Skip to main content
Review Article
Originally Published 21 May 2020
Free Access

Obesity Phenotypes, Diabetes, and Cardiovascular Diseases

This article has been corrected.
VIEW CORRECTION

Abstract

This review addresses the interplay between obesity, type 2 diabetes mellitus, and cardiovascular diseases. It is proposed that obesity, generally defined by an excess of body fat causing prejudice to health, can no longer be evaluated solely by the body mass index (expressed in kg/m2) because it represents a heterogeneous entity. For instance, several cardiometabolic imaging studies have shown that some individuals who have a normal weight or who are overweight are at high risk if they have an excess of visceral adipose tissue—a condition often accompanied by accumulation of fat in normally lean tissues (ectopic fat deposition in liver, heart, skeletal muscle, etc). On the other hand, individuals who are overweight or obese can nevertheless be at much lower risk than expected when faced with excess energy intake if they have the ability to expand their subcutaneous adipose tissue mass, particularly in the gluteal-femoral area. Hence, excessive amounts of visceral adipose tissue and of ectopic fat largely define the cardiovascular disease risk of overweight and moderate obesity. There is also a rapidly expanding subgroup of patients characterized by a high accumulation of body fat (severe obesity). Severe obesity is characterized by specific additional cardiovascular health issues that should receive attention. Because of the difficulties of normalizing body fat content in patients with severe obesity, more aggressive treatments have been studied in this subgroup of individuals such as obesity surgery, also referred to as metabolic surgery. On the basis of the above, we propose that we should refer to obesities rather than obesity.
Obesity, traditionally defined as an excess of body fat causing prejudice to health, is usually assessed in clinical practice by the body mass index (BMI), which is expressed as the ratio of body weight in kilograms divided by height in square meter (kg/m2).1 Since its introduction, many large population studies have reported a J-shaped relationship between the BMI and mortality/morbidity risk—a BMI above 30 kg/m2 (defining obesity in many guidelines) being clearly associated with increased morbidity/mortality risk.2–5 Despite its limitations, BMI has been adopted as a quick and simple clinical tool to first classify patients into risk categories and to monitor changes in adiposity over time both at the individual and population levels.
Despite considerable research efforts devoted to understanding the biology of obesity and energy balance, it has become obvious that available knowledge has to date been of little help to curb the obesity epidemic and that no part of the world has been spared from this phenomenon.6 It has been estimated by the Global Burden of Disease Obesity Collaborators that >603.7 million adult individuals are obese.7 The Global Burden of Disease group has also estimated that elevated BMI values were responsible for 4 million deaths in 2015, with two-thirds of this number attributed to cardiovascular disease (CVD).7 In the United States, the prevalence of obesity has reached almost 40% in 2015 to 2016 in a nationally representative survey (National Health and Nutrition Examination Survey).8 Furthermore, severe obesity (class III and above), defined by a BMI ≥40 kg/m2, has been reported to reach 7.7% with considerable disparities among ethnic groups.9
On the basis of such widespread growth, it is obvious that our evolving scientific knowledge about the etiology of obesity and its management at the individual level has not been translated into successful, large-scale clinical programs.10–12 The prescription of moving more and eating less has now been proven as a crude oversimplification of a complex interaction between physiology and the presence of obesity-promoting environments. Thus, beyond a better understanding of the biology of energy balance and obesity, the major social, environmental, and economic drivers of this epidemic must be understood, and holistic solutions targeting them should be evaluated.6,13 Potential intergenerational transmission of obesity, diabetes mellitus, or CVD susceptibility must also be taken into account when approaching interventions designed to reduce the burden of these diseases at the population level.14
Despite outstanding progress made in our ability to combat CVD, obesity remains the modifiable CVD risk factor that has not been adequately tackled either by lifestyle or pharmacotherapy, as opposed to hypertension, dyslipidemias, diabetes mellitus, and smoking. Furthermore, a previous study has revealed that the bulk of the CVD risk resulting from a high BMI or an elevated waist circumference was largely mediated by altered intermediate risk factors (atherogenic dyslipidemia, hypertension, and diabetes mellitus).15 Because physicians have powerful tools to target lipids, blood pressure, and glycemic control with drugs that have shown their clinical benefits through large randomized outcome trials, they may be more tempted to treat these established CVD risk factors with drugs that have shown their clinical efficacy rather than to target the upstream causes of these altered risk factors, that is, high-risk adiposity (Figure 1). In the present article, we propose that there are benefits to be gained from identifying the subgroups of the highest risk overweight/obese patients and targeting visceral adiposity and ectopic fat while also paying attention to the expanding population of patients with severe obesity.
Figure 1. Relationships between high-risk obesities, intermediate cardiometabolic risk factors, and cardiovascular outcomes. The majority of the association between adiposity and cardiovascular diseases is explained by altered cardiometabolic risk factors/comorbidities. However, high-risk obesities are the main drivers of altered cardiometabolic risk mediators.

Relationship of Body Weight/Fat to Diabetes Mellitus and CVD: From Population Studies to Individual Risk

Heterogeneity of Risk at Any Given BMI Value

The relationship of obesity with type 2 diabetes mellitus has been long recognized and clearly explains the high prevalence of type 2 diabetes mellitus achieved in many countries. Type 2 diabetes mellitus is a major risk factor for CVD. Because obesity is often associated with hypertension and dyslipidemia, many high-risk patients with obesity are characterized by a clustering of metabolic and cardiovascular risk factors.1,16,17 Thus, obesity (at least the high-risk forms described in this article) is a prevalent driver of altered metabolic and cardiovascular risk factors that can be targeted by pharmacotherapies but also modified by lifestyle. On this basis, it is not surprising that many studies have shown a progressive increase in CVD risk associated with higher BMI values.
There is, however, a controversy in cardiovascular medicine. Obesity may be associated with improved survival in patients with established CVD—a finding that is termed the obesity paradox.18–20 This concept is much debated because results from studies are inconsistent. There is a growing body of literature on potential methodological explanations for the obesity paradox, such as (1) misclassification bias caused by using BMI as a measure of obesity instead of body fat distribution, (2) unmeasured confounding factors (eg, cardiorespiratory fitness, sarcopenia, smoking), and (3) potential for bias due to illness-related weight loss. In addition, individuals with obesity often develop CVD at a younger age when they are generally healthier with fewer comorbid conditions than individuals with CVD who are not obese. Addressing this controversy is beyond the scope of this article, and the reader is referred to comprehensive reviews that have covered this topic.21–25
Although increased body mass has been associated with higher likelihood of finding altered cardiovascular risk factors, not every patient who is overweight or obese shows the alterations in cardiovascular risk factors that are expected from excess body fat. In the early 80’s, 2 pioneers, Ahmed Kissebah in the United States and Per Björntorp in Sweden, reported almost simultaneously that a high proportion of upper body (or abdominal) fat evaluated by the ratio of waist-to-hip circumferences (waist-to-hip ratio) was predictive of an increased risk of finding glucose intolerance, evidence of insulin resistance, hypertension, and hypertriglyceridemia.26–28 At any BMI level, increased proportion of abdominal fat as determined by an elevated waist-to-hip ratio was found to be associated with increased health risk, providing the first evidence of the limitations of the BMI in individual patient health risk assessment.
The development of imaging technologies has brought a revolution in the study of body adiposity and its relationship with health outcomes. Because of distinct properties of fat, muscle, and bone tissues, it has become possible to precisely assess body composition and body fat partitioning phenotypes.29–31 Early imaging studies first using computed tomography (CT)31,32 and later magnetic resonance imaging (MRI)33,34 revealed that the amount of adipose tissue located in the abdominal cavity, also called visceral adipose tissue (VAT), was a key correlate of the health risk resulting from a large waistline for a given BMI. When the cardiometabolic risk profile of individuals who were matched for their BMI (or matched for their level of total body fat) but with either a low versus a high accumulation of VAT was compared, it was found that subjects with excess VAT, irrespective of their BMI or total adiposity, had an altered cardiometabolic risk profile predictive of an increased risk of type 2 diabetes mellitus and cardiovascular outcomes.35–42 Since these early studies conducted in the late 80’s/early 90’s, many large cardiometabolic imaging studies have confirmed the notion that excess VAT is predictive of an increased health risk.40

Tale of the Tape

Because imaging is costly and requires expertise and expensive equipment, it was of interest to identify simple tools that could be used in primary care to identify patients at high risk of being viscerally obese. In 1994, we proposed the tape measurement of waist circumference as a simple index of abdominal adiposity.43 Despite the fact that there is a rather strong correlation between the BMI and waist circumference at the population level (r values ≥0.80 depending upon the cohort examined), there is nevertheless considerable variation in waist circumference at any given BMI value (Figure 2).44 Within each specific BMI unit, it has been shown that an elevated waist circumference was predictive of an increased accumulation of VAT.45 Unfortunately, current guidelines recommend single sex-specific waist circumference values (102 cm in men and 88 cm in women) to define abdominal obesity.4 Although these cutoff values have been shown to be helpful in identifying a subgroup of patients who are overweight or obese and at greater risk of being characterized by the features of the metabolic syndrome, they are not BMI specific. For instance, it is obvious that a waist circumference of 102 cm for a man with a BMI of 24.5 kg/m2 does not describe the same adiposity phenotype as another male individual with the same waistline but a BMI of 33 kg/m2. In this example, the man who presumably has normal weight actually has central obesity with a probable VAT excess, whereas the man who is obese with the same waistline is rather characterized by general obesity. These 2 simple clinical cases show why waist circumference should not be used in isolation to replace the BMI as an adiposity metric. Rather, waist should be added to the information provided by the BMI to further refine risk classification.46 Large prospective observational studies have confirmed that within every single BMI category, an elevated waistline is associated with increased mortality risk (Figure 3).15,47 As emphasized in a recent International Atherosclerosis Society–International Chair on Cardiometabolic Risk consensus statement on waist circumference,46 there is a need to identify BMI-specific waist circumference values associated with increased cardiometabolic and mortality risk.
Figure 2. Box and whisker plots showing the distribution of waist circumference values (age adjusted) per unit of body mass index (BMI) in the subsample of 64 624 men of the IDEA study (International Day for the Evaluation of Abdominal Obesity) who had BMI values ≥20 and <40 kg/m2. Data shown are medians, quartiles, and 10th and 90th percentiles. Reprinted from Després44 with permission. Copyright ©2011, Elsevier.
Figure 3. Hazard ratios (HRs) and 95% CIs for waist circumference in 5-cm increments* and all-cause mortality by body mass index (BMI) category (men and women combined), adjusted for education, marital status, smoking status, alcohol consumption, physical activity, and BMI in a sample of 650 000 individuals. *Waist circumference cutoff points (cm) for men: <90.0, 90.0 to 94.9, 95.0 to 99.9, 100.0 to 104.9, 105.0 to 109.9, 110.0+ and women: <70.0, 70.0 to 74.9, 75.0 to 79.9, 80.0 to 84.9, 85.0 to 89.9, 90.0+. Reprinted from Cerhan et al47 with permission. Copyright ©2014, Elsevier.
Despite its added value over the BMI, a large waistline may result from an excess of either subcutaneous or visceral abdominal adipose tissue. Several groups have suggested that blood triglyceride levels may represent a simple and useful marker of excess visceral adiposity when observed in the presence of a large waistline.48–52 For instance, using CT imaging, we have proposed that in the presence of increased triglyceride levels, some individuals may be viscerally obese if they have waist circumference values above 90 cm in men and 85 cm in women—a condition that we have described as hypertriglyceridemic waist.48,50 Prospective studies have shown that the presence of hypertriglyceridemic waist was, indeed, not only predictive of excess VAT but that this simple clinical phenotype was also associated with an altered cardiometabolic risk profile predictive of an increased CVD risk.51 Other approaches incorporating the information provided by waist circumference and blood triglyceride levels such as the lipid accumulation product53,54 and the Visceral Adiposity Index55 have also been reported to be useful in discriminating visceral obesity and related cardiometabolic risk.

Severe Obesity: a Rapidly Expanding Subgroup of High-Risk Patients

Severe obesity, defined by a BMI ≥40 kg/m2, has not only emerged as an important clinical problem but has rapidly become a preoccupying public health issue. Recent data show that the relative increase in the prevalence of severe obesity has been much greater than the relative increase in the prevalence of moderate obesity. For instance, between 1986 and 2010, the prevalence of the US population with a BMI above 40 kg/m2 has increased by 4-fold, whereas the prevalence of individuals with a BMI above 50 has increased by >10-fold. In comparison, the prevalence of a BMI above 30 kg/m2 has been reported to double during the same period. Severe obesity has a 50% greater prevalence in women than in men and is the highest among non-Hispanic black compared with other ethnic groups.56 Management of severe obesity is a challenge in clinical practice because this condition is associated with additional health issues compared with less-severe forms of obesity.57 Because of the health burden and risk associated with severe obesity, more invasive treatments may be considered including weight-loss surgery—a topic to be addressed in later sections.

From Obesity to Obesities

Because of the remarkable heterogeneity observed among individuals meeting the current definition of obesity (BMI ≥30 kg/m2), it is proposed that this condition is too complex to be considered as a homogeneous entity. First, among obese individuals with class I and II obesities (BMI between 30 and 40 kg/m2), the level of risk markedly differs depending upon factors such as regional body fat distribution, overall nutritional quality, level of physical activity, and cardiorespiratory fitness.39 Although the concept of metabolically healthy obesity has been debated, the term has been coined to refer to a lower risk subgroup of individuals who have low levels of VAT despite being obese, who eat well and who are fit because they are physically active.58 At the other end of the moderate obesity spectrum, some individuals are characterized by visceral obesity and show the features of the metabolic syndrome, putting them at high risk for cardiovascular events, particularly if they are sedentary and have poor dietary patterns.38 These observations reemphasize the notion that the BMI is not the appropriate index to assess health risk in class I and II obesities. Second, the rapidly expanding subgroup of patients with severe obesity represents another entity with additional health outcomes having special needs in terms of assessment and medical management. Thus, the singular term obesity does not properly describe the reality of having to deal with these different forms of obesities in terms of adipose tissue mass, body fat distribution, adipose tissue function, and patient lifestyle habits. On that basis, it is proposed that we should move away from the term obesity and rather refer to obesities that would better reflect the clinical reality and distinct treatment challenges associated with dealing with severe obesity versus more moderate forms of obesity.

