-derived growth factor

– Transforming growth factor-ν

– Tumor growth factor β

Interferon

Interleukins

Protein kinase C (PKC-delta)

Prostaglandins (PGF2)

Box 2. Examples of adipose tissue factors that affect free fatty acid metabolism.

Acetyl-coenzyme A carboxylase

Acetyl-coenzyme A synthetase

Acylation-stimulating protein

Adenosine

Adenosine monophosphate protein kinase (AMPK)

Adiponutrin

Adipophilin (adipose differentiation-related protein)

Adipsin (complement factor D)

Adrenomedullin

Agouti protein

Androgens

Angiotensin I and II

Angiotensinogen

Annexin

Apolipoprotein C1

Aquaporin 7

Carnitine palmitoyl transferase-1

Caveolin

Desnutrin (adipocyte triglyceride lipase [ATGL])

Estrogens

Fasting-induced adipocyte factor

Fatty acid-binding protein

Fatty acid synthase

Fatty acid translocase (CD36)

Fatty acid transport protein

Hormone sensitive lipase

Interleukins

Leptin

Lipin

Lipoprotein lipase

Perilipin

Phosphoenolpyruvate carboxykinase (PEPCK)

Prostaglandins

S3-12

Stearoyl-CoA desaturase

Tail interacting protein 47

Transcription factors

TNF-α

Box 3. Examples of adipose tissue properties that highlight its activity as a metabolic organ.

Receptors for traditional peptide and glycoprotein hormones

Adiponectin

Angiotensin II type 1 and 2

Gastrin/cholecystokinin

Glucagon

Glucagon-like peptide-1

Growth hormone

Insulin

Insulin-like growth factors

Thyroid stimulating hormone

Receptors for nuclear hormones

Androgens

Estrogens

Glucocorticoids

Nuclear factor-kB

Progesterone

Thyroid hormone

Vitamin D

Other nuclear receptors

Peroxisome proliferator-activated receptor (PPAR) α receptors

PPAR β receptors

PPAR δ receptors

PPAR γ receptors

Farnesoid X receptors

Liver X receptors

Retinoid X receptors

Receptors for cytokines or adipokines with cytokine-like activity

Adiponectin

Interleukins

Leptin

Transforming growth factor-β

TNF-α

Receptors for growth factors

Epidermal growth factor

Fibroblast growth factor

Hepatocyte growth factor

Insulin-like growth factor

Platelet-derived growth factor

Transforming growth factor-β

Tumor growth factor

Vascular endothelial growth factor

Catecholamine receptors

Catecholamines such as α1 and α2; β1, β2, β3

Muscarinic receptors

Nicotinic receptors

Other receptors

Adenosine

Cannabinoids

Lipoproteins (high-density lipoprotein, low-density lipoprotein, very-low-density lipoprotein)

Melanocortins

Neuropeptide Y

Prostaglandins

Box 4. Inflammatory factors associated with adipose tissue*.

Adipokines with cytokine activity

Adipsin

IL-1B, IL-6, IL-8, IL-17D, IL-18

Leptin

Macrophage colony-stimulating factor

Macrophage-inhibitor factor

Monocyte chemotactic protein-1

Regulated on activation, normal T-cell expressed and secreted (RANTES)

Resistin

TNF-α

Visceral adipose tissue-derived serpin

Acute phase reactants

α-1 acid glycoprotein

Amyloid A

Ceruloplasmin

C-reactive protein

Haptoglobin

IL-1 receptor antagonist

Lipocalins

Metallothionein

Pentraxin-3

Plasminogen activator inhibitor-1

Adipokines of the alternative complement system

Adipsin

Acylation-stimulating protein

Complements C3 and B

Chemotactic/chemoattractant adipokines

Eotaxin

Interferon inducible protein

Monocyte chemoattractant protein-1

Macrophage colony-stimulating factor

Macrophage migration inhibitory factor

Stromal derived factor-1

RANTES

Resistin

TNF-α

Vascular adhesion protein-1

Vascular cell adhesion molecule-1

Eicosanoids/prostaglandins

Prostaglandin E2

Anti-inflammatory adipose tissue factors

Adiponectin

Annexin-1

IL-10

IL-6

Nitric oxide.

Transforming growth factor-β

Key issues

Adipocyte hypertrophy and visceral adiposity may contribute to metabolic diseases, such as Type 2 diabetes mellitus, hypertension and dyslipidemia.

The pathogenic potential of adipose tissue is dependent upon genetic and environment factors.

Impaired adipogenesis during positive caloric balance may lead to adipocyte hypertrophy, which contributes to metabolic disease, especially if it occurs in the visceral region.

The pathogenic potential of adipose tissue is not only dependent upon how the fat is stored (hypertrophy versus hyperplasia), but also where the fat is stored (visceral versus subcutaneous distribution).

Adipose tissue has important endocrine and immune activities whose disruption may lead to metabolic disease.

The net release of free fatty acids is a potential adverse consequence of pathogenic adipose tissue, which may contribute to metabolic disease.

The pathogenic potential of adipose tissue is best viewed as a partnership with the inherited or acquired limitations in ‘cross-talk’ and/or impairments of other body organs.

Financial & competing interests disclosure

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

No writing assistance was utilized in the production of this manuscript.

Notes

*A net proinflammatory response that may contribute to metabolic disease occurs with increased secretion of adipose tissue proinflammatory factors, and a decrease in secretion of anti-inflammatory adipose tissue factors.

