Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Jan 1.
Published in final edited form as: Metabolism. 2015 Oct 23;65(1):73–80. doi: 10.1016/j.metabol.2015.10.019

Metabolic Health and Weight: Understanding metabolically unhealthy normal weight or metabolically healthy obese patients

Hannah Mathew 1,2, Olivia M Farr 2,3,*, Christos S Mantzoros 2,3
PMCID: PMC4750380  NIHMSID: NIHMS757071  PMID: 26683798

Obesity is most commonly defined as a BMI of over 30kg/m2. Typical classification is into categories of Class I (BMI >30 kg/m2), Class II (BMI equal to or over 35 kg/m2) and Class III (equal to or over 40 kg/m2), with the latter also known as severe obesity. While this method is most frequently utilized by clinicians, it has limitations-such as in those individuals with high muscle to fat ratios or those of Asian descent. Alternate methods for obesity definition and classification include data such as waist circumference, hip to waist ratio, or body fat percentage.

Using the BMI criteria for obesity, over 600 million people worldwide, including one third of adults in the United States meet criteria for obesity [1]. In the United States, this translates to mounting healthcare costs (estimated close to 128 billion dollars in 2008) and increased mortality compared to normal weight individuals [2]. Cardiovascular disease and secondarily malignancies has long been identified as the primary reason for these increases in mortality and costs. This in turn has been attributed to a worse metabolic profile which includes various combinations of impaired glucose tolerance/type 2 diabetes, dyslipidemia, hypertension and systemic inflammation.

However, hidden amongst traditional obesity related concerns, there lies a subset of patients without the expected sequelae of their weight. These patients circumvented the classic models of metabolic and cardiovascular risk, and are known as the “metabolically healthy obese” (MHO). Simultaneously, there are individuals who despite having “normal” weights, shoulder an increased burden of these risks. Accurate classifications and mechanistic understandings for individuals with these conditions would be required to ensure the best health care and appropriate treatments as well as to decrease health care costs due to improper treatments and requisite subsequent medical interventions. Here we discuss the current knowledge surrounding these two groups, and highlight important features for provider management.

Historically, the primary concern regarding obesity was due to the concurrent metabolic and cardiovascular risk. Yet, in recent years increased notice has been made of those individuals who do not fit into this traditional phenotype. Instead, metabolically healthy obese (MHO) and metabolically unhealthy normal weight (MUHNW) patients are generating important discussions regarding the classification of metabolic, and thus cardiovascular risk in patients. These cohorts have been previously highlighted in Metabolism, with discussions in recent years ranging from the association with diabetes, liver enzymes and vitamin D to the role of weight status and inflammation [39]. Here we discuss these two phenotypes, highlight current knowledge regarding their classification, development and management features for healthcare providers.

Presently, beyond cutoffs for surgical intervention, obesity guidelines do not distinguish between management of the various subclasses of obesity despite the fact that there has not been evidence for increased mortality in simple Class II Obesity [10]. In fact, while studies have shown that individuals with Grade II-III have greater mortality, there is evidence that Class I obesity patients may have lower all-cause mortality than normal weight patients [11]. Furthermore, existing guidelines also fail to individualize the management of MHO or metabolically unhealthy/abnormal obese (MUHO/MAO) patients. This is further complicated by a gap in the recognition and appropriate management of those normal weight individuals, who demonstrate high risk metabolic risk profiles.

Here we highlight these subtypes of obesity and metabolic profiles for providers, as well as ongoing research in the field.

Metabolically Healthy Obese (MHO)

Since 1982, there has been recognition of a group of patients who, despite meeting traditional BMI criteria for obesity, do not demonstrate high risk metabolic profiles. These individuals have been deemed as the “metabolically healthy obese” (MHO) [1214]. Broadly, this categorization is described as an absence of metabolic disorders such as insulin resistance, type 2 diabetes, dyslipidemia and hypertension in those patients with BMIs greater than 30kg/m2[15]. More specific classification schemes vary by study/research group, with variable cutoffs for blood pressure, and cholesterol (HDL, LDL, TC, TG or TG/HDL ratios) [1618]. Further conflict is introduced by variable definitions for insulin resistance, which can include fasting plasma glucose (FPG), hemoglobin A1c (HbA1c), and/or homeostasis model assessment (HOMA). For instance, one study showed that over 30% of patients were not correctly diagnosed with impaired glucose tolerance/T2DM when fasting plasma glucose was used as the primary criteria [19]. Thus, clearer and more consistent criteria are needed to determine whether an individual is MHO.

Metabolically Unhealthy Normal Weight (MUHNW)/Metabolically Obese Normal Weight (MONW)

In contrast to those metabolically healthy obese, there are also of subsets of patients who are considered “normal/healthy” weight, but demonstrate increased metabolic/cardiovascular risk. However, these patients have been even harder to define or characterize than the aforementioned group. First suggested by Ruderman in the 1980s, these individuals were described as hyperinsulinemic, insulin resistant, hypertriglyceremic and predisposed to subsequent development of type 2 diabetes mellitus and coronary artery disease [20, 21]. Broadly, these patients have been categorized by body mass indices of less than 25 kg/m2, but have metabolic abnormalities more commonly associated with their obese counterparts, including abdominal fat distribution and elevated blood pressure. Most studies set the cutoff as three or more metabolic derangements to fulfill “metabolic unhealthy” definition. More recently, Lee et al. [22] proposed utilizing the TyG index-a product of the fasting blood glucose and triglyceride levels to identify patients who are MONW.

Classification is further complicated by the limitations associated with utilizing BMI in definitions. The metabolically abnormal phenotype has been associated with increased waist circumference and body fat percentage in normal weight individuals, which may not be picked up with standard BMI measurements [23].

In coming years, it will be increasingly important to identify consistent criteria for these metabolic states/body phenotypes. With the ability to accurately and precisely classify these patients, healthcare providers will be better able to assess prevalence and study the predisposing factors for metabolic disease-and target therapies accordingly.

Prevalence

Given the inconsistency of MHO definitions, there is high degree of variability surrounding the estimated prevalence of this phenotype. In 2010, one analysis found that the prevalence of MHO varied from 3.3–32.1% in men and 12.2 to 57.5% in women, largely depending on which criteria for MHO patients was applied [18]. Another study found a prevalence of 53.7% for MHO amongst overweight adults when classified by visceral-to-subcutaneous fat ratio and this further related to lipoprotein subfraction analyses particularly for small dense LDL particles [7]. A study coming out of Korea found that the prevalence was 14.9% in the entire or 47.7% amongst obese individuals [4]. More recently, a systematic review by Rey-Lopez et al.[24] evaluated 27 prospective studies and found that prevalence ranged from six to 75%, depending on classification scheme used. The prevalence of MHO varies widely depending on how it has been defined, yet again underscoring the importance of establishing a clear definition and criteria.