Obesities and Cardiovascular Risk: Pathophysiological Aspects

As mentioned previously, many large cardiometabolic imaging studies (CT and MRI) have confirmed that excess visceral adiposity is strongly associated with metabolic abnormalities initially thought to be associated with excess fatness per se.40 Because excess visceral adiposity can be found not only in patients who are obese but also in those who are overweight or even have normal weight, and because excess visceral adiposity and ectopic fat accumulation are the main drivers of the insulin resistance syndrome, we can estimate the prevalence of visceral adiposity to be >20% in the overall population,38 although such prevalence is again dependent upon several factors such as age, sex, ethnicity, and lifestyle habits. Thus, even after control for BMI or the amount of total body fat, individuals with high levels of VAT show insulin resistance, hyperinsulinemia, glucose intolerance, a typical atherogenic dyslipidemia (high triglyceride and apolipoprotein B levels, increased proportion of small, dense LDL [low-density lipoprotein] particles, low HDL [high-density lipoprotein] cholesterol levels, and small HDL particles), inflammation, and increased blood pressure levels.37–39,42
Several factors contribute to explain the relationship of excess visceral adiposity to metabolic complications, and they have been discussed in many review articles.36–39,42 The concept of adipose tissue dysfunction has emerged in the past decade as a major determinant of these metabolic complications. Adipose tissue dysfunction can be defined as adipocyte hypertrophy, impaired adipogenesis, low free fatty acid uptake, reduced triglyceride synthesis, resistance to the inhibitory effect of insulin on lipolysis, fat tissue fibrosis, adipocyte autophagy stimulation,59,60 immune cell infiltration, and inflammatory cytokine secretion.42,61–64
Adipose tissue accretion takes place through the expansion of the lipid droplet in existing adipocytes (hypertrophy), the adipogenic differentiation of precursor cells to new lipid-storing adipocytes (hyperplasia), or a combination of both phenomena.65 Interestingly, there is a reciprocal relationship between adipocyte hypertrophy and hyperplasia, where low adipogenic capacity relates to adipocyte hypertrophy and high adipogenic capacity relates to smaller adipocytes.66 Accordingly, in both humans and animals, most studies report a reduction of the adipogenic potential in the obese state (reviewed in the study by Lessard and Tchernof67). In terms of regional differences, preadipocytes from the visceral fat depots such as the greater omentum are known to have lower adipogenic capacity compared with those from the subcutaneous fat compartments.67,68 In this regard, expansion of the visceral fat compartments in humans appears to take place mostly through adipocyte hypertrophy, which could partly explain the stronger association between visceral fat accumulation and obesity-related metabolic abnormalities. This is illustrated in Figure 4, showing that the number of metabolic syndrome features increases with adipocyte hypertrophy and fat mass accumulation, but that individuals with small adipocytes show a low number of metabolic syndrome features, even with an elevated body fat mass. This association is particularly pronounced in omental adipocytes (Figure 4), again most likely due to the predominantly hypertrophic expansion of this compartment.
Figure 4. Relationship between adipocyte size and features of the metabolic syndrome. Adipocyte size was measured by collagenase digestion of fat tissue in a sample of 125 women (age, 36.8–68.3 y; body mass index, 17.2–41.3 kg/m2) for whom visceral, omental (A), and subcutaneous (B) adipose tissues were obtained at the time of gynecological surgery. Features of the metabolic syndrome examined were high waist circumference, high fasting glycemia, hypertension, high fasting triglyceride concentrations, and low fasting HDL (high-density lipoprotein) cholesterol concentrations. Body fat mass was measured by dual-energy x-ray absorptiometry. The sample was divided according to tertiles of fat cell size and body fat mass. ANOVA P<0.05 in both fat compartments.
Low adipogenic capacity has been related to dramatically altered metabolic phenotypes in many mouse models of lipodystrophy (reviewed in the studies by Pap et al69 and Rochford70), as well as in genetic forms of partial lipodystrophy in humans (reviewed in the studies by Hussain and Garg71 and Garg72). We have performed studies on the expression of genes involved in lipodystrophy showing consistent results in patients who do not have genetic lipodystrophy.73 In vitro adipogenic potential of preadipocytes from the visceral and subcutaneous fat depots has also been examined in a small number of studies.68,74 These publications have shown that reduced adipogenic potential in preadipocytes obtained from the subcutaneous fat compartment is associated with increased prevalence of metabolic abnormalities,68,74 as well as visceral adipocyte hypertrophy and increased visceral fat accumulation measured by CT.68 Elegant studies have now identified cellular senescence as a key mechanism in the determination of interindividual variation in the adipogenic capacity of subcutaneous adipocytes.75,76
Altered adipose tissue extracellular matrix remodeling has also emerged as a key factor mediating adipose tissue dysfunction and limited expansion capability (reviewed in the studies by Ruiz-Ojeda et al77 and Lin et al78). Seminal studies using picrosirius red staining of collagens in adipose tissue had shown that collagen distribution patterns differed as a function of the fat compartment examined, as well as the presence of obesity.79 In particular, adipose tissue fibrosis could be quantified either through the presence of bundles of collagen fibers in bands of varying in thickness or as specific depots surrounding the adipocytes, termed pericellular fibrosis.79,80 In our study of visceral and subcutaneous adipose tissues in women, we found that pericellular fibrosis in both fat compartments was strongly related to macrophage infiltration in the corresponding depot, whereas pericellular fibrosis of visceral fat was a predictor of altered cardiometabolic risk variables.61 Interestingly, adipose tissue fibrosis in subcutaneous adipose tissue was related to reduced mobilization of adipose tissue up to 12 months after gastric bypass surgery.79 These results obviously need to be considered in the broader context of an emerging role for many other components of the extracellular matrix as markers of adipose tissue dysfunction.
Immune cell infiltration and alterations in adipose-derived inflammatory cytokines also represent critical aspects of adipose tissue dysfunction. Reviewing the plethora of studies on this topic is obviously beyond the scope of the present article. Briefly, we now know that adipose tissue homeostasis involves complex interactions of many immune cell types including not only macrophages but B cells, natural killer and regulatory T cells, eosinophils, and innate lymphoid cells.81 In a recent study of the stromal-vascular fraction of subcutaneous and VAT using single-cell RNA sequencing,82 we were able to detect up to 17 different immune cell type clusters, including 5 distinct clusters of macrophages. Some of these clusters showed depot-related differences and, most interestingly, a unique expression pattern of metallothionein genes, which are known to be involved in cellular matrix remodeling.82 There is strong evidence that infiltrating immune cells contribute to the low-grade inflammation pattern frequently associated with abdominal-visceral obesity and in turn alter the metabolic ability of adipose tissues to efficiently store lipids and expand.81
In the context of the increasing interest for the gut microbiota in obesity, we now know that gut bacteria and their fragments can translocate past the intestinal barrier, colonize, or accumulate in the blood and extraintestinal tissues.83,84 There, they can exert immunogenic roles that affect glucose homeostasis and other physiological parameters.85,86 Bacterial cell wall components such as peptidoglycans and lipopolysaccharides have been shown to alter glucose homeostasis in a detrimental87,88 or beneficial89,90 manner, suggesting that translocating bacteria or bacterial components can exert a complex modulatory role in metabolic tissues of the host. In a recent study, we have performed a comparative analysis of the microbial profile found in human plasma, liver, and 3 different adipose tissue depots (omental, mesenteric, and subcutaneous).91 Through the use of an optimized 16S metagenomic sequencing pipeline84 and extensive contamination controls, we identified reliable bacterial signatures in the liver, mesenteric, omental, and subcutaneous adipose tissues. Our analysis showed higher copy numbers of the 16S rRNA gene in omental adipose tissue and liver compared with mesenteric and subcutaneous adipose tissues. Microbial abundance of firmicutes, bacteroides, and proteobacteria in mesenteric adipose tissue was lower in participants with type 2 diabetes mellitus compared with BMI-matched individuals who did not have diabetes mellitus. Whether these results reflect actual bacterial translocation or sampling of bacterial nucleic acid sampling by infiltrating immune cells remains to be demonstrated. Yet, tissue microbial signatures may represent a new component of adipose tissue dysfunction.92
The above-cited alterations defining adipose tissue dysfunction collectively alter adipogenic differentiation, adipose tissue expansion capacity, and ultimately lipid storage and retention in adipose tissue. From the metabolic standpoint, nonesterified fatty acid spillover in the postprandial state resulting from poorly inhibited lipolysis and reduced triglyceride synthesis in hypertrophic lipid-storing adipocytes creates increased fatty acid flux to lean tissues, which in turn induces lipotoxicity and increased triglyceride storage in these ectopic sites (reviewed in the study by Noll and Carpentier93). Accordingly, excess VAT deposition has been shown to be frequently accompanied by accumulation of lipids in normally lean tissues such as the liver, the heart, the kidney, the pancreas, and skeletal muscles—a phenomenon described as ectopic fat deposition. For instance, many imaging studies have shown that excess liver fat accumulation is essentially associated with the same dysmetabolic state as visceral adiposity, leading some investigators to even suggest that liver fat was the key culprit for the abnormalities found in visceral adiposity.94,95 Because the liver plays a central role in glucose homeostasis and in lipid metabolism, fatty liver may obviously be a key driver of the full-blown dysmetabolic state found among insulin-resistant individuals. The question is rather whether excess liver fat found in isolation is associated with increased CVD risk. Mendelian randomization studies considering genetic variants associated with increased liver fat have not produced evidence of such an association with CVD.96 From a clinical standpoint, it is important to keep in mind that although liver steatosis can be found in the absence of visceral adiposity, it is rather infrequent as its most prevalent form also features an excess of VAT.95 In this regard, imaging studies have revealed that the shared variance between liver fat and visceral adiposity is about 25% to 30%97,98—a finding suggesting that there are substantial individual differences in the amount of liver fat for any given level of visceral adiposity. We know that susceptibility to visceral adiposity has a strong genetic basis.99,100 The factors explaining the considerable individual variation in liver fat content for any given adiposity level is a subject of considerable interest and of great clinical relevance. For instance, why would a subgroup of viscerally obese individuals be relatively protected against excess liver fat deposition? Are there additional genetic and environmental (eg, sugar content of the diet, lack of vigorous regular physical activity) factors involved? Some candidate genes for liver steatosis have been proposed, and this issue is the topic of great interest.95 One notion appears to emerge at this stage: although visceral adiposity and liver fat content are both independently associated with metabolic abnormalities, the most exacerbated dysmetabolic state is found among individuals with high levels of both VAT and liver fat.40,95
Regarding the genetic contribution to visceral obesity, Karlsson et al100 performed a genome-wide association study for predicted VAT mass. They identified 209 (including 102 novel) loci associated with VAT. In a sample of 325 153 white British participants of the UK Biobank, they confirmed that VAT mass was indeed associated with increased risk of hypertension, myocardial infarction/angina, type 2 diabetes mellitus, and dyslipidemia. Mendelian randomization analyses suggested that VAT was causally related to the development of these clinical outcomes. Accordingly, Ji et al101 found alleles associated with opposite effects on total adiposity and risk of type 2 diabetes mellitus, heart disease, and hypertension. For instance, alleles predictive of a lower risk of cardiometabolic diseases were found to be associated with more subcutaneous adipose tissue and lower levels of ectopic fat such as less liver fat. These results are concordant with the concept that the inherited capacity of some low-risk adipose depots (such as gluteal and femoral adipose tissue) to store excess triglycerides is an important driver (Figure 5) of the risk of CVD and type 2 diabetes mellitus associated with an excess of VAT and liver fat.102
Figure 5. Cardiovascular and metabolic consequences of high-risk obesities. The inherited inability of some low-risk adipose depots to store excess triglycerides is an important driver of excessive adipose tissue accumulation in visceral and ectopic sites. The related metabolic alterations impact negatively the cardiovascular system, increasing the risk of coronary artery disease, heart failure, atrial arrhythmias, and sudden death. HDL indicates high-density lipoprotein; and LDL, low-density lipoprotein.
Thus, according to the above model, the dysmetabolic environment of visceral adiposity can also be explained by considering the expanded VAT both as a driver and as a marker of cardiometabolic risk resulting from a dysfunctional subcutaneous adipose tissue. As for obesity, which is not a single condition, the large number of alterations defining adipose tissue dysfunction can no longer be considered to represent a homogeneous entity.

High-Risk Obesities and the Heart

Visceral Adiposity, Ectopic Fat, and Coronary Artery Disease

Obesity and related metabolic conditions/comorbidities negatively impact the cardiovascular system in several ways.103 Individuals with obesity are more likely to develop CVD and manifestations of CVD, particularly coronary artery disease, angina, myocardial infarction, heart failure, atrial fibrillation, and sudden cardiac death (Figure 5).104 Evidence also indicates that the distribution of excess adiposity is an important determinant of cardiovascular risk; visceral and ectopic adiposity confer a much higher risk than subcutaneous adiposity.105,106 Among obese individuals with coronary artery disease, it appears that the distribution of fat, rather than BMI itself, is more directly associated with mortality.107 Large cohort studies such as the Framingham Heart Study and the Jackson Heart Study have documented excess VAT as a predictor for the development of cardiovascular risk factors over time, independently of total body fat mass or subcutaneous adipose tissue levels.108,109 In a 10-year longitudinal study of Japanese American men, greater VAT levels were found in cases developing coronary artery disease compared with controls.110 Over the 10-year follow-up of study participants, excess visceral adiposity, regardless of BMI, has been documented as an independent precursor of coronary artery disease.110 In the Health, Aging and Body Composition Study, excess VAT in women aged 70 to 79 years was identified as an independent predictor of myocardial infarction over a 4.6-year follow-up.111
Excess adiposity, and especially VAT adiposity, accelerates the progression of atherosclerosis decades before the clinical manifestations of coronary artery disease, as shown in postmortem studies among young individuals.112 Importantly, greater vulnerability of atherosclerotic lesions within the coronary arteries and aorta was described for those with abdominal adiposity independently of other cardiovascular risk factors, indicating the major role of central adiposity in the pathogenesis of atherosclerotic diseases.113 Using coronary artery calcification as a subclinical predictor of coronary artery disease, data from the CARDIA study (Coronary Artery Risk Development in Young Adults) concluded that longer duration of abdominal adiposity was associated with subclinical coronary artery disease and its progression through midlife independent of the degree of adiposity.114
Similarly to VAT, ectopic cardiac adipose tissue accumulation was found to be associated with the development of metabolic and cardiac complications, including coronary artery disease.24,115 Although the long-term relationship between ectopic cardiac adipose tissue accumulation (in any depot) and coronary artery disease is largely unknown, a few cross-sectional studies have reported a potential association between excess ectopic cardiac adipose tissue and coronary artery disease. Cardiac fat depots, including pericardial and epicardial fat, have been associated with coronary events in the general population independent of traditional cardiovascular risk factors.116 However, the precise role of these localized cardiac fat depots in the development of the atherosclerotic process remains controversial.
In the MESA (Multi-Ethnic Study of Atherosclerosis), pericardial adipose tissue was associated with increased risk of all-cause CVD and atherosclerosis burden.116 Excess epicardial adipose tissue deposition measured by ultrasound, CT, and MRI has been related to abnormal myocardial flow reserve, coronary plaque vulnerability, coronary artery calcification, and coronary artery disease severity.117 Epicardial adipose tissue-derived adipocytokines, inflammation, and systemic mediators such as reactive oxygen species may favor the development of a local proatherogenic environment by paracrine and vasocrine mechanisms, thus promoting the pathogenesis of coronary artery disease.118
Besides effects on atherosclerosis in larger arteries, excess adiposity is linked to abnormalities in the coronary microvasculature—a key regulator of coronary flow reserve.119,120 Coronary microvascular disease is related to endothelial dysfunction and small vessel remodeling in responses to neural, mechanical, and metabolic factors. Microvascular disease is independently associated with BMI and provides prognostic information regarding cardiovascular risk among individuals with obesity.121 Importantly, the severity of coronary microvascular dysfunction has been correlated with the amount of VAT.122–124 In prospective studies, surgical weight-loss procedures have been associated with improvements in coronary microvascular function.125

Visceral Adiposity, Ectopic Fat, and Heart Failure

Obesity is a major risk factor for heart failure—an association not fully explained by obesity-related cardiovascular risk factors.126 Individuals with obesity are 2× more at risk of developing heart failure than those who have normal body weight.127,128 According to the Framingham Heart Study, each BMI increment by 1 unit is associated with a 5% increased heart failure risk in men and 7% in women after adjusting for other cardiovascular risk factors, and the risk of heart failure was found to be increased across the entire spectrum of BMI.128 In a meta-analysis looking at the association between obesity and heart failure incidence, each 10-cm increase in waist circumference was associated with an increased incidence of heart failure, with higher incidence for heart failure starting in the overweight BMI range.129
Approximately 50% of individuals with obesity have heart failure with preserved ejection fraction (HFpEF), which means that the usual clinical parameter (left ventricular ejection fraction) is normal, posing a substantial challenge in clinical practice. Although left ventricular ejection fraction has generally been found to be preserved in patients with obesity, many investigators have found evidence of subclinical myocardial dysfunction when sensitive cardiac imaging measurements such as myocardial strain are used.130,131 The prevalence of abnormal subclinical myocardial function (ie, abnormal myocardial strain) in obesity has been reported to range from 37% to 54%.131 Abnormal subclinical myocardial function in asymptomatic obese patients with diabetes mellitus has been associated with the progression of adverse CVD.131 Longer duration of obesity and its severity (BMI ≥40 kg/m2) have been identified as major factors predisposing to cardiac dysfunction and heart failure.132 Surgical treatment of obesity prevents development of heart failure and improves established heart failure, including HFpEF.133 Although the relationship between obesity and incident heart failure may involve hemodynamic and anatomic cardiac changes related to excess adiposity, evidence suggests that the relationship is also mediated by obesity-related metabolic, inflammatory, and hormonal changes (Figure 5).132
Only limited data are available regarding adiposity phenotypes or specific fat depots and the risk of heart failure or subsequent prognosis in individuals with heart failure. Recent evidence suggests that VAT plays a major role in the development, pathophysiology, and adverse outcomes of obese individuals with HFpEF.134 In the MESA cohort free of CVD, VAT levels (measured by CT scan) were independently associated with incident HFpEF but not heart failure with reduced ejection fraction.135 Among the 3310 participants from the TOPCAT trial (Treatment of Preserved Cardiac Function Heart Failure With an Aldosterone Antagonist) who had HFpEF, abdominal adiposity was associated with increases in all-cause mortality and CVD mortality during the 3.5-year follow-up.136 Heart failure is a major cause of morbidity and mortality in obesity; the cardiovascular prognosis of HFpEF is comparable to heart failure with reduced ejection fraction but remains without any known effective treatment.137 Compared with nonobese subjects with HFpEF, obese subjects with HFpEF display greater volume overload, more adverse biventricular remodeling, greater right ventricular dysfunction, and worse exercise capacity.138 Surgical weight loss prevents the onset of heart failure and improves heart failure symptoms and exercise tolerance.133,139 In a retrospective study, surgical weight reduction was found to reduce risk of developing heart failure and hospitalizations for heart failure in patients with a history of heart failure in both reduced ejection fraction and HFpEF.140 Interestingly, brain natriuretic peptide levels are much lower in obese patients with documented heart failure compared with normal-weight patients.141–144 Brain natriuretic peptide levels paradoxically increase following successful weight-loss therapy.145 A potential explanation includes the increase in the clearance of active natriuretic peptides by higher expression of clearance receptors on adipocytes.146,147

Visceral Adiposity, Ectopic Fat, and Arrhythmias

There is now compelling evidence to support the role of excess adiposity in determining arrhythmic risk, particularly focused on atrial fibrillation and sudden cardiac death.148–152 In a meta-analysis involving 125 000 individuals, those who were obese had an ≈50% increase in the prevalence of atrial fibrillation.153 Importantly, cohort studies have identified a significant dose relationship where increased risk of developing atrial fibrillation is observed with higher severity of obesity.148 Each 1-unit increase in BMI was associated with a 4% increased risk of new-onset atrial fibrillation during the follow-up in the Framingham Heart Study.152 Left atrial enlargement seen in patients with obesity appears to be mechanistically important as the association between BMI and atrial fibrillation was significantly weakened when adjustments were made for left atrial size.149 Every 5-unit increment in BMI confers an ≈29% greater risk of incident atrial fibrillation.154 Moreover, excess adiposity seems to be an important risk factor for the progression of paroxysmal atrial fibrillation to more permanent atrial fibrillation, which is related to higher morbidity and mortality.155 Progression of the disease has been documented in the context of obesity with a BMI in the range of 30 to 34.9 kg/m2 associated with a 54% increase in the likelihood of progression from paroxysmal to permanent atrial fibrillation and severe obesity (BMI ≥40 kg/m2) associated with an 87% increase in risk.155 Overweight and obesity increased the risk of atrial fibrillation through multiple mechanisms including atrial structural and electrical remodeling, which contribute to development of the arrhythmogenic substrate.
Besides overall adiposity, ectopic cardiac fat deposition (in any depot) has been associated with the prevalence and severity of atrial fibrillation (trends toward persistent atrial fibrillation and more symptoms).148 Epicardial adipose tissue has emerged as an important proarrhythmic substrate that may explain the excess risk of atrial fibrillation in obesity.156,157 The strength of this relationship is greater than for measures of abdominal and overall adiposity.158 In the Framingham Heart cohort including 3217 participants, CT-measured pericardial fat (but not VAT) was an independent predictor of prevalent atrial fibrillation even after adjusting for established atrial fibrillation risk factors (age, sex, systolic blood pressure, PR interval, and clinically significant valvular disease) and other measures of adiposity such as BMI.159 The amount of epicardial adipose tissue is a predictor for persistence of atrial fibrillation.160 Higher recurrence rate of atrial fibrillation after catheter ablation or electrical cardioversion was reported in relation with excess epicardial adipose tissue.161 The mechanisms by which increased epicardial adipose tissue contributes to the development and burden of atrial fibrillation are unclear. Epicardial adipose tissue may lead to atrial fibrillation via structural and electrical remodeling of the atrial. Pathophysiological mechanisms involve adipose tissue infiltration, profibrotic and proinflammatory epicardial adipose tissue paracrine effects, and oxidative stress.148,149,162 Increase in sympathetic or parasympathetic tone related to dense innervation of cardiac adipose tissue depots in contiguity with the atrial and pulmonary veins may also play a role.148,163,164

Visceral Adiposity, Ectopic Fat, and Sudden Death

Longitudinal studies have demonstrated the association between obesity and cardiac dysrhythmias with increased risk of sudden cardiac death.150–152,165 In the Framingham Heart Study, the annual rate of sudden cardiac death was ≈40-fold higher in individuals who were obese.166 Both mild and severe obesity are reported to associate with greater risk of ventricular tachycardia/fibrillation.167,168 The potential mechanisms for this association are multiple and may include increased cardiac electrical irritability, abnormal late potentials, and disrupted sympathovagal balance, leading to more frequent and complex ventricular dysrhythmias, even in the absence of clinically overt heart failure.169 Left ventricular hypertrophy as a result of obesity has been shown to give rise to increased ventricular ectopy.170 Obesity is also associated with delayed ventricular repolarization as evidenced by prolongation of the QT/QTc interval, which has been shown to independently predict cardiovascular mortality.171 Pathological changes in the cardiac conduction system have been shown from autopsy of young individuals with obesity and sudden cardiac death.172 Obesity was an independent risk factor for ventricular tachyarrhythmias in patients with ischemic cardiomyopathy treated with an implantable cardioverter-defibrillator in the MADIT II (Multicenter Automatic Defibrillator Implantation Trial II).167
Data suggest that there may be an important role for body fat partitioning, implicating abdominal adiposity as a marker of sudden cardiac death.173,174 In a multicenter, prospective study involving 14 941 patients, central adiposity was independently associated with sudden cardiac death independent of traditional risk factors.174 Epicardial adipose tissue was reported to be associated with increased incidence of premature ventricular contractions, ventricular tachycardia/fibrillation, and mortality due to sudden cardiac death.175,176