References

  • Bays H, Ballantyne C. Adiposopathy: why do adiposity and obesity cause metabolic disease? Future Lipidol.1(4), 389–420 (2006).
  • Hedley AA, Ogden CL, Johnson CL, Carroll MD, Curtin LR, Flegal KM. Prevalence of overweight and obesity among US children, adolescents, and adults, 1999–2002. JAMA291(23), 2847–2850 (2004).
  • Flegal KM, Carroll MD, Ogden CL, Johnson CL. Prevalence and trends in obesity among US adults, 1999–2000. JAMA288(14), 1723–1727 (2002).
  • Flegal KM, Graubard BI, Williamson DF, Gail MH. Cause-specific excess deaths associated with underweight, overweight, and obesity. JAMA298(17), 2028–2037 (2007).
  • Bays H, Chapman R. BMI and Frequency of Diabetes, Hypertension, and Dyslipidemia: Comparison of SHIELD and NHANES Data. Presented at the North American Association for the Study of Obesity (NAASO) 2005 Annual Scientific Meeting, Vancouver, British Columbia, October 15–19, 2005.
  • Bays HE, Chapman RH, Grandy S. The relationship of body mass index to diabetes mellitus, hypertension and dyslipidaemia: comparison of data from two national surveys. Int. J. Clin. Pract.61(5), 737–747 (2007).
  • Bays H, Dujovne CA. Adiposopathy is a more rational treatment target for metabolic disease than obesity alone. Curr. Atheroscler. Rep.8(2), 144–156 (2006).
  • Hossain P, Kawar B, El Nahas M. Obesity and diabetes in the developing world – a growing challenge. N. Engl. J. Med.356(3), 213–215 (2007).
  • Bays HE, Rodbard RW, Schorr AB, González-Campoy JM. Adiposopathy: treating pathogenic adipose tissue to reduce cardiovascular disease risk. Curr. Treat. Options Cardiovasc. Med.9(4), 259–271 (2007).
  • Schulz LO, Bennett PH, Ravussin E et al. Effects of traditional and western environments on prevalence of Type 2 diabetes in Pima Indians in Mexico and the U.S. Diabetes Care29(8), 1866–1871 (2006).
  • Nelson RG, Sievers ML, Knowler WC et al. Low incidence of fatal coronary heart disease in Pima Indians despite high prevalence of non-insulin-dependent diabetes. Circulation81(3), 987–995 (1990).
  • Ingelfinger JA, Bennett PH, Liebow IM, Miller M. Coronary heart disease in the Pima Indians. Electrocardiographic findings and postmortem evidence of myocardial infarction in a population with a high prevalence of diabetes mellitus. Diabetes25(7), 561–565 (1976).
  • Paolisso G, Tataranni PA, Foley JE, Bogardus C, Howard BV, Ravussin E. A high concentration of fasting plasma non-esterified fatty acids is a risk factor for the development of NIDDM. Diabetologia38(10), 1213–1217 (1995).
  • Weyer C, Foley JE, Bogardus C, Tataranni PA, Pratley RE. Enlarged subcutaneous abdominal adipocyte size, but not obesity itself, predicts Type II diabetes independent of insulin resistance. Diabetologia43(12), 1498–1506 (2000).
  • Abate N, Chandalia M, Snell PG, Grundy SM. Adipose tissue metabolites and insulin resistance in nondiabetic Asian Indian men. J. Clin. Endocrinol. Metab.89(6), 2750–2755 (2004).
  • Chandalia M, Abate N, Garg A, Stray-Gundersen J, Grundy SM. Relationship between generalized and upper body obesity to insulin resistance in Asian Indian men. J. Clin. Endocrinol. Metab.84(7), 2329–2335 (1999).
  • Smith JD, Al-Amri M, Sniderman AD, Cianflone K. Leptin and adiponectin in relation to body fat percentage, waist to hip ratio and the apoB/apoA1 ratio in Asian Indian and Caucasian men and women. Nutr. Metab. (Lond.)3(1), 18 (2006).
  • Zhu S, Heymsfield SB, Toyoshima H, Wang Z, Pietrobelli A, Heshka S. Race-ethnicity-specific waist circumference cutoffs for identifying cardiovascular disease risk factors. Am. J. Clin. Nutr.81(2), 409–415 (2005).
  • Chandalia M, Cabo-Chan AV Jr, Devaraj S, Jialal I, Grundy SM, Abate N. Elevated plasma high-sensitivity C-reactive protein concentrations in Asian Indians living in the United States. J. Clin. Endocrinol. Metab.88(8), 3773–3776 (2003).
  • Smith J, Al-Amri M, Dorairaj P, Sniderman A. The adipocyte life cycle hypothesis. Clin. Sci. (Lond.)110(1), 1–9 (2006).
  • Sniderman AD, Bhopal R, Prabhakaran D, Sarrafzadegan N, Tchernof A. Why might South Asians be so susceptible to central obesity and its atherogenic consequences? The adipose tissue overflow hypothesis. Int. J. Epidemiol.36(1), 220–225 (2007).
  • Chuang LM, Hsiung CA, Chen YD et al. Sibling-based association study of the PPARγ2 Pro12Ala polymorphism and metabolic variables in Chinese and Japanese hypertension families: a SAPPHIRe study. Stanford Asian-Pacific Program in Hypertension and Insulin Resistance. J. Mol. Med.79(11), 656–664 (2001).
  • WHO Expert Consultation. Appropriate body-mass index for Asian populations and its implications for policy and intervention strategies. Lancet363(9403), 157–163 (2004).
  • Calle C, Carranza MC, Simon MA, Torres A, Mayor P. Decreased insulin binding and antilipolytic response in adipocytes from patients with Cushing’s syndrome. Biosci. Rep.7(9), 713–719 (1987).
  • Rebuffe-Scrive M, Krotkiewski M, Elfverson J, Bjorntorp P. Muscle and adipose tissue morphology and metabolism in Cushing’s syndrome. J. Clin. Endocrinol. Metab.67(6), 1122–1128 (1988).
  • Pedersen SB. Studies on receptors and actions of steroid hormones in adipose tissue. Dan. Med. Bull.52(4), 258 (2005).
  • Tomlinson JW, Walker EA, Bujalska IJ et al. 11β-hydroxysteroid dehydrogenase type 1: a tissue-specific regulator of glucocorticoid response. Endocr. Rev.25(5), 831–866 (2004).
  • Darmon P, Dadoun F, Boullu-Ciocca S, Grino M, Alessi MC, Dutour A. Insulin resistance induced by hydrocortisone is increased in patients with abdominal obesity. Am. J. Physiol. Endocrinol. Metab.291(5), E995–E1002 (2006).
  • Wood IS, Trayhurn P. Adipokines and the signaling role of adipose tissue in inflammation and obesity. Future Lipidol.1(1), 81–89 (2006).
  • Smas CM, Sul HS. Control of adipocyte differentiation. Biochem. J.309 (Pt 3), 697–710 (1995).
  • Bays H, Mandarino L, DeFronzo RA. Role of the adipocyte, free fatty acids, and ectopic fat in pathogenesis of Type 2 diabetes mellitus: peroxisomal proliferator-activated receptor agonists provide a rational therapeutic approach. J. Clin. Endocrinol. Metab.89(2), 463–478 (2004).
  • Haque WA, Shimomura I, Matsuzawa Y, Garg A. Serum adiponectin and leptin levels in patients with lipodystrophies. J. Clin. Endocrinol. Metab.87 (5), 2395 (2002).
  • Gavrilova O, Marcus-Samuels B, Graham D et al. Surgical implantation of adipose tissue reverses diabetes in lipoatrophic mice. J. Clin. Invest.105(3), 271–278 (2000).
  • Nadler ST, Attie AD. Please pass the chips: genomic insights into obesity and diabetes. J. Nutr.131(8), 2078–2081 (2001).
  • Heilbronn L, Smith SR, Ravussin E. Failure of fat cell proliferation, mitochondrial function and fat oxidation results in ectopic fat storage, insulin resistance and Type II diabetes mellitus. Int. J. Obes. Relat. Metab. Disord.28(Suppl. 4), S12–S21 (2004).
  • Ravussin E, Smith SR. Increased fat intake, impaired fat oxidation, and failure of fat cell proliferation result in ectopic fat storage, insulin resistance, and Type 2 diabetes mellitus. Ann. NY Acad. Sci.967, 363–378 (2002).
  • Van, Harmelen V, Skurk T, Rohrig K et al. Effect of BMI and age on adipose tissue cellularity and differentiation capacity in women. Int. J. Obes. Relat. Metab. Disord.27(8), 889–895 (2003).
  • Marques BG, Hausman DB, Martin RJ. Association of fat cell size and paracrine growth factors in development of hyperplastic obesity. Am. J. Physiol.275(6 Pt 2), R1898–R1908 (1998).
  • Hausman DB, DiGirolamo M, Bartness TJ, Hausman GJ, Martin RJ. The biology of white adipocyte proliferation. Obes. Rev.2(4), 239–254 (2001).
  • Roche AF. The adipocyte-number hypothesis. Child Dev.52(1), 31–43 (1981).
  • Zhu X, He Q, Lin, Z. Human preadipocyte culture and the establishment of hyperplasia and hypertrophy model. Zhonghua Zheng. Xing.Shao Shang Wai Ke. Za Zhi.15(3), 199–201 (1999).
  • Dubois SG, Heilbronn LK, Smith SR, Albu JB, Kelley DE, Ravussin E. Decreased expression of adipogenic genes in obese subjects with Type 2 diabetes. Obesity14(9), 1543–1552 (2006).
  • Gregoire FM. Adipocyte differentiation: from fibroblast to endocrine cell. Exp. Biol. Med. (Maywood)226(11), 997–1002 (2001).
  • Haller H, Leonhardt W, Hanefeld M, Julius U. Relationship between adipocyte hypertrophy and metabolic disturbances. Endokrinologie74(1), 63–72 (1979).
