Next Article in Journal
An Evaluation of the Nutritional and Promotional Profile of Commercial Foods for Infants and Toddlers in the United States
Previous Article in Journal
Urinary Phosphate and Subclinical Atherosclerosis: The AWHS Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Update on Obesity and Cardiovascular Risk: From Pathophysiology to Clinical Management

by
Giovanna Gallo
1,
Giovambattista Desideri
2 and
Carmine Savoia
1,*
1
Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
2
Department of Clinical, Internal Medicine, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
*
Author to whom correspondence should be addressed.
Nutrients 2024, 16(16), 2781; https://doi.org/10.3390/nu16162781
Submission received: 25 July 2024 / Revised: 14 August 2024 / Accepted: 19 August 2024 / Published: 20 August 2024
(This article belongs to the Section Clinical Nutrition)

Abstract

:
Obesity is an epidemic worldwide. Overweight and multiple obesity-related mechanisms, including dysmetabolic alterations, contribute to cardiovascular deleterious effects. Hence, overweight and obesity have been independently associated with increased cardiovascular risk, whose assessment is crucial for preserving life quality and reducing mortality, and to address appropriate therapeutic strategies in obese patients. Beyond the standard of care in managing overweight and obesity in adults (i.e., diet and physical exercise), several relevant pharmacotherapies have been approved, and several procedures and device types for weight loss have been recommended. In such a contest, medical weight management remains one option for treating excess weight. Most drugs used for obesity reduce appetite and increase satiety and, secondarily, slow gastric emptying to reduce body weight and, therefore, act also to improve metabolic parameters. In this contest, agonists of the glucagon-like peptide-1 receptor (GLP-1RAs) modulate different metabolic pathways associated with glucose metabolism, energy homeostasis, antioxidation, and inflammation. Moreover, this class of drugs has shown efficacy in improving glycemic control, reducing the incidence of cardiovascular events in type 2 diabetic patients, and reducing body weight independently of the presence of diabetes. Recently, in overweight or obese patients with pre-existing cardiovascular disease but without diabetes, the GLP-1RA semaglutide reduced the incidence of cardiovascular and cerebrovascular events and death from cardiovascular causes. Thus, semaglutide has been approved for secondary prevention in obese people with cardiovascular disease. Nevertheless, whether this class of drugs is equally effective for primary prevention in obese people has to be demonstrated. In this review, we will summarize updates on the pathophysiology of obesity, the effects of obesity on cardiovascular risk, the impact of different obesity phenotypes on cardiovascular diseases, and the novelties in the clinical management of obesity for cardiovascular prevention.

1. Introduction

Obesity is an epidemic worldwide, accounting for 650 million adults in 2016 and an estimated prevalence of one billion in 2030. In Europe, 60% of inhabitants are either overweight or obese. In developed countries, nearly one-third of the population is predicted to be overweight or obese within the next few years [1,2].
Pathophysiological mechanisms involved in developing obesity and related dysmetabolic conditions are complex, including genetic, neurohormonal, environmental, pharmacological, and psychological factors.
Ectopic fat accumulation and dysregulated adipose tissue represent the main features of obesity and lead to insulin resistance, lipotoxicity, and activation of proinflammatory pathways, which are involved in the progression of vascular and heart damage and increase the risk of cardiovascular events [3].
The impact of obesity on cardiovascular morbidity is enhanced by the coexistence of several comorbidities in obese subjects. These include dysmetabolic conditions such as diabetes, hypertension, dyslipidemia, obstructive sleep apnea hypoventilation syndrome, nonalcoholic fatty liver disease, as well as other diseases including cardiovascular disease (CVD), cancer, chronic kidney disease, neuromuscular and orthopedic disorders with debilitating influence on the quality of life, prognosis, and health-care costs [4]. In this view, it is mandatory in clinical practice to reduce overweight and obesity-related cardiometabolic risk to decrease the burden of CVD [5]. Different pharmacological and non-pharmacological strategies have been proposed to reduce body weight effectively. In such a context, precision nutrition represents an emerging field of interest, consisting of tailored dietary and lifestyle recommendations. Regarding pharmacological strategies, glucagon-like peptide-1 receptor agonists (GLP1-RA) have shown significant results independent of diabetes in reducing body weight and cardiovascular risk [6].
In this narrative review, we will discuss an update on the pathophysiological mechanisms involved in developing obesity and obesity-related metabolic and CVD, the obesity phenotypes and their association with cardiovascular events, and available new pharmacological strategies in reducing body weight and cardiovascular risk.

2. Obesity, Inflammation and Adiposopathy

Historically, body mass index (BMI) has been indicated as the main parameter to define obesity (BMI ≥ 30 kg/m2). However, this anthropometric measure may be influenced by age, sex, and race/ethnicity. In this view, other parameters have been introduced in clinical practice, such as waist circumference (WC), waist-hip ratio (WHiR), waist-to-height ratio (WHtR), bioimpedance, 3D scanning and dual-energy X-ray absorptiometry (DEXA), with increasing attention to a qualitative rather than to a quantitative assessment of adipose tissue distribution [7,8].
Pathophysiological mechanisms involved in developing obesity and related metabolic conditions are complex, including genetic, neurohormonal, environmental, pharmacological, and psychological factors. Adipocyte hypertrophy in visceral adipose tissue (VAT), ectopic fat accumulation, and dysregulated adipokine production represent pillar pathophysiological processes in developing obesity and metabolic disorders as well as the progression of adiposopathy [9,10] (Figure 1). The latter condition involves functional changes of adipose tissue, including impaired adipogenesis, adipocyte lipolysis over lipogenesis, increased free fatty acid release, endocrinopathies (hypoadiponectinemia and hyperleptinemia), pathogenic adipose tissue immune responses with a proinflammatory milieu, and pathogenic “crosstalk” between adipose tissue and other organs which ultimately are associated with fat dysfunctional fat accumulation and increased cardiovascular risk.
Obesity is characterized by excess nutrient consumption that contributes to impaired interaction between adipocytes and the immune cells infiltrating the adipose tissue, causing the release of several proinflammatory mediators [11,12].
The effects of nutrients on adipose tissue depend on both quantitative and qualitative aspects. Dietary patterns high in glycemic load and energy density and low in fiber have been suggested to influence body composition, associated with greater fat mass and increased risk of excess adiposity, and to affect innate appetite control with greater energy consumption [13]. Also, polyunsaturated fatty acid consumption has been associated with childhood overweight and obesity. In contrast, a higher protein intake was not associated with a later risk of overweight [13].
Proinflammatory dietary patterns play an essential role in developing adiposopathy and obesity. These diets include low fiber and antioxidant intake, a few antioxidant polyphenols, and immunomodulatory fatty acids, such as eicosapentaenoic fatty acid (EPA) and docosahexaenoic fatty acid (DHA). These diets are also rich in lipoperoxidated vegetable oils, xenobiotics, excessive sodium, and high saturated and trans fats. Thus, proinflammatory diets may promote the accumulation of ceramides and diacylglycerols, as well as impair glucose uptake, PPARγ activity, and insulin signaling, as well as increase cytokine secretion [14].
An increase in monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor-alpha (TNF-α), interleukin (IL)-6, and leptin levels, and a decrease in adiponectin have been shown to enhance adipogenesis, atherogenesis, fibrosis, and inflammation [15,16].
VAT hypertrophy and inflammatory status are associated with limited oxygen diffusion, leading to mitochondrial dysfunction, reactive oxygen species (ROS) release, and oxidative stress, as well as inducing the activation of HIF-1α [17]. Thus, hypoxic conditions frequently occur in VAT, HIF-1α protein upregulation, influencing angiogenesis, glucose metabolism, cellular stress response, extracellular matrix remodeling, and inflammatory signaling [18].
Adipose tissue is also the primary source of circulating miRNAs in patients with obesity, which enhances autophagy, apoptosis, and inflammatory pathways [19]. Knockout of autophagy-related genes has been shown to improve insulin resistance and endoplasmic reticulum stress and to promote lipid metabolism through the browning of white adipose [20]. Moreover, in obese patients, chronic p53 activation increases M1 macrophage polarization via the NF-κB pathway and enhances the systemic inflammatory response [21].
Adipocytes also release chemerin, a newly characterized chemoattractant that induces the infiltration of innate and adaptive immunity cells and the activation of inflammatory pathways [22,23]. Indeed, inflammation plays a fundamental role in the promotion of obesity-related systemic diseases. Inflammatory mediators induce an M1 macrophage polarization and activate dendritic cells (DC), B, CD4 + T, and CD8 + T cells [22,23,24,25]. Functional overlap between macrophages and adipocytes has been described, since both cell types are activated by lipopolysaccharide and other bacterial products and can secrete cytokines [26]. In addition, pre-adipocytes can transdifferentiate into M1 macrophages. M1 phenotype shifting is associated with the production of IL-1β, IL-6, tumor necrosis factor α (TNFα), and ROS, which further reduce insulin signaling in adipocytes [27,28].
Healthy adipose tissue is characterized by the presence of CD4+ and T-Reg. In contrast, hypertrophic adipose tissue contains CD4+ and CD8+ cytotoxic effector T lymphocytes, which secrete perforin and granzyme B, eliminating damaged cells [29].
The accumulation of VAT is associated with the activation of the Th17 population in the adipose tissue by CD11c+ DC cells, suggesting the importance of innate/adaptive immune cell crosstalk in obesity-associated inflammation [30]. Proinflammatory Th1 CD4+ T cells have been shown in VAT to accelerate obesity and non-alcoholic fatty liver disease [31,32]. Accordingly, it has been hypothesized that VAT could serve as a reservoir of immune cells. Increased levels of circulating immunoglobulin G and A and B cell accumulation have been described in the adipose tissue of obese patients, contributing to insulin resistance [33]. This inflammatory and dysregulated energetic environment is characterized by increased glucose and free fatty acid levels associated with ROS, which activate a post-mitotic cell cycle and a senescent cell program, sustaining a low-grade chronic inflammation [34].
Obesity-associated insulin resistance leads to lipotoxicity and the accumulation of excess fat in non-fat tissues, promoting the activation of pattern recognition receptors (PRR) recognizing pathogen-associated molecular patterns (PAMPs) [35]. PRRs include different types of receptors, such as toll-like receptors (TLRs), nucleotide-binding oligomerization domain-like receptors, C-type lectin receptors, and retinoic acid-inducible gene I-like receptors [36]. TLR4 has been shown to recognize lipopolysaccharides (LPS) of the outer wall of Gram-negative bacteria [37]. In such a context, the role of the mother’s gut microbiota, transferred to the fetus through maternal nutrition, and of maternal nutrition and lifestyle habits are gaining increasing importance due to their epigenetic influence on DNA methylation, histone acetylation, and availability of enzymatic substrates, as well as their impact on early fetal and neonatal microbiome and offspring’s metabolism [38].
Patients with obesity often present increased gut permeability and absorption of bacterial endotoxins due to a predominance of Firmicutes phyla (i.e., Lactobacillus and Faecalibacterium) and Escherichia coli rather than Bacteroidetes [39]. In addition, this gut microbiota composition has been shown to produce fewer levels of short-chain fatty acids (SCFAs), including acetate, propionate, and butyrate, thus generating foam cell formation and favoring the mobilization of lipopolysaccharides, trimethylamine N-oxide, and phenylacetyl glutamine (PAGIn) in the general circulation, resulting in intestinal and systemic inflammation [40].
The prevalence of Actinobacteria in human milk has been associated with the adiposity of women before pregnancy and during lactation and has also influenced the offspring’s nutrition status [41]. Maternal hyperglycemia and obesity represent independent risk factors for childhood obesity, being associated with leptin secretion and food intake behaviors [42].