Severe Obesity and Cardiovascular Outcomes

Individuals with severe obesity (BMI ≥40 kg/m2) who are 100 or 200 pounds (45–90 kg) or more overweight have more complex CVD issues and encounter different challenges in the healthcare system.103 Among individuals with severe obesity, nearly one-third had clinical evidence of heart failure, and the probability of heart failure increased dramatically with increasing duration of severe obesity, with prevalence rates exceeding 70% at 20 years and 90% at 30 years.177 Severe obesity affects cardiac structure and function as it has been demonstrated to be associated with left ventricular remodeling and decreased left ventricular systolic function, as well as left ventricular diastolic dysfunction, which may eventually lead to heart failure.178,179 Additionally, several metabolic, inflammatory, and neurohormonal changes associated with severe obesity may contribute with impairment of the cardiovascular system.132 Histologically, cardiomyocyte hypertrophy, myocardial fat infiltration, and fibrosis have been described in severe obese hearts from postmortem studies.180 Increased pulmonary vascular resistance and pulmonary artery pressure with increase in right ventricular afterload may also occur in severe obesity, leading to the development of right ventricular dysfunction.126,181 Data suggest that weight loss achieved by any intervention may improve cardiac structure and function (through reverse remodeling) and decreased cardiovascular risk in this population. In addition, weight-loss surgery (ie, bariatric surgery) may reduce cardiac morbidity and mortality in patients with established cardiac disease.182 Although recent heart failure guidelines have not emphasized weight reduction, these guidelines recognize the high risk associated with severe obesity. Risk of developing atrial fibrillation has been reported to significantly increase with higher BMI, a BMI ≥30 kg/m2, increasing the lifetime risk of developing atrial fibrillation by 49%.127,153 Among individuals with severe obesity, those treated with weight-loss surgery had a lower rate of new-onset atrial fibrillation than did those receiving usual care.183 Severe obesity is also associated with increased mortality after in-hospital cardiac arrest due to either nonventricular fibrillation or ventricular fibrillation arrhythmias.184

Myocardial Metabolic Changes

Altered myocardial metabolism has been considered among the potential mechanisms leading to obesity-related cardiac disease.185 Obesity causes changes in myocardial metabolism, which include an increase in fatty acid oxidation and a decrease in glucose oxidation.186,187 These metabolic changes modify metabolism in the heart, culminating in abnormal cardiac substrate utilization, impaired cardiac efficiency, and decreased energy generation, producing functional consequences that are linked to the increased rate of heart failure in this population.188–190
Evidence for the role of the expansion of cardiac fat depots in the pathogenesis of myocardial metabolic diseases is accumulating.191–193 Advanced metabolic cardiac imaging (ie, magnetic resonance spectroscopy) provides a unique way of noninvasively investigating cardiometabolic consequences of excess adiposity on myocardial metabolism leading to cardiac disease.185 Using phosphorus magnetic resonance spectroscopy (31P-magnetic resonance spectroscopy), cardiac energy metabolism can be assessed noninvasively by quantifying the relative concentrations of phosphocreatine (PCr) and ATP in the myocardium to derive the PCr/ATP ratio—a sensitive index of the cardiac energetic state.188 Decreased PCr/ATP ratio is linked to cardiac dysfunction194 and is a predictor of mortality.188 In heart failure, the PCr/ATP ratio correlates with left ventricular systolic function and has been shown to be a better prognostic indicator than left ventricular ejection fraction.188,195 Using this index, a significant decline in the PCr/ATP ratio in association with increasing BMI was found.196,197 Visceral adiposity was also found to be associated with impaired myocardial energetics (ie, reduced cardiac PCr/ATP ratio) in obese diabetic men with no cardiovascular risk factors.192 Excess intramyocardial fat content (ie, cardiomyocyte accumulation of lipids) and impaired myocardial energetics have been identified as important contributors to the development of cardiac dysfunction in obese adults.198 Taken together, these results proposed interesting new areas of investigation to better understand obesity-related metabolic and cardiac diseases.

Myocardial Structure and Function Changes

Accumulating evidence shows the adverse effects of excess adiposity and weight gain on central and peripheral hemodynamics, as well as on cardiac structure and function.103,199,200 Facilitated by obesity-related comorbidities, as well as neurohormonal and metabolic alterations, these abnormalities may predispose to cardiac dysfunction and heart failure (Figure 5).57 Such maladaptation is most pronounced in individuals with visceral adiposity and severe obesity.132,201 These alterations have also been described in children and adolescents with obesity.126 Substantial weight loss from lifestyle intervention or weight-loss surgery is capable of reversing many of the alterations in cardiac performance and morphology associated with excess adiposity.132
Abnormalities of left ventricular diastolic performance are frequent in obesity; the prevalence of left ventricular diastolic dysfunction ranges from 23% to 75%, depending on the diagnostic criteria.131 As mentioned previously, left ventricular ejection fraction (ie, clinical parameter of left ventricular systolic function) has been reported to be usually preserved in obesity. However, novel sensitive measures of myocardial function with strain imaging by echocardiography and cardiac MRI have revealed subclinical alterations of cardiac function in individuals with obesity, even in the presence of a preserved left ventricular ejection fraction.202,203 The prevalence of abnormal subclinical cardiac function (abnormal myocardial strain) in obesity has been reported to range from 37% to 54%.131 Subclinical abnormalities of right ventricular function may also occur, leading to the development of right ventricular failure.204
Not only overall adiposity but also abnormal body fat distribution may lead to adverse cardiac adaptations. In the Dallas Heart Study, a cohort of 2710 participants without CVD, abdominal VAT levels measured by MRI were associated with concentric left ventricular remodeling and adverse hemodynamics.205 In a relatively healthy cohort of 1941 participants, the combination of excess VAT and lower skeletal muscle mass was associated with subclinical deterioration in left ventricular structure and left ventricular diastolic function.206 Excessive fat accumulation in visceral and ectopic sites results in local and systemic proinflammatory factors, which lead to left ventricular remodeling, concentric left ventricular hypertrophy, and ultimately diastolic and systolic cardiac failure.117,132,205 Recognition of high-risk obesities and the presence of subclinical cardiac structural/functional heart failure precursors are critical, because starting treatment at the preclinical stage may prevent heart failure progression and improve outcomes.

Double Hit: High-Risk Obesities, Type 2 Diabetes Mellitus, and Cardiovascular Outcomes

Obesity is an important driving factor for the development of type 2 diabetes mellitus.207 However, countless studies that have used either anthropometric indices of upper body/abdominal adiposity or direct imaging measurements (CT or MRI) have shown that while overall obesity increases the risk of type 2 diabetes mellitus, there is a substantial increase in risk associated with an augmented waistline, waist-to-hip ratio, or with higher levels of VAT, at any BMI level.36–40,42 Patients with type 2 diabetes mellitus have larger waistlines and more VAT than BMI-matched individuals without type 2 diabetes mellitus.208,209 Both visceral or severe obesities and type 2 diabetes mellitus predispose to cardiovascular abnormalities and CVD; their simultaneous presence should further increase the risk of cardiovascular outcomes.210 The increasing coexistence of type 2 diabetes mellitus and high-risk forms of obesity presents complex treatment challenges owing to the elevated risk of developing cardiovascular complications and heart failure. Ectopic and visceral adiposity are linked to insulin resistance, which may partly mediate the link between obesity, type 2 diabetes mellitus, and cardiovascular risk. The metabolic syndrome and insulin resistance have been recognized as a risk factor for cardiovascular morbidity and mortality.211,212 Studies have also shown that the presence of the metabolic syndrome increased to risk of developing cardiac dysfunction and heart failure.213 In the Uppsala Longitudinal Study of Adult Men, the presence of the metabolic syndrome (BMI >29.4 kg/m2 was used instead of waist circumference) increased the risk of developing heart failure >3-fold during the 20-year follow-up and remained significant after adjustment for established risk factors for heart failure.214 The MESA study also demonstrated that the metabolic syndrome predicts heart failure in a cohort free of CVD.215 Thus, the presence of features of the metabolic syndrome in type 2 diabetes mellitus further increases the risk of CVD found among people with chronically elevated glycemia and impaired plasma glucose homeostasis. As the main drivers for the presence of the metabolic syndrome are excessive amounts of VAT and of ectopic fat, it is proposed that particular attention should be given to the prevalent subgroup of patients with type 2 diabetes mellitus who are also viscerally obese and who show features of the metabolic syndrome. Type 2 diabetes mellitus accompanied by visceral adiposity and an excess of ectopic fat clearly represents a high-risk phenotype for cardiovascular outcomes.

Obesities: Implications for Assessment of Cardiometabolic Risk

Cardiometabolic imaging studies reviewed in this article suggest that in this world of precision medicine,216 we need to refine the current concept that obesity is a disease that can be assessed with the BMI. Measuring the patient’s BMI should be considered as the initial step to classify the patient into relative weight categories. Then, a waist circumference measurement has been shown to considerably refine cardiovascular risk assessment associated with a given BMI category by allowing the discrimination of patients with central, upper body, abdominal fat accumulation compared with individuals having a more uniformly distributed adipose tissue mass. As discussed above, an increased waistline for a given BMI along with the presence of metabolic syndrome features is predictive of visceral obesity and excess ectopic fat. As resources to manage obesity are not unlimited, it is proposed that attention should be given to the clinical management of this high-risk and prevalent group of overweight and moderately obese subjects. Furthermore, another rapidly expanding group of patients, individuals with severe obesity, should be highly prioritized considering the additional health burden associated with a large excess of body fat. Thus, applying the notions of precision or personalized medicine would imply that we refer to obesities rather than to obesity.

Obesities: Implications for Management by Lifestyle

Overweight/Moderate Obesity: Giving Lifestyle a Chance

Although environmental factors affecting human behaviors relevant to energy intake (overall caloric intake and diet quality, energy density, added sugar content, highly processed foods, food production, transformation and distribution systems, etc) and energy expenditure (walkability and safety of urban environments, parks, public facilities for physical activity, public transportation, etc) are not addressed by clinical weight-loss programs, many studies have shown that moderate weight loss (>5%) is associated with clinical benefits although long-term maintenance of a reduced body weight is a challenge because of the permissive obesogenic environments that we have designed for ourselves.6 In addition, because of substantial individual differences in regional adiposity, a moderate weight loss is likely to have markedly different consequences depending upon the patient’s form of obesity.217 As a general rule, lifestyle (diet and physical activity or exercise) intervention studies have shown that among individuals initially characterized by an excess of VAT and ectopic fat, there is a preferential and rapid loss of VAT and of, for instance, liver fat, associated with moderate weight loss.39,40 Furthermore, among individuals with type 2 diabetes mellitus who are viscerally obese, there is a selective loss of VAT with exercise training even in the absence of weight loss.218 This phenomenon is frequent as sedentary individuals with visceral obesity often show sarcopenia related to their sedentary behaviors. Under such circumstances, the absence of or moderate weight loss may mask or not properly capture substantial changes in body composition (increased in muscle mass and loss of VAT, liver fat, and mobilization of other ectopic fat depots).217,219 This phenomenon has been extensively studied, and for these reasons, the change in waist circumference has been recommended as a better therapeutic target than magnitude of weight loss as the former may be reduced even in the absence of weight loss.39,46 This notion is particularly relevant to patients with type 2 diabetes mellitus who are viscerally obese and who also often show liver steatosis. Change in body weight may not be sensitive enough to track changes in visceral/ectopic fat induced by a lifestyle modification program in these patients.
In addition to monitoring changes in waist, these high-risk, viscerally obese patients with features of the metabolic syndrome considerably benefit from increasing their level of cardiorespiratory fitness, which has been shown to be a powerful protective factor against CVD in individuals who are either obese or nonobese,220–223 leading to the introduction of the fat-and-fit concept.224,225 Thus, in the absence of weight loss, an increase in cardiorespiratory fitness and a reduction in waist circumference should be considered as highly desirable outcomes. Finally, both overall nutritional quality226 and level of physical activity227 have been shown to be negatively related to cardiovascular outcomes in a manner that is independent from weight loss.
In summary, among overweight and moderately obese individuals with excess visceral adiposity and ectopic fat, improving nutritional quality, increasing level of physical activity, improving cardiorespiratory fitness, and reducing waist circumference are highly desirable outcomes predictive of marked improvements in cardiometabolic risk (even in the absence of weight loss). It is, therefore, proposed to make these 4 indices lifestyle vital signs.228,229

Lifestyle Management of Severe Obesity: Does It Work?

Despite advances in medical management and lifestyle interventions, the clinical outcomes associated with cardiac disease and heart failure are worse for patients with severe obesity. There are 3 treatment options for individuals with severe obesity: (1) lifestyle modification therapy, (2) pharmacotherapy, and (3) bariatric surgery. Lifestyle modification therapy can be useful for improving cardiovascular risk factors and comorbid conditions but often fails to achieve sustainable weight loss and durable metabolic recovery. For example, individuals with severe obesity who underwent intensive lifestyle intervention in the Look AHEAD trial (Action for Health in Diabetes) and who achieved a 4-year weight loss ≥10% of initial body weight experienced significant improvements in several cardiovascular risk factors even within the context of persistent severe obesity.230 However, those who reached such an outcome represented only 25% of the sample characterized by severe obesity. The majority (75%) of individuals with severe obesity in that study did not maintain a weight loss ≥10% of initial body weight at 4 years.230 Pharmacological treatment is also of limited efficacy. Even when lifestyle modification231 or pharmacotherapy treatments are successful, the body weight loss is usually completely regained within 12 months. Lifestyle management may have a role in helping patients maintain long-term weight loss or expanding weight loss after bariatric surgery. Prospective studies have shown the effectiveness of lifestyle interventions carried out within 12 months of bariatric surgery, either pre- or postoperatively on weight loss and weight-loss maintenance.232,233 In a randomized controlled study, a structured lifestyle intervention program increased weight loss when initiated following bariatric surgery.234 Lifestyle modification thus represents a crucial aspect of obesity management, even in the context of bariatric surgery.

Pharmacological Management of Cardiometabolic Risk in Obesities

Additionally, on a background of lifestyle changes, newer pharmacological agents may facilitate both long-term weight loss and reduction in cardiovascular risk.235 Recently, new classes of glucose-lowering therapies originally developed for patients with type 2 diabetes mellitus have shown promising results for the treatment of obesity-related cardiac disease. GLP-1 (glucagon-like peptide-1) receptor agonists and SGLT2 (sodium-glucose co-transporter 2) inhibitors have demonstrated efficacy for weight loss and reduction of cardiovascular events.236,237 A meta-analysis showed that SGLT2 inhibitors, as a class, reduced the risk of cardiovascular death or hospitalization for heart failure by 23%, with a similar benefit in patients with and without atherosclerotic CVD and in patients with and without a history of heart failure.238 A 10% relative risk reduction in the 3-point major adverse CVD event (CVD death, nonfatal myocardial infarction, and nonfatal stroke) and a 13% risk reduction in cardiovascular mortality was reported in a meta-analysis of patients with type 2 diabetes mellitus using GLP-1 receptor agonists.239 The mechanisms by which SGLT2 inhibitors and GLP-1 receptor agonists modulate the cardiovascular system are unclear. The underlying mechanisms that have the greatest plausibility for both of these agents including the impact of ventricular loading conditions, direct effects on cardiac structure and function, myocardial energetics metabolism for SGLT2 inhibitors, and the antiatherosclerotic, anti-inflammatory, and modulation of endothelial function for GLP-1 receptor agonists.240 Recent studies evaluating the coadministration of SGLT2 inhibitors with other classes of drugs have also shown promising results.241
Little data are available describing the effects of these medications on visceral and ectopic fat depots. Data from small substudies within larger randomized trials of medications such as liraglutide (GLP-1 receptor agonist) have shown interesting results with greater reduction in VAT compared with subcutaneous adipose tissue. In the SUSTAIN 8 (Semaglutide Unabated Sustainability in Treatment of Type 2 Diabetes) substudy (n=178), reductions in visceral fat mass were observed in each treatment arm (GLP-1 receptor agonist semaglutide and SGLT2 inhibitor canagliflozin).242 In a prospective study of obese Japanese individuals treated with liraglutide over 24 weeks, decreases in visceral fat mass (mean reduction, 11.9%) and intrahepatic lipid content (mean reduction, 49.2%) were observed with no change in subcutaneous fat levels.243 Whether these reductions in visceral and ectopic adiposity translate to improvements in cardiovascular risk outcomes in high-risk individuals remains unknown.

Surgical Treatment of Obesities: Metabolic Surgeries

Cardiovascular Benefits

Cardiovascular benefits of weight-loss surgery, determined largely from nonrandomized studies, are being increasingly recognized. Surgical treatment of obesity is more effective than standard medical approaches to induce sustained weight loss and durable metabolic conditions/comorbidities remission.178,179,244,245 In the STAMPEDE randomized trial (Surgical Treatment and Medications Potentially Eradicate Diabetes Efficiently), weight-loss surgery was found to be superior to intensive medical therapy in terms of glycemic control, weight reduction, decreasing medication use (antidiabetic, antihypertensive, and lipid-lowering agents), and improvement in quality of life during the 5-year follow-up period. These beneficial effects were also observed among patients with mild obesity (BMI, 27–34 kg/m2).245 For severe obesity, surgery is now the preferred and currently only effective treatment modality. Surgical techniques differ in terms of morbidity and mortality rate, magnitude of weight loss, weight-loss maintenance, and rate of resolution of comorbidities over time.178 The various weight-loss surgery procedures promote weight loss through 2 general mechanisms according to the type of surgery: (1) restrictive surgeries limit the amount of food consumed by reducing the size of the stomach, (2) malabsorptive surgeries limit the absorption of nutrients by reorganizing or bypassing portions of the small intestine. Sleeve gastrectomy is the most popular restrictive procedure worldwide.246 It promotes weight loss through reduced meal volume and reduced appetite.178 Mechanistic studies have described a large number of metabolic improvements following malabsorptive surgeries, which have led to referring to these weight-loss surgery procedures as metabolic surgeries. Benefits of surgical treatment of obesity have been also demonstrated for the clinical manifestations of obesity-related cardiac disease.247–249 Both gastric bypass (ie, slightly malabsorptive surgery) and gastric sleeve (ie, restrictive surgery) significantly reduce visceral fat, with more modest reductions in subcutaneous fat levels.250
In the long term, surgical treatment of obesity is associated with reduced incidence of type 2 diabetes mellitus and with reduced overall mortality.251 In the Swedish Obese Subjects study, weight-loss surgery led to a 30% reduction in the incidence of cardiovascular events and a 50% reduction in cardiovascular deaths in individuals with severe obesity compared with those who received usual care after 15 years of follow-up.251 In a retrospective study including 6795 patients who underwent bariatric surgery, patients with severe obesity and coronary artery disease (n=249) were at greater risk of adverse cardiovascular outcomes compared with matched severely obese patients without coronary artery disease throughout the follow-up (mean, 7.4 years).252 A retrospective study of 20 235 surgical and nonsurgical patients shows that weight-loss surgery is associated to a lower incidence of coronary artery disease (acute myocardial infarction, unstable angina, percutaneous coronary intervention, or coronary artery bypass grafting).253 Observational analyses also suggest benefits after weight-loss surgery for individuals with heart failure (both preserved and reduced ejection fraction).132 In a retrospective study of 14 patients with heart failure with reduced ejection fraction who underwent weight-loss surgery, a significant reduction in BMI was found (50.8±2.04 to 36.8±1.72 kg/m2) with a significant improvement in left ventricular ejection fraction at 6 months (from 23±2% to 32±4%).254 Similarly, in another retrospective study, 12 patients with severe obesity and heart failure with reduced ejection fraction who underwent weight-loss surgery had improvement in left ventricular ejection fraction following surgery.255 In both of these reports, patients also had improvements in New York Heart Association functional class. Current guidelines proposed surgical treatment of obesity for adults with BMI ≥40 or ≥35 kg/m2 with obesity-related comorbidities such as systemic hypertension, type 2 diabetes mellitus, and obstructive sleep apnea that are difficult to control with lifestyle and pharmacotherapy. No guideline suggests a given procedure to be more appropriate than others for individuals with cardiac conditions.

Mechanistic Insight on the Impact of Weight-Loss Surgery

Our studies and collaborations on bariatric surgery over the past few years have allowed us to examine the dynamic nature of the relationship between severe obesity and cardiometabolic abnormalities. An interesting feature of bariatric surgery is the rapid remission of type 2 diabetes mellitus and improvement in insulin sensitivity, which typically occurs within days of the surgery, often in the absence of pronounced weight loss.256–258 A significant decrease in post-glucose or postprandial glycemia is observed a few days after surgery.259 This is accompanied by reduced insulin levels, as well as increased GLP-1, GIP (glucose-sensitive insulinotropic peptide), and pancreatic polypeptide secretion.259 Decreases in homeostasis model assessment–insulin resistance index and increases in glucose disposition index are also observed.259,260 However, peripheral insulin sensitivity measured with a euglycemic-hyperinsulinemic clamp is not significantly improved in the days following bariatric surgery.260 Interestingly, the blunted glucose excursion is particularly dramatic with malabsorptive surgeries such as the biliopancreatic diversion with duodenal switch, in which biliopancreatic secretions are channeled to the last 100 cm of ileum, allowing a much shorter time for digestion in a section of the gut that is initially nutrient naive.259,260 Hence, bypass of a significant portion of the gut represents one of the mechanisms driving the short-term improvements in glucose-insulin homeostasis. However, the most significant mechanism explaining such an early effect after surgery is fasting. For example, when unoperated individuals with severe obesity underwent a fasting protocol similar to that of patients who are operated, we found that the improvements in insulin sensitivity and β-cell function were recapitulated in large part.256 The significant increase in postprandial GLP-1 secretion, which is observed with both malabsorptive and restrictive surgeries,256,259,261 may mediate part of this effect. At such an early stage after surgery, weight loss is often minimal. Accordingly, early metabolic improvements are observed in the absence of changes in markers of adipose tissue function including subcutaneous adipocyte size and secretion of adipose-derived cytokines such as adiponectin and tumor necrosis factor-α.259,260 Despite this apparent dissociation between insulin resistance and adipose tissue dysfunction, important changes in substrate flux are observed in the early postsurgical period, in particular with nonesterified fatty acids. In a recent study performed 10 to 12 days after sleeve gastrectomy or biliopancreatic diversion with duodenal switch, Carreau et al262 used positron emission tomography coupled with CT imaging and showed a substantial reduction in cardiac fatty acid uptake over 6 hours in the postprandial state. Cardiac uptake values actually reached the range observed in healthy normal-weight individuals in all but 1 participant. At the same time, systemic fatty acid spillover was significantly decreased and uptake in the visceral fat compartment was increased. Interestingly, fatty acid uptake in the liver and subcutaneous adipose tissue, either in the thigh or abdomen, was unchanged.262
In the longer term after bariatric surgery, our studies and those of others have shown that surgery-induced weight loss is accompanied by extensive adipose tissue remodeling and improved adipose tissue-derived cytokine secretion, which can be measured starting around 3 months and up to 1 year after surgery and beyond.260,263 For example, our survey of the literature on adipose tissue and bariatric surgery indicated increases in anti-inflammatory factors such as adiponectin and omentin, as well as decreases in interleukin-6, C-reactive protein, visfatin, chemerin, and others.263 Our serial analysis of subcutaneous adipose tissue over the course of 1 year post-surgery indicates a reduction in the proportion of large subcutaneous adipocytes and median cell size, which can be measured 3 months and 1 year after biliopancreatic diversion with duodenal switch. Interestingly, smaller subcutaneous adipocyte size predicted better glycemic control 1 year after surgery.260
These results suggest that in the short term, even in the absence of extensive adipose tissue remodeling, bariatric surgery rapidly improves hepatic insulin sensitivity and glycemic control—a phenomenon that is due to changes in nutrient channeling and absorption, increased incretin effects, as well as acute caloric restriction (Figure 6). This is associated with improvements in fatty acid metabolism including a reduction in cardiac uptake and systemic spillover, as well as increased uptake in VAT.264 Longer term effects involve weight loss and extensive adipose tissue remodeling including reduced subcutaneous adipocyte size and improved adipose tissue-derived cytokines, which is associated with improved peripheral insulin sensitivity.260,263
Figure 6. Representation of the short- and long-term effects of bariatric surgery on glucose and insulin homeostasis and some of the underlying mediators. Short-term effects typically occur within days after bariatric surgery in the absence of pronounced weight loss, whereas long-term effects appear a few months to a year after surgery, in conjunction with weight loss and extensive adipose tissue remodeling, among other mechanisms.

High-Risk Obesities: From Precision Medicine to Precision Population Health?

Considerable knowledge relevant to the biology of obesities has been gained over the last 40 years. Unfortunately, such information has had little impact on obesity prevalence rates worldwide. It is, therefore, clear that although we better understand the biology of energy balance, we have not been in a position to properly decipher the complex interactions among environmental factors affecting behaviors (eg, choice and consumption of highly processed foods determined, among other factors, by food systems and their availability/marketing, level of physical activity affected by permissive environments and socioeconomic factors, psychosocial issues, quality of sleep, etc) modulating energy intake and expenditure. Thus, it is not surprising that classical clinical intervention studies focusing on the individual and not on his/her living environment have had limited long-term success. Large studies incorporating individual and environmental data are underway to hopefully design and evaluate community interventions that will not only target individuals but also features of their living environments and communities. By having access to massive data and with the help of artificial intelligence, it is hoped that the study of complex phenomena such as obesities will be facilitated, leading to the development of clinical solutions better aligned to population-based approaches—a concept described as precision lifestyle medicine,228 a notion embracing precision population health.265

Acknowledgments

We would like to acknowledge the support of many colleagues/students/ residents who have contributed to the work from their institution relevant to this article. We also want to acknowledge the generous help and trust of our study subjects and patients.

Footnote

Nonstandard Abbreviations and Acronyms

BMI
body mass index
CARDIA
Coronary Artery Risk Development in Young Adults
CT
computed tomography
CVD
cardiovascular disease
GIP
glucose-sensitive insulinotropic peptide
GLP-1
glucagon-like peptide-1HDLhigh-density lipoprotein
HFpEF
heart failure with preserved ejection fraction
LDL
low-density lipoprotein
Look AHEAD
Action for Health in Diabetes
MADIT II
Multicenter Automatic Defibrillator Implantation Trial II
MESA
Multi-Ethnic Study of Atherosclerosis
MRI
magnetic resonance imaging
PCr
phosphocreatine
SGLT2
sodium-glucose co-transporter 2
STAMPEDE
Surgical Treatment and Medications Potentially Eradicate Diabetes Efficiently
SUSTAIN
Semaglutide Unabated Sustainability in Treatment of Type 2 Diabetes
TOPCAT
Treatment of Preserved Cardiac Function Heart Failure With an Aldosterone Antagonist
VAT
visceral adipose tissue

Supplemental Material

File (res_despres_correction.pdf)