  • Smith U. Effect of cell size on lipid synthesis by human adipose tissue in vitro.J. Lipid Res.12(1), 65–70 (1971).
  • Rosen ED, Spiegelman BM. Molecular regulation of adipogenesis. Annu. Rev. Cell Dev. Biol.16, 145–171 (2000).
  • Bays H, Blonde L, Rosenson R. Adiposopathy: how do diet, exercise, weight loss and drug therapies improve metabolic disease? Expert Rev. Cardiovasc Ther.4(6), 871–895 (2006).
  • Ailhaud G. Adipose tissue as a secretory organ: from adipogenesis to the metabolic syndrome. CR Biol.329(8), 570–577 (2006).
  • Hissin PJ, Foley JE, Wardzala LJ et al. Mechanism of insulin-resistant glucose transport activity in the enlarged adipose cell of the aged, obese rat. J. Clin. Invest.70(4), 780–790 (1982).
  • Gregoire FM, Smas CM, Sul HS. Understanding adipocyte differentiation. Physiol. Rev.78(3), 783–809 (1998).
  • Nadler ST, Stoehr JP, Schueler KL, Tanimoto G, Yandell BS, Attie AD. The expression of adipogenic genes is decreased in obesity and diabetes mellitus. Proc. Natl Acad. Sci. USA97(21), 11371–11376 (2000).
  • Danforth E Jr. Failure of adipocyte differentiation causes Type II diabetes mellitus? Nat. Genet.26(1), 13(2000).
  • Salans LB, Bray GA, Cushman SW et al. Glucose metabolism and the response to insulin by human adipose tissue in spontaneous and experimental obesity. Effects of dietary composition and adipose cell size. J. Clin. Invest.53(3), 848–856 (1974).
  • Bray GA, Glennon JA, Salans LB, Horton ES, Danforth E Jr, Sims EA. Spontaneous and experimental human obesity: effects of diet and adipose cell size on lipolysis and lipogenesis. Metabolism26(7), 739–747 (1977).
  • Julius U, Leonhardt W, Schneider H et al. Basal and stimulated hyperinsulinemia in obesity: relationship to adipose-cell size. Endokrinologie73(2), 214–220 (1979).
  • Le Lay S, Krief S, Farnier C et al. Cholesterol, a cell size-dependent signal that regulates glucose metabolism and gene expression in adipocytes. J. Biol. Chem.276(20), 16904–16910 (2001).
  • Leonhardt W, Haller H, Hanefeld M. The adipocyte volume in human adipose tissue: II. Observations in diabetes mellitus, primary hyperlipoproteinemia and weight reduction. Int. J. Obes.2(4), 429–439 (1978).
  • Pausova Z. From big fat cells to high blood pressure: a pathway to obesity-associated hypertension. Curr. Opin. Nephrol. Hypertens.15(2), 173–178 (2006).
  • Jernas M, Palming J, Sjoholm K et al. Separation of human adipocytes by size: hypertrophic fat cells display distinct gene expression. FASEB J.20(9), 1540–1542 (2006).
  • Schneider BS, Faust IM, Hemmes R, Hirsch J. Effects of altered adipose tissue morphology on plasma insulin levels in the rat. Am. J. Physiol.240(4), E358–E362 (1981).
  • Morrison RF, Farmer SR. Hormonal signaling and transcriptional control of adipocyte differentiation. J. Nutr.130(12), 3116S–3121S (2000).
  • Hausman GJ, Wright JT, Richardson RL. The influence of extracellular matrix substrata on preadipocyte development in serum-free cultures of stromal-vascular cells. J. Anim. Sci.74(9), 2117–2128 (1996).
  • Rundhaug JE. Matrix metalloproteinases and angiogenesis. J. Cell Mol. Med.9(2), 267–285 (2005).
  • Lilla J, Stickens D, Werb Z. Metalloproteases and adipogenesis: a weighty subject. Am. J. Pathol.160(5), 1551–1554 (2002).
  • Kim KH, Song MJ, Chung J, Park H, Kim JB. Hypoxia inhibits adipocyte differentiation in a HDAC-independent manner. Biochem. Biophys. Res. Commun.333(4), 1178–1184 (2005).
  • Trayhurn P, Wood IS. Signalling role of adipose tissue: adipokines and inflammation in obesity. Biochem. Soc. Trans.33(Pt 5), 1078–1081 (2005).
  • Ye J, Gao Z, Yin J, He Q. Hypoxia is a potential risk factor for chronic inflammation and adiponectin reduction in adipose tissue of ob/ob and dietary obese mice. Am. J. Physiol. Endocrinol. Metab.293(4), E1118–E1128 (2007).
  • Wang B, Wood IS, Trayhurn P. Dysregulation of the expression and secretion of inflammation-related adipokines by hypoxia in human adipocytes. Pflugers Arch.455(3), 479–492 (2007).
  • Gregg EW, Cheng YJ, Cadwell BL et al. Secular trends in cardiovascular disease risk factors according to body mass index in US adults. JAMA293(15), 1868–1874 (2005).
  • St-Onge MP, Janssen I, Heymsfield SB. Metabolic syndrome in normal-weight Americans: new definition of the metabolically obese, normal-weight individual. Diabetes Care27(9), 2222–2228 (2004).
  • Ailhaud G, Fukamizu A, Massiera F, Negrel R, Saint-Marc P, Teboul M. Angiotensinogen, angiotensin II and adipose tissue development. Int. J. Obes. Relat. Metab. Disord.24(Suppl. 4), S33–S35 (2000).
  • Engeli S. Role of the renin–angiotensin –aldosterone system in the metabolic syndrome. Contrib. Nephrol.151, 122–134 (2006).
  • Engeli S, Schling P, Gorzelniak K et al. The adipose-tissue renin–angiotensin –aldosterone system: role in the metabolic syndrome? Int. J. Biochem. Cell Biol.35(6), 807–825 (2003).
  • Kershaw EE, Flier JS. Adipose tissue as an endocrine organ. J. Clin. Endocrinol. Metab.89(6), 2548–2556 (2004).
  • Strazzullo P, Galletti F. Impact of the renin–angiotensin system on lipid and carbohydrate metabolism. Curr. Opin. Nephrol. Hypertens.13(3), 325–332 (2004).
  • Saint-Marc P, Kozak LP, Ailhaud G, Darimont C, Negrel R. Angiotensin II as a trophic factor of white adipose tissue: stimulation of adipose cell formation. Endocrinology142(1), 487–492 (2001).
  • Janke J, Engeli S, Gorzelniak K, Luft FC, Sharma AM. Mature adipocytes inhibit in vitro differentiation of human preadipocytes via angiotensin type 1 receptors. Diabetes51(6), 1699–1707 (2002).
  • Simon MF, Daviaud D, Pradere JP et al. Lysophosphatidic acid inhibits adipocyte differentiation via lysophosphatidic acid 1 receptor-dependent down-regulation of peroxisome proliferator-activated receptor γ2. J. Biol. Chem.280(15), 14656–14662 (2005).
  • Ferry G, Tellier E, Try A et al. Autotaxin is released from adipocytes, catalyzes lysophosphatidic acid synthesis, and activates preadipocyte proliferation. Up-regulated expression with adipocyte differentiation and obesity. J. Biol. Chem.278(20), 18162–18169 (2003).
  • Zhu XH, He QL, Lin ZH. Effects of catecholamines on human preadipocyte proliferation and differentiation. Zhonghua Zheng. Xing.Wai Ke. Za Zhi.19(4), 282–284 (2003).
  • Gregoire F, Genart C, Hauser N, Remacle C. Glucocorticoids induce a drastic inhibition of proliferation and stimulate differentiation of adult rat fat cell precursors. Exp. Cell Res.196(2), 270–278 (1991).
  • Adachi H, Kurachi H, Homma H et al. Epidermal growth factor promotes adipogenesis of 3T3-L1 cell in vitro.Endocrinology135(5), 1824–1830 (1994).
  • Abuissa H, Jones PG, Marso SP, O’keefe JH Jr. Angiotensin-converting enzyme inhibitors or angiotensin receptor blockers for prevention of Type 2 diabetes a meta-analysis of randomized clinical trials. J. Am. Coll. Cardiol.46(5), 821–826 (2005).
  • Koh KK, Quon MJ, Han SH et al. Vascular and metabolic effects of combined therapy with ramipril and simvastatin in patients with Type 2 diabetes. Hypertension45(6), 1088–1093 (2005).
  • Benson SC, Pershadsingh HA, Ho CI et al. Identification of telmisartan as a unique angiotensin II receptor antagonist with selective PPARgγ-modulating activity. Hypertension43(5), 993–1002 (2004).
  • Prasad A, Quyyumi AA. Renin–angiotensin system and angiotensin receptor blockers in the metabolic syndrome. Circulation110(11), 1507–1512 (2004).
  • Ingelfinger JR, Solomon CG. Angiotensin-converting-enzyme inhibitors for impaired glucose tolerance – is there still hope? N. Engl. J. Med.355(15), 1608–1610 (2006).
  • Yvan-Charvet L, Even P, Bloch-Faure M et al. Deletion of the angiotensin type 2 receptor (AT2R) reduces adipose cell size and protects from diet-induced obesity and insulin resistance. Diabetes54(4), 991–999 (2005).
  • Beltowski J. Role of leptin in blood pressure regulation and arterial hypertension. J. Hypertens.24(5), 789–801 (2006).
  • Nonogaki K. New insights into sympathetic regulation of glucose and fat metabolism. Diabetologia43(5), 533–549 (2000).
  • Lebovitz HE, Banerji MA. Point: visceral adiposity is causally related to insulin resistance. Diabetes Care28(9), 2322–2325 (2005).