3. Obesity and Cardiovascular Diseases

A large body of evidence has demonstrated the association between obesity and CVD, including atherosclerotic events and heart failure (HF) and indeed, the secretion of growth factors, cytokines, and vasoactive peptides by VAT, neurohormonal dysfunction, insulin resistance and hyperinsulinemia, the activation of the renin–angiotensin system (RAAS) and inflammation induced cardiac fibrosis, cellular death, and microvascular dysfunction [3,43,44]. These conditions are associated with hypertension and other features of metabolic syndrome as well as coronary heart disease and heart failure development (Figure 2).
An association between obesity and hypertension has been described. Different mechanisms have been proposed, including vasoconstriction and renal sodium absorption through the activation of the sympathetic nervous system and the renin–angiotensin–aldosterone system by the VAT-released leptin. Moreover, the increased circulating free fatty acids also result in oxidative stress, inflammation, and metabolic dysregulation [8,45].
Up to 70% of patients with obesity have dyslipidemia, with elevated levels of triglycerides, low-density lipoprotein (LDL) cholesterol, apolipoprotein B, and non-high-density lipoprotein (HDL) cholesterol, as well as with an increase in small dense pro-atherogenic LDL particles [45,46]. A well-established relationship also exists between central/abdominal obesity and diabetes, recognized with the coined term “diabesity.” Moreover, direct vascular dysfunction and obesity-induced metabolic risk factors are associated with coronary atherosclerotic plaque formation. Higher central adiposity at each level of BMI is associated with an increased risk of coronary artery disease and cardiovascular mortality. The degree and duration of obesity (expressed as BMI-years and WC-years) have been shown to predict the risk of CAD [8,45]. Accordingly, favorable changes in body composition have been demonstrated to reduce cardiometabolic risk and morbidity [8,45].
Collagen deposition, fat accumulation, and inflammatory infiltration occur within the vascular wall, leading to progressive arterial thickening [46]. In physiological conditions, perivascular adipose tissue provides mechanical protection and regulates blood vessel tone, secreting adiponectin and angiotensin (1–7) with antithrombotic and vasodilating effects [47,48]. On the other hand, obesity induces a shift to a pro-oxidant and pro-inflammatory environment, leading to endothelial dysfunction, vasoconstriction, vascular stiffness, and atherogenesis [49]. In such a context, the volume of epicardial fat, which is increased in obesity, has been associated with coronary plaque instability, atrial fibrillation, and HFpEF [50]. It has been demonstrated that epicardial fat releases exosomes carrying lipids, proteins, and microRNAs, which are involved in adverse cardiac remodeling [51].
Also, ectopic fat accumulation in the liver, skeletal muscle, and kidney contribute to lipotoxicity, mitochondrial dysfunction, and hypoxia, leading to cytokine and growth factor unbalance and vascular and myocardial damage [52].
Cardiac fibrosis has been described in obese patients, representing a fundamental pathophysiological mechanism in developing cardiac remodeling and dysfunction and heart failure with preserved ejection fraction (HFpEF) [53].
Several pathways are involved in the fibrogenic process, including the cascades of transforming growth factor β (TGF-β), epidermal growth factor (EGF), insulin-like growth factor-1, growth differentiation factor-11, and connective tissue growth factor (CTGF) [54,55,56]. Also, matricellular proteins, including thrombospondins, secreted protein acidic and rich in cysteine (SPARC), and osteopontin, are upregulated in obesity and metabolic syndrome, inducing the deposition of collagen and the proliferation of cardiac fibroblasts [57]. In addition, the increased expression of neprilysin on the surface of mature adipocytes of obese subjects reduces natriuretic peptides, contributing to cardiac inflammation and fibrosis [58].
Obesity has also been associated with an increased risk of cancer as a consequence of metabolic dysfunction [59]. Although the pathophysiological mechanisms should be better clarified, it has been hypothesized that releasing cytokines and chemokines from the excess adipose tissue and insulin resistance might lead to a preneoplastic microenvironment. In addition, obesity-related alterations in the gut microbiome may also contribute to chronic inflammation and a higher cancer risk [59].
Based on available evidence, different phenotypes of obesity have been described including metabolic unhealthy normal weight (MUNW), metabolically healthy overweight/obese (MHO), metabolically unhealthy overweight/obese (MUO), and sarcopenic obesity (SO) [60] (Figure 1).
The critical factor characterizing MHO and MUO, representing the most common phenotypes, is alterations in body fat distribution [61]. MUO is characterized by increased WC, reduced subcutaneous fat, and a shift toward a visceral and pro-inflammatory hypertrophic adipose tissue distribution, with a dysfunctional deposition in the liver and skeletal muscle [62]. Ectopic fat and adipose tissue dysfunction lead to diabetogenic and atherogenic secretion patterns, contributing to MUO development and related CVD [63]. On the other hand, MHO is characterized by high BMI with a favorable lipid profile, low levels of inflammatory cytokines, and preserved insulin sensitivity [64], with no evidence of other dysmetabolic disorders (i.e., type 2 diabetes, hypertension, dyslipidemia) and atherosclerotic cardiovascular disease, as well as with a low rate of CVD development among obese patients [65]. To better classify MHO, Lavie and colleagues proposed the following diagnostic criteria: serum triglycerides ≤ 150 mg/dL), HDL-cholesterol > 40 mg/dL in men or >50 mg/dL in women, systolic blood pressure ≤ 130 mmHg, diastolic blood pressure ≤ 85 mmHg, no antihypertensive treatment as an alternative indicator, fasting blood glucose ≤ 100 mg/dL, and no drug treatment with glucose-lowering agents [66]. The risk of diabetes, CVD, and all-cause mortality is more significant in the MUO than in the MHO phenotype as well as the adverse outcomes are directly related to the number and severity of metabolic abnormalities [67]. This phenotype is more common among young, physically active females who are at lesser risk of CVD development [68]. It has been hypothesized that the differences between MUO and MHO might derive from different biological properties of adipocytes regarding oxidative stress metabolites, ceramides, sphingolipids, and amino-acid metabolism from visceral and subcutaneous adipose tissue rather than the location of the fat depot [69]. Defective adipogenesis has been proposed to play a pivotal role in developing adipose tissue dysfunction, systemic insulin resistance, inflammation, and related metabolic disorders [70]. Clinical studies have suggested that MHO is a transient phenotype in about one-third of obese individuals and that those who maintained an MHO phenotype had a risk of diabetes and CVD comparable to that of metabolically healthy, normal-weight subjects [71]. Longitudinal studies showed that up to 50% of subjects with MHO may convert to MUO within 20 years as a consequence of a decline in insulin sensitivity with increased fasting blood glucose and of the detrimental effect of prolonged excess adiposity [72]. The risk of transitioning from MHO to MUO is higher in subjects with high BMI, constant weight gain during the observational period, older age, and the presence of hepatic steatosis [73]. Despite the low estimated cardiovascular risk in MHO, the incidence of cardiovascular events, including coronary heart disease, cerebrovascular disease, HF, respiratory diseases, and all-cause mortality, is significantly higher in MHO individuals compared to age and sex-matched individuals with average weight [74].
Despite normal weight, the MUNW phenotype is characterized by increased visceral adiposity with an unbalanced fat/lean mass ratio. Changes in anthropometric parameters are also present. These include increased WC, WHiR, and WHtR [75]. MUNW is typically seen in older and sedentary individuals with a very low amount of gluteal-femoral fat mass compared with the visceral one [75]. Although MUNW is associated with an elevated cardiovascular risk [75], this obesity phenotype is often underdiagnosed since not uninvoked criteria are used for the diagnosis of the body fat mass, which ranges from 19 to 32% for men and 29 to 44% for women in different clinical settings [76].
Loss of skeletal muscle due to a sedentary lifestyle, chronic comorbidities, and impairment of VAT, associated with an absolute body fat gain, are the characteristics of the SO phenotype [77]. In such a context, impaired function of the growth differentiation factor myostatin and other myokines, including FNDC5/irisin, fibroblast growth factor 21, and brain-derived neurotrophic factor, has been proposed to contribute to the development of this phenotype, resulting in the inhibition of insulin signaling and lipid oxidation [78]. In addition, obesity and sarcopenia may enhance each other since muscle loss, with consequent reduced mobility, facilitates weight gain, which further worsens exercise capacity in a vicious circle, particularly in the elderly [79]. SO is associated with a poor prognosis due to the increased risk of falls and fractures, hospitalizations, and all-cause mortality [80,81].
All the different described obesity phenotypes have been shown to be related to CVD. Individuals with MHO have been shown to have an increased risk of HF and atrial fibrillation but without significant differences in myocardial infarction (MI), stroke, and cardiovascular death compared to healthy subjects [82].
A direct negative effect of energetic dysmetabolism on cardiac structure, mainly consisting of left ventricle hypertrophy (LVH) and HFpEF, has been reported in MUO and MUNW [82]. Different from age and exercise-related adaptative LVH, this pathological concentric LV remodeling develops in response to increased myocardial stress and arterial afterload and is associated with a change in cardiomyocyte phenotype and an excessive synthesis of extracellular matrix and fibrosis [83]. In addition, LVH represents an arrhythmic substrate with an additional risk of cardiac death triggered by co-factors such as hypoxia, hypercapnia, obstructive sleep apnea, and coronary artery disease [84].
Moreover, in MUNW individuals, a higher incidence of subclinical atherosclerosis, with soft atherosclerotic plaques and an increased risk of plaque rupture and ischemic events, has been reported compared to healthy individuals [85].
A higher incidence of cardiovascular events, including MI, HF, and atrial fibrillation, in subjects with SO compared to those with normal body composition has been described [86]. Other studies demonstrated a poor prognosis after MI in SO patients, characterized by an increased rate of all-cause death, recurrent MI, ischemic stroke, and hospitalization for HF [87]. Indeed, sarcopenic obesity is characterized by several pathological alterations that may be associated with CVD, including increased oxidative stress and mitochondrial dysfunction [88]. The loss of skeletal muscle mass with the accumulation of intramuscular fat and the impairment of contractile function is associated with insulin resistance and metabolic abnormalities, which play a role in developing endothelial dysfunction, vascular remodeling, and cardiovascular events [89].