References

1.
González-Muniesa P, Mártinez-González MA, Hu FB, Després JP, Matsuzawa Y, Loos RJF, Moreno LA, Bray GA, Martinez JA. Obesity. Nat Rev Dis Primers. 2017;3:17034. doi: 10.1038/nrdp.2017.34
2.
WHO Expert Consultation. Appropriate body-mass index for Asian populations and its implications for policy and intervention strategies. Lancet. 2004;363:157–163. doi: 10.1016/S0140-6736(03)15268-3
3.
Di Angelantonio E, Bhupathiraju ShN, Wormser D, Gao P, Kaptoge S, Berrington de Gonzalez A, Cairns BJ, Huxley R, Jackson ChL, Joshy G, et al; Global, B. M. I. Mortality Collaboration. Body-mass index and all-cause mortality: individual-participant-data meta-analysis of 239 prospective studies in four continents. Lancet. 2016;388:776–786. doi: 10.1016/S0140-6736(16)30175-1
4.
Jensen MD, Ryan DH, Apovian CM, Ard JD, Comuzzie AG, Donato KA, Hu FB, Hubbard VS, Jakicic JM, Kushner RF, et al; American College of Cardiology/American Heart Association Task Force on Practice Guidelines; Obesity Society. 2013 AHA/ACC/TOS guideline for the management of overweight and obesity in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and The Obesity Society. Circulation. 2014;129:S102–S138. doi: 10.1161/01.cir.0000437739.71477.ee
5.
Keys A, Fidanza F, Karvonen MJ, Kimura N, Taylor HL. Indices of relative weight and obesity. J Chronic Dis. 1972;25:329–343. doi: 10.1016/0021-9681(72)90027-6
6.
Blüher M. Obesity: global epidemiology and pathogenesis. Nat Rev Endocrinol. 2019;15:288–298. doi: 10.1038/s41574-019-0176-8
7.
Afshin A, Forouzanfar MH, Reitsma MB, Sur P, Estep K, Lee A, Marczak L, Mokdad AH, Moradi-Lakeh M, Naghavi M, et al; GBD 2015 Obesity Collaborators. Health effects of overweight and obesity in 195 countries over 25 years. N Engl J Med. 2017;377:13–27. doi: 10.1056/NEJMoa1614362
8.
Hales CM, Fryar CD, Carroll MD, Freedman DS, Ogden CL. Trends in obesity and severe obesity prevalence in US youth and adults by sex and age, 2007-2008 to 2015-2016. JAMA. 2018;319:1723–1725. doi: 10.1001/jama.2018.3060
9.
Flegal KM, Kruszon-Moran D, Carroll MD, Fryar CD, Ogden CL. Trends in obesity among adults in the United States, 2005 to 2014. JAMA. 2016;315:2284–2291. doi: 10.1001/jama.2016.6458
10.
Curioni CC, Lourenço PM. Long-term weight loss after diet and exercise: a systematic review. Int J Obes (Lond). 2005;29:1168–1174. doi: 10.1038/sj.ijo.0803015
11.
Dombrowski SU, Knittle K, Avenell A, Araújo-Soares V, Sniehotta FF. Long term maintenance of weight loss with non-surgical interventions in obese adults: systematic review and meta-analyses of randomised controlled trials. BMJ. 2014;348:g2646. doi: 10.1136/bmj.g2646
12.
MacLean PS, Wing RR, Davidson T, Epstein L, Goodpaster B, Hall KD, Levin BE, Perri MG, Rolls BJ, Rosenbaum M, et al. NIH working group report: innovative research to improve maintenance of weight loss. Obesity (Silver Spring). 2015;23:7–15. doi: 10.1002/oby.20967
13.
Swinburn BA, Sacks G, Hall KD, McPherson K, Finegood DT, Moodie ML, Gortmaker SL. The global obesity pandemic: shaped by global drivers and local environments. Lancet. 2011;378:804–814. doi: 10.1016/S0140-6736(11)60813-1
14.
Portha B, Grandjean V, Movassat J. Mother or father: who is in the front line? Mechanisms underlying the non-genomic transmission of obesity/diabetes via the maternal or the paternal line. Nutrients. 2019;11:E233. doi: 10.3390/nu11020233
15.
Wormser D, Kaptoge S, Di Angelantonio E, Wood AM, Pennells L, Thompson A, Sarwar N, Kizer JR, Lawlor DA, Nordestgaard BG, et al. Separate and combined associations of body-mass index and abdominal adiposity with cardiovascular disease: collaborative analysis of 58 prospective studies. Lancet. 2011;377:1085–1095. doi: 10.1016/S0140-6736(11)60105-0
16.
Bastien M, Poirier P, Lemieux I, Després JP. Overview of epidemiology and contribution of obesity to cardiovascular disease. Prog Cardiovasc Dis. 2014;56:369–381. doi: 10.1016/j.pcad.2013.10.016
17.
Cornier MA, Després JP, Davis N, Grossniklaus DA, Klein S, Lamarche B, Lopez-Jimenez F, Rao G, St-Onge MP, Towfighi A, et al; American Heart Association Obesity Committee of the Council on Nutrition, Physical Activity and Metabolism; Council on Arteriosclerosis, Thrombosis and Vascular Biology; Council on Cardiovascular Disease in the Young; Council on Cardiovascular Radiology and Intervention; Council on Cardiovascular Nursing; Council on Epidemiology and Prevention; Council on the Kidney in Cardiovascular Disease, and Stroke Council. Assessing adiposity: a scientific statement from the American Heart Association. Circulation. 2011;124:1996–2019. doi: 10.1161/CIR.0b013e318233bc6a
18.
Lavie CJ, De Schutter A, Patel DA, Romero-Corral A, Artham SM, Milani RV. Body composition and survival in stable coronary heart disease: impact of lean mass index and body fat in the “obesity paradox”. J Am Coll Cardiol. 2012;60:1374–1380. doi: 10.1016/j.jacc.2012.05.037
19.
Romero-Corral A, Montori VM, Somers VK, Korinek J, Thomas RJ, Allison TG, Mookadam F, Lopez-Jimenez F. Association of bodyweight with total mortality and with cardiovascular events in coronary artery disease: a systematic review of cohort studies. Lancet. 2006;368:666–678. doi: 10.1016/S0140-6736(06)69251-9
20.
Romero-Corral A, Somers VK, Sierra-Johnson J, Jensen MD, Thomas RJ, Squires RW, Allison TG, Korinek J, Lopez-Jimenez F. Diagnostic performance of body mass index to detect obesity in patients with coronary artery disease. Eur Heart J. 2007;28:2087–2093. doi: 10.1093/eurheartj/ehm243
21.
Antonopoulos AS, Oikonomou EK, Antoniades C, Tousoulis D. From the BMI paradox to the obesity paradox: the obesity-mortality association in coronary heart disease. Obes Rev. 2016;17:989–1000. doi: 10.1111/obr.12440
22.
Elagizi A, Kachur S, Lavie CJ, Carbone S, Pandey A, Ortega FB, Milani RV. An overview and update on obesity and the obesity paradox in cardiovascular diseases. Prog Cardiovasc Dis. 2018;61:142–150. doi: 10.1016/j.pcad.2018.07.003
23.
Lavie CJ, De Schutter A, Milani RV. Healthy obese versus unhealthy lean: the obesity paradox. Nat Rev Endocrinol. 2015;11:55–62. doi: 10.1038/nrendo.2014.165
24.
Neeland IJ, Poirier P, Després JP. Cardiovascular and metabolic heterogeneity of obesity: clinical challenges and implications for management. Circulation. 2018;137:1391–1406. doi: 10.1161/CIRCULATIONAHA.117.029617
25.
Xia JY, Lloyd-Jones DM, Khan SS. Association of body mass index with mortality in cardiovascular disease: new insights into the obesity paradox from multiple perspectives. Trends Cardiovasc Med. 2019;29:220–225. doi: 10.1016/j.tcm.2018.08.006
26.
Kalkhoff RK, Hartz AH, Rupley D, Kissebah AH, Kelber S. Relationship of body fat distribution to blood pressure, carbohydrate tolerance, and plasma lipids in healthy obese women. J Lab Clin Med. 1983;102:621–627.
27.
Kissebah AH, Vydelingum N, Murray R, Evans DJ, Hartz AJ, Kalkhoff RK, Adams PW. Relation of body fat distribution to metabolic complications of obesity. J Clin Endocrinol Metab. 1982;54:254–260. doi: 10.1210/jcem-54-2-254
28.
Krotkiewski M, Björntorp P, Sjöström L, Smith U. Impact of obesity on metabolism in men and women. Importance of regional adipose tissue distribution. J Clin Invest. 1983;72:1150–1162. doi: 10.1172/JCI111040
29.
Borkan GA, Gerzof SG, Robbins AH, Hults DE, Silbert CK, Silbert JE. Assessment of abdominal fat content by computed tomography. Am J Clin Nutr. 1982;36:172–177. doi: 10.1093/ajcn/36.1.172
30.
Sjöström L, Kvist H, Cederblad A, Tylén U. Determination of total adipose tissue and body fat in women by computed tomography, 40K, and tritium. Am J Physiol. 1986;250:E736–E745. doi: 10.1152/ajpendo.1986.250.6.E736
31.
Tokunaga K, Matsuzawa Y, Ishikawa K, Tarui S. A novel technique for the determination of body fat by computed tomography. Int J Obes. 1983;7:437–445.
32.
Ferland M, Després JP, Tremblay A, Pinault S, Nadeau A, Moorjani S, Lupien PJ, Thériault G, Bouchard C. Assessment of adipose tissue distribution by computed axial tomography in obese women: association with body density and anthropometric measurements. Br J Nutr. 1989;61:139–148. doi: 10.1079/bjn19890104
33.
Ross R, Goodpaster B, Kelley D, Boada F. Magnetic resonance imaging in human body composition research. From quantitative to qualitative tissue measurement. Ann N Y Acad Sci. 2000;904:12–17. doi: 10.1111/j.1749-6632.2000.tb06415.x
34.
Seidell JC, Bakker CJ, van der Kooy K. Imaging techniques for measuring adipose-tissue distribution–a comparison between computed tomography and 1.5-T magnetic resonance. Am J Clin Nutr. 1990;51:953–957. doi: 10.1093/ajcn/51.6.953
35.
Després JP, Nadeau A, Tremblay A, Ferland M, Moorjani S, Lupien PJ, Thériault G, Pinault S, Bouchard C. Role of deep abdominal fat in the association between regional adipose tissue distribution and glucose tolerance in obese women. Diabetes. 1989;38:304–309. doi: 10.2337/diab.38.3.304
36.
Després JP, Moorjani S, Lupien PJ, Tremblay A, Nadeau A, Bouchard C. Regional distribution of body fat, plasma lipoproteins, and cardiovascular disease. Arteriosclerosis. 1990;10:497–511. doi: 10.1161/01.atv.10.4.497
37.
Després JP, Lemieux I. Abdominal obesity and metabolic syndrome. Nature. 2006;444:881–887. doi: 10.1038/nature05488
38.
Després JP, Lemieux I, Bergeron J, Pibarot P, Mathieu P, Larose E, Rodés-Cabau J, Bertrand OF, Poirier P. Abdominal obesity and the metabolic syndrome: contribution to global cardiometabolic risk. Arterioscler Thromb Vasc Biol. 2008;28:1039–1049. doi: 10.1161/ATVBAHA.107.159228
39.
Després JP. Body fat distribution and risk of cardiovascular disease: an update. Circulation. 2012;126:1301–1313. doi: 10.1161/CIRCULATIONAHA.111.067264
40.
Neeland IJ, Ross R, Després JP, Matsuzawa Y, Yamashita S, Shai I, Seidell J, Magni P, Santos RD, Arsenault B, et al; International Atherosclerosis Society; International Chair on Cardiometabolic Risk Working Group on Visceral Obesity. Visceral and ectopic fat, atherosclerosis, and cardiometabolic disease: a position statement. Lancet Diabetes Endocrinol. 2019;7:715–725. doi: 10.1016/S2213-8587(19)30084-1
41.
Smith U. Abdominal obesity: a marker of ectopic fat accumulation. J Clin Invest. 2015;125:1790–1792. doi: 10.1172/JCI81507
42.
Tchernof A, Després JP. Pathophysiology of human visceral obesity: an update. Physiol Rev. 2013;93:359–404. doi: 10.1152/physrev.00033.2011
43.
Pouliot MC, Després JP, Lemieux S, Moorjani S, Bouchard C, Tremblay A, Nadeau A, Lupien PJ. Waist circumference and abdominal sagittal diameter: best simple anthropometric indexes of abdominal visceral adipose tissue accumulation and related cardiovascular risk in men and women. Am J Cardiol. 1994;73:460–468. doi: 10.1016/0002-9149(94)90676-9
44.
Després JP. Excess visceral adipose tissue/ectopic fat the missing link in the obesity paradox? J Am Coll Cardiol. 2011;57:1887–1889. doi: 10.1016/j.jacc.2010.10.063
45.
Nazare JA, Smith J, Borel AL, Aschner P, Barter P, Van Gaal L, Tan CE, Wittchen HU, Matsuzawa Y, Kadowaki T, et al; INSPIRE ME IAA Investigators. Usefulness of measuring both body mass index and waist circumference for the estimation of visceral adiposity and related cardiometabolic risk profile (from the INSPIRE ME IAA study). Am J Cardiol. 2015;115:307–315. doi: 10.1016/j.amjcard.2014.10.039
46.
Ross R, Neeland IJ, Yamashita S, Shai I, Seidell J, Magni P, Santos RD, Arsenault B, Cuevas A, Hu FB, et al. Waist circumference as a vital sign in clinical practice: a consensus statement from the IAS and ICCR Working Group on Visceral Obesity. Nat Rev Endocrinol. 2020;16:177–189. doi: 10.1038/s41574-019-0310-7
47.
Cerhan JR, Moore SC, Jacobs EJ, Kitahara CM, Rosenberg PS, Adami HO, Ebbert JO, English DR, Gapstur SM, Giles GG, et al. A pooled analysis of waist circumference and mortality in 650,000 adults. Mayo Clin Proc. 2014;89:335–345. doi: 10.1016/j.mayocp.2013.11.011
48.
Arsenault BJ, Lemieux I, Després JP, Wareham NJ, Kastelein JJ, Khaw KT, Boekholdt SM. The hypertriglyceridemic-waist phenotype and the risk of coronary artery disease: results from the EPIC-Norfolk prospective population study. CMAJ. 2010;182:1427–1432. doi: 10.1503/cmaj.091276
49.
LeBlanc S, Coulombe F, Bertrand OF, Bibeau K, Pibarot P, Marette A, Alméras N, Lemieux I, Després JP, Larose E. Hypertriglyceridemic waist: a simple marker of high-risk atherosclerosis features associated with excess visceral adiposity/ectopic fat. J Amer Heart Assoc. 2018;7:e008139. doi: 10.1161/JAHA.117.008139
50.
Lemieux I, Pascot A, Couillard C, Lamarche B, Tchernof A, Alméras N, Bergeron J, Gaudet D, Tremblay G, Prud’homme D, et al. Hypertriglyceridemic waist: a marker of the atherogenic metabolic triad (hyperinsulinemia; hyperapolipoprotein B; small, dense LDL) in men? Circulation. 2000;102:179–184. doi: 10.1161/01.cir.102.2.179
51.
Lemieux I, Poirier P, Bergeron J, Alméras N, Lamarche B, Cantin B, Dagenais GR, Després JP. Hypertriglyceridemic waist: a useful screening phenotype in preventive cardiology? Can J Cardiol. 2007;23 Suppl B:23B–31B. doi: 10.1016/s0828-282x(07)71007-3
52.
Ma CM, Liu XL, Lu N, Wang R, Lu Q, Yin FZ. Hypertriglyceridemic waist phenotype and abnormal glucose metabolism: a system review and meta-analysis. Endocrine. 2019;64:469–485. doi: 10.1007/s12020-019-01945-6
53.
Kahn HS. The lipid accumulation product is better than BMI for identifying diabetes: a population-based comparison. Diabetes Care. 2006;29:151–153. doi: 10.2337/diacare.29.1.151
54.
Kahn HS. The “lipid accumulation product” performs better than the body mass index for recognizing cardiovascular risk: a population-based comparison. BMC Cardiovasc Disord. 2005;5:26. doi: 10.1186/1471-2261-5-26
55.
Amato MC, Giordano C, Galia M, Criscimanna A, Vitabile S, Midiri M, Galluzzo A; AlkaMeSy Study Group. Visceral adiposity index: a reliable indicator of visceral fat function associated with cardiometabolic risk. Diabetes Care. 2010;33:920–922. doi: 10.2337/dc09-1825
56.
Sturm R, Hattori A. Morbid obesity rates continue to rise rapidly in the United States. Int J Obes (Lond). 2013;37:889–891. doi: 10.1038/ijo.2012.159
57.
Piché ME, Poirier P, Lemieux I, Després JP. Overview of epidemiology and contribution of obesity and body fat distribution to cardiovascular disease: an update. Prog Cardiovasc Dis. 2018;61:103–113. doi: 10.1016/j.pcad.2018.06.004
58.
Smith GI, Mittendorfer B, Klein S. Metabolically healthy obesity: facts and fantasies. J Clin Invest. 2019;129:3978–3989. doi: 10.1172/JCI129186
59.
Blüher M, Bashan N, Shai I, Harman-Boehm I, Tarnovscki T, Avinaoch E, Stumvoll M, Dietrich A, Klöting N, Rudich A. Activated ask1-MKK4-p38MAPK/JNK stress signaling pathway in human omental fat tissue may link macrophage infiltration to whole-body Insulin sensitivity. J Clin Endocrinol Metab. 2009;94:2507–2515. doi: 10.1210/jc.2009-0002
60.
Kovsan J, Blüher M, Tarnovscki T, Klöting N, Kirshtein B, Madar L, Shai I, Golan R, Harman-Boehm I, Schön MR, et al. Altered autophagy in human adipose tissues in obesity. J Clin Endocrinol Metab. 2011;96:E268–E277. doi: 10.1210/jc.2010-1681
61.
Michaud A, Tordjman J, Pelletier M, Liu Y, Laforest S, Noël S, Le Naour G, Bouchard C, Clément K, Tchernof A. Relevance of omental pericellular adipose tissue collagen in the pathophysiology of human abdominal obesity and related cardiometabolic risk. Int J Obes (Lond). 2016;40:1823–1831. doi: 10.1038/ijo.2016.173
62.
Laforest S, Labrecque J, Michaud A, Cianflone K, Tchernof A. Adipocyte size as a determinant of metabolic disease and adipose tissue dysfunction. Crit Rev Clin Lab Sci. 2015;52:301–313. doi: 10.3109/10408363.2015.1041582
63.
Michaud A, Drolet R, Noël S, Paris G, Tchernof A. Visceral fat accumulation is an indicator of adipose tissue macrophage infiltration in women. Metabolism. 2012;61:689–698. doi: 10.1016/j.metabol.2011.10.004
64.
Harman-Boehm I, Blüher M, Redel H, Sion-Vardy N, Ovadia S, Avinoach E, Shai I, Klöting N, Stumvoll M, Bashan N, et al. Macrophage infiltration into omental versus subcutaneous fat across different populations: effect of regional adiposity and the comorbidities of obesity. J Clin Endocrinol Metab. 2007;92:2240–2247. doi: 10.1210/jc.2006-1811
65.
Spalding KL, Arner E, Westermark PO, Bernard S, Buchholz BA, Bergmann O, Blomqvist L, Hoffstedt J, Näslund E, Britton T, et al. Dynamics of fat cell turnover in humans. Nature. 2008;453:783–787. doi: 10.1038/nature06902
66.
Arner E, Westermark PO, Spalding KL, Britton T, Rydén M, Frisén J, Bernard S, Arner P. Adipocyte turnover: relevance to human adipose tissue morphology. Diabetes. 2010;59:105–109. doi: 10.2337/db09-0942
67.
Lessard J, Tchernof A. Depot- and obesity-related differences in adipogenesis. Clin Lipidol. 2012;7:587–596.
68.
Lessard J, Laforest S, Pelletier M, Leboeuf M, Blackburn L, Tchernof A. Low abdominal subcutaneous preadipocyte adipogenesis is associated with visceral obesity, visceral adipocyte hypertrophy, and a dysmetabolic state. Adipocyte. 2014;3:197–205. doi: 10.4161/adip.29385
69.
Pap A, Cuaranta-Monroy I, Peloquin M, Nagy L. Is the mouse a good model of human PPARgamma-related metabolic diseases? Int J Mol Sci. 2016;17:E1236. doi: 10.3390/ijms17081236
70.
Rochford JJ. Mouse models of lipodystrophy and their significance in understanding fat regulation. Curr Top Dev Biol. 2014;109:53–96. doi: 10.1016/B978-0-12-397920-9.00005-6
71.
Hussain I, Garg A. Lipodystrophy syndromes. Endocrinol Metab Clin North Am. 2016;45:783–797. doi: 10.1016/j.ecl.2016.06.012
72.
Garg A. Lipodystrophies. Am J Med. 2000;108:143–152. doi: 10.1016/s0002-9343(99)00414-3
73.
Nadeau M, Noël S, Laberge PY, Hurtubise J, Tchernof A. Adipose tissue lamin A/C messenger RNA expression in women. Metabolism. 2010;59:1106–1114. doi: 10.1016/j.metabol.2009.09.034
74.
Park HT, Lee ES, Cheon YP, Lee DR, Yang KS, Kim YT, Hur JY, Kim SH, Lee KW, Kim T. The relationship between fat depot-specific preadipocyte differentiation and metabolic syndrome in obese women. Clin Endocrinol (Oxf). 2012;76:59–66. doi: 10.1111/j.1365-2265.2011.04141.x
75.
Gustafson B, Nerstedt A, Smith U. Reduced subcutaneous adipogenesis in human hypertrophic obesity is linked to senescent precursor cells. Nat Commun. 2019;10:2757. doi: 10.1038/s41467-019-10688-x
76.
Palmer AK, Gustafson B, Kirkland JL, Smith U. Cellular senescence: at the nexus between ageing and diabetes. Diabetologia. 2019;62:1835–1841. doi: 10.1007/s00125-019-4934-x
77.
Ruiz-Ojeda FJ, Mendez-Gutierrez A, Aguilera CM, Plaza-Diaz J. Extracellular matrix remodeling of adipose tissue in obesity and metabolic diseases. Int J Mol Sci. 2019;20:4888. doi: 10.3390/ijms20194888
78.
Lin D, Chun TH, Kang L. Adipose extracellular matrix remodelling in obesity and insulin resistance. Biochem Pharmacol. 2016;119:8–16. doi: 10.1016/j.bcp.2016.05.005
79.
Divoux A, Tordjman J, Lacasa D, Veyrie N, Hugol D, Aissat A, Basdevant A, Guerre-Millo M, Poitou C, Zucker JD, et al. Fibrosis in human adipose tissue: composition, distribution, and link with lipid metabolism and fat mass loss. Diabetes. 2010;59:2817–2825. doi: 10.2337/db10-0585
80.
Divoux A, Clément K. Architecture and the extracellular matrix: the still unappreciated components of the adipose tissue. Obes Rev. 2011;12:e494–e503. doi: 10.1111/j.1467-789X.2010.00811.x
81.
Kohlgruber A, Lynch L. Adipose tissue inflammation in the pathogenesis of type 2 diabetes. Curr Diab Rep. 2015;15:92. doi: 10.1007/s11892-015-0670-x
82.
Vijay J, Gauthier MF, Biswell RL, Louiselle DA, Johnston JJ, Cheung WA, Belden B, Pramatarova A, Biertho L, Gibson M, et al; Multiple Tissue Human Expression Resource Consortium. Single-cell analysis of human adipose tissue identifies depot and disease specific cell types. Nat Metab. 2020;2:97–109. doi: 10.1038/s42255-019-0152-6
83.
Païssé S, Valle C, Servant F, Courtney M, Burcelin R, Amar J, Lelouvier B. Comprehensive description of blood microbiome from healthy donors assessed by 16S targeted metagenomic sequencing. Transfusion. 2016;56:1138–1147. doi: 10.1111/trf.13477
84.
Lluch J, Servant F, Païssé S, Valle C, Valière S, Kuchly C, Vilchez G, Donnadieu C, Courtney M, Burcelin R, et al. The characterization of novel tissue microbiota using an optimized 16S metagenomic sequencing pipeline. PLoS One. 2015;10:e0142334. doi: 10.1371/journal.pone.0142334
85.
Amar J, Chabo C, Waget A, Klopp P, Vachoux C, Bermúdez-Humarán LG, Smirnova N, Bergé M, Sulpice T, Lahtinen S, et al. Intestinal mucosal adherence and translocation of commensal bacteria at the early onset of type 2 diabetes: molecular mechanisms and probiotic treatment. EMBO Mol Med. 2011;3:559–572. doi: 10.1002/emmm.201100159
86.
Lelouvier B, Servant F, Païssé S, Brunet AC, Benyahya S, Serino M, Valle C, Ortiz MR, Puig J, Courtney M, et al. Changes in blood microbiota profiles associated with liver fibrosis in obese patients: a pilot analysis. Hepatology. 2016;64:2015–2027. doi: 10.1002/hep.28829
87.
Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, Burcelin R. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes. 2008;57:1470–1481. doi: 10.2337/db07-1403
88.
Schertzer JD, Tamrakar AK, Magalhães JG, Pereira S, Bilan PJ, Fullerton MD, Liu Z, Steinberg GR, Giacca A, Philpott DJ, et al. NOD1 activators link innate immunity to insulin resistance. Diabetes. 2011;60:2206–2215. doi: 10.2337/db11-0004
89.
Cavallari JF, Fullerton MD, Duggan BM, Foley KP, Denou E, Smith BK, Desjardins EM, Henriksbo BD, Kim KJ, Tuinema BR, et al. Muramyl dipeptide-based postbiotics mitigate obesity-induced insulin resistance via IRF4. Cell Metab. 2017;25:1063–1074.e3. doi: 10.1016/j.cmet.2017.03.021
90.
Denou E, Lolmède K, Garidou L, Pomie C, Chabo C, Lau TC, Fullerton MD, Nigro G, Zakaroff-Girard A, Luche E, et al. Defective NOD2 peptidoglycan sensing promotes diet-induced inflammation, dysbiosis, and insulin resistance. EMBO Mol Med. 2015;7:259–274. doi: 10.15252/emmm.201404169
91.
Anhê FF, Jensen BAH, Varin TV, et al. Type 2 diabetes influences bacterial tissue compartmentalisation in human obesity. Nature Metab. 2020;2:233–242.
92.
Cani PD, Van Hul M. Microbial signatures in metabolic tissues: a novel paradigm for obesity and diabetes? Nature Metab. 2020;2:211–212.
93.
Noll C, Carpentier AC. Dietary fatty acid metabolism in prediabetes. Curr Opin Lipidol. 2017;28:1–10. doi: 10.1097/MOL.0000000000000369
94.
Fabbrini E, Magkos F, Mohammed BS, Pietka T, Abumrad NA, Patterson BW, Okunade A, Klein S. Intrahepatic fat, not visceral fat, is linked with metabolic complications of obesity. Proc Natl Acad Sci U S A. 2009;106:15430–15435. doi: 10.1073/pnas.0904944106
95.
Stefan N, Häring HU, Cusi K. Non-alcoholic fatty liver disease: causes, diagnosis, cardiometabolic consequences, and treatment strategies. Lancet Diabetes Endocrinol. 2019;7:313–324. doi: 10.1016/S2213-8587(18)30154-2