  • Karelis AD, St-Pierre DH, Conus F, Rabasa-Lhoret R, Poehlman ET. Metabolic and body composition factors in subgroups of obesity: what do we know? J. Clin. Endocrinol. Metab.89(6), 2569–2575 (2004).
  • Sims EA. Are there persons who are obese, but metabolically healthy? Metabolism50(12), 1499–1504 (2001).
  • Tchkonia T, Giorgadze N, Pirtskhalava T et al. Fat depot origin affects adipogenesis in primary cultured and cloned human preadipocytes. Am. J. Physiol. Regul. Integr. Comp. Physiol.282(5), R1286–R1296 (2002).
  • Tchkonia T, Giorgadze N, Pirtskhalava T et al. Fat depot-specific characteristics are retained in strains derived from single human preadipocytes. Diabetes55(9), 2571–2578 (2006).
  • Mohamed-Ali V, Goodrick S, Rawesh A et al. Subcutaneous adipose tissue releases interleukin-6, but not tumor necrosis factor-α, in vivo.J. Clin. Endocrinol. Metab.82(12), 4196–4200 (1997).
  • Raz I, Eldor R, Cernea S, Shafrir E. Diabetes: insulin resistance and derangements in lipid metabolism. Cure through intervention in fat transport and storage. Diabetes Metab. Res. Rev.21(1), 3–14 (2005).
  • Reynisdottir S, Dauzats M, Thorne A, Langin D. Comparison of hormone-sensitive lipase activity in visceral and subcutaneous human adipose tissue. J. Clin. Endocrinol. Metab.82(12), 4162–4166 (1997).
  • Arner P. Regional differences in protein production by human adipose tissue. Biochem. Soc. Trans.29(Pt 2), 72–75 (2001).
  • Bjorntorp P. The regulation of adipose tissue distribution in humans. Int. J. Obes. Relat. Metab. Disord.20(4), 291–302 (1996).
  • Arner P. Regional adipocity in man. J. Endocrinol.155(2), 191–192 (1997).
  • Bujalska IJ, Kumar S, Stewart PM. Does central obesity reflect “Cushing’s disease of the omentum”? Lancet349(9060), 1210–1213 (1997).
  • Carr MC, Brunzell JD. Abdominal obesity and dyslipidemia in the metabolic syndrome: importance of Type 2 diabetes and familial combined hyperlipidemia in coronary artery disease risk. J. Clin. Endocrinol. Metab.89(6), 2601–2607 (2004).
  • Catalano KJ, Bergman RN, Ader M. Increased susceptibility to insulin resistance associated with abdominal obesity in aging rats. Obes. Res.13(1), 11–20 (2005).
  • Despres JP. Intra-abdominal obesity: an untreated risk factor for Type 2 diabetes and cardiovascular disease. J. Endocrinol. Invest.29(3 Suppl.), 77–82 (2006).
  • Janssen I, Katzmarzyk PT, Ross R. Waist circumference and not body mass index explains obesity-related health risk. Am. J. Clin. Nutr.79(3), 379–384 (2004).
  • Bergman RN, Kim SP, Catalano KJ et al. Why visceral fat is bad: mechanisms of the metabolic syndrome. Obesity14(Suppl. 1), 16S–19S (2006).
  • Grundy SM. Obesity, metabolic syndrome, and cardiovascular disease. J. Clin. Endocrinol. Metab.89(6), 2595–2600 (2004).
  • Gomez-Ambrosi J, Catalan V, diez-Caballero A et al. Gene expression profile of omental adipose tissue in human obesity. FASEB J.18(1), 215–217 (2004).
  • Couillard C, Bergeron N, Prud’homme D et al. Postprandial triglyceride response in visceral obesity in men. Diabetes47(6), 953–960 (1998).
  • Abate N, Garg A, Peshock RM, Stray-Gundersen J, Adams-Huet B, Grundy SM. Relationship of generalized and regional adiposity to insulin sensitivity in men with NIDDM. Diabetes45(12), 1684–1693 (1996).
  • McCarty MF. A paradox resolved: the postprandial model of insulin resistance explains why gynoid adiposity appears to be protective. Med. Hypotheses61(2), 173–176 (2003).
  • Tchkonia T, Lenburg M, Thomou T et al. Identification of depot-specific human fat cell progenitors through distinct expression profiles and developmental gene patterns. Am. J. Physiol. Endocrinol. Metab.292(1), E298–E307 (2007).
  • Lebovitz HE. The relationship of obesity to the metabolic syndrome. Int. J. Clin. Pract. Suppl.134, 18–27 (2003).
  • Dusserre E, Moulin P, Vidal H. Differences in mRNA expression of the proteins secreted by the adipocytes in human subcutaneous and visceral adipose tissues. Biochim. Biophys. Acta1500(1), 88–96 (2000).
  • Wang Y, Sullivan S, Trujillo M et al. Perilipin expression in human adipose tissues: effects of severe obesity, gender, and depot. Obes. Res.11(8), 930–936 (2003).
  • Goodpaster BH, Krishnaswami S, Harris TB et al. Obesity, regional body fat distribution, and the metabolic syndrome in older men and women. Arch. Intern. Med.165(7), 777–783 (2005).
  • Rexrode KM, Buring JE, Manson JE. Abdominal and total adiposity and risk of coronary heart disease in men. Int. J. Obes. Relat. Metab. Disord.25(7), 1047–1056 (2001).
  • Kitabchi AE, Buffington CK. Body fat distrubution, hyperandrogenicity and health risk. Semin. Reprod. Endocrinol.12(1), 6–14 (1994).
  • Bjorntorp P. Adipose tissue distribution and function. Int. J. Obes.15(Suppl. 2), 67–81 (1991).
  • Rebuffe-Scrive M, Andersson B, Olbe L, Bjorntorp P. Metabolism of adipose tissue in intraabdominal depots of nonobese men and women. Metabolism38(5), 453–458 (1989).
  • Rexrode KM, Carey VJ, Hennekens CH et al. Abdominal adiposity and coronary heart disease in women. JAMA280(21), 1843–1848 (1998).
  • Vague J. La differenciation sexuelle, facteur determinant des formes de l’obesite. Presse Med.30, 339–340 (1947).
  • Krotkiewski M, Blohme B, Lindholm N, Bjorntorp P. The effects of adrenal corticosteroids on regional adipocyte size in man. J. Clin. Endocrinol. Metab.42(1), 91–97 (1976).
  • Kirschner MA, Samojlik E, Drejka M, Szmal E, Schneider G, Ertel N. Androgen-estrogen metabolism in women with upper body versus lower body obesity. J. Clin. Endocrinol. Metab.70(2), 473–479 (1990).
  • Cohen PG. The hypogonadal-obesity cycle: role of aromatase in modulating the testosterone-estradiol shunt – a major factor in the genesis of morbid obesity. Med. Hypotheses.52(1), 49–51 (1999).
  • Tan GD, Goossens GH, Humphreys SM, Vidal H, Karpe F. Upper and lower body adipose tissue function: a direct comparison of fat mobilization in humans. Obes. Res.12(1), 114–118 (2004).
  • Misra A, Garg A, Abate N, Peshock RM, Stray-Gundersen J, Grundy SM. Relationship of anterior and posterior subcutaneous abdominal fat to insulin sensitivity in nondiabetic men. Obes. Res.5(2), 93–99 (1997).
  • Goodpaster BH, Thaete FL, Simoneau JA, Kelley DE. Subcutaneous abdominal fat and thigh muscle composition predict insulin sensitivity independently of visceral fat. Diabetes46(10), 1579–1585 (1997).
  • Smith SR, Lovejoy JC, Greenway F et al. Contributions of total body fat, abdominal subcutaneous adipose tissue compartments, and visceral adipose tissue to the metabolic complications of obesity. Metabolism50(4), 425–435 (2001).
  • Imbeault P, Lemieux S, Prud’homme D et al. Relationship of visceral adipose tissue to metabolic risk factors for coronary heart disease: is there a contribution of subcutaneous fat cell hypertrophy? Metabolism48(3), 355–362 (1999).
  • Wajchenberg BL. Subcutaneous and visceral adipose tissue: their relation to the metabolic syndrome. Endocr. Rev.21(6), 697–738 (2000).
  • Baker AR, Silva NF, Quinn DW et al. Human epicardial adipose tissue expresses a pathogenic profile of adipocytokines in patients with cardiovascular disease. Cardiovasc. Diabetol.5, 1 (2006).
  • Higuchi ML, Gutierrez PS, Bezerra HG et al. Comparison between adventitial and intimal inflammation of ruptured and nonruptured atherosclerotic plaques in human coronary arteries. Arq. Bras. Cardiol.79(1), 20–24 (2002).
  • Mazurek T, Zhang L, Zalewski A et al. Human epicardial adipose tissue is a source of inflammatory mediators. Circulation108(20), 2460–2466 (2003).
  • Torriani M, Grinspoon S. Racial differences in fat distribution: the importance of intermuscular fat. Am. J. Clin. Nutr.81(4), 731–732 (2005).
  • Engeli S. Is there a pathophysiological role for perivascular adipocytes? Am. J. Physiol. Heart Circ. Physiol.289(5), H1794–H1795 (2005).
  • Bray GA. Medical consequences of obesity. J. Clin. Endocrinol. Metab.89(6), 2583–2589 (2004).
  • Kushner RF, Roth JL. Assessment of the obese patient. Endocrinol. Metab. Clin. North Am.32(4), 915–933 (2003).
  • Brennan AM, Mantzoros CS. Drug insight: the role of leptin in human physiology and pathophysiology – emerging clinical applications. Nat. Clin. Pract. Endocrinol. Metab.2(6), 318–327 (2006).
  • Jequier E. Leptin signaling, adiposity, and energy balance. Ann. NY Acad. Sci.967379–388 (2002).