4. Therapeutic Strategies in Obesity Management

Although a primary objective should be the prevention of obesity before its development, integrated treatment initiatives should be encouraged and intensified when this pathology has already occurred [4]. Different pharmacological and non-pharmacological strategies have been proposed to reduce body weight effectively.
Excessive eating habits and proinflammatory dietary patterns should be carefully modulated and tailored to each individual. In particular, a diet low in calories and with nutrients with anti-inflammatory and immunomodulatory properties should be encouraged to reduce VAT accumulation with inflammatory phenotype [14,15]. Therefore, there should be recommendations for food rich in fibers with antioxidant polyphenols—such as eicosapentaenoic fatty acid (EPA) and docosahexaenoic fatty acid (DHA)—food rich in lipoperoxidated vegetable oils and xenobiotics, and low in salt and high saturated and trans fats. Diet suggestions should be associated with regular physical activity, particularly in childhood. The impact of excessive eating habits and physical activity restriction is particularly relevant during childhood, since in this period, the speed of formation of new fat cells is high, their number being three-fold in obese children compared to those with normal weight [90]. During this life period, environmental factors are of great importance. They might influence physiological feedback mechanisms that regulate food intake, consisting of short-term gastrointestinal stimuli and long-term signals from the adipose tissue [91].
In addition, a food intake at unusual times may produce an internal desynchronization between central and peripheral clocks, resulting in impaired secretion of hormones such as insulin, glucagon, corticosterone, leptin, adiponectin, visfatin, chemerin, and lipocalin, which regulate satiation, and appetite [92]. Also, oscillations in the microbiota composition have been described, with circadian rhythms, contributing to reduced hunger during sleep [93]. On this basis, it has been shown that chrono-disruption in food intake is associated with adverse metabolic effects, such as impaired glucose tolerance and diabetes and an increased risk of obesity [94]. In obese women, longer eating time during the day and quicker eating are described [95]. In addition, night eating has been related to increased BMI, with a reduction of equivalent energy expenditure, an increased risk for weight gain, and an impaired glycemic/insulinemic response [96]. Consistently, it has been shown that more intake of calories in the morning rather than in the evening improves glucose tolerance, particularly in diabetic patients [97].
Regarding other strategies to achieve weight loss, in addition to quantitative interventions on food intake and energy restriction, the consumption of low-energy-dense foods and whole-grain-containing diets represents a fundamental tool, increasing fullness, decreasing the intake of saturated fat and regulating metabolic functions and proinflammatory states [98,99]. Common dietetic strategies consist of hypocaloric diets, Mediterranean diets, high-protein diets (to preserve lean muscle mass and enhance satiety), low or moderate carbohydrate diets, low-fat diets, intermittent fasting, or time-restricted diets [87].
An emerging field of interest is represented by precision nutrition, which consists of tailored dietary and lifestyle recommendations based on nutrigenetics, metabolomics, and metagenomics, including anthropometric parameters, clinical biomarkers, genetic testing of single nucleotide polymorphisms, and analysis of gut microbiome [100].
When associated with dietary interventions, moderate to vigorous physical activity contributes to additional long-term weight loss (up to 20%), which may contribute to improve cardiovascular outcomes. In this regard, physical activity accumulated in bouts of at least 10 min duration is inversely associated with BMI and other parameters of body fatness [101].
Bariatric/metabolic surgery (BMS), defined as the procedures inducing loss of body weight throughout the modification of gastrointestinal physiology, still represents an effective and safe tool for patients with BMI ≥ 40 kg/m2 or BMI ≥ 35 kg/m2 with comorbidities, being associated with a 14–25% body weight reduction and a significant decrease in the risk of hypertension, diabetes, non-alcoholic fatty liver disease, and mortality [100]. Sleeve gastrectomy and Roux-en-Y gastric bypass account for approximately 90% of all operations performed worldwide with mid- and long-term efficacy. Biliopancreatic diversion with duodenal switch, one-anastomosis gastric bypass, and the less invasive adjustable gastric banding represents an alternative approach [100].
Different pharmacological strategies with varying safety profiles have been proposed to control body weight. These anti-obesity drugs improve weight and metabolic parameters, with different potency and effects. Nevertheless, the currently available data do not support unique and conclusive evidence for reducing hard cardiovascular outcomes. The first drugs consisted of sympathomimetic and serotonin-releasing drugs aimed to suppress appetite and increase energy expenditure, such as phentermine, sibutramine, and dexfenfluramine [102]. However, due to the release of norepinephrine, these agents were associated with an increased risk of cardiovascular adverse effects such as tachycardia, dyspnea, dysrhythmias, MI, and sudden cardiac death. Thus, their use has been progressively abandoned [102]. Other pharmacological agents with limited use in clinical practice are mazindol, which exerts an anorexigenic effect through a central action in the hypothalamus, and orlistat (tetrahydrolipstatin), which reversibly inhibits gastric and pancreatic lipases, avoiding the absorption of free fatty acids [102].
In the last few years, other effective pharmacological strategies with a better efficacy/safety profile have been introduced in clinical practice (Table 1).
The association of bupropion, used for addiction to opioids and alcohol, with naltrexone, used for the treatment of depression and nicotine addiction, has been shown to reduce body weight through the action of hypothalamic nucleus arcuatus and on the dopaminergic mesolimbic system, respectively stimulating pro-opiomelanocortin (POMC) neurons and blocking the negative feedback of β-endorphins [103].
More recently, GLP1-RA has shown significant results independently from diabetes, stimulating insulin secretion and glucose-lowering, slowing gastric emptying, and increasing hypothalamic sense of satiety through stimulating POMC neurons [104,105] (Figure 3).
Furthermore, GLP1-RA modulates lipid metabolism in white and brown adipose tissues, promoting brown adipose tissue thermogenesis and white adipose tissue browning [106,107]. The activation of the AMPK pathway in brown adipose tissue triggers lipolysis through the release of free fatty acids and the upregulation of uncoupling protein 1 (UCP1), which generates thermogenesis through the release of electrons during oxidative phosphorylation in the inner mitochondrial membrane [108]. Different studies have demonstrated that GLP1-RA decreases ROS production in endothelial cells and cardiomyocytes and reduces the expression of adhesion molecules, such as monocyte chemotactic protein-1 (MCP-1), E-selectin, intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1), and the accumulation of inflammatory cells, as well as inducing the expression of NO synthase-3 (NOS-3) leading to vasorelaxation and slowing the progression of atherosclerotic plaques [108].
In addition, ectopic adipose tissue depots have been reduced by GLP1-RA, which contributes to the favorable effects on the vascular endothelium and myocardium [109]. In the SCALE Obesity and Prediabetes study, liraglutide produced a body weight loss of 7.8 kg, which was maintained for three years and was associated with the improvement of metabolic profile and blood pressure control and with an 80% reduction of the risk of diabetes [110]. The STEP (Semaglutide Treatment Effect in People with Obesity) study and its sub-analyses showed that treatment with semaglutide produced a 5% reduction of body weight in >90% of subjects and a 20% reduction in about 35% [111]. In the STEP-HF study, 529 obese patients with HFpEF were randomized to receive once-weekly semaglutide (2.4 mg) or placebo for 52 weeks to investigate the effects on body weight and quality of life evaluated with the Kansas City Cardiomyopathy Questionnaire clinical summary score (KCCQ-CSS) [111]. Semaglutide produced a more significant improvement of KCCQ-CSS (+7.8 points), a greater reduction in body weight of 10.7%, and a 20.3 m improvement of the 6-min walking test compared to the placebo. No significant differences were reported in the risk of adverse events [112].
Besides their effects on weight reduction and glycemic control, GLP1-RA has been shown to reduce cardiovascular outcomes in obese subjects independently from diabetes. More recently, the SELECT (Semaglutide Effects on Cardiovascular Outcomes in People with Overweight or Obesity) study investigated the effects of semaglutide on the incidence of major adverse cardiovascular events in participants without diabetes and with established CVD, including prior MI, prior stroke, or symptomatic peripheral arterial disease (PAD), and overweight or obesity [113]. The study included 17,604 patients randomly assigned (1:1) to receive once-weekly subcutaneous semaglutide 2.4 mg or placebo [114]. The main inclusion criteria were age ≥  45 years, BMI ≥  27 kg/m2, prior MI, stroke, or symptomatic PAD. Exclusion criteria were a history of diabetes mellitus, treatment with glucose-lowering agents within the past 90 days, and HF in New York Heart Association (NYHA) class IV. The primary efficacy endpoint was a composite of death from cardiovascular causes, nonfatal MI, or nonfatal stroke, assessed in a time-to-first-event analysis. Confirmatory secondary endpoints, tested in hierarchical order, were cardiovascular death, a composite HF endpoint consisting of cardiovascular death, hospitalizations or urgent medical visits for HF, and death from any cause. After about 40 months of follow-up, a once-weekly dose of subcutaneous semaglutide produced a 20% reduction of the composite outcome of death from cardiovascular causes, nonfatal MI, or nonfatal stroke [113]. The trial was not powered to analyze statistical significance for mortality and other separate secondary endpoints. However, a trend toward reducing these outcomes has been reported [112]. In the semaglutide group, body weight was reduced by 9.4%, with a steady state reached at ten weeks and maintained throughout the follow-up period. In addition, semaglutide was associated with the reduction of systolic blood pressure, incidence of diabetes, LDL-cholesterol, and C-reactive protein. The effects of semaglutide were independent of the baseline BMI range and were also observed in overweight patients with a BMI < 30 kg/m2 [113,114,115].
In this regard, soluble guanylate cyclase stimulators, by improving sensitivity to NO and promoting the cGMP-PKG pathway, might represent a future potential synergistic therapeutic strategy that enhances the anti-fibrotic, anti-inflammatory, and anti-proliferative properties of GLP1-RA [116]. This association may exert favorable effects, particularly on cardiovascular remodeling in obese individuals.
Retatrutide is a novel triple agonist of the glucose-dependent insulinotropic polypeptide, glucagon-like peptide 1, and glucagon receptors. A phase 2 study investigated the effects of retatrutide in 338 obese subjects, showing a body weight reduction of up to 17.5% at 24 weeks and 24.2% at 48 weeks [117].
Tirzepatide is a once-weekly subcutaneous injectable peptide engineered from the native GIP sequence, with agonist activity at both the GIP and GLP-1 receptors, and it has been approved for type 2 diabetes. Tirzepatide also induces a delay of gastric emptying, reduces fasting and postprandial glucose concentration, and decreases food intake, with sustained weight reduction in obese adults [118]. In the ongoing SURPASS-CVOT (comparison of tirzepatide and dulaglutide on major adverse cardiovascular events in participants with type 2 diabetes and atherosclerotic cardiovascular disease), 13,299 diabetic subjects with established atherosclerotic CVD were randomized to receive tirzepatide, a once-weekly GIP/GLP-1 receptor agonist, and dulaglutide, a GLP1-RA, to investigate cardiovascular safety and efficacy [119].
Finally, taking into account the abnormal distribution of adipose tissue in obese patients and the different phenotypes, drug metabolism and elimination may be significantly impacted. Therefore, dose adjustments could be required [120]. This aspect may contribute to the failure of therapeutic strategies and the need to switch an agent to another or prescribe combination treatments to maintain the weight loss rate. Next-generation multi-omics might provide novel targets and predict the success of each therapeutic approach.