96.
Lauridsen BK, Stender S, Kristensen TS, Kofoed KF, Køber L, Nordestgaard BG, Tybjærg-Hansen A. Liver fat content, non-alcoholic fatty liver disease, and ischaemic heart disease: mendelian randomization and meta-analysis of 279 013 individuals. Eur Heart J. 2018;39:385–393. doi: 10.1093/eurheartj/ehx662
97.
Kelley DE, McKolanis TM, Hegazi RA, Kuller LH, Kalhan SC. Fatty liver in type 2 diabetes mellitus: relation to regional adiposity, fatty acids, and insulin resistance. Am J Physiol Endocrinol Metab. 2003;285:E906–E916. doi: 10.1152/ajpendo.00117.2003
98.
Liu J, Fox CS, Hickson D, Bidulescu A, Carr JJ, Taylor HA. Fatty liver, abdominal visceral fat, and cardiometabolic risk factors: the Jackson Heart Study. Arterioscler Thromb Vasc Biol. 2011;31:2715–2722. doi: 10.1161/ATVBAHA.111.234062
99.
Bouchard C, Tremblay A, Després JP, Nadeau A, Lupien PJ, Thériault G, Dussault J, Moorjani S, Pinault S, Fournier G. The response to long-term overfeeding in identical twins. N Engl J Med. 1990;322:1477–1482. doi: 10.1056/NEJM199005243222101
100.
Karlsson T, Rask-Andersen M, Pan G, Höglund J, Wadelius C, Ek WE, Johansson Å. Contribution of genetics to visceral adiposity and its relation to cardiovascular and metabolic disease. Nat Med. 2019;25:1390–1395. doi: 10.1038/s41591-019-0563-7
101.
Ji Y, Yiorkas AM, Frau F, Mook-Kanamori D, Staiger H, Thomas EL, Atabaki-Pasdar N, Campbell A, Tyrrell J, Jones SE, et al. Genome-wide and abdominal MRI data provide evidence that a genetically determined favorable adiposity phenotype is characterized by lower ectopic liver fat and lower risk of type 2 diabetes, heart disease, and hypertension. Diabetes. 2019;68:207–219. doi: 10.2337/db18-0708
102.
Loos RJF, Kilpeläinen TO. Genes that make you fat, but keep you healthy. J Intern Med. 2018;284:450–463. doi: 10.1111/joim.12827
103.
Poirier P, Giles TD, Bray GA, Hong Y, Stern JS, Pi-Sunyer FX, Eckel RH; American Heart Association; Obesity Committee of the Council on Nutrition, Physical Activity, and Metabolism. Obesity and cardiovascular disease: pathophysiology, evaluation, and effect of weight loss: an update of the 1997 American Heart Association scientific statement on obesity and heart disease from the Obesity Committee of the Council on Nutrition, Physical Activity, and Metabolism. Circulation. 2006;113:898–918. doi: 10.1161/CIRCULATIONAHA.106.171016
104.
Auclair A, Martin J, Bastien M, Bonneville N, Biertho L, Marceau S, Hould FS, Biron S, Lebel S, Lescelleur O, et al. Is there a role for visceral adiposity in inducing type 2 diabetes remission in severely obese patients following biliopancreatic diversion with duodenal switch surgery? Obes Surg. 2016;26:1717–1727. doi: 10.1007/s11695-015-1973-3
105.
Fantuzzi G, Mazzone T. Adipose tissue and atherosclerosis: exploring the connection. Arterioscler Thromb Vasc Biol. 2007;27:996–1003. doi: 10.1161/ATVBAHA.106.131755
106.
Okura T, Nakata Y, Yamabuki K, Tanaka K. Regional body composition changes exhibit opposing effects on coronary heart disease risk factors. Arterioscler Thromb Vasc Biol. 2004;24:923–929. doi: 10.1161/01.ATV.0000125702.26272.f6
107.
Coutinho T, Goel K, Corrêa de Sá D, Kragelund C, Kanaya AM, Zeller M, Park JS, Kober L, Torp-Pedersen C, Cottin Y, et al. Central obesity and survival in subjects with coronary artery disease: a systematic review of the literature and collaborative analysis with individual subject data. J Am Coll Cardiol. 2011;57:1877–1886. doi: 10.1016/j.jacc.2010.11.058
108.
Abraham TM, Pedley A, Massaro JM, Hoffmann U, Fox CS. Association between visceral and subcutaneous adipose depots and incident cardiovascular disease risk factors. Circulation. 2015;132:1639–1647. doi: 10.1161/CIRCULATIONAHA.114.015000
109.
Liu J, Fox CS, Hickson DA, May WD, Hairston KG, Carr JJ, Taylor HA. Impact of abdominal visceral and subcutaneous adipose tissue on cardiometabolic risk factors: the Jackson Heart Study. J Clin Endocrinol Metab. 2010;95:5419–5426. doi: 10.1210/jc.2010-1378
110.
Fujimoto WY, Bergstrom RW, Boyko EJ, Chen KW, Leonetti DL, Newell-Morris L, Shofer JB, Wahl PW. Visceral adiposity and incident coronary heart disease in Japanese-American men. The 10-year follow-up results of the Seattle Japanese-American Community Diabetes Study. Diabetes Care. 1999;22:1808–1812. doi: 10.2337/diacare.22.11.1808
111.
Nicklas BJ, Penninx BW, Cesari M, Kritchevsky SB, Newman AB, Kanaya AM, Pahor M, Jingzhong D, Harris TB; Health, Aging and Body Composition Study. Association of visceral adipose tissue with incident myocardial infarction in older men and women: the Health, Aging and Body Composition Study. Am J Epidemiol. 2004;160:741–749. doi: 10.1093/aje/kwh281
112.
McGill HCMcMahan CA, Malcom GT, Oalmann MC, Strong JP. Relation of glycohemoglobin and adiposity to atherosclerosis in youth. Pathobiological Determinants of Atherosclerosis in Youth (PDAY) Research Group. Arterioscler Thromb Vasc Biol. 1995;15:431–440. doi: 10.1161/01.atv.15.4.431
113.
McGill HCMcMahan CA, Herderick EE, Zieske AW, Malcom GT, Tracy RE, Strong JP; Pathobiological Determinants of Atherosclerosis in Youth (PDAY) Research Group. Obesity accelerates the progression of coronary atherosclerosis in young men. Circulation. 2002;105:2712–2718. doi: 10.1161/01.cir.0000018121.67607.ce
114.
Reis JP, Loria CM, Lewis CE, Powell-Wiley TM, Wei GS, Carr JJ, Terry JG, Liu K. Association between duration of overall and abdominal obesity beginning in young adulthood and coronary artery calcification in middle age. JAMA. 2013;310:280–288. doi: 10.1001/jama.2013.7833
115.
Piché ME, Poirier P. Obesity, ectopic fat and cardiac metabolism. Expert Rev Endocrinol Metab. 2018;13:213–221. doi: 10.1080/17446651.2018.1500894
116.
Shah RV, Anderson A, Ding J, Budoff M, Rider O, Petersen SE, Jensen MK, Koch M, Allison M, Kawel-Boehm N, et al. Pericardial, but not hepatic, fat by CT is associated with CV outcomes and structure: the Multi-Ethnic Study of Atherosclerosis. JACC Cardiovasc Imaging. 2017;10:1016–1027. doi: 10.1016/j.jcmg.2016.10.024
117.
Packer M. Epicardial adipose tissue may mediate deleterious effects of obesity and inflammation on the myocardium. J Am Coll Cardiol. 2018;71:2360–2372. doi: 10.1016/j.jacc.2018.03.509
118.
Guglielmi V, Sbraccia P. Epicardial adipose tissue: at the heart of the obesity complications. Acta Diabetol. 2017;54:805–812. doi: 10.1007/s00592-017-1020-z
119.
Schindler TH, Schelbert HR, Quercioli A, Dilsizian V. Cardiac PET imaging for the detection and monitoring of coronary artery disease and microvascular health. JACC Cardiovasc Imaging. 2010;3:623–640. doi: 10.1016/j.jcmg.2010.04.007
120.
Taqueti VR, Di Carli MF. Coronary microvascular disease pathogenic mechanisms and therapeutic options: JACC state-of-the-art review. J Am Coll Cardiol. 2018;72:2625–2641. doi: 10.1016/j.jacc.2018.09.042
121.
Bajaj NS, Osborne MT, Gupta A, Tavakkoli A, Bravo PE, Vita T, Bibbo CF, Hainer J, Dorbala S, Blankstein R, et al. Coronary microvascular dysfunction and cardiovascular risk in obese patients. J Am Coll Cardiol. 2018;72:707–717. doi: 10.1016/j.jacc.2018.05.049
122.
Eringa EC, Bakker W, van Hinsbergh VW. Paracrine regulation of vascular tone, inflammation and insulin sensitivity by perivascular adipose tissue. Vascul Pharmacol. 2012;56:204–209. doi: 10.1016/j.vph.2012.02.003
123.
Hall ME, Brinkley TE, Chughtai H, Morgan TM, Hamilton CA, Jordan JH, Stacey RB, Soots S, Hundley WG. Adiposity is associated with gender-specific reductions in left ventricular myocardial perfusion during dobutamine stress. PLoS One. 2016;11:e0146519. doi: 10.1371/journal.pone.0146519
124.
Sorop O, Olver TD, van de Wouw J, Heinonen I, van Duin RW, Duncker DJ, Merkus D. The microcirculation: a key player in obesity-associated cardiovascular disease. Cardiovasc Res. 2017;113:1035–1045. doi: 10.1093/cvr/cvx093
125.
Quercioli A, Montecucco F, Pataky Z, Thomas A, Ambrosio G, Staub C, Di Marzo V, Ratib O, Mach F, Golay A, et al. Improvement in coronary circulatory function in morbidly obese individuals after gastric bypass-induced weight loss: relation to alterations in endocannabinoids and adipocytokines. Eur Heart J. 2013;34:2063–2073. doi: 10.1093/eurheartj/eht085
126.
Alpert MA, Lavie CJ, Agrawal H, Aggarwal KB, Kumar SA. Obesity and heart failure: epidemiology, pathophysiology, clinical manifestations, and management. Transl Res. 2014;164:345–356. doi: 10.1016/j.trsl.2014.04.010
127.
Bozkurt B, Aguilar D, Deswal A, Dunbar SB, Francis GS, Horwich T, Jessup M, Kosiborod M, Pritchett AM, Ramasubbu K, et al; American Heart Association Heart Failure and Transplantation Committee of the Council on Clinical Cardiology; Council on Cardiovascular Surgery and Anesthesia; Council on Cardiovascular and Stroke Nursing; Council on Hypertension; and Council on Quality and Outcomes Research. Contributory risk and management of comorbidities of hypertension, obesity, diabetes mellitus, hyperlipidemia, and metabolic syndrome in chronic heart failure: a scientific statement from the American Heart Association. Circulation. 2016;134:e535–e578. doi: 10.1161/CIR.0000000000000450
128.
Kenchaiah S, Evans JC, Levy D, Wilson PW, Benjamin EJ, Larson MG, Kannel WB, Vasan RS. Obesity and the risk of heart failure. N Engl J Med. 2002;347:305–313. doi: 10.1056/NEJMoa020245
129.
Aune D, Sen A, Norat T, Janszky I, Romundstad P, Tonstad S, Vatten LJ. Body mass index, abdominal fatness, and heart failure incidence and mortality: a systematic review and dose-response meta-analysis of prospective studies. Circulation. 2016;133:639–649. doi: 10.1161/CIRCULATIONAHA.115.016801
130.
Barbosa JA, Rodrigues AB, Mota CC, Barbosa MM, Simões e Silva AC. Cardiovascular dysfunction in obesity and new diagnostic imaging techniques: the role of noninvasive image methods. Vasc Health Risk Manag. 2011;7:287–295. doi: 10.2147/VHRM.S17801
131.
Kosmala W, Sanders P, Marwick TH. Subclinical myocardial impairment in metabolic diseases. JACC Cardiovasc Imaging. 2017;10:692–703. doi: 10.1016/j.jcmg.2017.04.001
132.
Rodriguez Flores M, Aguilar Salinas C, Piché ME, Auclair A, Poirier P. Effect of bariatric surgery on heart failure. Expert Rev Cardiovasc Ther. 2017;15:567–579. doi: 10.1080/14779072.2017.1352471
133.
Miranda WR, Batsis JA, Sarr MG, Collazo-Clavell ML, Clark MM, Somers VK, Lopez-Jimenez F. Impact of bariatric surgery on quality of life, functional capacity, and symptoms in patients with heart failure. Obes Surg. 2013;23:1011–1015. doi: 10.1007/s11695-013-0953-8
134.
Haykowsky MJ, Nicklas BJ, Brubaker PH, Hundley WG, Brinkley TE, Upadhya B, Becton JT, Nelson MD, Chen H, Kitzman DW. Regional adipose distribution and its relationship to exercise intolerance in older obese patients who have heart failure with preserved ejection fraction. JACC Heart Fail. 2018;6:640–649. doi: 10.1016/j.jchf.2018.06.002
135.
Rao VN, Zhao D, Allison MA, Guallar E, Sharma K, Criqui MH, Cushman M, Blumenthal RS, Michos ED. Adiposity and incident heart failure and its subtypes: MESA (Multi-Ethnic Study of Atherosclerosis). JACC Heart Fail. 2018;6:999–1007. doi: 10.1016/j.jchf.2018.07.009
136.
Tsujimoto T, Kajio H. Abdominal obesity is associated with an increased risk of all-cause mortality in patients with HFpEF. J Am Coll Cardiol. 2017;70:2739–2749. doi: 10.1016/j.jacc.2017.09.1111
137.
Loop MS, Van Dyke MK, Chen L, Brown TM, Durant RW, Safford MM, Levitan EB. Comparison of length of stay, 30-day mortality, and 30-day readmission rates in medicare patients with heart failure and with reduced versus preserved ejection fraction. Am J Cardiol. 2016;118:79–85. doi: 10.1016/j.amjcard.2016.04.015
138.
Obokata M, Reddy YNV, Pislaru SV, Melenovsky V, Borlaug BA. Evidence supporting the existence of a distinct obese phenotype of heart failure with preserved ejection fraction. Circulation. 2017;136:6–19. doi: 10.1161/CIRCULATIONAHA.116.026807
139.
Sjöström L, Peltonen M, Jacobson P, Sjöström CD, Karason K, Wedel H, Ahlin S, Anveden Å, Bengtsson C, Bergmark G, et al. Bariatric surgery and long-term cardiovascular events. JAMA. 2012;307:56–65. doi: 10.1001/jama.2011.1914
140.
Shimada YJ, Tsugawa Y, Brown DF, Hasegawa K. Bariatric surgery and emergency department visits and hospitalizations for heart failure exacerbation: population-based, self-controlled series. J Am Coll Cardiol. 2016;67:895–903. doi: 10.1016/j.jacc.2015.12.016
141.
Horwich TB, Hamilton MA, Fonarow GC. B-type natriuretic peptide levels in obese patients with advanced heart failure. J Am Coll Cardiol. 2006;47:85–90. doi: 10.1016/j.jacc.2005.08.050
142.
Lavie CJ, Milani RV, Ventura HO. Adipose composition and heart failure prognosis: paradox or not? J Am Coll Cardiol. 2017;70:2750–2751. doi: 10.1016/j.jacc.2017.10.017
143.
Pandey A, Berry JD, Drazner MH, Fang JC, Tang WHW, Grodin JL. Body mass index, natriuretic peptides, and risk of adverse outcomes in patients with heart failure and preserved ejection fraction: analysis from the TOPCAT trial. J Amer Heart Assoc. 2018;7:e009664. doi: 10.1161/JAHA.118.009664
144.
Wang TJ, Larson MG, Levy D, Benjamin EJ, Leip EP, Wilson PW, Vasan RS. Impact of obesity on plasma natriuretic peptide levels. Circulation. 2004;109:594–600. doi: 10.1161/01.CIR.0000112582.16683.EA
145.
Bertoni AG, Wagenknecht LE, Kitzman DW, Marcovina SM, Rushing JT, Espeland MA; Brain Natriuretic Peptide Subgroup of the Look AHEAD Research Group. Impact of the look AHEAD intervention on NT-pro brain natriuretic peptide in overweight and obese adults with diabetes. Obesity (Silver Spring). 2012;20:1511–1518. doi: 10.1038/oby.2011.296
146.
Martin J, Bergeron S, Pibarot P, Bastien M, Biertho L, Lescelleur O, Bertrand F, Simard S, Poirier P. Impact of bariatric surgery on N-terminal fragment of the prohormone brain natriuretic peptide and left ventricular diastolic function. Can J Cardiol. 2013;29:969–975. doi: 10.1016/j.cjca.2012.11.010
147.
Sarzani R, Dessì-Fulgheri P, Paci VM, Espinosa E, Rappelli A. Expression of natriuretic peptide receptors in human adipose and other tissues. J Endocrinol Invest. 1996;19:581–585. doi: 10.1007/bf03349021
148.
Lavie CJ, Pandey A, Lau DH, Alpert MA, Sanders P. Obesity and atrial fibrillation prevalence, pathogenesis, and prognosis: effects of weight loss and exercise. J Am Coll Cardiol. 2017;70:2022–2035. doi: 10.1016/j.jacc.2017.09.002
149.
Pathak RK, Mahajan R, Lau DH, Sanders P. The implications of obesity for cardiac arrhythmia mechanisms and management. Can J Cardiol. 2015;31:203–210. doi: 10.1016/j.cjca.2014.10.027
150.
Plourde B, Sarrazin JF, Nault I, Poirier P. Sudden cardiac death and obesity. Expert Rev Cardiovasc Ther. 2014;12:1099–1110. doi: 10.1586/14779072.2014.952283
151.
Tedrow UB, Conen D, Ridker PM, Cook NR, Koplan BA, Manson JE, Buring JE, Albert CM. The long- and short-term impact of elevated body mass index on the risk of new atrial fibrillation the WHS (women’s health study). J Am Coll Cardiol. 2010;55:2319–2327. doi: 10.1016/j.jacc.2010.02.029
152.
Wang TJ, Parise H, Levy D, D’Agostino RBWolf PA, Vasan RS, Benjamin EJ. Obesity and the risk of new-onset atrial fibrillation. JAMA. 2004;292:2471–2477. doi: 10.1001/jama.292.20.2471
153.
Wanahita N, Messerli FH, Bangalore S, Gami AS, Somers VK, Steinberg JS. Atrial fibrillation and obesity–results of a meta-analysis. Am Heart J. 2008;155:310–315. doi: 10.1016/j.ahj.2007.10.004
154.
Wong CX, Sullivan T, Sun MT, Mahajan R, Pathak RK, Middeldorp M, Twomey D, Ganesan AN, Rangnekar G, Roberts-Thomson KC, et al. Obesity and the risk of incident, post-operative, and post-ablation atrial fibrillation: a meta-analysis of 626,603 individuals in 51 studies. JACC Clin Electrophysiol. 2015;1:139–152. doi: 10.1016/j.jacep.2015.04.004
155.
Tsang TS, Barnes ME, Miyasaka Y, Cha SS, Bailey KR, Verzosa GC, Seward JB, Gersh BJ. Obesity as a risk factor for the progression of paroxysmal to permanent atrial fibrillation: a longitudinal cohort study of 21 years. Eur Heart J. 2008;29:2227–2233. doi: 10.1093/eurheartj/ehn324
156.
Mahajan R, Nelson A, Pathak RK, Middeldorp ME, Wong CX, Twomey DJ, Carbone A, Teo K, Agbaedeng T, Linz D, et al. Electroanatomical remodeling of the atria in obesity: impact of adjacent epicardial fat. JACC Clin Electrophysiol. 2018;4:1529–1540. doi: 10.1016/j.jacep.2018.08.014
157.
Wong CX, Abed HS, Molaee P, Nelson AJ, Brooks AG, Sharma G, Leong DP, Lau DH, Middeldorp ME, Roberts-Thomson KC, et al. Pericardial fat is associated with atrial fibrillation severity and ablation outcome. J Am Coll Cardiol. 2011;57:1745–1751. doi: 10.1016/j.jacc.2010.11.045
158.
Wong CX, Sun MT, Odutayo A, Emdin CA, Mahajan R, Lau DH, Pathak RK, Wong DT, Selvanayagam JB, Sanders P, et al. Associations of epicardial, abdominal, and overall adiposity with atrial fibrillation. Circ Arrhythm Electrophysiol. 2016;9:e004378. doi: 10.1161/CIRCEP.116.004378
159.
Thanassoulis G, Massaro JM, O’Donnell CJ, Hoffmann U, Levy D, Ellinor PT, Wang TJ, Schnabel RB, Vasan RS, Fox CS, et al. Pericardial fat is associated with prevalent atrial fibrillation: the Framingham Heart Study. Circ Arrhythm Electrophysiol. 2010;3:345–350. doi: 10.1161/CIRCEP.109.912055
160.
Hatem SN, Redheuil A, Gandjbakhch E. Cardiac adipose tissue and atrial fibrillation: the perils of adiposity. Cardiovasc Res. 2016;109:502–509. doi: 10.1093/cvr/cvw001
161.
Masuda M, Mizuno H, Enchi Y, Minamiguchi H, Konishi S, Ohtani T, Yamaguchi O, Okuyama Y, Nanto S, Sakata Y. Abundant epicardial adipose tissue surrounding the left atrium predicts early rather than late recurrence of atrial fibrillation after catheter ablation. J Interv Card Electrophysiol. 2015;44:31–37. doi: 10.1007/s10840-015-0031-3
162.
Venteclef N, Guglielmi V, Balse E, Gaborit B, Cotillard A, Atassi F, Amour J, Leprince P, Dutour A, Clément K, et al. Human epicardial adipose tissue induces fibrosis of the atrial myocardium through the secretion of adipo-fibrokines. Eur Heart J. 2015;36:795–805a. doi: 10.1093/eurheartj/eht099
163.
Gaborit B, Sengenes C, Ancel P, Jacquier A, Dutour A. Role of epicardial adipose tissue in health and disease: a matter of fat? Compr Physiol. 2017;7:1051–1082. doi: 10.1002/cphy.c160034
164.
Zhu W, Zhang H, Guo L, Hong K. Relationship between epicardial adipose tissue volume and atrial fibrillation: a systematic review and meta-analysis. Herz. 2016;41:421–427. doi: 10.1007/s00059-015-4387-z
165.
Noheria A, Teodorescu C, Uy-Evanado A, Reinier K, Mariani R, Gunson K, Jui J, Chugh SS. Distinctive profile of sudden cardiac arrest in middle-aged vs. older adults: a community-based study. Int J Cardiol. 2013;168:3495–3499. doi: 10.1016/j.ijcard.2013.04.207
166.
Kannel WB, Plehn JF, Cupples LA. Cardiac failure and sudden death in the Framingham Study. Am Heart J. 1988;115:869–875. doi: 10.1016/0002-8703(88)90891-5
167.
Pietrasik G, Goldenberg I, McNitt S, Moss AJ, Zareba W. Obesity as a risk factor for sustained ventricular tachyarrhythmias in MADIT II patients. J Cardiovasc Electrophysiol. 2007;18:181–184. doi: 10.1111/j.1540-8167.2006.00680.x
168.
Sabbag A, Goldenberg I, Moss AJ, McNitt S, Glikson M, Biton Y, Jackson L, Polonsky B, Zareba W, Kutyifa V. Predictors and risk of ventricular tachyarrhythmias or death in black and white cardiac patients: a MADIT-CRT Trial Substudy. JACC Clin Electrophysiol. 2016;2:448–455. doi: 10.1016/j.jacep.2016.03.003
169.
Klein S, Burke LE, Bray GA, Blair S, Allison DB, Pi-Sunyer X, Hong Y, Eckel RH; American Heart Association Council on Nutrition, Physical Activity, and Metabolism. Clinical implications of obesity with specific focus on cardiovascular disease: a statement for professionals from the American Heart Association Council on Nutrition, Physical Activity, and Metabolism: endorsed by the American College of Cardiology Foundation. Circulation. 2004;110:2952–2967. doi: 10.1161/01.CIR.0000145546.97738.1E
170.
Messerli FH, Nunez BD, Ventura HO, Snyder DW. Overweight and sudden death. Increased ventricular ectopy in cardiopathy of obesity. Arch Intern Med. 1987;147:1725–1728. doi: 10.1001/archinte.147.10.1725
171.
el-Gamal A, Gallagher D, Nawras A, Gandhi P, Gomez J, Allison DB, Steinberg JS, Shumacher D, Blank R, Heymsfield SB. Effects of obesity on QT, RR, and QTc intervals. Am J Cardiol. 1995;75:956–959. doi: 10.1016/s0002-9149(99)80700-0
172.
Bharati S, Lev M. Cardiac conduction system involvement in sudden death of obese young people. Am Heart J. 1995;129:273–281. doi: 10.1016/0002-8703(95)90008-x
173.
Empana JP, Ducimetiere P, Charles MA, Jouven X. Sagittal abdominal diameter and risk of sudden death in asymptomatic middle-aged men: the Paris Prospective Study I. Circulation. 2004;110:2781–2785. doi: 10.1161/01.CIR.0000146395.64065.BA
174.
Adabag S, Huxley RR, Lopez FL, Chen LY, Sotoodehnia N, Siscovick D, Deo R, Konety S, Alonso A, Folsom AR. Obesity related risk of sudden cardiac death in the atherosclerosis risk in communities study. Heart. 2015;101:215–221. doi: 10.1136/heartjnl-2014-306238
175.
Fuller B, Garland J, Anne S, Beh R, McNevin D, Tse R. Increased epicardial fat thickness in sudden death from stable coronary artery atherosclerosis. Am J Forensic Med Pathol. 2017;38:162–166. doi: 10.1097/PAF.0000000000000310
176.
Wu CK, Tsai HY, Su MY, Wu YF, Hwang JJ, Tseng WY, Lin JL, Lin LY. Pericardial fat is associated with ventricular tachyarrhythmia and mortality in patients with systolic heart failure. Atherosclerosis. 2015;241:607–614. doi: 10.1016/j.atherosclerosis.2015.05.025
177.
Alpert MA, Terry BE, Mulekar M, Cohen MV, Massey CV, Fan TM, Panayiotou H, Mukerji V. Cardiac morphology and left ventricular function in normotensive morbidly obese patients with and without congestive heart failure, and effect of weight loss. Am J Cardiol. 1997;80:736–740. doi: 10.1016/s0002-9149(97)00505-5
178.
Piché MÈ, Auclair A, Harvey J, Marceau S, Poirier P. How to choose and use bariatric surgery in 2015. Can J Cardiol. 2015;31:153–166. doi: 10.1016/j.cjca.2014.12.014
179.
Poirier P, Cornier MA, Mazzone T, Stiles S, Cummings S, Klein S, McCullough PA, Ren Fielding C, Franklin BA; American Heart Association Obesity Committee of the Council on Nutrition, Physical Activity, and Metabolism. Bariatric surgery and cardiovascular risk factors: a scientific statement from the American Heart Association. Circulation. 2011;123:1683–1701. doi: 10.1161/CIR.0b013e3182149099
180.
Warnes CA, Roberts WC. The heart in massive (more than 300 pounds or 136 kilograms) obesity: analysis of 12 patients studied at necropsy. Am J Cardiol. 1984;54:1087–1091. doi: 10.1016/s0002-9149(84)80149-6
181.