Without one standardized definition, however, the true prevalence of the MUHNW phenotype is difficult to quantify. Using criteria of two or more metabolic abnormalities, Wildman et al. [25] reviewed NHANES data from 1999–2004 to find that 23.5% of normal weight adults were metabolically abnormal. Similarly, a Korean study, using data from the third national Korean National Health and Nutrition survey, found a prevalence of 8.7% for the MUHNW phenotype [26]. There is some evidence that race may contribute to MUHNW, as for certain races, central adiposity is high despite low overall BMI. One study has shown that Asian Americans have a 5× prevalence of being lean as compared to obese with diabetes mellitus, suggesting that they are more likely to be MUHNW than typically obese [27]. African and Latino Americans had the highest overall prevalence of lean body weight with diabetes in the same study, suggesting higher overall levels of MUHNW, although they showed similar rates with obesity and diabetes, suggesting higher levels of metabolic dysfunction regardless of body weight [27]. These differences are likely due to differences in central adiposity. For instance, lean (defined by BMI) Asian Indians show lower insulin sensitivity than other races of Asian descent but also higher body fat percentage and waist circumference [28]. Similarly, lean Chinese participants have NAFLD which also correlates with higher waist circumference and poorer metabolic outcomes such as blood glucose, blood pressure, and insulin levels [29]. One should keep in mind that for a given BMI, Chinese subjects tend to accumulate fat intra-abdominally and thus even lean subjects may have ectopic fat deposition and intra-abdominal obesity. With varying prevalence, it becomes more difficult to identify at risk populations and subsequently dedicate research into the genetic and lifestyle factors that contribute to their presentations.

Genetic Factors

Obesity as a whole has and continues to undergo extensive study into the underlying genetic mechanisms contributing towards its development. Notable findings have been monogenic obesity presentations secondary to disruptions of the POMC, leptin and MCR4 loci [30] in addition to minor contributions by several other genes [31]. In general, overall obesity is a polygenic disorder and gene-environment interactions play a very important role. The genetic and pathophysiologic development of metabolically healthy obesity remains an area of continued study. As suggested before, MHO patients are noted to have less central and visceral obesity. These studies have also alluded to reduced adipocyte hypertrophy, fibrosis and stress as potential contributors to this presentation.

Implicated genes include those involved in transcription related to adipogenesis [32]. With the knowledge that white adipose tissue plays a role both lipid and glucose regulation, Das et al. [33] looked to identify difference in RNA transcripts of MHO and MAO patients. They found that those patients with metabolic derangement had upregulation of 141 genes and downregulation of 17 genes. Highlighted differences included the upregulation of matrix metallopeptidase 9 (MMP 9 and osteopontin (SPP1), as well as downregulation of NDRG4 (N-myc downstream-regulated gene family member 4) and GINS3 (GINS complex subunit 3) [33]. Another study revealed that MHO as compared to metabolically unhealthy obese patients had differential gene expression for amino acids and for branched-chain amino acid catabolism in subcutaneous adipose tissue [34]. Particularly, levels of glutamic acid, valine and isoleucine correlated with HOMA-IR where MHO had less expression and lower HOMA-IR [34]. Further underscoring the role in genetic variation within adipogenesis in MUHNW patients, Yaghootkar et al. looked to compare genetic risk scores against disease outcomes, and were able to identify 11 genetic variants/risk alleles which were found to lead to increased hypertension, type 2 diabetes, and coronary artery disease. The identified loci were primarily associated with transcription factors involved in adipogenesis [35]. With specific focus on the development of T2DM, Scott et al. tested associations between genetic risk scores and incident diabetes, finding that a genetic score for insulin resistance mirrored monogenic models for lipodystrophy, and was predictive, regardless of weight status [36]. Additionally, adipocyte storage capacity has been a complementary avenue for study, as this may hold the key in subsequent development of metabolic risk.. Previously, to explain the transition from normal adipose tissue to that which leads to metabolic derangements, studies proposed the “adipose tissue expandability hypothesis.” This postulated that once adipocytes reached a threshold capacity for storage, they begin to promote insulin resistance with lipotoxicity and adipokine release. This was supported by knockout studies with PPARγ, lipodystrophy models, alterations in adipokine secretion following saturation of adipose tissue [37]. Investigating this led to further characterization of genetic contributors to the pathways of adipogenesis (SFRP-1, Wnt, S14), apoptosis (TRAIL, TWEAK, BCL2, CASP 3/7), and angiogenesis (VEGF-A,B,C,D). Expanding the understanding of adipose tissue in the context of MHO patients, Tinahones et al. looked at genes associated with both lipolysis and lipogenesis and BMI, insulin and HOMA-IR. They found a positive correlation in PPARγ, DGAT1, AQP7, GK, ATGL, HSL, and perilipin and the BMI, insulin, and HOMA-IR in both subcutaneous and visceral fat tissue. Additionally, they demonstrated a negative correlation between genes-ACC1, PEPCK, ACSS2, FABP4, and the aforementioned measures of metabolic risk [38]. Future studies should examine these in more depth to determine their influence over and/or interaction with typical obesity.

Mouse models for MHO

There are several mouse models which may serve as models for MHO. For instance, adiponectin transgenic mice on an ob/ob background become obese on a normal diet but remain insulin sensitive and show fewer inflammatory markers without liver steatosis [39]. Indeed, elevated adiponectin has been observed in humans with MHO [40]. Similarly, TBP2 knockout mice show weight gain and high adiposity but remain insulin sensitive [41]. HcB19 mice crossed with ob/ob become very obese, but also retain peripheral insulin sensitivity and do not show β-cell apoptosis [42]. Additionally, a leaky knockout of the Brd2 gene in mice do show liver steatosis and elevated levels of circulating insulin, but they have less inflammatory markers, hypoglycemia and normal glucose infusion rates [43, 44].

While study of MUHNW mouse models is more limited, work in Goto-Kakizaki rats has implicated Major Histocompatibility Complex (MHC) genes RT1-Ba, Bb, and Db1 in development of insulin resistance and inflammation with a lack of obesity [32] and may serve as models for further study. Careful study of appropriate animal models is needed to propel this field forward.

Dietary and Lifestyle Contributions

Along with ongoing research into genetic determinants, there remain questions surrounding the environmental or external factors leading to MHO presentations. Most commonly, studies have examined diet and fitness for potential contributions, but also for consideration of age, alcohol, tobacco and other factors.

Drawing from National Health and Nutrition Examination Surveys (NHANES) data from two separate time periods, Camhi et al. examined dietary quality (using the Healthy Eating Index) in obese patients, and found that adolescents and MHO women 19–44 years of age, had higher dietary quality with increased fruit, grain, meat and bean intake [45]. Previously, data had not shown marked differences, other than increased adherence to food pyramid recommendation in MHO individuals [10].