5. Conclusions

Obesity is a chronic disease with a heavy burden of metabolic and cardiovascular sequelae. Proinflammatory dietary patterns play an essential role in developing adiposopathy and obesity and promote insulin resistance and cytokine secretion, leading to vascular and cardiac damage. In this view, structured pharmacological and non-pharmacological interventions are needed to reduce the risk of cardiovascular events and improve quality of life. Different pharmacological strategies have been introduced to reduce body weight and control cardiometabolic sequelae. Notably, GLP1-RA has been shown to decrease body weight, improve glycemic control, and reduce cardiovascular outcomes, at least in secondary prevention, in obese subjects independently from diabetes. Thus, this class of drugs represents a new effective tool whose prescription should be implemented in clinical practice for cardiovascular protection. Different obesity phenotypes have been described, and further studies are needed to investigate the safety and efficacy profiles of available therapeutic tools in each clinical scenario, particularly for primary prevention.

Author Contributions

Conceptualization, G.G. and C.S.; methodology, G.G. and C.S.; investigation, G.G. and C.S.; writing—original draft preparation, G.G.; writing—review and editing, C.S., G.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. GBD 2015 Obesity Collaborators; Afshin, A.; Forouzanfar, M.H.; Reitsma, M.B.; Sur, P.; Estep, K.; Lee, A.; Marczak, L.; Mokdad, A.H.; Moradi-Lakeh, M.; et al. Health Effects of Overweight and Obesity in 195 Countries over 25 Years. N. Engl. J. Med. 2017, 377, 13–27. [Google Scholar]
  2. Koliaki, C.; Dalamaga, M.; Liatis, S. Update on the Obesity Epidemic: After the Sudden Rise, Is the Upward Trajectory Beginning to Flatten? Curr. Obes. Rep. 2023, 12, 514–527. [Google Scholar] [CrossRef] [PubMed]
  3. Bianchettin, R.G.; Lavie, C.J.; Lopez-Jimenez, F. Challenges in Cardiovascular Evaluation and Management of Obese Patients: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2023, 81, 490–504. [Google Scholar] [CrossRef]
  4. Powell-Wiley, T.M.; Poirier, P.; Burke, L.E.; Després, J.P.; Gordon-Larsen, P.; Lavie, C.J.; Lear, S.A.; Ndumele, C.E.; Neeland, I.J.; Sanders, P.; et al. Obesity and Cardiovascular Disease: A Scientific Statement from the American Heart Association. Circulation 2021, 143, e984–e1010. [Google Scholar] [CrossRef] [PubMed]
  5. Volpe, M.; Gallo, G. Obesity and cardiovascular disease: An executive document on pathophysiological and clinical links promoted by the Italian Society of Cardiovascular Prevention (SIPREC). Front. Cardiovasc. Med. 2023, 13, 1136340. [Google Scholar] [CrossRef]
  6. Alsaqaaby, M.S.; Cooney, S.; le Roux, C.W.; Pournaras, D.J. Sex, race, and BMI in clinical trials of medications for obesity over the past three decades: A systematic review. Lancet Diabetes Endocrinol. 2024, 12, 414–421. [Google Scholar] [CrossRef]
  7. Ross, R.; Neeland, I.J.; Yamashita, S.; Shai, I.; Seidell, J.; Magni, P.; Santos, R.D.; Arsenault, B.; Cuevas, A.; Hu, F.B.; 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. [Google Scholar] [CrossRef] [PubMed]
  8. Lopez-Jimenez, F.; Almahmeed, W.; Bays, H.; Cuevas, A.; Di Angelantonio, E.; le Roux, C.W.; Sattar, N.; Sun, M.C.; Wittert, G.; Pinto, F.J.; et al. Obesity and cardiovascular disease: Mechanistic insights and management strategies. A joint position paper by the World Heart Federation and World Obesity Federation. Eur. J. Prev. Cardiol. 2022, 29, 2218–2237. [Google Scholar] [CrossRef] [PubMed]
  9. Bays, H. Central obesity as a clinical marker of adiposopathy; increased visceral adiposity as a surrogate marker for global fat dysfunction. Curr. Opin. Endocrinol. Diabetes Obes. 2014, 21, 345–351. [Google Scholar] [CrossRef] [PubMed]
  10. Heinonen, S.; Saarinen, L.; Naukkarinen, J.; Rodríguez, A.; Frühbeck, G.; Hakkarainen, A.; Lundbom, J.; Lundbom, N.; Vuolteenaho, K.; Moilanen, E.; et al. Adipocyte morphology and implications for metabolic derangements in acquired obesity. Int. J. Obes. 2014, 38, 1423–1431. [Google Scholar] [CrossRef]
  11. Stern, J.H.; Rutkowski, J.M.; Scherer, P.E. Adiponectin, Leptin, and Fatty Acids in the Maintenance of Metabolic Homeostasis through Adipose Tissue Crosstalk. Cell Metab. 2016, 23, 770–784. [Google Scholar] [CrossRef]
  12. Aida, X.M.U.; Ivan, T.V.; Juan, G.J.R. Adipose Tissue Immunometabolism: Unveiling the Intersection of Metabolic and Immune Regulation. Rev. Investig. Clin. 2024, 76, 65–79. [Google Scholar]
  13. Tristan Asensi, M.; Napoletano, A.; Sofi, F.; Dinu, M. Low-Grade Inflammation and Ultra-Processed Foods Consumption: A Review. Nutrients 2023, 15, 1546. [Google Scholar] [CrossRef]
  14. Buechler, C.; Feder, S.; Haberl, E.M.; Aslanidis, C. Chemerin Isoforms and Activity in Obesity. Int. J. Mol. Sci. 2019, 20, 1128. [Google Scholar] [CrossRef] [PubMed]
  15. Amano, S.U.; Cohen, J.L.; Vangala, P.; Tencerova, M.; Nicoloro, S.M.; Yawe, J.C.; Shen, Y.; Czech, M.P.; Aouadi, M. Local proliferation of macrophages contributes to obesity-associated adipose tissue inflammation. Cell Metab. 2014, 19, 162–171. [Google Scholar] [CrossRef] [PubMed]
  16. Coats, B.R.; Schoenfelt, K.Q.; Barbosa-Lorenzi, V.C.; Peris, E.; Cui, C.; Hoffman, A.; Zhou, G.; Fernandez, S.; Zhai, L.; Hall, B.A.; et al. Metabolically Activated Adipose Tissue Macrophages Perform Detrimental and Beneficial Functions during Diet-Induced Obesity. Cell Rep. 2017, 20, 3149–3161. [Google Scholar] [CrossRef] [PubMed]
  17. Reilly, S.M.; Saltiel, A.R. Adapting to obesity with adipose tissue inflammation. Nat. Rev. Endocrinol. 2017, 13, 633–643. [Google Scholar]
  18. Karshovska, E.; Wei, Y.; Subramanian, P.; Mohibullah, R.; Geißler, C.; Baatsch, I.; Popal, A.; Corbalán Campos, J.; Exner, N.; Schober, A. HIF-1α (Hypoxia-Inducible Factor-1α) Promotes Macrophage Necroptosis by Regulating miR-210 and miR-383. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 583–596. [Google Scholar] [CrossRef]
  19. Liu, X.; Sun, H.; Zheng, L.; Zhang, J.; Su, H.; Li, B.; Wu, Q.; Liu, Y.; Xu, Y.; Song, X.; et al. Adipose-derived miRNAs as potential biomarkers for predicting adulthood obesity and its complications: A systematic review and bioinformatic analysis. Obes. Rev. 2024, 25, e13748. [Google Scholar] [CrossRef]
  20. Tong, M.; Saito, T.; Zhai, P.; Oka, S.I.; Mizushima, W.; Nakamura, M.; Ikeda, S.; Shirakabe, A.; Sadoshima, J. Mitophagy Is Essential for Maintaining Cardiac Function During High Fat Diet-Induced Diabetic Cardiomyopathy. Circ. Res. 2019, 124, 1360–1371. [Google Scholar] [CrossRef] [PubMed]
  21. Shimizu, I.; Yoshida, Y.; Katsuno, T.; Tateno, K.; Okada, S.; Moriya, J.; Yokoyama, M.; Nojima, A.; Ito, T.; Zechner, R.; et al. p53-induced adipose tissue inflammation is critically involved in the development of insulin resistance in heart failure. Cell Metab. 2012, 15, 51–64. [Google Scholar] [CrossRef] [PubMed]
  22. Soedono, S.; Cho, K.W. Adipose Tissue Dendritic Cells: Critical Regulators of Obesity-Induced Inflammation and Insulin Resistance. Int. J. Mol. Sci. 2021, 22, 8666. [Google Scholar] [CrossRef] [PubMed]