Ashrafian H, le Roux CW, Darzi A, Athanasiou T. Effects of bariatric surgery on cardiovascular function. Circulation. 2008;118:2091–2102. doi: 10.1161/CIRCULATIONAHA.107.721027
182.
Aggarwal R, Harling L, Efthimiou E, Darzi A, Athanasiou T, Ashrafian H. The effects of bariatric surgery on cardiac structure and function: a systematic review of cardiac imaging outcomes. Obes Surg. 2016;26:1030–1040. doi: 10.1007/s11695-015-1866-5
183.
Jamaly S, Carlsson L, Peltonen M, Jacobson P, Sjöström L, Karason K. Bariatric surgery and the risk of new-onset atrial fibrillation in Swedish obese subjects. J Am Coll Cardiol. 2016;68:2497–2504. doi: 10.1016/j.jacc.2016.09.940
184.
Shahreyar M, Dang G, Waqas Bashir M, Kumar G, Hussain J, Ahmad S, Pandey B, Thakur A, Bhandari S, Thandra K, et al. Outcomes of in-hospital cardiopulmonary resuscitation in morbidly obese patients. JACC Clin Electrophysiol. 2017;3:174–183. doi: 10.1016/j.jacep.2016.08.011
185.
Taegtmeyer H, Young ME, Lopaschuk GD, Abel ED, Brunengraber H, Darley-Usmar V, Des Rosiers C, Gerszten R, Glatz JF, Griffin JL, et al; American Heart Association Council on Basic Cardiovascular Sciences. Assessing cardiac metabolism: a scientific statement from the American Heart Association. Circ Res. 2016;118:1659–1701. doi: 10.1161/RES.0000000000000097
186.
Lopaschuk GD, Folmes CD, Stanley WC. Cardiac energy metabolism in obesity. Circ Res. 2007;101:335–347. doi: 10.1161/CIRCRESAHA.107.150417
187.
Zlobine I, Gopal K, Ussher JR. Lipotoxicity in obesity and diabetes-related cardiac dysfunction. Biochim Biophys Acta. 2016;1861:1555–1568. doi: 10.1016/j.bbalip.2016.02.011
188.
Neubauer S. The failing heart–an engine out of fuel. N Engl J Med. 2007;356:1140–1151. doi: 10.1056/NEJMra063052
189.
Rider OJ, Cox P, Tyler D, Clarke K, Neubauer S. Myocardial substrate metabolism in obesity. Int J Obes (Lond). 2013;37:972–979. doi: 10.1038/ijo.2012.170
190.
Scheuermann-Freestone M, Madsen PL, Manners D, Blamire AM, Buckingham RE, Styles P, Radda GK, Neubauer S, Clarke K. Abnormal cardiac and skeletal muscle energy metabolism in patients with type 2 diabetes. Circulation. 2003;107:3040–3046. doi: 10.1161/01.CIR.0000072789.89096.10
191.
Iozzo P. Metabolic toxicity of the heart: insights from molecular imaging. Nutr Metab Cardiovasc Dis. 2010;20:147–156. doi: 10.1016/j.numecd.2009.08.011
192.
Levelt E, Pavlides M, Banerjee R, Mahmod M, Kelly C, Sellwood J, Ariga R, Thomas S, Francis J, Rodgers C, et al. Ectopic and visceral fat deposition in lean and obese patients with type 2 diabetes. J Am Coll Cardiol. 2016;68:53–63. doi: 10.1016/j.jacc.2016.03.597
193.
McGavock JM, Lingvay I, Zib I, Tillery T, Salas N, Unger R, Levine BD, Raskin P, Victor RG, Szczepaniak LS. Cardiac steatosis in diabetes mellitus: a 1H-magnetic resonance spectroscopy study. Circulation. 2007;116:1170–1175. doi: 10.1161/CIRCULATIONAHA.106.645614
194.
Ingwall JS, Weiss RG. Is the failing heart energy starved? On using chemical energy to support cardiac function. Circ Res. 2004;95:135–145. doi: 10.1161/01.RES.0000137170.41939.d9
195.
Neubauer S, Horn M, Cramer M, Harre K, Newell JB, Peters W, Pabst T, Ertl G, Hahn D, Ingwall JS, et al. Myocardial phosphocreatine-to-ATP ratio is a predictor of mortality in patients with dilated cardiomyopathy. Circulation. 1997;96:2190–2196. doi: 10.1161/01.cir.96.7.2190
196.
Perseghin G, Lattuada G, De Cobelli F, Esposito A, Belloni E, Ntali G, Ragogna F, Canu T, Scifo P, Del Maschio A, et al. Increased mediastinal fat and impaired left ventricular energy metabolism in young men with newly found fatty liver. Hepatology. 2008;47:51–58. doi: 10.1002/hep.21983
197.
Rider OJ, Francis JM, Tyler D, Byrne J, Clarke K, Neubauer S. Effects of weight loss on myocardial energetics and diastolic function in obesity. Int J Cardiovasc Imaging. 2013;29:1043–1050. doi: 10.1007/s10554-012-0174-6
198.
Rayner JJ, Banerjee R, Holloway CJ, Lewis AJM, Peterzan MA, Francis JM, Neubauer S, Rider OJ. The relative contribution of metabolic and structural abnormalities to diastolic dysfunction in obesity. Int J Obes (Lond). 2018;42:441–447. doi: 10.1038/ijo.2017.239
199.
Alpert MA, Lambert CR, Panayiotou H, Terry BE, Cohen MV, Massey CV, Hashimi MW, Mukerji V. Relation of duration of morbid obesity to left ventricular mass, systolic function, and diastolic filling, and effect of weight loss. Am J Cardiol. 1995;76:1194–1197. doi: 10.1016/s0002-9149(99)80338-5
200.
Lavie CJ, Arena R, Alpert MA, Milani RV, Ventura HO. Management of cardiovascular diseases in patients with obesity. Nat Rev Cardiol. 2018;15:45–56. doi: 10.1038/nrcardio.2017.108
201.
Morricone L, Malavazos AE, Coman C, Donati C, Hassan T, Caviezel F. Echocardiographic abnormalities in normotensive obese patients: relationship with visceral fat. Obes Res. 2002;10:489–498. doi: 10.1038/oby.2002.67
202.
Barbosa MM, Beleigoli AM, de Fatima Diniz M, Freire CV, Ribeiro AL, Nunes MC. Strain imaging in morbid obesity: insights into subclinical ventricular dysfunction. Clin Cardiol. 2011;34:288–293. doi: 10.1002/clc.20907
203.
Tumuklu MM, Etikan I, Kisacik B, Kayikcioglu M. Effect of obesity on left ventricular structure and myocardial systolic function: assessment by tissue doppler imaging and strain/strain rate imaging. Echocardiography. 2007;24:802–809. doi: 10.1111/j.1540-8175.2007.00484.x
204.
Wong CY, O’Moore-Sullivan T, Leano R, Hukins C, Jenkins C, Marwick TH. Association of subclinical right ventricular dysfunction with obesity. J Am Coll Cardiol. 2006;47:611–616. doi: 10.1016/j.jacc.2005.11.015
205.
Neeland IJ, Gupta S, Ayers CR, Turer AT, Rame JE, Das SR, Berry JD, Khera A, McGuire DK, Vega GL, et al. Relation of regional fat distribution to left ventricular structure and function. Circ Cardiovasc Imaging. 2013;6:800–807. doi: 10.1161/CIRCIMAGING.113.000532
206.
Park J, Kim NH, Kim SH, Kim JS, Kim YH, Lim HE, Kim EJ, Na JO, Cho GY, Baik I, et al. Visceral adiposity and skeletal muscle mass are independently and synergistically associated with left ventricular structure and function: the Korean Genome and Epidemiology Study. Int J Cardiol. 2014;176:951–955. doi: 10.1016/j.ijcard.2014.08.108
207.
Flegal KM, Graubard BI, Williamson DF, Gail MH. Cause-specific excess deaths associated with underweight, overweight, and obesity. JAMA. 2007;298:2028–2037. doi: 10.1001/jama.298.17.2028
208.
Balkau B, Deanfield JE, Després JP, Bassand JP, Fox KA, Smith SCBarter P, Tan CE, Van Gaal L, Wittchen HU, et al. International Day for the Evaluation of Abdominal Obesity (IDEA): a study of waist circumference, cardiovascular disease, and diabetes mellitus in 168,000 primary care patients in 63 countries. Circulation. 2007;116:1942–1951. doi: 10.1161/CIRCULATIONAHA.106.676379
209.
Gallagher D, Kelley DE, Yim JE, Spence N, Albu J, Boxt L, Pi-Sunyer FX, Heshka S; MRI Ancillary Study Group of the Look AHEAD Research Group. Adipose tissue distribution is different in type 2 diabetes. Am J Clin Nutr. 2009;89:807–814. doi: 10.3945/ajcn.2008.26955
210.
Haffner SM, Lehto S, Rönnemaa T, Pyörälä K, Laakso M. Mortality from coronary heart disease in subjects with type 2 diabetes and in nondiabetic subjects with and without prior myocardial infarction. N Engl J Med. 1998;339:229–234. doi: 10.1056/NEJM199807233390404
211.
Lakka HM, Laaksonen DE, Lakka TA, Niskanen LK, Kumpusalo E, Tuomilehto J, Salonen JT. The metabolic syndrome and total and cardiovascular disease mortality in middle-aged men. JAMA. 2002;288:2709–2716. doi: 10.1001/jama.288.21.2709
212.
Malik S, Wong ND, Franklin SS, Kamath TV, L’Italien GJ, Pio JR, Williams GR. Impact of the metabolic syndrome on mortality from coronary heart disease, cardiovascular disease, and all causes in United States adults. Circulation. 2004;110:1245–1250. doi: 10.1161/01.CIR.0000140677.20606.0E
213.
Li C, Ford ES, McGuire LC, Mokdad AH. Association of metabolic syndrome and insulin resistance with congestive heart failure: findings from the Third National Health and Nutrition Examination Survey. J Epidemiol Community Health. 2007;61:67–73. doi: 10.1136/jech.2006.048173
214.
Ingelsson E, Arnlöv J, Lind L, Sundström J. Metabolic syndrome and risk for heart failure in middle-aged men. Heart. 2006;92:1409–1413. doi: 10.1136/hrt.2006.089011
215.
Bahrami H, Bluemke DA, Kronmal R, Bertoni AG, Lloyd-Jones DM, Shahar E, Szklo M, Lima JA. Novel metabolic risk factors for incident heart failure and their relationship with obesity: the MESA (Multi-Ethnic Study of Atherosclerosis) study. J Am Coll Cardiol. 2008;51:1775–1783. doi: 10.1016/j.jacc.2007.12.048
216.
Joyner MJ, Paneth N. Promises, promises, and precision medicine. J Clin Invest. 2019;129:946–948. doi: 10.1172/JCI126119
217.
Després JP. Obesity and cardiovascular disease: weight loss is not the only target. Can J Cardiol. 2015;31:216–222. doi: 10.1016/j.cjca.2014.12.009
218.
Ross R, Rissanen J, Pedwell H, Clifford J, Shragge P. Influence of diet and exercise on skeletal muscle and visceral adipose tissue in men. J Appl Physiol (1985). 1996;81:2445–2455. doi: 10.1152/jappl.1996.81.6.2445
219.
Ross R, Bradshaw AJ. The future of obesity reduction: beyond weight loss. Nat Rev Endocrinol. 2009;5:319–325. doi: 10.1038/nrendo.2009.78
220.
Artero EG, Jackson AS, Sui X, Lee DC, O’Connor DP, Lavie CJ, Church TS, Blair SN. Longitudinal algorithms to estimate cardiorespiratory fitness: associations with nonfatal cardiovascular disease and disease-specific mortality. J Am Coll Cardiol. 2014;63:2289–2296. doi: 10.1016/j.jacc.2014.03.008
221.
Berry JD, Willis B, Gupta S, Barlow CE, Lakoski SG, Khera A, Rohatgi A, de Lemos JA, Haskell W, Lloyd-Jones DM. Lifetime risks for cardiovascular disease mortality by cardiorespiratory fitness levels measured at ages 45, 55, and 65 years in men. The Cooper Center Longitudinal Study. J Am Coll Cardiol. 2011;57:1604–1610. doi: 10.1016/j.jacc.2010.10.056
222.
Farrell SW, Kampert JB, Kohl HWBarlow CE, Macera CA, Paffenbarger RSGibbons LW, Blair SN. Influences of cardiorespiratory fitness levels and other predictors on cardiovascular disease mortality in men. Med Sci Sports Exerc. 1998;30:899–905. doi: 10.1097/00005768-199806000-00019
223.
Ross R, Blair SN, Arena R, Church TS, Després JP, Franklin BA, Haskell WL, Kaminsky LA, Levine BD, Lavie CJ, et al; American Heart Association Physical Activity Committee of the Council on Lifestyle and Cardiometabolic Health; Council on Clinical Cardiology; Council on Epidemiology and Prevention; Council on Cardiovascular and Stroke Nursing; Council on Functional Genomics and Translational Biology; Stroke Council. Importance of assessing cardiorespiratory fitness in clinical practice: a case for fitness as a clinical vital sign: a scientific statement from the American Heart Association. Circulation. 2016;134:e653–e699. doi: 10.1161/CIR.0000000000000461
224.
Lee CD, Blair SN, Jackson AS. Cardiorespiratory fitness, body composition, and all-cause and cardiovascular disease mortality in men. Am J Clin Nutr. 1999;69:373–380. doi: 10.1093/ajcn/69.3.373
225.
Lee DC, Sui X, Church TS, Lee IM, Blair SN. Associations of cardiorespiratory fitness and obesity with risks of impaired fasting glucose and type 2 diabetes in men. Diabetes Care. 2009;32:257–262. doi: 10.2337/dc08-1377
226.
Estruch R, Ros E, Salas-Salvadó J, Covas MI, Corella D, Arós F, Gómez-Gracia E, Ruiz-Gutiérrez V, Fiol M, Lapetra J, et al; PREDIMED Study Investigators. Primary prevention of cardiovascular disease with a mediterranean diet supplemented with extra-virgin olive oil or nuts. N Engl J Med. 2018;378:e34. doi: 10.1056/NEJMoa1800389
227.
Broekhuizen LN, Boekholdt SM, Arsenault BJ, Després JP, Stroes ES, Kastelein JJ, Khaw KT, Wareham NJ. Physical activity, metabolic syndrome, and coronary risk: the EPIC-Norfolk prospective population study. Eur J Cardiovasc Prev Rehabil. 2011;18:209–217. doi: 10.1177/1741826710389397
228.
Després JP. Predicting longevity using metabolomics: a novel tool for precision lifestyle medicine? Nat Rev Cardiol. 2020;17:67–68. doi: 10.1038/s41569-019-0310-2
229.
Lévesque V, Poirier P, Després JP, Alméras N. Relation between a simple lifestyle risk score and established biological risk factors for cardiovascular disease. Am J Cardiol. 2017;120:1939–1946. doi: 10.1016/j.amjcard.2017.08.008
230.
Unick JL, Beavers D, Bond DS, Clark JM, Jakicic JM, Kitabchi AE, Knowler WC, Wadden TA, Wagenknecht LE, Wing RR; Look AHEAD Research Group. The long-term effectiveness of a lifestyle intervention in severely obese individuals. Am J Med. 2013;126:236–42, 242.e1. doi: 10.1016/j.amjmed.2012.10.010
231.
Unick JL, Beavers D, Jakicic JM, Kitabchi AE, Knowler WC, Wadden TA, Wing RR; Look AHEAD Research Group. Effectiveness of lifestyle interventions for individuals with severe obesity and type 2 diabetes: results from the Look AHEAD trial. Diabetes Care. 2011;34:2152–2157. doi: 10.2337/dc11-0874
232.
Livhits M, Mercado C, Yermilov I, Parikh JA, Dutson E, Mehran A, Ko CY, Gibbons MM. Does weight loss immediately before bariatric surgery improve outcomes: a systematic review. Surg Obes Relat Dis. 2009;5:713–721. doi: 10.1016/j.soard.2009.08.014
233.
Papalazarou A, Yannakoulia M, Kavouras SA, Komesidou V, Dimitriadis G, Papakonstantinou A, Sidossis LS. Lifestyle intervention favorably affects weight loss and maintenance following obesity surgery. Obesity (Silver Spring). 2010;18:1348–1353. doi: 10.1038/oby.2009.346
234.
Kalarchian MA, Marcus MD, Courcoulas AP, Lutz C, Cheng Y, Sweeny G. Structured dietary intervention to facilitate weight loss after bariatric surgery: a randomized, controlled pilot study. Obesity (Silver Spring). 2016;24:1906–1912. doi: 10.1002/oby.21591
235.
Bray GA, Heisel WE, Afshin A, Jensen MD, Dietz WH, Long M, Kushner RF, Daniels SR, Wadden TA, Tsai AG, et al. The science of obesity management: an endocrine society scientific statement. Endocr Rev. 2018;39:79–132. doi: 10.1210/er.2017-00253
236.
Ghosh RK, Ghosh GC, Gupta M, Bandyopadhyay D, Akhtar T, Deedwania P, Lavie CJ, Fonarow GC, Aneja A. Sodium glucose co-transporter 2 inhibitors and heart failure. Am J Cardiol. 2019;124:1790–1796. doi: 10.1016/j.amjcard.2019.08.038
237.
Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jódar E, Leiter LA, Lingvay I, Rosenstock J, Seufert J, Warren ML, et al; SUSTAIN-6 Investigators. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med. 2016;375:1834–1844. doi: 10.1056/NEJMoa1607141
238.
Zelniker TA, Wiviott SD, Raz I, Im K, Goodrich EL, Bonaca MP, Mosenzon O, Kato ET, Cahn A, Furtado RHM, et al. SGLT2 inhibitors for primary and secondary prevention of cardiovascular and renal outcomes in type 2 diabetes: a systematic review and meta-analysis of cardiovascular outcome trials. Lancet. 2019;393:31–39. doi: 10.1016/S0140-6736(18)32590-X
239.
Bethel MA, Patel RA, Merrill P, Lokhnygina Y, Buse JB, Mentz RJ, Pagidipati NJ, Chan JC, Gustavson SM, Iqbal N, et al; EXSCEL Study Group. Cardiovascular outcomes with glucagon-like peptide-1 receptor agonists in patients with type 2 diabetes: a meta-analysis. Lancet Diabetes Endocrinol. 2018;6:105–113. doi: 10.1016/S2213-8587(17)30412-6
240.
Garg V, Verma S, Connelly K. Mechanistic insights regarding the role of SGLT2 inhibitors and GLP1 agonist drugs on cardiovascular disease in diabetes. Prog Cardiovasc Dis. 2019;62:349–357. doi: 10.1016/j.pcad.2019.07.005
241.
Frías JP, Guja C, Hardy E, Ahmed A, Dong F, Öhman P, Jabbour SA. Exenatide once weekly plus dapagliflozin once daily versus exenatide or dapagliflozin alone in patients with type 2 diabetes inadequately controlled with metformin monotherapy (DURATION-8): a 28 week, multicentre, double-blind, phase 3, randomised controlled trial. Lancet Diabetes Endocrinol. 2016;4:1004–1016. doi: 10.1016/S2213-8587(16)30267-4
242.
McCrimmon RJ, Catarig AM, Frias JP, Lausvig NL, le Roux CW, Thielke D, Lingvay I. Effects of once-weekly semaglutide vs once-daily canagliflozin on body composition in type 2 diabetes: a substudy of the SUSTAIN 8 randomised controlled clinical trial. Diabetologia. 2020;63:473–485. doi: 10.1007/s00125-019-05065-8
243.
Ishii S, Nagai Y, Sada Y, Fukuda H, Nakamura Y, Matsuba R, Nakagawa T, Kato H, Tanaka Y. Liraglutide reduces visceral and intrahepatic fat without significant loss of muscle mass in obese patients with type 2 diabetes: a prospective case series. J Clin Med Res. 2019;11:219–224. doi: 10.14740/jocmr3647
244.
Padwal R, Klarenbach S, Wiebe N, Birch D, Karmali S, Manns B, Hazel M, Sharma AM, Tonelli M. Bariatric surgery: a systematic review and network meta-analysis of randomized trials. Obes Rev. 2011;12:602–621. doi: 10.1111/j.1467-789X.2011.00866.x
245.
Schauer PR, Bhatt DL, Kirwan JP, Wolski K, Brethauer SA, Navaneethan SD, Aminian A, Pothier CE, Kim ES, Nissen SE, et al; STAMPEDE Investigators. Bariatric surgery versus intensive medical therapy for diabetes–3-year outcomes. N Engl J Med. 2014;370:2002–2013. doi: 10.1056/NEJMoa1401329
246.
Angrisani L, Santonicola A, Iovino P, Vitiello A, Zundel N, Buchwald H, Scopinaro N. Bariatric surgery and endoluminal procedures: IFSO worldwide survey 2014. Obes Surg. 2017;27:2279–2289. doi: 10.1007/s11695-017-2666-x
247.
Piché MÈ, Martin J, Cianflone K, Bastien M, Marceau S, Biron S, Hould FS, Poirier P. Changes in predicted cardiovascular disease risk after biliopancreatic diversion surgery in severely obese patients. Metabolism. 2014;63:79–86. doi: 10.1016/j.metabol.2013.09.004
248.
Poirier P, Alpert MA, Fleisher LA, Thompson PD, Sugerman HJ, Burke LE, Marceau P, Franklin BA; American Heart Association Obesity Committee of Council on Nutrition, Physical Activity and Metabolism; Council on Cardiopulmonary Perioperative and Critical Care; Council on Cardiovascular Surgery and Anesthesia; Council on Cardiovascular Disease in the Young; Council on Cardiovascular Nursing; Council on Clinical Cardiology. Cardiovascular evaluation and management of severely obese patients undergoing surgery: a science advisory from the American Heart Association. Circulation. 2009;120:86–95. doi: 10.1161/CIRCULATIONAHA.109.192575
249.
Sjöström L, Narbro K, Sjöström CD, Karason K, Larsson B, Wedel H, Lystig T, Sullivan M, Bouchard C, Carlsson B, et al; Swedish Obese Subjects Study. Effects of bariatric surgery on mortality in Swedish obese subjects. N Engl J Med. 2007;357:741–752. doi: 10.1056/NEJMoa066254
250.
Dadson P, Landini L, Helmiö M, Hannukainen JC, Immonen H, Honka MJ, Bucci M, Savisto N, Soinio M, Salminen P, et al. Effect of bariatric surgery on adipose tissue glucose metabolism in different depots in patients with or without type 2 diabetes. Diabetes Care. 2016;39:292–299. doi: 10.2337/dc15-1447
251.
Sjöström L. Review of the key results from the Swedish Obese Subjects (SOS) trial - a prospective controlled intervention study of bariatric surgery. J Intern Med. 2013;273:219–234. doi: 10.1111/joim.12012
252.
Pirlet C, Biertho L, Poirier P, Marceau S, Marceau P, Biron S, Hould FS, Lebel S, Lescelleur O, Julien F, et al. Comparison of short and long term cardiovascular outcomes after bariatric surgery in patients with vs without coronary artery disease. Am J Cardiol. 2020;125:40–47. doi: 10.1016/j.amjcard.2019.09.041
253.
Fisher DP, Johnson E, Haneuse S, Arterburn D, Coleman KJ, O’Connor PJ, O’Brien R, Bogart A, Theis MK, Anau J, et al. Association between bariatric surgery and macrovascular disease outcomes in patients with type 2 diabetes and severe obesity. JAMA. 2018;320:1570–1582. doi: 10.1001/jama.2018.14619
254.
McCloskey CA, Ramani GV, Mathier MA, Schauer PR, Eid GM, Mattar SG, Courcoulas AP, Ramanathan R. Bariatric surgery improves cardiac function in morbidly obese patients with severe cardiomyopathy. Surg Obes Relat Dis. 2007;3:503–507. doi: 10.1016/j.soard.2007.05.006
255.
Ramani GV, McCloskey C, Ramanathan RC, Mathier MA. Safety and efficacy of bariatric surgery in morbidly obese patients with severe systolic heart failure. Clin Cardiol. 2008;31:516–520. doi: 10.1002/clc.20315
256.
Plourde CÉ, Grenier-Larouche T, Caron-Dorval D, Biron S, Marceau S, Lebel S, Biertho L, Tchernof A, Richard D, Carpentier AC. Biliopancreatic diversion with duodenal switch improves insulin sensitivity and secretion through caloric restriction. Obesity (Silver Spring). 2014;22:1838–1846. doi: 10.1002/oby.20771
257.
Guidone C, Manco M, Valera-Mora E, Iaconelli A, Gniuli D, Mari A, Nanni G, Castagneto M, Calvani M, Mingrone G. Mechanisms of recovery from type 2 diabetes after malabsorptive bariatric surgery. Diabetes. 2006;55:2025–2031. doi: 10.2337/db06-0068
258.
Mari A, Manco M, Guidone C, Nanni G, Castagneto M, Mingrone G, Ferrannini E. Restoration of normal glucose tolerance in severely obese patients after bilio-pancreatic diversion: role of insulin sensitivity and beta cell function. Diabetologia. 2006;49:2136–2143. doi: 10.1007/s00125-006-0337-x
259.
Michaud A, Grenier-Larouche T, Caron-Dorval D, Marceau S, Biertho L, Simard S, Richard D, Tchernof A, Carpentier AC. Biliopancreatic diversion with duodenal switch leads to better postprandial glucose level and beta cell function than sleeve gastrectomy in individuals with type 2 diabetes very early after surgery. Metabolism. 2017;74:10–21. doi: 10.1016/j.metabol.2017.06.005
260.
Grenier-Larouche T, Carreau AM, Geloën A, Frisch F, Biertho L, Marceau S, Lebel S, Hould FS, Richard D, Tchernof A, et al. Fatty acid metabolic remodeling during type 2 diabetes remission after bariatric surgery. Diabetes. 2017;66:2743–2755. doi: 10.2337/db17-0414
261.
Bryant EJ, King NA, Falkén Y, Hellström PM, Holst JJ, Blundell JE, Näslund E. Relationships among tonic and episodic aspects of motivation to eat, gut peptides, and weight before and after bariatric surgery. Surg Obes Relat Dis. 2013;9:802–808. doi: 10.1016/j.soard.2012.09.011
262.
Carreau AM, Noll C, Blondin DP, Frisch F, Nadeau M, Pelletier M, Phoenix S, Cunnane SC, Guérin B, Turcotte EE, et al. Bariatric surgery rapidly decreases cardiac dietary fatty acid partitioning and hepatic insulin resistance through increased intra-abdominal adipose tissue storage and reduced spillover in type 2 diabetes. Diabetes. 2020;69:567–577. doi: 10.2337/db19-0773
263.
Labrecque J, Laforest S, Michaud A, Biertho L, Tchernof A. Impact of bariatric surgery on white adipose tissue inflammation. Can J Diabetes. 2017;41:407–417. doi: 10.1016/j.jcjd.2016.12.003
264.
Grenier-Larouche T, Carreau AM, Carpentier AC. Early metabolic improvement after bariatric surgery: the first steps toward remission of type 2 diabetes. Can J Diabetes. 2017;41:418–425. doi: 10.1016/j.jcjd.2016.10.013
265.
Ma J, Rosas LG, Lv N. Precision lifestyle medicine: a new frontier in the science of behavior change and population health. Am J Prev Med. 2016;50:395–397. doi: 10.1016/j.amepre.2015.09.035