  • Havel PJ. Update on adipocyte hormones: regulation of energy balance and carbohydrate/lipid metabolism. Diabetes53(Suppl. 1), S143–S151 (2004).
  • Wang MY, Orci L, Ravazzola M, Unger RH. Fat storage in adipocytes requires inactivation of leptin’s paracrine activity: implications for treatment of human obesity. Proc. Natl Acad. Sci. USA102(50), 18011–18016 (2005).
  • Franks PW, Brage S, Luan J et al. Leptin predicts a worsening of the features of the metabolic syndrome independently of obesity. Obes. Res.13(8), 1476–1484 (2005).
  • Ruano M, Silvestre V, Castro R et al. Morbid obesity, hypertensive disease and the renin–angiotensin–aldosterone axis. Obes. Surg.15(5), 670–676 (2005).
  • Morse SA, Bravo PE, Morse MC, Reisin E. The heart in obesity-hypertension. Expert Rev. Cardiovasc. Ther.3(4), 647–658 (2005).
  • Coatmellec-Taglioni, G, Ribiere C. Factors that influence the risk of hypertension in obese individuals. Curr. Opin. Nephrol. Hypertens.12(3), 305–308 (2003).
  • Dubinski, A, Zdrojewicz, Z. The role of leptin in the development of hypertension. Postepy Hig. Med. Dosw. (Online)60447–452 (2006).
  • Bravo PE, Morse S, Borne DM, Aguilar EA, Reisin E. Leptin and hypertension in obesity. Vasc. Health Risk Manag.2(2), 163–169 (2006).
  • Van, Harmelen V, Reynisdottir S, Eriksson P et al. Leptin secretion from subcutaneous and visceral adipose tissue in women. Diabetes47(6), 913–917 (1998).
  • Couillard C, Mauriege P, Imbeault P et al. Hyperleptinemia is more closely associated with adipose cell hypertrophy than with adipose tissue hyperplasia. Int. J. Obes. Relat. Metab. Disord.24(6), 782–788 (2000).
  • Acree LS, Montgomery PS, Gardner AW. The influence of obesity on arterial compliance in adult men and women. Vasc. Med.12(3), 183–188 (2007).
  • Jensen MD. Is visceral fat involved in the pathogenesis of the metabolic syndrome? Human model. Obesity14(Suppl. 1), 20S–24S (2006).
  • Johnson JA, Fried SK, Pi-Sunyer FX, Albu JB. Impaired insulin action in subcutaneous adipocytes from women with visceral obesity. Am. J. Physiol. Endocrinol. Metab.280(1), E40–E49 (2001).
  • Shen W, Wang Z, Punyanita M et al. Adipose tissue quantification by imaging methods: a proposed classification. Obes. Res.11(1), 5–16 (2003).
  • Abate, N, Garg A. Heterogeneity in adipose tissue metabolism: causes, implications and management of regional adiposity. Prog. Lipid Res.34(1), 53–70 (1995).
  • Rockall AG, Sohaib SA, Evans D et al. Computed tomography assessment of fat distribution in male and female patients with Cushing’s syndrome. Eur. J. Endocrinol.149(6), 561–567 (2003).
  • Abate N, Burns D, Peshock RM, Garg A, Grundy SM. Estimation of adipose tissue mass by magnetic resonance imaging: validation against dissection in human cadavers. J. Lipid Res.35(8), 1490–1496 (1994).
  • Rockall AG, Sohaib SA, Evans D et al. Hepatic steatosis in Cushing’s syndrome: a radiological assessment using computed tomography. Eur. J. Endocrinol.149(6), 543–548 (2003).
  • Monziols M, Collewet G, Bonneau M, Mariette F, Davenel A, Kouba M. Quantification of muscle, subcutaneous fat and intermuscular fat in pig carcasses and cuts by magnetic resonance imaging. Meat science.72, 146–154 (2006).
  • Klein S, Allison DB, Heymsfield SB et al. Waist circumference and cardiometabolic risk: a Consensus Statement from Shaping America’s Health: Association for Weight Management and Obesity Prevention; NAASO, the Obesity Society; the American Society for Nutrition; and the American Diabetes Association. Obesity15(5), 1061–1067 (2007).
  • Farin HM, Abbasi F, Reaven GM. Comparison of body mass index versus waist circumference with the metabolic changes that increase the risk of cardiovascular disease in insulin-resistant individuals. Am. J. Cardiol.98(8), 1053–1056 (2006).
  • Brown CD, Higgins M, Donato KA et al. Body mass index and the prevalence of hypertension and dyslipidemia. Obes. Res.8(9), 605–619 (2000).
  • DeFronzo RA. Dysfunctional fat cells, lipotoxicity and Type 2 diabetes. Int. J. Clin. Pract. Suppl.143, 9–21 (2004).
  • Lewis GF, Carpentier A, Adeli K, Giacca A. Disordered fat storage and mobilization in the pathogenesis of insulin resistance and Type 2 diabetes. Endocr. Rev.23(2), 201–229 (2002).
  • Savage DB, Petersen KF, Shulman GI. Mechanisms of insulin resistance in humans and possible links with inflammation. Hypertension45(5), 828–833 (2005).
  • de Jongh RT, Serne EH, Ijzerman RG, de Vries G, Stehouwer CD. Free fatty acid levels modulate microvascular function: relevance for obesity-associated insulin resistance, hypertension, and microangiopathy. Diabetes53(11), 2873–2882 (2004).
  • Fagot-Campagna A, Balkau B, Simon D et al. High free fatty acid concentration: an independent risk factor for hypertension in the Paris Prospective Study. Int. J. Epidemiol.27(5), 808–813 (1998).
  • Bays H, Abate N, Chandalia M. Adiposopathy: sick fat causes high blood sugar, high blood pressure, and dyslipidemia. Future Cardiol.1(1), 39–59 (2005).
  • Yu YH, Ginsberg HN. Adipocyte signaling and lipid homeostasis: sequelae of insulin-resistant adipose tissue. Circ. Res.96(10), 1042–1052 (2005).
  • Jazet IM, Pijl H, Meinders AE. Adipose tissue as an endocrine organ: impact on insulin resistance. Neth. J. Med.61(6), 194–212 (2003).
  • Miner JL. The adipocyte as an endocrine cell. J. Anim. Sci.82(3), 935–941 (2004).
  • Ahima RS. Adipose tissue as an endocrine organ. Obesity14(Suppl. 5), 242S–249S (2006).
  • Schaffler A, Muller-Ladner U, Scholmerich J, Buchler C. Role of adipose tissue as an inflammatory organ in human diseases. Endocr. Rev.27(5), 449–467 (2006).
  • Caspar-Bauguil S, Cousin B, Galinier A et al. Adipose tissues as an ancestral immune organ: site-specific change in obesity. FEBS Lett.579(17), 3487–3492 (2005).
  • Fantuzzi G. Adipose tissue, adipokines, and inflammation. J. Allergy Clin. Immunol.115(5), 911–919 (2005).
  • Trayhurn P, Wood S. Adipokines: inflammation and the pleiotropic role of white adipose tissue. Br. J. Nutr.92347–355 (2004).
  • Wellen KE, Hotamisligil GS. Obesity-induced inflammatory changes in adipose tissue. J. Clin. Invest.112(12), 1785–1788 (2003).
  • Wisse BE. The inflammatory syndrome: the role of adipose tissue cytokines in metabolic disorders linked to obesity. J. Am. Soc. Nephrol.15(11), 2792–2800 (2004).
  • Kougias P, Chai H, Lin PH, Yao Q, Lumsden AB, Chen C. Effects of adipocyte-derived cytokines on endothelial functions: implication of vascular disease. J. Surg. Res.126(1), 121–129 (2005).
  • Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW Jr. Obesity is associated with macrophage accumulation in adipose tissue. J. Clin. Invest.112(12), 1796–1808 (2003).
  • Xu H, Barnes GT, Yang Q et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J. Clin. Invest.112(12), 1821–1830 (2003).
  • Bastard JP, Maachi M, Lagathu C et al. Recent advances in the relationship between obesity, inflammation, and insulin resistance. Eur. Cytokine Netw.17(1), 4–12 (2006).
  • Bo S, Gambino R, Pagani A et al. Relationships between human serum resistin, inflammatory markers and insulin resistance. Int. J. Obes. (Lond.)29(11), 1315–1320 (2005).
  • Garanty-Bogacka B, Syrenicz M, Syrenicz A, Gebala A, Lulka D, Walczak M. Serum markers of inflammation and endothelial activation in children with obesity-related hypertension. Neuro. Endocrinol. Lett.26(3), 242–246 (2005).
  • Pittas AG, Joseph NA, Greenberg AS. Adipocytokines and insulin resistance. J. Clin. Endocrinol. Metab.89(2), 447–452 (2004).
  • Lau DC, Dhillon B, Yan H, Szmitko PE, Verma S. Adipokines: molecular links between obesity and atheroslcerosis. Am. J. Physiol. Heart Circ. Physiol.288(5), H2031–H2041 (2005).
  • Fain JN, Bahouth SW, Madan AK. TNFa release by the nonfat cells of human adipose tissue. Int. J. Obes. Relat. Metab. Disord.28(4), 616–622 (2004).
  • Fain JN, Tichansky DS, Madan AK. Most of the interleukin 1 receptor antagonist, cathepsin S, macrophage migration inhibitory factor, nerve growth factor, and interleukin 18 release by explants of human adipose tissue is by the non-fat cells, not by the adipocytes. Metabolism55(8), 1113–1121 (2006).
  • Maeda K, Okubo K, Shimomura I, Mizuno K, Matsuzawa Y, Matsubara K. Analysis of an expression profile of genes in the human adipose tissue. Gene190(2), 227–235 (1997).