  23. Li, C.; Wang, G.; Sivasami, P.; Ramirez, R.N.; Zhang, Y.; Benoist, C.; Mathis, D. Interferon-α-producing plasmacytoid dendritic cells drive the loss of adipose tissue regulatory T cells during obesity. Cell Metab. 2021, 33, 1610–1623.e5. [Google Scholar] [CrossRef] [PubMed]
  24. Valentine, Y.; Nikolajczyk, B.S. T cells in obesity-associated inflammation: The devil is in the details. Immunol. Rev. 2024, 324, 25–41. [Google Scholar] [CrossRef]
  25. Fabbrini, E.; Cella, M.; McCartney, S.A.; Fuchs, A.; Abumrad, N.A.; Pietka, T.A.; Chen, Z.; Finck, B.N.; Han, D.H.; Magkos, F.; et al. Association between specific adipose tissue CD4+ T-cell populations and insulin resistance in obese individuals. Gastroenterology 2013, 145, e1–e3. [Google Scholar] [CrossRef]
  26. Chung, S.; Lapoint, K.; Martinez, K.; Kennedy, A.; Boysen Sandberg, M.; McIntosh, M.K. Preadipocytes mediate lipopolysaccharide-induced inflammation and insulin resistance in primary cultures of newly differentiated human adipocytes. Endocrinology 2006, 147, 5340–5351. [Google Scholar] [CrossRef]
  27. Odegaard, J.I.; Chawla, A. Alternative macrophage activation and metabolism. Annu. Rev. Pathol. 2011, 6, 275–297. [Google Scholar] [CrossRef]
  28. Huber, J.; Kiefer, F.W.; Zeyda, M.; Ludvik, B.; Silberhumer, G.R.; Prager, G.; Zlabinger, G.J.; Stulnig, T.M. CC chemokine and CC chemokine receptor profiles in visceral and subcutaneous adipose tissue are altered in human obesity. J. Clin. Endocrinol. Metab. 2008, 93, 3215–3221. [Google Scholar] [CrossRef] [PubMed]
  29. Saetang, J.; Sangkhathat, S. Role of innate lymphoid cells in obesity and metabolic disease (Review). Mol. Med. Rep. 2018, 17, 1403–1412. [Google Scholar] [CrossRef] [PubMed]
  30. Bertola, A.; Ciucci, T.; Rousseau, D.; Bourlier, V.; Duffaut, C.; Bonnafous, S.; Blin-Wakkach, C.; Anty, R.; Iannelli, A.; Gugenheim, J.; et al. Identification of adipose tissue dendritic cells correlated with obesity-associated insulin-resistance and inducing Th17 responses in mice and patients. Diabetes 2012, 61, 2238–2247. [Google Scholar] [CrossRef] [PubMed]
  31. Rizk, N.M.; Fadel, A.; AlShammari, W.; Younes, N.; Bashah, M. The Immunophenotyping Changes of Peripheral CD4+ T Lymphocytes and Inflammatory Markers of Class III Obesity Subjects After Laparoscopic Gastric Sleeve Surgery—A Follow-Up Study. J. Inflamm. Res. 2021, 14, 1743–1757. [Google Scholar] [CrossRef]
  32. Zou, J.; Lai, B.; Zheng, M.; Chen, Q.; Jiang, S.; Song, A.; Huang, Z.; Shi, P.; Tu, X.; Wang, D.; et al. CD4+ T cells memorize obesity and promote weight regain. Cell Mol. Immunol. 2018, 15, 630–639. [Google Scholar] [CrossRef] [PubMed]
  33. Turbitt, W.J.; Buchta Rosean, C.; Weber, K.S.; Norian, L.A. Obesity and CD8 T cell metabolism: Implications for anti-tumor immunity and cancer immunotherapy outcomes. Immunol. Rev. 2020, 295, 203–219. [Google Scholar] [CrossRef] [PubMed]
  34. Li, Q.; Hagberg, C.E.; Silva Cascales, H.; Lang, S.; Hyvönen, M.T.; Salehzadeh, F.; Chen, P.; Alexandersson, I.; Terezaki, E.; Harms, M.J.; et al. Obesity and hyperinsulinemia drive adipocytes to activate a cell cycle program and senesce. Nat. Med. 2021, 27, 1941–1953. [Google Scholar] [CrossRef] [PubMed]
  35. Bergheim, I.; Moreno-Navarrete, J.M. The relevance of intestinal barrier dysfunction, antimicrobial proteins and bacterial endotoxin in metabolic dysfunction-associated steatotic liver disease. Eur. J. Clin. Investig. 2024, 54, e14224. [Google Scholar] [CrossRef]
  36. Fan, Y.; Li, Y.; Chu, Y.; Liu, J.; Cui, L.; Zhang, D. Toll-Like Receptors Recognize Intestinal Microbes in Liver Cirrhosis. Front. Immunol. 2021, 12, 608498. [Google Scholar] [CrossRef] [PubMed]
  37. Lu, Y.C.; Yeh, W.C.; Ohashi, P.S. LPS/TLR4 signal transduction pathway. Cytokine 2008, 42, 145–151. [Google Scholar] [CrossRef]
  38. Faienza, M.F.; Urbano, F.; Anaclerio, F.; Moscogiuri, L.A.; Konstantinidou, F.; Stuppia, L.; Gatta, V. Exploring Maternal Diet-Epigenetic-Gut Microbiome Crosstalk as an Intervention Strategy to Counter Early Obesity Programming. Curr. Issues Mol. Biol. 2024, 46, 4358–4378. [Google Scholar] [CrossRef]
  39. Liu, B.N.; Liu, X.T.; Liang, Z.H.; Wang, J.H. Gut microbiota in obesity. World J. Gastroenterol. 2021, 27, 3837–3850. [Google Scholar] [CrossRef]
  40. Bastin, M.; Andreelli, F. The gut microbiota and diabetic cardiomyopathy in humans. Diabetes Metab. 2020, 46, 197–202. [Google Scholar] [CrossRef]
  41. Chavoya-Guardado, M.A.; Vasquez-Garibay, E.M.; Ruiz-Quezada, S.L.; Ramírez-Cordero, M.I.; Larrosa-Haro, A.; Castro-Albarran, J. Firmicutes, Bacteroidetes and Actinobacteria in Human Milk and Maternal Adiposity. Nutrients 2022, 14, 2887. [Google Scholar] [CrossRef] [PubMed]
  42. Lowe, W.L.; Lowe, L.P.; Kuang, A.; Catalano, P.M.; Nodzenski, M.; Talbot, O.; Tam, W.H.; Sacks, D.A.; McCance, D.; Linder, B.; et al. Maternal glucose levels during pregnancy and childhood adiposity in the Hyperglycemia and Adverse Pregnancy Outcome Follow-up Study. Diabetologia 2019, 62, 598–610. [Google Scholar] [CrossRef] [PubMed]
  43. Savoia, C.; Schiffrin, E.L. Reduction of C-reactive protein and the use of anti-hypertensives. Vasc. Health Risk Manag. 2007, 3, 975–983. [Google Scholar]
  44. Touyz, R.M.; Savoia, C.; He, Y.; Endemann, D.; Pu, Q.; Ko, E.A.; De Ciuceis, C.; Montezano, A.; Schiffrin, E.L. Increased inflammatory biomarkers in hypertensive type 2 diabetic patients: Improvement after angiotensin II type 1 receptor blockade. J. Am. Soc. Hypertens. 2007, 1, 189–199. [Google Scholar] [CrossRef] [PubMed]
  45. Welsh, A.; Hammad, M.; Piña, I.L.; Kulinski, J. Obesity and cardiovascular health. Eur. J. Prev. Cardiol. 2024, 31, 1026–1035. [Google Scholar] [CrossRef] [PubMed]
  46. Koenen, M.; Hill, M.A.; Cohen, P.; Sowers, J.R. Obesity, Adipose Tissue and Vascular Dysfunction. Circ. Res. 2021, 128, 951–968. [Google Scholar] [CrossRef]
  47. Chang, L.; Garcia-Barrio, M.T.; Chen, Y.E. Perivascular adipose tissue regulates vascular function by targeting vascular smooth muscle cells. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 1094–1109. [Google Scholar] [CrossRef] [PubMed]
  48. Savoia, C.; Arrabito, A.; Parente, R.; Nicoletti, C.; Madaro, L.; Battistoni, A.; Filippini, A.; Steckelings, U.M.; Touyz, R.M.; Volpe, M. Mas Receptor Activation Contributes to the Improvement of Nitric Oxide Bioavailability and Vascular Remodeling during Chronic AT1R (Angiotensin Type-1 Receptor) Blockade in Experimental Hypertension. Hypertension 2020, 76, 1753–1761. [Google Scholar] [CrossRef]
  49. Gil-Ortega, M.; Stucchi, P.; Guzman-Ruiz, R.; Cano, V.; Arribas, S.; Gonzalez, M.C.; Ruiz-Gayo, M.; Fernández-Alfonso, M.S.; Somoza, B. Adaptative nitric oxide overproduction in perivascular adipose tissue during early diet-induced obesity. Endocrinology 2010, 151, 3299–3306. [Google Scholar] [CrossRef]
  50. Lechner, K.; McKenzie, A.L.; Kränkel, N.; Von Schacky, C.; Worm, N.; Nixdorff, U.; Lechner, B.; Scherr, J.; Weingärtner, O.; Krauss, R.M. High-Risk Atherosclerosis and Metabolic Phenotype: The Roles of Ectopic Adiposity, Atherogenic Dyslipidemia, and Inflammation. Metab. Syndr. Relat. Disord. 2020, 18, 176–185. [Google Scholar] [CrossRef]