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.

Information & Authors

Information

Published In

Go to Circulation Research
Go to Circulation Research
Circulation Research
Pages: 1477 - 1500
PubMed: 32437302

Versions

You are viewing the most recent version of this article.

History

Published online: 21 May 2020
Published in print: 22 May 2020

Permissions

Request permissions for this article.

Keywords

  1. body fat distribution
  2. cardiovascular diseases
  3. diabetes mellitus
  4. heart failure
  5. obesity

Subjects

Authors

Affiliations

Marie-Eve Piché
From the Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec – Université Laval (M.-E.P., A.T., J.-P.D.), Université Laval, Québec, QC, Canada.
Department of Medicine, Faculty of Medicine (M.-E.P.), Université Laval, Québec, QC, Canada.
André Tchernof
From the Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec – Université Laval (M.-E.P., A.T., J.-P.D.), Université Laval, Québec, QC, Canada.
School of Nutrition (A.T.), Université Laval, Québec, QC, Canada.
From the Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec – Université Laval (M.-E.P., A.T., J.-P.D.), Université Laval, Québec, QC, Canada.
Vitam – Centre de recherche en santé durable, CIUSSS – Capitale-Nationale (J.-P.D.), Université Laval, Québec, QC, Canada.
Department of Kinesiology, Faculty of Medicine (J.-P.D.), Université Laval, Québec, QC, Canada.

Notes

For Sources of Funding and Disclosures, see page 1493.
Correspondence to: Jean-Pierre Després, CQ, PhD, FAHA, FIAS, Vitam – Centre de recherche en santé durable, CIUSSS – Capitale-Nationale, 2525 Chemin de la Canardière, Pavillon Landry-Poulin, A-2419, Québec QC G1J 0A4, Canada. Email [email protected]

Disclosures

A. Tchernof receives research funding from Johnson & Johnson Medical Companies and Medtronic for studies related to bariatric surgery. The other authors report no conflicts.

Sources of Funding

M.-E. Piché is a recipient of Early Career Research Grants from the Fonds de recherche du Québec-Santé and the Fondation of the Québec Heart and Lung Institute. J.-P. Després is the Scientific Director of the International Chair of Cardiometabolic Risk supported by the Fondation de l’Université Laval. Research from J.-P. Després discussed in this article has been and is currently supported by the Canadian Institutes of Health Research (Foundation grant, FDN-167278), as well as by the Fondation of the Québec Heart and Lung Institute. A. Tchernof is a codirector of the Research Chair in bariatric and metabolic surgery.

Metrics & Citations

Metrics

Citations

Download Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.

  1. Huanglian-Jiedu decoction promotes adipose thermogenesis in obese mice by suppressing the expression of HDAC3, Journal of Ethnopharmacology, 336, (118760), (2025).https://doi.org/10.1016/j.jep.2024.118760
    Crossref
  2. Intussusception and Gut Dysmotility: A Systematic Review Assessing Unexpected Complications of Bariatric Surgery, Cureus, (2024).https://doi.org/10.7759/cureus.58086
    Crossref
  3. Association between BMI and health-related physical fitness in adolescents with hearing impairment in China: a cross-sectional study, PeerJ, 12, (e17995), (2024).https://doi.org/10.7717/peerj.17995
    Crossref
  4. Cyy-287, a novel pyrimidine-2,4-diamine derivative, efficiently mitigates inflammatory responses, fibrosis, and lipid synthesis in obesity-induced cardiac and hepatic dysfunction, PeerJ, 12, (e17009), (2024).https://doi.org/10.7717/peerj.17009
    Crossref
  5. Testicular fat deposition attenuates reproductive performance via decreased follicle-stimulating hormone level and sperm meiosis and testosterone synthesis in mouse, Animal Bioscience, 37, 1, (50-60), (2024).https://doi.org/10.5713/ab.23.0175
    Crossref
  6. ANÁLISE DA PERDA DE PESO EM PACIENTES SUBMETIDOS A CIRURGIA BARIÁTRICA: REVISÃO SISTEMÁTICA DE LITERATURA, Revista Contemporânea, 4, 5, (e4260), (2024).https://doi.org/10.56083/RCV4N5-053
    Crossref
  7. The inhibitory effects of Xiao-Gao-Jiang-Zhuo-containing serum on adipogenesis in 3T3-L1 preadipocytes, Biomedical Papers, 168, 1, (25-34), (2024).https://doi.org/10.5507/bp.2023.016
    Crossref
  8. Healthcare Management of an Obese Person, DIET FACTOR (Journal of Nutritional and Food Sciences), (10-18), (2024).https://doi.org/10.54393/df.v5i01.119
    Crossref
  9. Prediabetes: An overlooked risk factor for major adverse cardiac and cerebrovascular events in atrial fibrillation patients, World Journal of Diabetes, 15, 1, (24-33), (2024).https://doi.org/10.4239/wjd.v15.i1.24
    Crossref
  10. Determination of SRPA and adiposity measures and its association with glycemic status in type 2 diabetics having high mean HbA1c in a private clinic of a city in west India, Journal of Family Medicine and Primary Care, 13, 9, (3897-3901), (2024).https://doi.org/10.4103/jfmpc.jfmpc_205_24
    Crossref
  11. See more
Loading...

View Options

View options

PDF and All Supplements

Download PDF and All Supplements

PDF/ePub

View PDF/ePub

Get Access

Login options

Check if you have access through your login credentials or your institution to get full access on this article.

Personal login Institutional Login
Purchase Options

Purchase this article to access the full text.

Purchase access to this article for 24 hours

Obesity Phenotypes, Diabetes, and Cardiovascular Diseases
Circulation Research
  • Vol. 126
  • No. 11

Purchase access to this journal for 24 hours

Circulation Research
  • Vol. 126
  • No. 11
Restore your content access

Enter your email address to restore your content access:

Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

Media

Figures

Other

Tables

Share

Share

Share article link

Share

Comment Response