  • Bastard JP, Jardel C, Delattre J, Hainque B, Bruckert E, Oberlin F. Evidence for a link between adipose tissue interleukin-6 content and serum C-reactive protein concentrations in obese subjects. Circulation99(16), 2221–2222 (1999).
  • Matsuzawa Y. Adipocytokines: emerging therapeutic targets. Curr. Atheroscler. Rep.7(1), 58–62 (2005).
  • Festa A, D’Agostino R Jr, Tracy RP, Haffner SM. Elevated levels of acute-phase proteins and plasminogen activator inhibitor-1 predict the development of Type 2 diabetes: the insulin resistance atherosclerosis study. Diabetes51(4), 1131–1137 (2002).
  • Bluher M, Fasshauer M, Tonjes A, Kratzsch J, Schon MR, Paschke R. Association of interleukin-6, C-reactive protein, interleukin-10 and adiponectin plasma concentrations with measures of obesity, insulin sensitivity and glucose metabolism. Exp. Clin. Endocrinol. Diabetes113(9), 534–537 (2005).
  • Forouhi NG, Sattar N, McKeigue PM. Relation of C-reactive protein to body fat distribution and features of the metabolic syndrome in Europeans and South Asians. Int. J. Obes. Relat. Metab. Disord.25(9), 1327–1331 (2001).
  • Ronti T, Lupattelli G, Mannarino E. The endocrine function of adipose tissue: an update. Clin. Endocrinol. (Oxf.)64(4), 355–365 (2006).
  • Bahceci M, Gokalp D, Bahceci S, Tuzcu A, Atmaca S, Arikan S. The correlation between adiposity and adiponectin, tumor necrosis factor a, interleukin-6 and high sensitivity C-reactive protein levels. Is adipocyte size associated with inflammation in adults? J. Endocrinol. Invest.30(3), 210–214 (2007).
  • Zvonic S, Baugh JE Jr, Arbour-Reily P, Mynatt RL, Stephens JM. Cross-talk among gp130 cytokines in adipocytes. J. Biol. Chem.280(40), 33856–33863 (2005).
  • Schling, P, Loffler G. Cross talk between adipose tissue cells: impact on pathophysiology. News Physiol. Sci.1799–104 (2002).
  • Costa JL, Hochgeschwender U, Brennan M. The role of melanocyte-stimulating hormone in insulin resistance and Type 2 diabetes mellitus. Treat. Endocrinol.5(1), 7–13 (2006).
  • Sell H, Dietze-Schroeder D, Eckel J. The adipocyte–myocyte axis in insulin resistance. Trends Endocrinol. Metab.17(10), 416–422 (2006).
  • Bartness TJ, Kay, Song C, Shi H, Bowers RR, Foster MT. Brain-adipose tissue cross talk. Proc. Nutr. Soc.64(1), 53–64 (2005).
  • Argiles JM, Lopez-Soriano J, Almendro V, Busquets S, Lopez-Soriano FJ. Cross-talk between skeletal muscle and adipose tissue: a link with obesity? Med. Res. Rev.25(1), 49–65 (2005).
  • Samec S, Seydoux J, Dulloo AG. Interorgan signaling between adipose tissue metabolism and skeletal muscle uncoupling protein homologs: is there a role for circulating free fatty acids? Diabetes47(11), 1693–1698 (1998).
  • Plomgaard P, Bouzakri K, Krogh-Madsen R, Mittendorfer B, Zierath JR, Pedersen BK. Tumor necrosis factor-a induces skeletal muscle insulin resistance in healthy human subjects via inhibition of Akt substrate 160 phosphorylation. Diabetes54(10), 2939–2945 (2005).
  • Storz P, Doppler H, Wernig A, Pfizenmaier K, Muller G. Cross-talk mechanisms in the development of insulin resistance of skeletal muscle cells palmitate rather than tumour necrosis factor inhibits insulin-dependent protein kinase B (PKB)/Akt stimulation and glucose uptake. Eur. J. Biochem.266(1), 17–25 (1999).
  • Alpert MA, Fraley MA, Birchem JA, Senkottaiyan N. Management of obesity cardiomyopathy. Expert Rev. Cardiovasc. Ther.3(2), 225–230 (2005).
  • Berg AH, Scherer PE. Adipose tissue, inflammation, and cardiovascular disease. Circ. Res.96(9), 939–949 (2005).
  • Fruhbeck G. The adipose tissue as a source of vasoactive factors. Curr. Med. Chem. Cardiovasc. Hematol. Agents2(3), 197–208 (2004).
  • Matsuzawa Y. Therapy Insight: adipocytokines in metabolic syndrome and related cardiovascular disease. Nat. Clin. Pract.3(1), 35–42 (2006).
  • Orshal JM, Khalil RA. Interleukin-6 impairs endothelium-dependent NO-cGMP-mediated relaxation and enhances contraction in systemic vessels of pregnant rats. Am. J. Physiol. Regul. Integr. Comp. Physiol.286(6), R1013–R1023 (2004).
  • Paquot N, Tappy L. Adipocytolines: link between obesity, Type 2 diabetes and atherosclerosis. Rev. Med. Liege60(5–6), 369–373 (2005).
  • Bays H. Adiposopathy: role of adipocyte factors in a new paradigm. Expert Rev. Cardiovasc.Ther.3(2), 187–189 (2005).
  • Uno K, Katagiri H, Yamada T et al. Neuronal pathway from the liver modulates energy expenditure and systemic insulin sensitivity. Science312(5780), 1656–1659 (2006).
  • Schaffler A, Scholmerich J, Buchler C. Mechanisms of disease: adipocytokines and visceral adipose tissue – emerging role in nonalcoholic fatty liver disease. Nat. Clin. Pract. Gastroenterol. Hepatol.2(6), 273–280 (2005).
  • Schaffler A, Scholmerich J, Buchler C. Mechanisms of disease: adipocytokines and visceral adipose tissue – emerging role in intestinal and mesenteric diseases. Nat. Clin. Pract. Gastroenterol. Hepatol.2(2), 103–111 (2005).
  • Rossi GP, Sticchi D, Giuliani L et al. Adiponectin receptor expression in the human adrenal cortex and aldosterone-producing adenomas. Int. J. Mol. Med.17(6), 975–980 (2006).
  • Ghizzoni L, Mastorakos G, Ziveri M et al. Interactions of leptin and thyrotropin 24-hour secretory profiles in short normal children. J. Clin. Endocrinol. Metab.86(5), 2065–2072 (2001).
  • Porte D Jr. Central regulation of energy homeostasis. Diabetes55(Suppl. 2), S155–S160 (2006).
  • Perez-Tilve D, Stern JE, Tschop M. The brain and the metabolic syndrome: not a wireless connection. Endocrinology147(3), 1136–1139 (2006).
  • Egan BM. Insulin resistance and the sympathetic nervous system. Curr. Hypertens. Rep.5(3), 247–254 (2003).
  • Song CK, Jackson RM, Harris RB, Richard D, Bartness TJ. Melanocortin-4 receptor mRNA is expressed in sympathetic nervous system outflow neurons to white adipose tissue. Am. J. Physiol. Regul. Integr. Comp. Physiol.289(5), R1467–R1476 (2005).
  • Bays H. The melanocortin system as a therapeutic treatment target for adiposity and adiposopathy. Drugs RD7(5), 289–302 (2006).
  • Kalra SP, Dube MG, Pu S, Xu B, Horvath TL, Kalra PS. Interacting appetite-regulating pathways in the hypothalamic regulation of body weight. Endocr. Rev.20(1), 68–100 (1999).
  • Konturek PC, Konturek JW, Czesnikiewicz-Guzik M, Brzozowski T, Sito E, Konturek PC. Neuro-hormonal control of food intake; basic mechanisms and clinical implications. J. Physiol. Pharmacol.56(Suppl. 6), 5–25 (2005).
  • Schwartz MW, Woods SC, Porte D Jr, Seeley RJ, Baskin DG. Central nervous system control of food intake. Nature404(6778), 661–671 (2000).
  • Baskin DG, Hahn TM, Schwartz MW. Leptin sensitive neurons in the hypothalamus. Horm. Metab. Res.31(5), 345–350 (1999).
  • Benoit SC, Air EL, Coolen LM et al. The catabolic action of insulin in the brain is mediated by melanocortins. J. Neurosci.22(20), 9048–9052 (2002).
  • Breen TL, Conwell IM, Wardlaw SL. Effects of fasting, leptin, and insulin on AGRP and POMC peptide release in the hypothalamus. Brain Res.1032(1–2), 141–148 (2005).
  • Bruning JC, Gautam D, Burks DJ et al. Role of brain insulin receptor in control of body weight and reproduction. Science289(5487), 2122–2125 (2000).
  • Havel PJ. Role of adipose tissue in body-weight regulation: mechanisms regulating leptin production and energy balance. Proc. Nutr. Soc.59(3), 359–371 (2000).
  • Havrankova J, Brownstein M, Roth J. Insulin and insulin receptors in rodent brain. Diabetologia20(Suppl.), 268–273 (1981).
  • Liu J, da Silva AA, Tallam LS, Hall JE. Chronic central nervous system hyperinsulinemia and regulation of arterial pressure and food intake. J. Hypertens.24(7), 1391–1395 (2006).
  • Obici S, Feng Z, Karkanias G, Baskin DG, Rossetti L. Decreasing hypothalamic insulin receptors causes hyperphagia and insulin resistance in rats. Nat. Neurosci.5(6), 566–572 (2002).
  • Plum L, Belgardt BF, Bruning JC. Central insulin action in energy and glucose homeostasis. J. Clin. Invest.116(7), 1761–1766 (2006).
  • Buono P, Pasanisi F, Nardelli C et al. Six novel mutations in the proopiomelanocortin and melanocortin receptor 4 genes in severely obese adults living in southern Italy. Clin. Chem.51(8), 1358–1364 (2005).
  • Li G, Mobbs CV, Scarpace PJ. Central pro-opiomelanocortin gene delivery results in hypophagia, reduced visceral adiposity, and improved insulin sensitivity in genetically obese Zucker rats. Diabetes52(8), 1951–1957 (2003).
  • Adan RA, Kas MJ. Inverse agonism gains weight. Trends Pharmacol. Sci.24(6), 315–321 (2003).
  • Cowley MA, Pronchuk N, Fan W, Dinulescu DM, Colmers WF, Cone RD. Integration of NPY, AGRP, and melanocortin signals in the hypothalamic paraventricular nucleus: evidence of a cellular basis for the adipostat. Neuron24(1), 155–163 (1999).
  • Hwa JJ, Witten MB, Williams P et al. Activation of the NPY Y5 receptor regulates both feeding and energy expenditure. Am. J. Physiol.277(5 Pt 2), R1428–R1434 (1999).
  • Marsh DJ, Hollopeter G, Kafer KE, Palmiter RD. Role of the Y5 neuropeptide Y receptor in feeding and obesity. Nat. Med.4(6), 718–721 (1998).
  • Palmiter RD, Erickson JC, Hollopeter G, Baraban SC, Schwartz MW. Life without neuropeptide Y. Recent Prog. Horm. Res.53, 163–199 (1998).
  • Govaerts C, Srinivasan S, Shapiro A et al. Obesity-associated mutations in the melanocortin 4 receptor provide novel insights into its function. Peptides26(10), 1909–1919 (2005).
  • la-Fera MA, Baile CA. Roles for melanocortins and leptin in adipose tissue apoptosis and fat deposition. Peptides26(10), 1782–1787 (2005).
  • MacKenzie RG. Obesity-associated mutations in the human melanocortin-4 receptor gene. Peptides27(2), 395–403 (2006).
  • Obici S, Feng Z, Tan J, Liu L, Karkanias G, Rossetti L. Central melanocortin receptors regulate insulin action. J. Clin. Invest.108(7), 1079–1085 (2001).
  • Savontaus E, Breen TL, Kim A, Yang LM, Chua SC Jr, Wardlaw SL. Metabolic effects of transgenic melanocyte-stimulating hormone overexpression in lean and obese mice. Endocrinology145(8), 3881–3891 (2004).
  • Yeo GS, Farooqi IS, Challis BG, Jackson RS, O’Rahilly S. The role of melanocortin signalling in the control of body weight: evidence from human and murine genetic models. QJM93(1), 7–14 (2000).
  • Joseph-Bravo P. Hypophysiotropic thyrotropin-releasing hormone neurons as transducers of energy homeostasis. Endocrinology145(11), 4813–4815 (2004).
  • Richard D, Huang Q, Timofeeva E. The corticotropin-releasing hormone system in the regulation of energy balance in obesity. Int. J. Obes. Relat. Metab. Disord.24(Suppl. 2), S36–S39 (2000).
  • Bensaid M, Gary-Bobo M, Esclangon A et al. The cannabinoid CB1 receptor antagonist SR141716 increases Acrp30 mRNA expression in adipose tissue of obese fa/fa rats and in cultured adipocyte cells. Mol. Pharmacol.63(4), 908–914 (2003).
  • Cota D, Marsicano G, Tschop M et al. The endogenous cannabinoid system affects energy balance via central orexigenic drive and peripheral lipogenesis. J. Clin. Invest.112(3), 423–431 (2003).
  • Huffman JW. CB2 receptor ligands. Mini Rev. Med. Chem.5(7), 641–649 (2005).
  • Lichtman AH, Cravatt BF. Food for thought: endocannabinoid modulation of lipogenesis. J. Clin. Invest.115(5), 1130–1133 (2005).
  • Osei-Hyiaman D, Harvey-White J, Batkai S, Kunos G. The role of the endocannabinoid system in the control of energy homeostasis. Int. J. Obes. (Lond.)30(Suppl. 1), S33–S38 (2006).
  • Pagotto U, Vicennati V, Pasquali R. The endocannabinoid system and the treatment of obesity. Ann. Med.37(4), 270–275 (2005).
  • Pagotto U, Marsicano G, Cota D, Lutz B, Pasquali R. The emerging role of the endocannabinoid system in endocrine regulation and energy balance. Endocr. Rev.27(1), 73–100 (2006).
  • Pagotto U, Cervino C, Vicennati V, Marsicano G, Lutz B, Pasquali R. How many sites of action for endocannabinoids to control energy metabolism? Int. J. Obes. (Lond.)30(Suppl. 1), S39–S43 (2006).
  • Roche R, Hoareau L, Bes-Houtmann S et al. Presence of the cannabinoid receptors, CB1 and CB2, in human omental and subcutaneous adipocytes. Histochem. Cell Biol.126(2), 177–187 (2006).
  • Tomas F, Kelly M, Xiang X et al. Metabolic and hormonal interactions between muscle and adipose tissue. Proc. Nutr. Soc.63(2), 381–385 (2004).
  • Batterham RL, Cowley MA, Small CJ et al. Gut hormone PYY(3–36) physiologically inhibits food intake. Nature418(6898), 650–654 (2002).
  • Ellacott KL, Halatchev IG, Cone RD. Interactions between gut peptides and the central melanocortin system in the regulation of energy homeostasis. Peptides27(2), 340–349 (2006).
  • Storlien L, Oakes ND, Kelley DE. Metabolic flexibility. Proc. Nutr. Soc.63(2), 363–368 (2004).
  • Adams JM, Pratipanawatr T, Berria R et al. Ceramide content is increased in skeletal muscle from obese insulin-resistant humans. Diabetes53(1), 25–31 (2004).
  • Chavez JA, Holland WL, Bar J, Sandhoff K, Summers SA. Acid ceramidase overexpression prevents the inhibitory effects of saturated fatty acids on insulin signaling. J. Biol. Chem.280(20), 20148–20153 (2005).
  • Kelley DE, Goodpaster BH. Skeletal muscle triglyceride. An aspect of regional adiposity and insulin resistance. Diabetes Care24(5), 933–941 (2001).
  • Kelley DE, Mandarino LJ. Fuel selection in human skeletal muscle in insulin resistance: a re-examination. Diabetes49, 677–683 (2000).
  • Cha BS, Ciaraldi TP, Park KS, Carter L, Mudaliar SR, Henry RR. Impaired fatty acid metabolism in Type 2 diabetic skeletal muscle cells is reversed by PPARgγ agonists. Am. J. Physiol. Endocrinol. Metab.289(1), E151–E159 (2005).
  • Grill V, Persson G, Carlsson S et al. Family history of diabetes in middle-aged Swedish men is a gender unrelated factor which associates with insulinopenia in newly diagnosed diabetic subjects. Diabetologia42(1), 15–23 (1999).
  • Petersen KF, Befroy D, Dufour S et al. Mitochondrial dysfunction in the elderly: possible role in insulin resistance. Science300(5622), 1140–1142 (2003).
  • Wolk R, Shamsuzzaman AS, Somers VK. Obesity, sleep apnea, and hypertension. Hypertension42(6), 1067–1074 (2003).
  • Zhang R, Reisin E. Obesity-hypertension: the effects on cardiovascular and renal systems. Am. J. Hypertens.13(12), 1308–1314 (2000).
  • McGavock JM, Victor RG, Unger RH, Szczepaniak LS. Adiposity of the heart, revisited. Ann. Intern. Med.144(7), 517–524 (4–4-2006).
  • Bays H. Adiposopathy, metabolic syndrome, quantum physics, general relativity, chaos and the theory of everything. Expert Rev. Cardiovasc.Ther.3(3), 393–404 (2005).
  • Kim SH, Reaven GM. The metabolic syndrome: one step forward, two steps back. Diab. Vasc. Dis. Res.1(2), 68–75 (2004).
  • Reaven GM. The metabolic syndrome: is this diagnosis necessary? Am. J. Clin. Nutr.83(6), 1237–1247 (2006).
  • Reaven G. The metabolic syndrome or the insulin resistance syndrome? Different names, different concepts, and different goals. Endocrinol. Metab. Clin. North Am.33(2), 283–303 (2004).
  • Stern MP, Williams K, Gonzalez-Villalpando C, Hunt KJ, Haffner SM. Does the metabolic syndrome improve identification of individuals at risk of Type 2 diabetes and/or cardiovascular disease? Diabetes Care27(11), 2676–2681 (2004).
  • Goldfarb B. ADA/EASD statement casts critical eye on metabolic syndrome. DOC News2(10), 1 (2006).
  • Iribarren C, Go AS, Husson G et al. Metabolic syndrome and early-onset coronary artery disease: is the whole greater than its parts? J. Am. Coll. Cardiol.48(9), 1800–1807 (2006).
  • Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III) final report. Circulation106(25), 3143–3421 (2002).
  • Alberti KG, Zimmet PZ. Definition, diagnosis and classification of diabetes mellitus and its complications. Part 1: diagnosis and classification of diabetes mellitus provisional report of a WHO consultation. Diabet. Med.15(7), 539–553 (1998).
  • Kahn R, Buse J, Ferrannini E, Stern M. The metabolic syndrome: time for a critical appraisal: joint statement from the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care28(9), 2289–2304 (2005).
  • Grundy SM. Does the metabolic syndrome exist? Diabetes Care29(7), 1689–1692 (2006).
  • Grundy SM. Does a diagnosis of metabolic syndrome have value in clinical practice? Am. J. Clin. Nutr.83(6), 1248–1251 (2006).
  • Davidson MB. Does the metabolic syndrome exist: response to Grundy. Diabetes Care29(11), 2565–2566 (2006).
  • Bays H, Chapman R, Klingman D, Fanning K, Grandy S. High prevalence of misdiagnosis of the metabolic syndrome in a self-reported survey: possible confusion with having “a metabolism problem”. NAASO 2006 Annual Meeting.Boston Massachusetts USA. October 22, 2006 (Abstract 605).