  51. Del Campo, C.V.; Liaw, N.Y.; Gunadasa-Rohling, M.; Matthaei, M.; Braga, L.; Kennedy, T.; Salinas, G.; Voigt, N.; Giacca, M.; Zimmermann, W.H.; et al. Regenerative potential of epicardium-derived extracellular vesicles mediated by conserved miRNA transfer. Cardiovasc. Res. 2022, 118, 597–611. [Google Scholar] [CrossRef] [PubMed]
  52. Ferrara, D.; Montecucco, F.; Dallegri, F.; Carbone, F. Impact of different ectopic fat depots on cardiovascular and metabolic diseases. J. Cell Physiol. 2019, 234, 21630–21641. [Google Scholar] [CrossRef] [PubMed]
  53. Kruszewska, J.; Cudnoch-Jedrzejewska, A.; Czarzasta, K. Remodeling and Fibrosis of the Cardiac Muscle in the Course of Obesity-Pathogenesis and Involvement of the Extracellular Matrix. Int. J. Mol. Sci. 2022, 23, 4195. [Google Scholar] [CrossRef] [PubMed]
  54. Dadson, K.; Chasiotis, H.; Wannaiampikul, S.; Tungtrongchitr, R.; Xu, A.; Sweeney, G. Adiponectin mediated APPL1-AMPK signaling induces cell migration, MMP activation, and collagen remodeling in cardiac fibroblasts. J. Cell Biochem. 2014, 115, 785–793. [Google Scholar] [CrossRef] [PubMed]
  55. Paredes, S.; Fonseca, L.; Ribeiro, L.; Ramos, H.; Oliveira, J.C.; Palma, I. Novel and traditional lipid profiles in Metabolic Syndrome reveal a high atherogenicity. Sci. Rep. 2019, 9, 11792. [Google Scholar] [CrossRef] [PubMed]
  56. De Paolis, P.; Porcellini, A.; Savoia, C.; Lombardi, A.; Gigante, B.; Frati, G.; Rubattu, S.; Musumeci, B.; Volpe, M. Functional cross-talk between angiotensin II and epidermal growth factor receptors in NIH3T3 fibroblasts. J. Hypertens. 2002, 20, 693–699. [Google Scholar] [CrossRef]
  57. Raman, P.; Krukovets, I.; Marinic, T.E.; Bornstein, P.; Stenina, O.I. Glycosylation mediates up-regulation of a potent antiangiogenic and proatherogenic protein, thrombospondin-1, by glucose in vascular smooth muscle cells. J. Biol. Chem. 2007, 282, 5704–5714. [Google Scholar] [CrossRef]
  58. Volpe, M.; Gallo, G.; Rubattu, S. Endocrine functions of the heart: From bench to bedside. Eur. Heart J. 2023, 44, 643–655. [Google Scholar] [CrossRef] [PubMed]
  59. Iyengar, N.M.; Gucalp, A.; Dannenberg, A.J.; Hudis, C.A. Obesity and Cancer Mechanisms: Tumor Microenvironment and Inflammation. J. Clin. Oncol. 2016, 34, 4270–4276. [Google Scholar] [CrossRef]
  60. Preda, A.; Carbone, F.; Tirandi, A.; Montecucco, F.; Liberale, L. Obesity phenotypes and cardiovascular risk: From pathophysiology to clinical management. Rev. Endocr. Metab. Disord. 2023, 24, 901–919. [Google Scholar] [CrossRef] [PubMed]
  61. Badoud, F.; Perreault, M.; Zulyniak, M.A.; Mutch, D.M. Molecular insights into the role of white adipose tissue in metabolically unhealthy normal weight and metabolically healthy obese individuals. FASEB J. 2015, 29, 748–758. [Google Scholar] [CrossRef]
  62. Goossens, G.H. The Metabolic Phenotype in Obesity: Fat Mass, Body Fat Distribution, and Adipose Tissue Function. Obes. Facts. 2017, 10, 207–215. [Google Scholar] [CrossRef]
  63. Iacobini, C.; Pugliese, G.; Blasetti Fantauzzi, C.; Federici, M.; Menini, S. Metabolically healthy versus metabolically unhealthy obesity. Metabolism 2019, 92, 51–60. [Google Scholar] [CrossRef]
  64. Stefan, N.; Häring, H.U.; Hu, F.B.; Schulze, M.B. Metabolically healthy obesity: Epidemiology, mechanisms, and clinical implications. Lancet Diabetes Endocrinol. 2013, 1, 152–162. [Google Scholar] [CrossRef] [PubMed]
  65. Kramer, C.K.; Zinman, B.; Retnakaran, R. Are metabolically healthy overweight and obesity benign conditions?: A systematic review and meta-analysis. Ann. Intern. Med. 2013, 159, 758–769. [Google Scholar] [CrossRef] [PubMed]
  66. Lavie, C.J.; Laddu, D.; Arena, R.; Ortega, F.B.; Alpert, M.A.; Kushner, R.F. Healthy Weight and Obesity Prevention: JACC Health Promotion Series. J. Am. Coll. Cardiol. 2018, 72, 1506–1531. [Google Scholar] [CrossRef] [PubMed]
  67. Zheng, R.; Zhou, D.; Zhu, Y. The long-term prognosis of cardiovascular disease and all-cause mortality for metabolically healthy obesity: A systematic review and meta-analysis. J. Epidemiol. Community Health 2016, 70, 1024–1031. [Google Scholar] [CrossRef]
  68. Després, J.P. Body fat distribution and risk of cardiovascular disease: An update. Circulation 2012, 126, 1301–1313. [Google Scholar] [CrossRef]
  69. Candi, E.; Tesauro, M.; Cardillo, C.; Lena, A.M.; Schinzari, F.; Rodia, G.; Sica, G.; Gentileschi, P.; Rovella, V.; Annicchiarico-Petruzzelli, M.; et al. Metabolic profiling of visceral adipose tissue from obese subjects with or without metabolic syndrome. Biochem. J. 2018, 475, 1019–1035. [Google Scholar] [CrossRef] [PubMed]
  70. McLaughlin, T.; Deng, A.; Yee, G.; Lamendola, C.; Reaven, G.; Tsao, P.S.; Cushman, S.W.; Sherman, A. Inflammation in subcutaneous adipose tissue: Relationship to adipose cell size. Diabetologia 2010, 53, 369–377. [Google Scholar] [CrossRef]
  71. Appleton, S.L.; Seaborn, C.J.; Visvanathan, R.; Hill, C.L.; Gill, T.K.; Taylor, A.W.; Adams, R.J.; North West Adelaide Health Study Team. Diabetes and cardiovascular disease outcomes in the metabolically healthy obese phenotype: A cohort study. Diabetes Care 2013, 36, 2388–2394. [Google Scholar] [CrossRef]
  72. Mongraw-Chaffin, M.; Foster, M.C.; Anderson, C.A.M.; Burke, G.L.; Haq, N.; Kalyani, R.R.; Ouyang, P.; Sibley, C.T.; Tracy, R.; Woodward, M.; et al. Metabolically Healthy Obesity, Transition to Metabolic Syndrome, and Cardiovascular Risk. J. Am. Coll. Cardiol. 2018, 71, 1857–1865. [Google Scholar] [CrossRef]
  73. Cui, Z.; Truesdale, K.P.; Bradshaw, P.T.; Cai, J.; Stevens, J. Three-year weight change and cardiometabolic risk factors in obese and normal weight adults who are metabolically healthy: The atherosclerosis risk in communities study. Int. J. Obes. 2015, 39, 1203–1208. [Google Scholar] [CrossRef] [PubMed]
  74. Zhou, Z.; Macpherson, J.; Gray, S.R.; Gill, J.M.R.; Welsh, P.; Celis-Morales, C.; Sattar, N.; Pell, J.P.; Ho, F.K. Are people with metabolically healthy obesity really healthy? A prospective cohort study of 381,363 UK Biobank participants. Diabetologia 2021, 64, 1963–1972. [Google Scholar] [CrossRef]
  75. Stefan, N.; Schick, F.; Häring, H.U. Causes, Characteristics, and Consequences of Metabolically Unhealthy Normal Weight in Humans. Cell Metab. 2017, 26, 292–300. [Google Scholar] [CrossRef] [PubMed]
  76. Wijayatunga, N.N.; Dhurandhar, E.J. Normal weight obesity and unaddressed cardiometabolic health risk-a narrative review. Int. J. Obes. 2021, 45, 2141–2155. [Google Scholar] [CrossRef] [PubMed]
  77. Stenholm, S.; Harris, T.B.; Rantanen, T.; Visser, M.; Kritchevsky, S.B.; Ferrucci, L. Sarcopenic obesity: Definition, cause and consequences. Curr. Opin. Clin. Nutr. Metab. Care 2008, 11, 693–700. [Google Scholar] [CrossRef]
  78. Consitt, L.A.; Clark, B.C. The Vicious Cycle of Myostatin Signaling in Sarcopenic Obesity: Myostatin Role in Skeletal Muscle Growth, Insulin Signaling and Implications for Clinical Trials. J. Frailty Aging 2018, 7, 21–27. [Google Scholar] [CrossRef] [PubMed]