Similarly, investigation into exercise/fitness regimens has been conducted. One cross sectional study from the Cork and Kerry Diabetes and Heart Disease Study, did not show any effect of physical activity, smoking, alcohol intake or calorie intake on development of the MHO phenotype, whereas there was positive evidence for dietary qualityi [10]. Alternately, data from the Korean National Health Study demonstrated the MHO phenotype was less likely with advanced age, male gender, and with any history of tobacco smoking [26].

With regards to factors influencing the development of metabolic syndrome/unhealthy metabolic profiles, several studies have examined the influence of lifestyle contributors. While multiple studies identify age, alcohol intake and activity levels as likely contributors, there is differing evidence surrounding the influence of gender [46, 47]. Smoking status and lower education levels have also been identified as potential contributors to this phenotype [26]. This will need to be better defined by large epidemiology studies in the future.

Metabolic Outcomes

Given concern surrounding obesity as a public health epidemic lies within cardiovascular risk and disease, this has also been an important question for MHO patients. However, the data surrounding this has been conflicting and inconsistent.

Despite variable criteria, certain anthropometric and other characteristics have been associated with the MHO phenotype. Most notably, distinct fat distribution patterns (with less ectopic and visceral fat), and favorable inflammatory profiles (with lower concentrations of inflammatory markers) are seen with MHO [48]. Furthermore, in another study, MHO women had lower blood pressure, triglycerides, and glucose but higher HDL cholesterol, adiponectin and LDL size than MUHNW women [6]. Phillips et al. found that MHO patients had lower levels of complement component 3, TNF-a, IL-6 and CRP, as well as a reduced white blood cell count and higher adiponectin [49]. Multiple studies have suggested this “favorable” inflammatory profile in MHO development [5052], though the degree of profile is not universally agreed upon [19]. MHO show higher free fatty acid (FFA), IL-6, and CD4+ T cell levels and macrophage infiltration into adipose tissue [9]. Liver enzymes, but not vitamin D, have been shown to be independently associated with MHO in the Korean population [4]. Another study also showed that MHO individuals have lower levels of liver enzymes such as ALT, AST, and GGT in addition to less fat in the liver [53]. This was further supported by Chen et al., who stratified insulin resistant and sensitive groups, subsequently finding that lower visceral adiposity, liver fat, blood sugar levels, and blood pressures were all associated with insulin sensitive obesity [54]. Altogether, there appears to be a normal/healthy metabolic profile that sets MHO patients apart from their metabolically unhealthy obese counterparts.

To evaluate cardiovascular risk, Pimentel et al. [55] applied the well validated Framingham Risk Score (FRS) in a Brazilian population, and found that there was some indication that metabolically unhealthy obese patients were at increased cardiovascular risk as compared to MHO patients. However, this was specifically noted to hold true when ATP III criteria (alone or in conjunction with HOMA), but not with HOMA model alone [55]. This was in opposition to data gleaned from the London based Whitehall II cohort, which showed decreased when development of T2DM in MHO patients, but also demonstrated an increased risk of cardiovascular disease compared to their metabolically unhealthy counterparts [56]. To compare MHO patients to their non-obese counterparts, Hamer et al. [57] followed MHO patients over a 7-year time period. They found that in this case, MHO patients were not at comparatively increased cardiovascular risk [57]. Most recently, and potentially most comprehensively-another prospective study, the Nord-Trøndelag Health Study (HUNT) specifically evaluated risk of acute myocardial infarctions (AMIs), as well as heart failure (HF). After initial metabolic and anthropometric measurements, patients were followed for an average of 12.2 years, with AMIs and HF being identified by hospitalization data. Using this data, Morkedal et al. [58] found that these patients were not at significantly increased risk of AMI compared to their normal weight counterparts, but heart failure was increased in obese patients regardless of metabolic status. Another study found that MHO were at less risk of angiographic coronary artery disease as compared to metabolically unhealthy obese and even MUHNW [8]. To comprehensively assess both clinical and subclinical cardiovascular disease, Stefan et al. [59] performed a systematic review of the literature, which further emphasized variable evidence in this area. Of note, on analysis of MHO patients, they were found to have significant increase in subclinical markers, such as carotid intimal thickness. More recently, Kim et al. found that lipid and LDL particle subfraction profiles were significantly different between MHO and non-MHO in a Korean population. Specifically, small dense LDL particles were associated with increased metabolic abnormalities in obese adults [7]. The prospective Framingham Heart Study suggested lower risk of obesity-related cancers in MHO patients with normal glucose levels as compared to those with elevated glucose levels [60].

Limited studies have looked at longer-term outcomes in MUHNW patients. One study observed higher waist circumference, higher HbA1c, triglyceride, and CRP levels, and lower HDL and adiponectin in individuals who were MUHNW [61]. Some studies have found less cardiovascular disease risk in MUHNW than MHO [57, 6264], while others have not [65, 66]. In an elderly Korean population over an average of 10 years, MUHNW patients had significantly higher all cause and cardiovascular mortality than MHO patients [67]. These patients have also been found to have increased markers of atherosclerosis [68]. Another study has shown that MUHNW participants have worse cardiovascular outcomes than MHO including higher levels of fatty liver and of subclinical systolic and diastolic dysfunction, suggesting that these outcomes are not dependent on BMI but rather on overall metabolic health [62]. MUHNW women have been shown to have increased risk for cardiovascular disease with higher blood pressure, triglycerides and glucose as well as lower adiponectin, HDL, and LDL size than MHO women [6]. MONW has also been shown to be associated with angiographic coronary artery disease after controlling for potential confounders [8]. There are several mechanisms through which this may be occurring. Fatty liver has been associated with increased fat in the pericardium [6971] which in turn leads to lipotoxicity and apoptosis of cardiac myocytes [72]. MUHNW individuals also show higher levels of inflammatory markers which is associated with cardiovascular disease risk [73, 74]. Future studies will be needed to explore this more fully.

Conversion of MHO to metabolically unhealthy obesity

There remains suggestion that despite “healthy” metabolic profiles, these patients may still be at increased risk for adverse long term outcomes, though data behind this does not account for confounding factors on smoking, physical activity, age and sex [11]. Further confounding considerations include the possibility that as many as 30% of these patients may convert to a MAO/MUHO phenotype, with resultant increase cardiovascular risk [48]. Additionally, while MHO patients have been shown to have better vascular reactivity than patients with metabolic syndrome, they have poorer reactivity compared to metabolically healthy people of normal weight [75], further suggesting deficits that may evolve over time. This too, has been conflicting in its evidence, with an underlying question being whether MHO presentations simply represent an early snapshot in the timeline of metabolic health. One group found that nearly 50% of MHO patients transitioned to metabolically unhealthy phenotypes when followed longitudinally for 10 years [76]. A theory then could be that MHO would eventually transition to being metabolically unhealthy given enough time, but large, longitudinal studies would be required to determine this possibility. By studying a large longitudinal sample of individuals who are MHO at baseline and monitoring the development of diabetes, cardiovascular disease, hypertension, and fatty liver disease over a long period of time, it would be possible to determine whether there are indications that individuals who are MHO would develop the typical metabolic complications of obesity.

Treatments and Interventions

With contradicting evidence surrounding the factors leading to, and longitudinal risks of, MHO, there remains limited data surrounding appropriate long-term management and interventions for patients with MHO. On review, a handful of trials have been conducted in these patients, with the majority focusing on exercise/lifestyle changes as the intervention modality. In 2009, Arsenault et al.[77] examined metabolically healthy overweight or obese postmenopausal women with elevated blood pressures. This cohort underwent a six month intensive exercise program, after which there was no significant change noted in metabolic profile [77]. Janiszewski [78] expanded weight loss interventions to also evaluate diet along with exercise interventions in MHO men and women, as well as MAO patients. While all patients had improvements in anthropometric measurements, only insulin sensitivity/fasting insulin were noted as cardiometabolic improvements in MHO patients [78]. A similar study did not show this improvement in insulin sensitivity, though did find it in metabolically unhealthy individuals [79]. Recently, a 12-week energy restricted diet intervention demonstrated significant improvements in insulin, as well as hepatic enzyme, fatty liver and leptin measurements [80]. Thus, these studies suggest that exercise and lifestyle modifications might be therapeutic for MHO individuals in terms of long-term insulin sensitivity, but as there are conflicting results, future, larger studies will need to determine if this is indeed the case.

Specialized treatments specific for the MUHNW phenotype do not exist, but they are instead treated as metabolically unhealthy obese patients with lifestyle modification and exercise in addition to the appropriate therapeutics for their metabolic abnormalities. Indeed, there are a number of pharmaceutical therapies effective for the comorbidities typically associated with obesity, such as diabetes, hypertension, and cardiovascular disease, which may be effective in treating the metabolic dysfunction associated with MUHNW [8185]. There are also dietary and exercise therapies generally recommended for obesity which may benefit this phenotype as well [8691]. Indeed, even small changes in weight and/or waist circumference seem to benefit biomarkers for cardiometabolic risk [92]. Upon more complete definitions of the MUHNW phenotype, targeted therapies may be warranted.

Future directions

Altogether, there is clear evidence towards the existence of MHO and MUHNW individuals with distinct phenotypes and metabolic outcomes. Clear definitions are needed in order to classify these individuals across research and clinical studies. Additionally, time relationships should be studied in terms of the possibility that MHO may all eventually develop metabolic dysfunctions typically associated with obesity. Regardless, it appears that MHO individuals cross-sectionally and over brief periods of time are at less risk of metabolic dysfunction, including diabetes, cardiovascular disease, and obesity-related cancers; while individuals who are MUHNW display increased risks for these conditions despite a normal weight. After these conditions are well-defined, targeted therapies should be developed to target these unique metabolic disorders.

Acknowledgments

Olivia M. Farr is supported by a NICHD 5T32HD052961.

Abbreviations

BMI

Body Mass Index

T2DM

type 2 Diabetes Mellitus

MHO

Metabolically healthy but obese

MUHNW

Metabolically unhealthy normal weight

MUHO

Metabolically Unhealthy Obese

MAO

Metabolically Abnormal Obese

LDL

Low density lipoprotein

HDL

High densisty lipoprotein

TC

Total cholesterol

TG

Triglycerides

FPG

Fasting plasma glucose

HbA1c

Hemoglobin A1c

TyG

Triglycerides

MONW

Metabolically obese but normal weight

POMC

Pro-opiomelanocortin

MCR4

Melanocortin receptor 4

RNA

Ribonucleic acid

SPP1

Osteopontin

NDRG4

N-myc downstream-regulated gene family member 4

GINS3

GINS complex subunit 3

SFRP1

secreted frizzled-related protein 1

Wnt

Wingless-type mammary tumor virus integration site

TRAIL

TNF-related apoptosis-inducing ligand

TWEAK

TNF-related weak inducer of apoptosis

BCL2

B-cell lymphoma-2

CASP3/7

Caspase 3/7

VEGF

Vascular Endothelial Growth Factor

HOMA-IR

Homeostatic model assessment for insulin resistance

PPARγ

peroxisome proliferator-activated receptor alpha

DGAT1

Diacylglycerol-O-Acyltransferase

AQP7

Aquaporin 7

GK

glycerol kinase

ATGL

Adipose triglyceride lipase

HSL

Hormone sensitive lipase

ACC1

Acetyl-CoA carboxylase 1

PEPCK

Phosphenolpyruvate carboxykinase

ACSS2

Acyl-CoA synthetase short chain family member 2

FABP4

Fatty acid binding protein 4

TBP2

Thioredoxin-binding protein-2

HcB19

HcB19/Dem mouse strain

Brd2

Bromodomain-containing protein 2

RT1-Ba, Bb

Rat1 locuse Ba/Bb

Db1

didemnin-B 1

Footnotes

Disclosure Statement: The authors have nothing to disclose.

Conflicts of Interest

The authors have no conflicts to disclose.

Author Contributions

HM, OMF, and CSM wrote the manuscript.

i

With closer adherence to current food pyramid recommendations

References

  • 1.Ogden CL, Carroll MD, Kit BK, Flegal KM. Prevalence of childhood and adult obesity in the united states, 2011–2012. JAMA. 2014;311(8):806–814. doi: 10.1001/jama.2014.732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Finkelstein EA, Trogdon JG, Cohen JW, Dietz W. Annual medical spending attributable to obesity: Payer-and service-specific estimates. Health Aff (Millwood) 2009;28(5):w822–w831. doi: 10.1377/hlthaff.28.5.w822. [DOI] [PubMed] [Google Scholar]
  • 3.Eckel N, Mühlenbruch K, Meidtner K, Boeing H, Stefan N, Schulze MB. Characterization of metabolically unhealthy normal-weight individuals: Risk factors and their associations with type 2 diabetes. Metabolism. (0) doi: 10.1016/j.metabol.2015.03.009. [DOI] [PubMed] [Google Scholar]
  • 4.Hong HC, Lee JS, Choi HY, Yang SJ, Yoo HJ, Seo JA, et al. Liver enzymes and vitamin d levels in metabolically healthy but obese individuals: Korean national health and nutrition examination survey. Metabolism. 2013;62(9):1305–1312. doi: 10.1016/j.metabol.2013.04.002. [DOI] [PubMed] [Google Scholar]
  • 5.Jankovic A, Korac A, Srdic-Galic B, Buzadzic B, Otasevic V, Stancic A, et al. Differences in the redox status of human visceral and subcutaneous adipose tissues--relationships to obesity and metabolic risk. Metabolism. 2014;63(5):661–671. doi: 10.1016/j.metabol.2014.01.009. [DOI] [PubMed] [Google Scholar]
  • 6.Kim M, Paik JK, Kang R, Kim SY, Lee SH, Lee JH. Increased oxidative stress in normal-weight postmenopausal women with metabolic syndrome compared with metabolically healthy overweight/obese individuals. Metabolism. 2013;62(4):554–560. doi: 10.1016/j.metabol.2012.10.006. [DOI] [PubMed] [Google Scholar]
  • 7.Kim S, Lee H, Lee DC, Lee HS, Lee JW. Predominance of small dense ldl differentiates metabolically unhealthy from metabolically healthy overweight adults in korea. Metabolism. 2014;63(3):415–421. doi: 10.1016/j.metabol.2013.11.015. [DOI] [PubMed] [Google Scholar]
  • 8.Kwon BJ, Kim DW, Her SH, Kim DB, Jang SW, Cho EJ, et al. Metabolically obese status with normal weight is associated with both the prevalence and severity of angiographic coronary artery disease. Metabolism. 2013;62(7):952–960. doi: 10.1016/j.metabol.2013.01.006. [DOI] [PubMed] [Google Scholar]
  • 9.van Beek L, Lips MA, Visser A, Pijl H, Ioan-Facsinay A, Toes R, et al. Increased systemic and adipose tissue inflammation differentiates obese women with t2dm from obese women with normal glucose tolerance. Metabolism. 2014;63(4):492–501. doi: 10.1016/j.metabol.2013.12.002. [DOI] [PubMed] [Google Scholar]
  • 10.Phillips CM, Dillon C, Harrington JM, McCarthy VJ, Kearney PM, Fitzgerald AP, et al. Defining metabolically healthy obesity: Role of dietary and lifestyle factors. PLoS One. 2013;8(10):e76188. doi: 10.1371/journal.pone.0076188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Kramer CK, Zinman B, Retnakaran R. Metabolically healthy overweight and obesity. Ann Intern Med. 2014;160(7):516. doi: 10.7326/L14-5007-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Herrmann W, Reuter W, Schutz C, Lindhofer HG, Wurzberger G, Schneider P. the behavior of specific parameters of lipid and lipoprotein metabolism in metabolically healthy and obese subjects. Z Gesamte Inn Med. 1982;37(2):43–50. [PubMed] [Google Scholar]
  • 13.Bluher S, Schwarz P. Metabolically healthy obesity from childhood to adulthood - does weight status alone matter? Metabolism. 2014;63(9):1084–1092. doi: 10.1016/j.metabol.2014.06.009. [DOI] [PubMed] [Google Scholar]
  • 14.Sims EA. Are there persons who are obese, but metabolically healthy? Metabolism. 2001;50(12):1499–1504. doi: 10.1053/meta.2001.27213. [DOI] [PubMed] [Google Scholar]
  • 15.Bluher M. Are metabolically healthy obese individuals really healthy? Eur J Endocrinol. 2014;171(6):R209–R219. doi: 10.1530/EJE-14-0540. [DOI] [PubMed] [Google Scholar]
  • 16.Achilike I, Hazuda HP, Fowler SP, Aung K, Lorenzo C. Predicting the development of the metabolically healthy obese phenotype. Int J Obes (Lond) 2015;39(2):228–234. doi: 10.1038/ijo.2014.113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Phillips CM. Metabolically healthy obesity: Definitions, determinants and clinical implications. Rev Endocr Metab Disord. 2013;14(3):219–227. doi: 10.1007/s11154-013-9252-x. [DOI] [PubMed] [Google Scholar]
  • 18.Velho S, Paccaud F, Waeber G, Vollenweider P, Marques-Vidal P. Metabolically healthy obesity: Different prevalences using different criteria. Eur J Clin Nutr. 2010;64(10):1043–1051. doi: 10.1038/ejcn.2010.114. [DOI] [PubMed] [Google Scholar]
  • 19.Gomez-Ambrosi J, Catalan V, Rodriguez A, Andrada P, Ramirez B, Ibanez P, et al. Increased cardiometabolic risk factors and inflammation in adipose tissue in obese subjects classified as metabolically healthy. Diabetes Care. 2014;37(10):2813–2821. doi: 10.2337/dc14-0937. [DOI] [PubMed] [Google Scholar]
  • 20.Ruderman NB, Schneider SH, Berchtold P. The "metabolically-obese," normal-weight individual. The American Journal of Clinical Nutrition. 1981;34(8):1617–1621. doi: 10.1093/ajcn/34.8.1617. [DOI] [PubMed] [Google Scholar]
  • 21.Ruderman N, Chisholm D, Pi-Sunyer X, Schneider S. The metabolically obese, normalweight individual revisited. Diabetes. 1998;47(5):699–713. doi: 10.2337/diabetes.47.5.699. [DOI] [PubMed] [Google Scholar]
  • 22.Lee SH, Han K, Yang HK, Kim HS, Cho JH, Kwon HS, et al. A novel criterion for identifying metabolically obese but normal weight individuals using the product of triglycerides and glucose. Nutrition & Diabetes. 2015;5(4):e149. doi: 10.1038/nutd.2014.46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Shea JL, King MT, Yi Y, Gulliver W, Sun G. Body fat percentage is associated with cardiometabolic dysregulation in bmi-defined normal weight subjects. Nutr Metab Cardiovasc Dis. 2012;22(9):741–747. doi: 10.1016/j.numecd.2010.11.009. [DOI] [PubMed] [Google Scholar]
  • 24.Rey-Lopez JP, de Rezende LF, Pastor-Valero M, Tess BH. The prevalence of metabolically healthy obesity: A systematic review and critical evaluation of the definitions used. Obes Rev. 2014;15(10):781–790. doi: 10.1111/obr.12198. [DOI] [PubMed] [Google Scholar]
  • 25.Wildman RP, Muntner P, Reynolds K, McGinn AP, Rajpathak S, Wylie-Rosett J, et al. The obese without cardiometabolic risk factor clustering and the normal weight with cardiometabolic risk factor clustering: Prevalence and correlates of 2 phenotypes among the us population (nhanes 1999–2004) Arch Intern Med. 2008;168(15):1617–1624. doi: 10.1001/archinte.168.15.1617. [DOI] [PubMed] [Google Scholar]
  • 26.Lee K. Metabolically obese but normal weight (monw) and metabolically healthy but obese (mho) phenotypes in koreans: Characteristics and health behaviors. Asia Pac J Clin Nutr. 2009;18(2):280–284. [PubMed] [Google Scholar]
  • 27.Coleman NJ, Miernik J, Philipson L, Fogelfeld L. Lean versus obese diabetes mellitus patients in the united states minority population. J Diabetes Complications. 2014;28(4):500–505. doi: 10.1016/j.jdiacomp.2013.11.010. [DOI] [PubMed] [Google Scholar]
  • 28.Tan VM, Lee YS, Venkataraman K, Khoo EY, Tai ES, Chong YS, et al. Ethnic differences in insulin sensitivity and beta-cell function among asian men. Nutr Diabetes. 2015;5:e173. doi: 10.1038/nutd.2015.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Feng RN, Du SS, Wang C, Li YC, Liu LY, Guo FC, et al. Lean-non-alcoholic fatty liver disease increases risk for metabolic disorders in a normal weight chinese population. World J Gastroenterol. 2014;20(47):17932–17940. doi: 10.3748/wjg.v20.i47.17932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Navarro E, Funtikova AN, Fito M, Schroder H. Can metabolically healthy obesity be explained by diet, genetics, and inflammation? Mol Nutr Food Res. 2015;59(1):75–93. doi: 10.1002/mnfr.201400521. [DOI] [PubMed] [Google Scholar]
  • 31.Speakman JR, Levitsky DA, Allison DB, Bray MS, de Castro JM, Clegg DJ, et al. Set points, settling points and some alternative models: Theoretical options to understand how genes and environments combine to regulate body adiposity. Dis Model Mech. 2011;4(6):733–745. doi: 10.1242/dmm.008698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Denis GV, Obin MS. 'Metabolically healthy obesity': Origins and implications. Mol Aspects Med. 2013;34(1):59–70. doi: 10.1016/j.mam.2012.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Das SK, Ma L, Sharma NK. Adipose tissue gene expression and metabolic health of obese adults. Int J Obes (Lond) 2015;39(5):869–873. doi: 10.1038/ijo.2014.210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Badoud F, Lam KP, DiBattista A, Perreault M, Zulyniak MA, Cattrysse B, et al. Serum and adipose tissue amino acid homeostasis in the metabolically healthy obese. J Proteome Res. 2014;13(7):3455–3466. doi: 10.1021/pr500416v. [DOI] [PubMed] [Google Scholar]
  • 35.Yaghootkar H, Scott RA, White CC, Zhang W, Speliotes E, Munroe PB, et al. Genetic evidence for a normal-weight "metabolically obese" phenotype linking insulin resistance, hypertension, coronary artery disease, and type 2 diabetes. Diabetes. 2014;63(12):4369–4377. doi: 10.2337/db14-0318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Scott RA, Fall T, Pasko D, Barker A, Sharp SJ, Arriola L, et al. Common genetic variants highlight the role of insulin resistance and body fat distribution in type 2 diabetes, independent of obesity. Diabetes. 2014;63(12):4378–4387. doi: 10.2337/db14-0319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Tan CY, Vidal-Puig A. Adipose tissue expandability: The metabolic problems of obesity may arise from the inability to become more obese. Biochem Soc Trans. 2008;36(Pt 5):935–940. doi: 10.1042/BST0360935. [DOI] [PubMed] [Google Scholar]
  • 38.Tinahones FJ, Garrido-Sanchez L, Miranda M, Garcia-Almeida JM, Macias-Gonzalez M, Ceperuelo V, et al. Obesity and insulin resistance-related changes in the expression of lipogenic and lipolytic genes in morbidly obese subjects. Obes Surg. 2010;20(11):1559–1567. doi: 10.1007/s11695-010-0194-z. [DOI] [PubMed] [Google Scholar]
  • 39.Kim JY, van de Wall E, Laplante M, Azzara A, Trujillo ME, Hofmann SM, et al. Obesity-associated improvements in metabolic profile through expansion of adipose tissue. J Clin Invest. 2007;117(9):2621–2637. doi: 10.1172/JCI31021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kloting N, Fasshauer M, Dietrich A, Kovacs P, Schon MR, Kern M, et al. Insulin-sensitive obesity. Am J Physiol Endocrinol Metab. 2010;299(3):E506–E515. doi: 10.1152/ajpendo.00586.2009. [DOI] [PubMed] [Google Scholar]
  • 41.Chutkow WA, Birkenfeld AL, Brown JD, Lee HY, Frederick DW, Yoshioka J, et al. Deletion of the alpha-arrestin protein txnip in mice promotes adiposity and adipogenesis while preserving insulin sensitivity. Diabetes. 2010;59(6):1424–1434. doi: 10.2337/db09-1212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Chen J, Hui ST, Couto FM, Mungrue IN, Davis DB, Attie AD, et al. Thioredoxin-interacting protein deficiency induces akt/bcl-xl signaling and pancreatic beta-cell mass and protects against diabetes. Faseb j. 2008;22(10):3581–3594. doi: 10.1096/fj.08-111690. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Wang F, Deeney JT, Denis GV. Brd2 gene disruption causes "metabolically healthy" obesity: Epigenetic and chromatin-based mechanisms that uncouple obesity from type 2 diabetes. Vitam Horm. 2013;91:49–75. doi: 10.1016/B978-0-12-407766-9.00003-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Wang F, Liu H, Blanton WP, Belkina A, Lebrasseur NK, Denis GV. Brd2 disruption in mice causes severe obesity without type 2 diabetes. Biochem J. 2010;425(1):71–83. doi: 10.1042/BJ20090928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Camhi SM, Whitney Evans E, Hayman LL, Lichtenstein AH, Must A. Healthy eating index and metabolically healthy obesity in u.S. Adolescents and adults. Prev Med. 2015;77:23–27. doi: 10.1016/j.ypmed.2015.04.023. [DOI] [PubMed] [Google Scholar]
  • 46.Bradshaw PT, Monda KL, Stevens J. Metabolic syndrome in healthy obese, overweight, and normal weight individuals: The atherosclerosis risk in communities study. Obesity (Silver Spring) 2013;21(1):203–209. doi: 10.1002/oby.20248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Du T, Zhang J, Yuan G, Zhang M, Zhou X, Liu Z, et al. Nontraditional risk factors for cardiovascular disease and visceral adiposity index among different body size phenotypes. Nutr Metab Cardiovasc Dis. 2015;25(1):100–107. doi: 10.1016/j.numecd.2014.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Samocha-Bonet D, Dixit VD, Kahn CR, Leibel RL, Lin X, Nieuwdorp M, et al. Metabolically healthy and unhealthy obese--the 2013 stock conference report. Obes Rev. 2014;15(9):697–708. doi: 10.1111/obr.12199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Phillips CM, Perry IJ. Does inflammation determine metabolic health status in obese and nonobese adults? The Journal of Clinical Endocrinology & Metabolism. 2013;98(10):E1610–E1619. doi: 10.1210/jc.2013-2038. [DOI] [PubMed] [Google Scholar]
  • 50.Karelis AD, Faraj M, Bastard JP, St-Pierre DH, Brochu M, Prud'homme D, et al. The metabolically healthy but obese individual presents a favorable inflammation profile. J Clin Endocrinol Metab. 2005;90(7):4145–4150. doi: 10.1210/jc.2005-0482. [DOI] [PubMed] [Google Scholar]
  • 51.Esser N, L'Homme L, De Roover A, Kohnen L, Scheen AJ, Moutschen M, et al. Obesity phenotype is related to nlrp3 inflammasome activity and immunological profile of visceral adipose tissue. Diabetologia. 2013;56(11):2487–2497. doi: 10.1007/s00125-013-3023-9. [DOI] [PubMed] [Google Scholar]
  • 52.Bednarek-Tupikowska G, Zdrojowy-Welna A, Stachowska B, Kuliczkowska-Plaksej J, Matczak-Giemza M, Kubicka E, et al. Accumulation of abdominal fat in relation to selected proinflammatory cytokines concentrations in non-obese wroclaw inhabitants. Endokrynol Pol. 2014;65(6):449–455. doi: 10.5603/EP.2014.0062. [DOI] [PubMed] [Google Scholar]
  • 53.Messier V, Karelis AD, Robillard ME, Bellefeuille P, Brochu M, Lavoie JM, et al. Metabolically healthy but obese individuals: Relationship with hepatic enzymes. Metabolism. 2010;59(1):20–24. doi: 10.1016/j.metabol.2009.06.020. [DOI] [PubMed] [Google Scholar]
  • 54.Chen DL, Liess C, Poljak A, Xu A, Zhang J, Thoma C, et al. Phenotypic characterization of insulin-resistant and insulin-sensitive obesity. J Clin Endocrinol Metab. 2015:jc20152712. doi: 10.1210/jc.2015-2712. [DOI] [PubMed] [Google Scholar]
  • 55.Pimentel Ade C, Scorsatto M, de Oliveira GM, Rosa G, Luiz RR. Racharacterization of metabolically healthy obese brazilians and cardiovascular risk prediction. Nutrition. 2015;31(6):827–833. doi: 10.1016/j.nut.2014.12.024. [DOI] [PubMed] [Google Scholar]
  • 56.Hinnouho GM, Czernichow S, Dugravot A, Nabi H, Brunner EJ, Kivimaki M, et al. Metabolically healthy obesity and the risk of cardiovascular disease and type 2 diabetes: The whitehall ii cohort study. Eur Heart J. 2015;36(9):551–559. doi: 10.1093/eurheartj/ehu123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Hamer M, Stamatakis E. Metabolically healthy obesity and risk of all-cause and cardiovascular disease mortality. J Clin Endocrinol Metab. 2012;97(7):2482–2488. doi: 10.1210/jc.2011-3475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Morkedal B, Vatten LJ, Romundstad PR, Laugsand LE, Janszky I. Risk of myocardial infarction and heart failure among metabolically healthy but obese individuals: Hunt (nordtrondelag health study), norway. J Am Coll Cardiol. 2014;63(11):1071–1078. doi: 10.1016/j.jacc.2013.11.035. [DOI] [PubMed] [Google Scholar]
  • 59.Roberson LL, Aneni EC, Maziak W, Agatston A, Feldman T, Rouseff M, et al. Beyond bmi: The "metabolically healthy obese" phenotype & its association with clinical/subclinical cardiovascular disease and all-cause mortality -- a systematic review. BMC Public Health. 2014;14:14. doi: 10.1186/1471-2458-14-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Moore LL, Chadid S, Singer MR, Kreger BE, Denis GV. Metabolic health reduces risk of obesity-related cancer in framingham study adults. Cancer Epidemiol Biomarkers Prev. 2014;23(10):2057–2065. doi: 10.1158/1055-9965.EPI-14-0240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Eckel N, Muhlenbruch K, Meidtner K, Boeing H, Stefan N, Schulze MB. Characterization of metabolically unhealthy normal-weight individuals: Risk factors and their associations with type 2 diabetes. Metabolism. 2015;64(8):862–871. doi: 10.1016/j.metabol.2015.03.009. [DOI] [PubMed] [Google Scholar]
  • 62.Dobson R, Burgess MI, Sprung VS, Irwin A, Hamer M, Jones J, et al. Metabolically healthy and unhealthy obesity: Differential effects on myocardial function according to metabolic syndrome, rather than obesity. Int J Obes (Lond) 2015 doi: 10.1038/ijo.2015.151. [DOI] [PubMed] [Google Scholar]
  • 63.Appleton SL, Seaborn CJ, Visvanathan R, Hill CL, Gill TK, Taylor AW, et al. Diabetes and cardiovascular disease outcomes in the metabolically healthy obese phenotype: A cohort study. Diabetes Care. 2013;36(8):2388–2394. doi: 10.2337/dc12-1971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Calori G, Lattuada G, Piemonti L, Garancini MP, Ragogna F, Villa M, et al. Prevalence, metabolic features, and prognosis of metabolically healthy obese italian individuals: The cremona study. Diabetes Care. 2011;34(1):210–215. doi: 10.2337/dc10-0665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Aung K, Lorenzo C, Hinojosa MA, Haffner SM. Risk of developing diabetes and cardiovascular disease in metabolically unhealthy normal-weight and metabolically healthy obese individuals. J Clin Endocrinol Metab. 2014;99(2):462–468. doi: 10.1210/jc.2013-2832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kramer CK, Zinman B, Retnakaran R. Are metabolically healthy overweight and obesity benign conditions?: A systematic review and meta-analysis. Ann Intern Med. 2013;159(11):758–769. doi: 10.7326/0003-4819-159-11-201312030-00008. [DOI] [PubMed] [Google Scholar]
  • 67.Choi KM, Cho HJ, Choi HY, Yang SJ, Yoo HJ, Seo JA, et al. Higher mortality in metabolically obese normal-weight people than in metabolically healthy obese subjects in elderly koreans. Clin Endocrinol (Oxf) 2013;79(3):364–370. doi: 10.1111/cen.12154. [DOI] [PubMed] [Google Scholar]
  • 68.Yoo HJ, Hwang SY, Hong HC, Choi HY, Seo JA, Kim SG, et al. Association of metabolically abnormal but normal weight (manw) and metabolically healthy but obese (mho) individuals with arterial stiffness and carotid atherosclerosis. Atherosclerosis. 2014;234(1):218–223. doi: 10.1016/j.atherosclerosis.2014.02.033. [DOI] [PubMed] [Google Scholar]
  • 69.Hallsworth K, Hollingsworth KG, Thoma C, Jakovljevic D, MacGowan GA, Anstee QM, et al. Cardiac structure and function are altered in adults with non-alcoholic fatty liver disease. J Hepatol. 2013;58(4):757–762. doi: 10.1016/j.jhep.2012.11.015. [DOI] [PubMed] [Google Scholar]