  • Pladevall M, Singal B, Williams LK et al. A single factor underlies the metabolic syndrome: a confirmatory factor analysis. Diabetes Care29(1), 113–122 (2006).
  • Lebovitz HE. Insulin resistance – a common link between Type 2 diabetes and cardiovascular disease. Diabetes Obes. Metab.8(3), 237–249 (2006).
  • Stern MP. Diabetes and cardiovascular disease. The “common soil” hypothesis. Diabetes44(4), 369–374 (1995).
  • Bays H. Adiposopathy: the endocannabinoid system as a therapeutic treatment target for dysfunctional “sick” fat. CJHP19(1), 32–39 (2007).
  • Bays H. Adiposopathy - Defining, diagnosing, and establishing indications to treat “sick fat”: what are the regulatory considerations? US Endocrine Dis.2, 12–14 (2006).
  • Bays HE. Current and investigational antiobesity agents and obesity therapeutic treatment targets. Obes. Res.12(8), 1197–1211 (2004).
  • Iwen KA, Perwitz N, Kraus D, Fasshauer M, Klein J. Putting fat cells onto the road map to novel therapeutic strategies. Discov. Med.6(32), 75–81 (2006).
  • Golay A, Ybarra J. Link between obesity and Type 2 diabetes. Best Pract. Res. Clin. Endocrinol. Metab.19(4), 649–663 (2005).
  • Shafrir E. Development and consequences of insulin resistance: lessons from animals with hyperinsulinaemia. Diabetes Metab.22(2), 122–131 (1996).
  • Kannisto K, Pietilainen KH, Ehrenborg E et al. Overexpression of 11β-hydroxysteroid dehydrogenase-1 in adipose tissue is associated with acquired obesity and features of insulin resistance: studies in young adult monozygotic twins. J. Clin. Endocrinol. Metab.89(9), 4414–4421 (2004).
  • Wake DJ, Rask E, Livingstone DE, Soderberg S, Olsson T, Walker BR. Local and systemic impact of transcriptional up-regulation of 11β-hydroxysteroid dehydrogenase type 1 in adipose tissue in human obesity. J. Clin. Endocrinol. Metab.88(8), 3983–3988 (2003).
  • Walker BR. 11β-hydroxysteroid dehydrogenase type 1 in obesity. Obes. Res.12(1), 1–3 (2004).
  • Putignano P, Pecori, Giraldi F, Cavagnini F. Tissue-specific dysregulation of 11β-hydroxysteroid dehydrogenase type 1 and pathogenesis of the metabolic syndrome. J. Endocrinol. Invest.27(10), 969–974 (2004).
  • Wolf G. Glucocorticoids in adipocytes stimulate visceral obesity. Nutr. Rev.60(5 Pt 1), 148–151 (2002).
  • Masuzaki, H, Flier JS. Tissue-specific glucocorticoid reactivating enzyme, 11 β-hydroxysteroid dehydrogenase type 1 (11 β-HSD1) – a promising drug target for the treatment of metabolic syndrome. Curr. Drug Targets. Immune Endocr.Metabol. Disord.3(4), 255–262 (2003).
  • Fujimoto WY, Jablonski KA, Bray GA et al. Body size and shape changes and the risk of diabetes in the diabetes prevention program. Diabetes56(6), 1680–1685 (2007).
  • Pi-Sunyer X, Blackburn G, Brancati FL et al. Reduction in weight and cardiovascular disease risk factors in individuals with Type 2 diabetes: one-year results of the look AHEAD trial. Diabetes Care30(6), 1374–1383 (2007).
  • Anderson JW, Kendall CW, Jenkins DJ. Importance of weight management in Type 2 diabetes: review with meta-analysis of clinical studies. J. Am. Coll. Nutr.22(5), 331–339 (2003).
  • Henry RR, Gumbiner B. Benefits and limitations of very-low-calorie diet therapy in obese NIDDM. Diabetes Care14(9), 802–823 (1991).
  • Tsai AG, Wadden TA. Systematic review: an evaluation of major commercial weight loss programs in the United States. Ann. Intern. Med.142(1), 56–66 (2005).
  • Wadden TA, Berkowitz RI, Womble LG et al. Randomized trial of lifestyle modification and pharmacotherapy for obesity. N. Engl. J. Med.353(20), 2111–2120 (2005).
  • Gardner CD, Kiazand A, Alhassan S et al. Comparison of the Atkins, Zone, Ornish, and LEARN diets for change in weight and related risk factors among overweight premenopausal women: the A TO Z Weight Loss Study: a randomized trial. JAMA297(9), 969–977 (2007).
  • Viguerie N, Vidal H, Arner P et al. Adipose tissue gene expression in obese subjects during low-fat and high-fat hypocaloric diets. Diabetologia48(1), 123–131 (2005).
  • Hamman RF, Wing RR, Edelstein SL et al. Effect of weight loss with lifestyle intervention on risk of diabetes. Diabetes Care29, 2102–2107 (2006).
  • Diaz VA, Player MS, Mainous AG 3rd, Carek PJ, Geesey ME. Competing impact of excess weight versus cardiorespiratory fitness on cardiovascular risk. Am. J. Cardiol.98(11), 1468–1471 (2006).
  • Larson-Meyer DE, Heilbronn LK, Redman LM et al. Effect of calorie restriction with or without exercise on insulin sensitivity, β-cell function, fat cell size, and ectopic lipid in overweight subjects. Diabetes Care29(6), 1337–1344 (2006).
  • Norris SL, Zhang X, Avenell A et al. Long-term effectiveness of lifestyle and behavioral weight loss interventions in adults with Type 2 diabetes: a meta-analysis. Am. J. Med.117(10), 762–774 (2004).
  • Aucott L, Poobalan A, Smith WC et al. Weight loss in obese diabetic and non-diabetic individuals and long-term diabetes outcomes – a systematic review. Diabetes Obes. Metab.6(2), 85–94 (2004).
  • Boden G, Sargrad K, Homko C, Mozzoli M, Stein TP. Effect of a low-carbohydrate diet on appetite, blood glucose levels, and insulin resistance in obese patients with Type 2 diabetes. Ann. Intern. Med.142(6), 403–411 (2005).
  • Bruun JM, Helge JW, Richelsen B, Stallknecht B. Diet and exercise reduce low-grade inflammation and macrophage infiltration in adipose tissue but not in skeletal muscle in severely obese subjects. Am. J. Physiol. Endocrinol. Metab. (2005).
  • Straznicky NE, Louis WJ, McGrade P, Howes LG. The effects of dietary lipid modification on blood pressure, cardiovascular reactivity and sympathetic activity in man. J. Hypertens.11(4), 427–437 (1993).
  • Aucott L, Poobalan A, Smith WC, Avenell A, Jung R, Broom J. Effects of weight loss in overweight/obese individuals and long-term hypertension outcomes: a systematic review. Hypertension45(6), 1035–1041 (2005).
  • Stone NJ, Kushner R. Effects of dietary modification and treatment of obesity. Emphasis on improving vascular outcomes. Med. Clin. North Am.84(1), 95–122 (2000).
  • Chen AK, Roberts CK, Barnard RJ. Effect of a short-term diet and exercise intervention on metabolic syndrome in overweight children. Metabolism55(7), 871–878 (2006).
  • Bassuk SS, Manson JE. Epidemiological evidence for the role of physical activity in reducing risk of Type 2 diabetes and cardiovascular disease. J. Appl. Physiol.99(3), 1193–1204 (2005).
  • Astrup A, Finer N. Redefining Type 2 diabetes: ‘diabesity’ or ‘obesity dependent diabetes mellitus’? Obes. Rev.1(2), 57–59 (2000).
  • Avenell A, Brown TJ, McGee MA et al. What are the long-term benefits of weight reducing diets in adults? A systematic review of randomized controlled trials. J. Hum. Nutr. Diet17(4), 317–335 (2004).
  • Deeg MA, Buse JB, Goldberg RB et al. Pioglitazone and rosiglitazone have different effects on serum lipoprotein particle concentrations and sizes in patients with Type 2 diabetes and dyslipidemia. Diabetes Care (2007).
  • Chilton R, Chiquette E. Thiazolidinediones and cardiovascular disease. Curr. Atheroscler. Rep.7(2), 115–120 (2005).
  • Bosch J, Yusuf S, Gerstein HC Wareham NJ. Results of the DREAM trial (Diabetes Reduction Assessment with Ramipril and Rosiglitazone Medication). Presented at: European Association for the Study of Diabetes Annual Meeting. Copenhagen, Denmark, September 14–17, 2006.
  • Clasen R, Schupp M, Foryst-Ludwig A et al. PPARγ-activating angiotensin type-1 receptor blockers induce adiponectin. Hypertension46(1), 137–143 (2005).
  • Erol A. The role of fat tissue in the cholesterol lowering and the pleiotropic effects of statins – statins activate the generation of metabolically more capable adipocytes. Med. Hypotheses64(1), 69–73 (2005).
  • Bray GA. The underlying basis for obesity: relationship to cancer. J. Nutr.132(11 Suppl.), S3451–S3455 (2002).
  • Franks S. Polycystic ovary syndrome. N. Engl. J. Med.333(13), 853–861 (1995).
  • Silfen ME, Denburg MR, Manibo AM et al. Early endocrine, metabolic, and sonographic characteristics of polycystic ovary syndrome (PCOS): comparison between nonobese and obese adolescents. J. Clin. Endocrinol. Metab.88(10), 4682–4688 (2003).
  • Mai K, Bobbert T, Kullmann V et al. Free fatty acids increase androgen precursors in vivo.J. Clin. Endocrinol. Metab.91(4), 1501–1507 (2006).

Websites

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.