  79. Pinel, A.; Guillet, C.; Capel, F.; Pouget, M.; De Antonio, M.; Pereira, B.; Topinkova, E.; Eglseer, D.; Barazzoni, R.; Cruz-Jentoft, A.J. Identification of factors associated with sarcopenic obesity development: Literature review and expert panel voting. Clin. Nutr. 2024, 43, 1414–1424. [Google Scholar] [CrossRef] [PubMed]
  80. Ballin, M.; Nordström, P.; Niklasson, J.; Nordström, A. Associations of Visceral Adipose Tissue and Skeletal Muscle Density with Incident Stroke, Myocardial Infarction, and All-Cause Mortality in Community-Dwelling 70-Year-Old Individuals: A Prospective Cohort Study. J. Am. Heart Assoc. 2021, 10, e020065. [Google Scholar] [CrossRef]
  81. Atkins, J.L.; Wannamathee, S.G. Sarcopenic obesity in ageing: Cardiovascular outcomes and mortality. Br. J. Nutr. 2020, 124, 1102–1113. [Google Scholar] [CrossRef] [PubMed]
  82. Fauchier, G.; Bisson, A.; Bodin, A.; Herbert, J.; Semaan, C.; Angoulvant, D.; Ducluzeau, P.H.; Lip, G.Y.H.; Fauchier, L. Metabolically healthy obesity and cardiovascular events: A nationwide cohort study. Diabetes Obes. Metab. 2021, 23, 2492–2501. [Google Scholar] [CrossRef]
  83. Neeland, I.J.; Gupta, S.; Ayers, C.R.; Turer, A.T.; Rame, J.E.; Das, S.R.; Berry, J.D.; Khera, A.; McGuire, D.K.; Vega, G.L.; et al. Relation of Regional Fat Distribution to Left Ventricular Structure and Function. Circ. Cardiovasc. Imaging 2013, 6, 800–807. [Google Scholar] [CrossRef]
  84. Cordola Hsu, A.R.; Xie, B.; Peterson, D.V.; LaMonte, M.J.; Garcia, L.; Eaton, C.B.; Going, S.B.; Phillips, L.S.; Manson, J.E.; Anton-Culver, H.; et al. Metabolically Healthy/Unhealthy Overweight/Obesity Associations with Incident Heart Failure in Postmenopausal Women: The Women’s Health Initiative. Circ. Heart Fail. 2021, 14, e007297. [Google Scholar] [CrossRef] [PubMed]
  85. Kim, S.; Kyung, C.; Park, J.S.; Lee, S.P.; Kim, H.K.; Ahn, C.W.; Kim, K.R.; Kang, S. Normal-weight obesity is associated with increased risk of subclinical atherosclerosis. Cardiovasc. Diabetol. 2015, 14, 58. [Google Scholar] [CrossRef]
  86. Baumgartner, R.N.; Wayne, S.J.; Waters, D.L.; Janssen, I.; Gallagher, D.; Morley, J.E. Sarcopenic obesity predicts instrumental activities of daily living disability in the elderly. Obes. Res. 2004, 12, 1995–2004. [Google Scholar] [CrossRef]
  87. Sato, R.; Okada, K.; Akiyama, E.; Konishi, M.; Matsuzawa, Y.; Nakahashi, H.; Minamimoto, Y.; Kimura, Y.; Maejima, N.; Iwahashi, N. Impact of sarcopenic obesity on long-term clinical outcomes after ST-segment elevation myocardial infarction. Atherosclerosis 2021, 335, 135–141. [Google Scholar] [CrossRef]
  88. Nishikawa, H.; Asai, A.; Fukunishi, S.; Nishiguchi, S.; Higuchi, K. Metabolic Syndrome and Sarcopenia. Nutrients 2021, 13, 3519. [Google Scholar] [CrossRef] [PubMed]
  89. Rubio-Ruiz, M.E.; Guarner-Lans, V.; Pérez-Torres, I.; Soto, M.E. Mechanisms underlying metabolic syndrome-related sarcopenia and possible therapeutic measures. Int. J. Mol. Sci. 2019, 20, 647. [Google Scholar] [CrossRef]
  90. Palacios-Marin, I.; Serra, D.; Jiménez-Chillarón, J.C.; Herrero, L.; Todorčević, M. Childhood obesity: Implications on adipose tissue dynamics and metabolic health. Obes. Rev. 2023, 24, e13627. [Google Scholar] [CrossRef]
  91. Kim, H.J.; Kwon, O. Nutrition and exercise: Cornerstones of health with emphasis on obesity and type 2 diabetes management-A narrative review. Obes. Rev. 2024, 25, e13762. [Google Scholar] [CrossRef]
  92. Peters, B.; Vahlhaus, J.; Pivovarova-Ramich, O. Meal timing and its role in obesity and associated diseases. Front. Endocrinol. 2024, 15, 1359772. [Google Scholar] [CrossRef] [PubMed]
  93. Collado, M.C.; Engen, P.A.; Bandín, C.; Cabrera-Rubio, R.; Voigt, R.M.; Green, S.J.; Naqib, A.; Keshavarzian, A.; Scheer, F.A.J.L.; Garaulet, M. Timing of food intake impacts daily rhythms of human salivary microbiota: A randomized, crossover study. FASEB J. 2018, 32, 2060–2072. [Google Scholar] [CrossRef]
  94. Shi, S.Q.; Ansari, T.S.; McGuinness, O.P.; Wasserman, D.H.; Johnson, C.H. Circadian disruption leads to insulin resistance and obesity. Curr. Biol. 2013, 23, 372–381. [Google Scholar] [CrossRef]
  95. Corbalán-Tutau, M.D.; Madrid, J.A.; Garaulet, M. Timing and duration of sleep and meals in obese and normal weight women. Association with increase blood pressure. Appetite 2012, 59, 9–16. [Google Scholar] [CrossRef] [PubMed]
  96. Bo, S.; Fadda, M.; Castiglione, A.; Ciccone, G.; De Francesco, A.; Fedele, D.; Guggino, A.; Parasiliti Caprino, M.; Ferrara, S. Is the timing of caloric intake associated with variation in diet-induced thermogenesis and in the metabolic pattern? A randomized cross-over study. Int. J. Obes. 2015, 39, 1689–1695. [Google Scholar] [CrossRef]
  97. Baron, K.G.; Reid, K.J.; Kern, A.S.; Zee, P.C. Role of sleep timing in caloric intake and BMI. Obesity 2011, 19, 1374–1381. [Google Scholar] [CrossRef]
  98. Hannon, S.C.; Hillier, S.E.; Thondre, P.S.; Clegg, M.E. Lower Energy-Dense Ready Meal Consumption Affects Self-Reported Appetite Ratings with No Effect on Subsequent Food Intake in Women. Nutrients 2021, 13, 4505. [Google Scholar] [CrossRef] [PubMed]
  99. Cheng, W.-Y.; Yeh, W.-J.; Ko, J.; Huang, Y.-L.; Yang, H.-Y. Consumption ofDehulledAdlay Improved LipidMetabolismand Inflammation in Overweight and Obese Individuals after a 6-Week Single-ArmPilot Study. Nutrients 2022, 14, 2250. [Google Scholar] [CrossRef]
  100. Eisenberg, D.; Shikora, S.A.; Aarts, E.; Aminian, A.; Angrisani, L.; Cohen, R.V.; de Luca, M.; Faria, S.L.; Goodpaster, K.P.S.; Haddad, A.; et al. 2022 American Society of Metabolic and Bariatric Surgery (ASMBS) and International Federation for the Surgery of Obesity and Metabolic Disorders (IFSO) Indications for Metabolic and Bariatric Surgery. Obes. Surg. 2023, 33, 3–14. [Google Scholar] [CrossRef]
  101. Jakicic, J.M.; Rogers, R.J.; Davis, K.K.; Collins, K.A. Role of Physical Activity and Exercise in Treating Patients with Overweight and Obesity. Clin. Chem. 2018, 64, 99–107. [Google Scholar] [CrossRef]
  102. Chakhtoura, M.; Haber, R.; Ghezzawi, M.; Rhayem, C.; Tcheroyan, R.; Mantzoros, C.S. Pharmacotherapy of obesity: An update on the available medications and drugs under investigation. EClinicalMedicine 2023, 20, 101882. [Google Scholar] [CrossRef] [PubMed]
  103. Nissen, S.E.; Wolski, K.E.; Prcela, L.; Wadden, T.; Buse, J.B.; Bakris, G.; Perez, A.; Smith, S.R. Effect of Naltrexone-Bupropion on Major Adverse Cardiovascular Events in Overweight and Obese Patients with Cardiovascular Risk Factors: A Randomized Clinical Trial. JAMA 2016, 315, 990–1004. [Google Scholar] [CrossRef] [PubMed]
  104. Michos, E.D.; Lopez-Jimenez, F.; Gulati, M. Role of Glucagon-Like Peptide-1 Receptor Agonists in Achieving Weight Loss and Improving Cardiovascular Outcomes in People with Overweight and Obesity. J. Am. Heart Assoc. 2023, 12, e029282. [Google Scholar] [CrossRef]
  105. Gallo, G.; Volpe, M. Potential Mechanisms of the Protective Effects of the Cardiometabolic Drugs Type-2 Sodium-Glucose Transporter Inhibitors and Glucagon-like Peptide-1 Receptor Agonists in Heart Failure. Int. J. Mol. Sci. 2024, 25, 2484. [Google Scholar] [CrossRef]
  106. Beiroa, D.; Imbernon, M.; Gallego, R.; Senra, A.; Herranz, D.; Villarroya, F.; Serrano, M.; Fernø, J.; Salvador, J.; Escalada, J.; et al. GLP-1 agonism stimulates brown adipose tissue thermogenesis and browning through hypothalamic AMPK. Diabetes 2014, 63, 3346–3358. [Google Scholar] [CrossRef]