  • 70.Perseghin G, Lattuada G, De Cobelli F, Esposito A, Belloni E, Ntali G, et al. Increased mediastinal fat and impaired left ventricular energy metabolism in young men with newly found fatty liver. Hepatology. 2008;47(1):51–58. doi: 10.1002/hep.21983. [DOI] [PubMed] [Google Scholar]
  • 71.Perseghin G, Ntali G, De Cobelli F, Lattuada G, Esposito A, Belloni E, et al. Abnormal left ventricular energy metabolism in obese men with preserved systolic and diastolic functions is associated with insulin resistance. Diabetes Care. 2007;30(6):1520–1526. doi: 10.2337/dc06-2429. [DOI] [PubMed] [Google Scholar]
  • 72.Szczepaniak LS, Victor RG, Orci L, Unger RH. Forgotten but not gone: The rediscovery of fatty heart, the most common unrecognized disease in america. Circ Res. 2007;101(8):759–767. doi: 10.1161/CIRCRESAHA.107.160457. [DOI] [PubMed] [Google Scholar]
  • 73.Edgley AJ, Krum H, Kelly DJ. Targeting fibrosis for the treatment of heart failure: A role for transforming growth factor-beta. Cardiovasc Ther. 2012;30(1):e30–e40. doi: 10.1111/j.1755-5922.2010.00228.x. [DOI] [PubMed] [Google Scholar]
  • 74.Tamaki S, Mano T, Sakata Y, Ohtani T, Takeda Y, Kamimura D, et al. Interleukin-16 promotes cardiac fibrosis and myocardial stiffening in heart failure with preserved ejection fraction. PLoS One. 2013;8(7):e68893. doi: 10.1371/journal.pone.0068893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Francesca Schinzari MI, Umberto Campia, Nadia Mores, Valentina Rovella, Manfredi Tesauro, Nicola Di Daniele, Carmine Cardillo. Vasodilator responses and endothelin-dependent vasoconstriction in metabolically healthy obesity and the metabolic syndrome. Am J Physiol Endocrinol Metab. 2015 doi: 10.1152/ajpendo.00278.2015. published ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Schroder H, Ramos R, Baena-Diez JM, Mendez MA, Canal DJ, Fito M, et al. Determinants of the transition from a cardiometabolic normal to abnormal overweight/obese phenotype in a spanish population. Eur J Nutr. 2014;53(6):1345–1353. doi: 10.1007/s00394-013-0635-2. [DOI] [PubMed] [Google Scholar]
  • 77.Arsenault BJ, Cote M, Cartier A, Lemieux I, Despres JP, Ross R, et al. Effect of exercise training on cardiometabolic risk markers among sedentary, but metabolically healthy overweight or obese post-menopausal women with elevated blood pressure. Atherosclerosis. 2009;207(2):530–533. doi: 10.1016/j.atherosclerosis.2009.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Janiszewski PM, Ross R. Effects of weight loss among metabolically healthy obese men and women. Diabetes Care. 2010;33(9):1957–1959. doi: 10.2337/dc10-0547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Kantartzis K, Machann J, Schick F, Rittig K, Machicao F, Fritsche A, et al. Effects of a lifestyle intervention in metabolically benign and malign obesity. Diabetologia. 2011;54(4):864–868. doi: 10.1007/s00125-010-2006-3. [DOI] [PubMed] [Google Scholar]
  • 80.Ruiz JR, Ortega FB, Labayen I. A weight loss diet intervention has a similar beneficial effect on both metabolically abnormal obese and metabolically healthy but obese premenopausal women. Ann Nutr Metab. 2013;62(3):223–230. doi: 10.1159/000345026. [DOI] [PubMed] [Google Scholar]
  • 81.de Mello AH, Pra M, Cardoso LC, de Bona Schraiber R, Rezin GT. Incretin-based therapies for obesity treatment. Metabolism. 2015;64(9):967–981. doi: 10.1016/j.metabol.2015.05.012. [DOI] [PubMed] [Google Scholar]
  • 82.Fu L, Bruckbauer A, Li F, Cao Q, Cui X, Wu R, et al. Interaction between metformin and leucine in reducing hyperlipidemia and hepatic lipid accumulation in diet-induced obese mice. Metabolism. 2015 doi: 10.1016/j.metabol.2015.07.006. [DOI] [PubMed] [Google Scholar]
  • 83.Kizer JR. Adiponectin, cardiovascular disease, and mortality: Parsing the dual prognostic implications of a complex adipokine. Metabolism. 2014;63(9):1079–1083. doi: 10.1016/j.metabol.2014.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Patel D. Pharmacotherapy for the management of obesity. Metabolism. 2015 doi: 10.1016/j.metabol.2015.08.001. [DOI] [PubMed] [Google Scholar]
  • 85.Xia SF, Duan XM, Hao LY, Li LT, Cheng XR, Xie ZX, et al. Role of thyroid hormone homeostasis in obesity-prone and obesity-resistant mice fed a high-fat diet. Metabolism. 2015;64(5):566–579. doi: 10.1016/j.metabol.2014.12.010. [DOI] [PubMed] [Google Scholar]
  • 86.Bluher S, Petroff D, Wagner A, Warich K, Gausche R, Klemm T, et al. The one year exercise and lifestyle intervention program klaks: Effects on anthropometric parameters, cardiometabolic risk factors and glycemic control in childhood obesity. Metabolism. 2014;63(3):422–430. doi: 10.1016/j.metabol.2013.11.016. [DOI] [PubMed] [Google Scholar]
  • 87.Disanzo BL, You T. Effects of exercise training on indicators of adipose tissue angiogenesis and hypoxia in obese rats. Metabolism. 2014;63(4):452–455. doi: 10.1016/j.metabol.2013.12.004. [DOI] [PubMed] [Google Scholar]
  • 88.Ko BJ, Park KH, Mantzoros CS. Diet patterns, adipokines, and metabolism: Where are we and what is next? Metabolism. 2014;63(2):168–177. doi: 10.1016/j.metabol.2013.11.004. [DOI] [PubMed] [Google Scholar]
  • 89.Slusher AL, Mock JT, Whitehurst M, Maharaj A, Huang CJ. The impact of obesity on pentraxin 3 and inflammatory milieu to acute aerobic exercise. Metabolism. 2015;64(2):323–329. doi: 10.1016/j.metabol.2014.10.022. [DOI] [PubMed] [Google Scholar]
  • 90.Mathieson RA, Walberg JL, Gwazdauskas FC, Hinkle DE, Gregg JM. The effect of varying carbohydrate content of a very-low-caloric diet on resting metabolic rate and thyroid hormones. Metabolism. 1986;35(5):394–398. doi: 10.1016/0026-0495(86)90126-5. [DOI] [PubMed] [Google Scholar]
  • 91.Song MK, Rosenthal MJ, Song AM, Yang H, Ao Y, Yamaguchi DT. Raw vegetable food containing high cyclo (his-pro) improved insulin sensitivity and body weight control. Metabolism. 2005;54(11):1480–1489. doi: 10.1016/j.metabol.2005.05.014. [DOI] [PubMed] [Google Scholar]
  • 92.Valsamakis G, McTernan PG, Chetty R, Al Daghri N, Field A, Hanif W, et al. Modest weight loss and reduction in waist circumference after medical treatment are associated with favorable changes in serum adipocytokines. Metabolism. 2004;53(4):430–434. doi: 10.1016/j.metabol.2003.11.022. [DOI] [PubMed] [Google Scholar]

RESOURCES