  107. Zhu, E.; Yang, Y.; Zhang, J.; Li, Y.; Li, C.; Chen, L.; Sun, B. Liraglutide suppresses obesity and induces brown fat-like phenotype via Soluble Guanylyl Cyclase mediated pathway in vivo and in vitro. Oncotarget 2016, 7, 81077–81089. [Google Scholar] [CrossRef] [PubMed]
  108. Luna-Marco, C.; de Marañon, A.M.; Hermo-Argibay, A.; Rodriguez-Hernandez, Y.; Hermenejildo, J.; Fernandez-Reyes, M.; Apostolova, N.; Vila, J.; Sola, E.; Morillas, C.; et al. Effects of GLP-1 receptor agonists on mitochondrial function, inflammatory markers and leukocyte-endothelium interactions in type 2 diabetes. Redox Biol. 2023, 66, 102849. [Google Scholar] [CrossRef]
  109. Liu, F.; Yang, Q.; Zhang, H.; Zhang, Y.; Yang, G.; Ban, B.; Li, Y.; Zhang, M. The effects of glucagon-like peptide-1 receptor agonists on adipose tissues in patients with type 2 diabetes: A meta-analysis of randomised controlled trials. PLoS ONE 2022, 17, e0270899. [Google Scholar] [CrossRef] [PubMed]
  110. Davies, M.J.; Bergenstal, R.; Bode, B.; Kushner, R.F.; Lewin, A.; Skjøth, T.V.; Andreasen, A.H.; Jensen, C.B.; DeFronzo, R.A. NN8022-1922 Study Group. Efficacy of Liraglutide for Weight Loss Among Patients with Type 2 Diabetes: The SCALE Diabetes Randomized Clinical Trial. JAMA 2015, 314, 687–699. [Google Scholar] [CrossRef]
  111. Wilding, J.P.H.; Batterham, R.L.; Calanna, S.; Davies, M.; Van Gaal, L.F.; Lingvay, I.; McGowan, B.M.; Rosenstock, J.; Tran, M.T.D.; Wadden, T.A.; et al. Once-Weekly Semaglutide in Adults with Overweight or Obesity. N. Engl. J. Med. 2021, 384, 989–1002. [Google Scholar] [CrossRef] [PubMed]
  112. Kosiborod, M.N.; Abildstrøm, S.Z.; Borlaug, B.A.; Butler, J.; Rasmussen, S.; Davies, M.; Hovingh, G.K.; Kitzman, D.W.; Lindegaard, M.L.; Møller, D.V.; et al. Semaglutide in Patients with Heart Failure with Preserved Ejection Fraction and Obesity. N. Engl. J. Med. 2023, 389, 1069–1084. [Google Scholar] [CrossRef] [PubMed]
  113. Lincoff, A.M.; Brown-Frandsen, K.; Colhoun, H.M.; Deanfield, J.; Emerson, S.S.; Esbjerg, S.; Hardt-Lindberg, S.; Hovingh, G.K.; Kahn, S.E.; Kushner, R.F.; et al. Semaglutide and Cardiovascular Outcomes in Obesity without Diabetes. N. Engl. J. Med. 2023, 389, 2221–2232. [Google Scholar] [CrossRef] [PubMed]
  114. Ferrera, A.; Gallo, G.; Volpe, M. Obesity and Overweight: The “Elephant in the Room” That We can No Longer Ignore: Time to SELECT Treatments. High Blood Press. Cardiovasc. Prev. 2024, 31, 3–6. [Google Scholar] [CrossRef]
  115. Gallo, G.; Rubattu, S.; Volpe, M. Targeting Cyclic Guanylate Monophosphate in Resistant Hypertension and Heart Failure: Are Sacubitril/Valsartan and Vericiguat Synergistic and Effective in Both Conditions? High Blood Press. Cardiovasc. Prev. 2021, 28, 541–545. [Google Scholar] [CrossRef]
  116. Liuzzo, G.; Patrono, C. Reducing cardiovascular outcomes with semaglutide: A metabolic route for a SELECT few. Eur. Heart J. 2024, 45, 570–571. [Google Scholar] [CrossRef] [PubMed]
  117. Jastreboff, A.M.; Kaplan, L.M.; Frías, J.P.; Wu, Q.; Du, Y.; Gurbuz, S.; Coskun, T.; Haupt, A.; Milicevic, Z.; Hartman, M.L.; et al. Triple-Hormone-Receptor Agonist Retatrutide for Obesity—A Phase 2 Trial. N. Engl. J. Med. 2023, 389, 514–526. [Google Scholar] [CrossRef] [PubMed]
  118. Jastreboff, A.M.; Aronne, L.J.; Ahmad, N.N.; Wharton, S.; Connery, L.; Alves, B.; Kiyosue, A.; Zhang, S.; Liu, B.; Bunck, M.C.; et al. Tirzepatide Once Weekly for the Treatment of Obesity. N. Engl. J. Med. 2022, 387, 205–216. [Google Scholar] [CrossRef]
  119. Nicholls, S.J.; Bhatt, D.L.; Buse, J.B.; Prato, S.D.; Kahn, S.E.; Lincoff, A.M.; McGuire, D.K.; Nauck, M.A.; Nissen, S.E.; Sattar, N.; et al. SURPASS-CVOT investigators. Comparison of tirzepatide and dulaglutide on major adverse cardiovascular events in participants with type 2 diabetes and atherosclerotic cardiovascular disease: SURPASS-CVOT design and baseline characteristics. Am. Heart J. 2024, 267, 1–11. [Google Scholar] [CrossRef]
  120. Gouju, J.; Legeay, S. Pharmacokinetics of obese adults: Not only an increase in weight. Biomed. Pharmacother. 2023, 166, 115281. [Google Scholar] [CrossRef]
Figure 1. Pathophysiological mechanisms of obesity. MUNW: metabolic unhealthy normal weight; MHO: metabolically healthy overweight/obese; MUO: metabolically unhealthy overweight/obese; SO: sarcopenic obesity.
Figure 1. Pathophysiological mechanisms of obesity. MUNW: metabolic unhealthy normal weight; MHO: metabolically healthy overweight/obese; MUO: metabolically unhealthy overweight/obese; SO: sarcopenic obesity.
Nutrients 16 02781 g001
Figure 2. Obesity and development of cardiovascular disease RAAS; renin–angiotensin–aldosterone system.
Figure 2. Obesity and development of cardiovascular disease RAAS; renin–angiotensin–aldosterone system.
Nutrients 16 02781 g002
Figure 3. Mechanisms of action of GLP1-RA in obesity.
Figure 3. Mechanisms of action of GLP1-RA in obesity.
Nutrients 16 02781 g003
Table 1. Modern pharmacological treatment of obesity.
Table 1. Modern pharmacological treatment of obesity.
Pharmacological ClassMechanism of ActionEffects
Bupropion/naltrexoneAction on hypothalamic nucleus arcuatus and dopaminergic mesolimbic system.Body weight reduction up to 5%.
GLP1-RAStimulation of insulin secretion and glucose-lowering; slowing gastric emptying and increasing hypothalamic sense of satiety through stimulating POMC neurons.Greater reduction in body weight (+10.7%) compared to placebo. Reduction of the composite outcome of death from cardiovascular causes, nonfatal MI, or nonfatal stroke (−20%) in secondary prevention.
Tirzepatide Stimulation of first- and second-phase insulin secretion and reduction of glucagon levels, both in a glucose-dependent manner; it also induces a delay of gastric emptying, reduces fasting and postprandial glucose concentration, and decreases food intake. Sustained weight reduction in obese adults (up to 24%).
Retatrutide Novel triple agonist of the glucose-dependent insulinotropic polypeptide, glucagon-like peptide 1 and, glucagon receptors.Body weight reduction up to 17.5% at 24 weeks and to 24.2% at 48 weeks.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gallo, G.; Desideri, G.; Savoia, C. Update on Obesity and Cardiovascular Risk: From Pathophysiology to Clinical Management. Nutrients 2024, 16, 2781. https://doi.org/10.3390/nu16162781

AMA Style

Gallo G, Desideri G, Savoia C. Update on Obesity and Cardiovascular Risk: From Pathophysiology to Clinical Management. Nutrients. 2024; 16(16):2781. https://doi.org/10.3390/nu16162781

Chicago/Turabian Style

Gallo, Giovanna, Giovambattista Desideri, and Carmine Savoia. 2024. "Update on Obesity and Cardiovascular Risk: From Pathophysiology to Clinical Management" Nutrients 16, no. 16: 2781. https://doi.org/10.3390/nu16162781

APA Style

Gallo, G., Desideri, G., & Savoia, C. (2024). Update on Obesity and Cardiovascular Risk: From Pathophysiology to Clinical Management. Nutrients, 16(16), 2781. https://doi.org/10.3390/nu16162781

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop