Abstract
Insulin resistance, dyslipidemia, hypertension, and visceral adiposity are the leading causes of the growing worldwide health burden associated with metabolic syndrome, obesity, and cardiovascular diseases (CVDs). Despite the “obesity paradox,” which emphasizes the varied cardiovascular outcomes among obese people, obesity is now acknowledged as an active contributor to cardiometabolic dysfunction through endocrine, inflammatory, and metabolic pathways. Growing evidence indicates that nutrition is a key determinant of cardiometabolic risk, highlighting the need to understand diet-mediated mechanisms linking adipose tissue to cardiac function. Adipokines, including adiponectin, leptin, TNF-α, and resistin, which regulate systemic inflammation, metabolic homeostasis, and myocardial physiology, are secreted by adipose tissue, which is no longer thought of as passive energy storage. Its heterogeneous phenotypes, white, brown, and beige adipose tissue, exhibit distinct metabolic profiles that influence cardiac energetics and inflammatory status. Nutrient-driven transitions between these phenotypes further underscore the intricate interplay between diet, adipose biology, and cardiac metabolism. Central nutrient-sensing pathways, including mTOR, AMPK, SIRT1, PPAR-γ, and LKB1, integrate macronutrient and micronutrient signals to regulate adipose tissue remodeling and systemic metabolic flexibility. These pathways interact with hormonal mediators such as insulin, leptin, and adiponectin, forming a complex regulatory network that shapes the adipose-cardiac axis. This review synthesises current knowledge on how nutrient inputs modulate adipose tissue phenotypes and signaling pathways to influence cardiac function. By elucidating these mechanisms, we highlight emerging opportunities for precision nutrition and targeted therapeutics to restore metabolic balance, strengthen cardiac resilience, and reduce the burden of cardiometabolic disease.
1. Introduction
Cardiovascular diseases (CVDs), obesity, and metabolic syndrome together constitute a major worldwide health concern that dramatically raises rates of morbidity and mortality [1]. Up to 65% of patients presenting with acute coronary syndrome in some populations have metabolic syndrome, according to recent epidemiological investigations that indicate concerning prevalence rates globally [2,3]. Insulin resistance, hypertension, increased blood lipids (dyslipidemia), and abdominal obesity are the main causes of this epidemic. With distinct processes connecting signals from adipose tissue to heart dysfunction, obesity has been found to be an independent and controllable risk factor for CVD [4]. Nevertheless, the scientific community also acknowledges the so-called “obesity paradox”, unexpected findings that suggest some obese individuals maintain relatively preserved heart function and a lower mortality risk [5,6]. Despite these exceptions, dietary interventions remain pivotal in modifying cardiovascular risk, thereby underscoring the fundamental role of nutrition in both disease prevention and treatment [7,8].
The understanding of adipose-cardiac interactions has shifted from viewing adipose tissue merely as a passive energy store to recognising its active endocrine function. The discovery of adipokines such as adiponectin, tumour necrosis factor-alpha (TNF-α), leptin, and resistin marked a paradigm shift, revealing how adipose tissue dysfunction contributes to systemic inflammation and cardiac pathology [9,10]. This interdisciplinary insight transformed perspectives from organ-centric pathophysiology to integrative, system-level frameworks encompassing metabolic, inflammatory, and neurohumoral pathways that interlink adipose tissue and cardiac function [11].
Adipose tissue exhibits heterogeneous phenotypes with distinct metabolic roles [12]. White adipose tissue (WAT) predominantly displays a metabolically inflexible and pro-inflammatory profile associated with lipid storage and insulin resistance. Brown adipose tissue (BAT), conversely, is thermogenic, metabolically flexible, and exerts anti-inflammatory effects through oxidative metabolism and non-shivering thermogenesis. Beige adipose tissue exhibits an intermediate phenotype, capable of WAT-to-BAT-like transitions in response to nutrient and hormonal stimuli [13,14]. These nutrient-mediated phenotypic shifts have crucial implications for cardiac metabolism, influencing substrate availability and inflammatory milieu in the adipose-cardiac axis [15].
Adipose tissue integrates nutrient signals through master regulators such as mechanistic target of rapamycin (mTOR), AMP-activated protein kinase (AMPK), sirtuin-1 (SIRT1), peroxisome proliferator-activated receptor gamma (PPAR-γ), and liver kinase B1 (LKB1) [12,16]. Macronutrients differentially modulate these pathways—glucose primarily activates mTOR, amino acids engage mTOR and related sensors, and lipids influence PPAR-γ activity [17]. Micronutrients serve as essential cofactors for energy-metabolism enzymes, further refining the cellular response to nutrient availability. This signaling network interfaces with endocrine hormones, including insulin, leptin, and adiponectin, orchestrating metabolic flexibility and systemic energy balance [18,19].
While recent reviews have examined individual aspects of adipose-cardiac communication, metabolic flexibility, or dietary effects on cardiovascular health, an integrative framework connecting these elements remains lacking. Current literature often addresses macronutrient and micronutrient effects in isolation, without linking dietary inputs to the full spectrum of adipose phenotypes and their cardiac consequences. Furthermore, emerging mediators such as adipose-derived extracellular vesicles and gut microbiota metabolites are rarely discussed alongside classical adipokine pathways. This review addresses these gaps by synthesizing nutrient-sensing mechanisms, adipose tissue remodeling, and inter-organ communication within a unified framework that bridges molecular insights with clinical biomarkers and therapeutic targets.
Understanding the adipose-cardiac axis is vital for precision medicine approaches targeting cardiometabolic diseases. Current knowledge gaps persist regarding how different nutrients and dietary patterns remodel this axis at the molecular and systemic levels. This review aims to elucidate these processes and their implications for developing targeted nutritional and pharmacological interventions to restore metabolic balance, enhance cardiac resilience, and reduce disease burden.
2. Molecular Mechanisms of Nutrient Sensing and Signaling in Adipose Tissue and Heart
The molecular mechanisms that govern nutrient sensing and signaling across adipose tissue and the heart form an integrated regulatory network essential for maintaining metabolic homeostasis [20]. Adipose tissue acts as both a nutrient reservoir and an endocrine organ, translating fluctuations in energy availability into cellular and systemic responses. These processes are shaped by the anatomical distribution of adipose depots, subcutaneous, visceral, perivascular, and epicardial, which differ in metabolic activity, vascularization, innervation, and cellular composition [21]. Within each depot, adipocytes interact with stromal vascular cells and immune populations to coordinate nutrient uptake, lipid storage, and endocrine output. Through the synthesis and secretion of adipokines, adipose tissue communicates nutrient status to peripheral organs, including the heart, thereby influencing cardiac substrate selection, oxidative capacity, and inflammatory tone [22,23].
Among the adipokines involved in nutrient-dependent communication, adiponectin represents a key metabolic checkpoint [24]. Its circulating levels are determined by the molecular structure encoded by the ADIPOQ gene and by nutritional conditions that modulate its transcription and multimerization [25]. Adiponectin signals primarily through AdipoR1, AdipoR2, and T-cadherin, activating downstream pathways that enhance cellular energy efficiency. Central to its action is robust AMPK stimulation, which, in turn, activates PGC-1α, promoting mitochondrial biogenesis and improved oxidative metabolism. In cardiomyocytes, adiponectin facilitates glucose handling by enhancing GLUT4 translocation to the cell surface, while simultaneously increasing fatty acid oxidation capacity [26,27]. Beyond metabolic effects, adiponectin generates strong anti-inflammatory and antioxidative responses by suppressing NF-κB signaling and enhancing mitochondrial reactive oxygen species scavenging. Dietary composition directly influences adiponectin secretion, with unsaturated fats, fiber-rich diets, and caloric moderation promoting higher adiponectin levels and greater receptor responsiveness [28,29].
In contrast, nutrient excess and adipose tissue dysfunction lead to increased production of pro-inflammatory adipokines such as resistin, TNF-α, and IL-6 [30,31]. These cytokines exert both paracrine effects within adipose depots and systemic actions that reach the heart. By activating TLR4 and the NF-κB pathway in cardiac tissue, they promote oxidative stress, impair myocardial insulin signaling, and drive pathological remodeling, including hypertrophy and fibrosis [32,33]. In parallel, endothelial dysfunction arises from inflammatory signaling and elevated circulating lipids. High glucose load, saturated fatty acids, and nutrient oversupply are potent drivers of these pro-inflammatory secretory programs, linking dietary patterns to cardiometabolic dysfunction [34,35].
Leptin also plays a critical role in nutrient-derived signaling between adipose tissue and the heart [36,37]. Cardiomyocytes express functional leptin receptors through which leptin modulates substrate metabolism, fatty acid utilization, and myocardial contractility [38]. Under physiological conditions, leptin helps maintain metabolic balance; however, chronic energy surplus leads to leptin resistance, blunting its regulatory effects. As a result, cardiac tissue becomes exposed to persistent nutrient overload and inadequate metabolic adaptation, which contributes to lipotoxicity, impaired contractile function, and progression toward cardiometabolic disease [38,39,40].
Metabolic byproducts originating from adipose tissue provide an additional layer of nutrient-dependent communication [41,42]. Free fatty acids (FFAs) function both as essential nutrient signals and as lipotoxins when present in excess, influencing cardiac energy metabolism and mitochondrial integrity [43]. Glycerol, released during lipolysis, serves as a substrate for hepatic gluconeogenesis and indirectly affects cardiac substrate supply [44]. Ketone bodies, particularly β-hydroxybutyrate and acetoacetate, produced during enhanced lipolytic flux, are readily utilized by the heart as efficient oxidative fuels [45,46]. Moreover, adipose tissue releases extracellular vesicles enriched in microRNAs, such as miR-223 and miR-122, which modulate cardiac gene expression, metabolic pathways, and inflammatory signaling [47].
The heart integrates these diverse adipose-derived nutrient cues through a network of receptors and transcriptional regulators [16,41]. PPAR-α acts as the central transcriptional controller of fatty acid oxidation genes, adjusting myocardial substrate use in response to lipid availability [48]. PPAR-γ contributes to metabolic and anti-inflammatory regulation, whereas AMPK serves as the primary cellular energy sensor, responding to shifts in ATP demand and substrate supply. Sirtuins (SIRT1, SIRT3, SIRT7) link nutrient state to gene expression via NAD+-dependent deacetylation reactions, influencing mitochondrial function, redox status, and cardiac stress responses. AdipoR1 and AdipoR2, as key receptors for adiponectin, represent major integration points for nutrient-sensitive endocrine signaling, coordinating AMPK and PPAR pathways to optimize cardiac metabolic adaptation [49,50].
Collectively, these adipokine pathways do not operate in isolation but converge on shared intracellular nodes, particularly AMPK, mTOR, and NF-κB, where the balance between cardioprotective and cardiotoxic signals determines disease trajectory [51,52]. This mechanistic interdependence implies that targeting a single pathway may yield limited benefit if upstream adipose dysfunction persists. Multi-targeted strategies, including lifestyle modifications that simultaneously reduce pro-inflammatory adipokine secretion, enhance adiponectin output, and restore leptin sensitivity, are therefore more likely to achieve durable cardioprotection. These pathway interactions also inform biomarker development; composite panels reflecting both protective (adiponectin) and harmful (resistin, TNF-α, ceramides) signals may improve cardiovascular risk prediction beyond single markers [53].
Through these interconnected pathways, the heart “interprets” adipose-derived signals and adjusts its metabolic priorities. When nutrient sensing across tissues is balanced, this axis supports metabolic flexibility, efficient substrate use, and protection against oxidative and inflammatory stress. However, chronic nutrient excess, depot-specific adipose dysfunction, and endocrine imbalance disrupt this communication network, accelerating the transition from adaptive metabolic remodeling to cardiometabolic disease [12,51].
3. Dietary Patterns and Their Impact on Adipose Tissue and Cardiac Function
3.1. Mediterranean Diet (MedDiet)
The Mediterranean diet is characterized by a nutrient profile rich in monounsaturated fatty acids from olive oil, high-quality complex carbohydrates from whole grains, moderate protein intake with an emphasis on fish over red meat, abundant polyphenols from fruits and vegetables, and moderate wine consumption. These components synergistically modulate adipose tissue biology and cardiometabolic function [54,55,56].
Within adipose tissue, polyphenol-mediated PPAR-γ activation enhances insulin sensitivity and supports the maintenance of a metabolically healthy adipocyte phenotype. MedDiet adherence promotes the preservation and recruitment of brown adipose tissue (BAT) characteristics while reducing white adipose tissue (WAT) inflammation by inhibiting NF-κB [57,58]. A consistent finding across mechanistic and clinical studies is the upregulation of adiponectin, accompanied by the suppression of pro-inflammatory adipokines. These processes converge on increased mitochondrial biogenesis, primarily driven by PGC-1α activation, thereby improving oxidative capacity in adipocytes [59,60].
Cardiac tissue benefits from these systemic improvements through greater metabolic flexibility, enhanced endothelial function, and modulation of vascular tone. MedDiet interventions have been associated with reduced atrial fibrillation burden, improvements in diastolic function, particularly in HFpEF populations, and attenuation of prothrombotic pathways. The diet’s influence on cardiac autonomic regulation, reflected by increased parasympathetic and reduced sympathetic activity, further contributes to cardioprotection [61].
Polyphenols serve as central mediators of these effects. Resveratrol activates SIRT1, improving mitochondrial resilience to oxidative stress; catechins exhibit potent antioxidant and anti-inflammatory actions; and anthocyanins enhance endothelial nitric oxide production and vascular reactivity. These molecular actions correspond with observed reductions in LDL oxidation, improved SCORE2-based cardiovascular risk estimates, and enhanced endothelial-dependent vasodilation. Landmark trials such as PREDIMED and multiple meta-analyses consistently validate the MedDiet as one of the most effective dietary patterns for reducing cardiovascular morbidity and mortality [62,63,64,65].
3.2. Ketogenic Diet
The ketogenic diet imposes a profound metabolic shift characterized by carbohydrate restriction and reliance on ketone bodies derived from fatty acids. Increased adipose lipolysis produces elevated acetyl-CoA, which drives hepatic ketogenesis [66,67]. The heart efficiently oxidizes β-hydroxybutyrate, which can serve as a preferred oxidative substrate under ketogenic conditions. Alterations in the NADH/NAD+ ratio and increased reliance on fatty acid oxidation (FAO) constitute key metabolic adaptations [68,69].
In adipose tissue, ketogenic feeding enhances lipolytic capacity, stimulates BAT activation and beige adipocyte differentiation through β-3 adrenergic signaling, and reduces WAT mass alongside infiltration of pro-inflammatory immune cells. These changes translate into a more favorable adipokine profile, notably with increased adiponectin secretion [70,71]. Cardiac adaptations include enhanced mitochondrial efficiency, improved substrate switching, and reduced susceptibility to delayed afterdepolarizations (DADs) associated with calcium overload, thereby potentially lowering arrhythmogenic risk [72]. However, ketogenic diets may cause a rise in LDL-cholesterol, driven by increased lipoprotein turnover, reduced LDL receptor activity, and enhanced hepatic VLDL secretion. The clinical significance of this elevation varies between individuals and remains an area of ongoing investigation [73]. Despite short-term benefits, particularly weight loss and improved glycemic control, long-term adherence is challenging due to dietary monotony, nutrient deficiencies, increased risk of kidney stones, and socioeconomic constraints. These limitations underscore the importance of tailored patient selection and careful monitoring in clinical applications [74,75].
3.3. Intermittent Fasting (IF) and Time-Restricted Eating (TRE)
Intermittent fasting and time-restricted eating induce characteristic metabolic cascades centered on energy sensing. Fasting intervals activate AMPK and inhibit mTORC1, promoting autophagy, mitophagy, and enhanced lipid oxidation. These processes elevate circulating ketone bodies and improve mitochondrial quality across metabolic tissues [76,77]. In adipose tissue, IF fosters a shift toward a metabolically healthier phenotype, with increased BAT activation via sympathetic nervous system engagement and β-3 adrenergic signaling. Adipokine secretion profiles improve, and feeding-fasting cycles reinforce circadian rhythm alignment, which is essential for optimal adipose and systemic metabolic function [78,79]. Cardiac tissue similarly benefits from increased autophagy and improved mitochondrial turnover, along with reduced oxidative stress and systemic inflammation [80]. However, specific fasting protocols may predispose susceptible individuals to arrhythmias, potentially via autonomic shifts, electrolyte fluctuations, or ketone-mediated electrophysiological changes. Protocols such as 16:8, 5:2, and alternate-day fasting differ in metabolic impact and adherence profiles. Early time-restricted eating (TRE) protocols show superior effects on reducing visceral adipose tissue compared to late TRE, which is particularly important for cardiometabolic protection. However, the optimal fasting window varies by genotype and chronotype, highlighting the need for personalized fasting schedules [81,82]. Population-specific responses are increasingly recognized: women, older adults, and individuals with advanced metabolic impairment may require modified fasting windows to optimize safety and efficacy [83,84].
Chrononutrition, distinct from time-restricted eating, emphasizes the importance of synchronizing food intake with endogenous circadian rhythms, which are governed by peripheral clocks in adipose tissue, the heart, the liver, and other organs. Each individual has a unique optimal timing for food consumption aligned with these rhythms. Late meals, a common concern, can directly disrupt gene expression in adipose tissue and cardiac muscle, adversely affecting metabolic regulation and circadian alignment. Therefore, timing meals to align with one’s circadian biology is critical for maintaining metabolic homeostasis and preventing dysfunction.
3.4. Caloric Restriction (CR)
Caloric restriction (CR) induces a coordinated systemic metabolic response [85]. Reduced caloric intake lowers hepatic glucose production, enhances whole-body insulin sensitivity, and decreases circulating FFAs by modulating adipose lipolysis. In adipose tissue, CR leads to a reduction in WAT mass, improved insulin signaling, increased adiponectin levels, and decreased macrophage infiltration, all characteristics of a more insulin-sensitive and anti-inflammatory adipose phenotype [86,87,88]. Cardiac-specific benefits arise through activation of the SIRT1–PGC-1α axis, culminating in enhanced mitochondrial biogenesis and improved myocardial energetic capacity [89,90]. CR also augments endothelial function and nitric oxide availability, improves coronary flow reserve, and supports diastolic performance [91,92]. Redox homeostasis is a central feature of CR. Reduced caloric intake lowers electron flux through the mitochondrial electron transport chain, decreasing ROS generation. Concurrently, antioxidant enzyme expression increases, NAD+ availability rises, and SIRT1 activity is enhanced, jointly contributing to improved mitochondrial efficiency and stress resistance [93,94]. While CR reliably induces weight loss, its long-term feasibility is limited, and excessive restriction may jeopardize lean mass. Strategies incorporating resistance training, periodic refeeding, and protein adequacy can help preserve skeletal muscle while maintaining metabolic benefits [95].
3.5. DASH Diet (Dietary Approaches to Stop Hypertension)
The DASH diet emphasizes fruits, vegetables, whole grains, lean proteins, and low-fat dairy while limiting sodium, saturated fat, and added sugars. Originally developed for hypertension management, DASH has demonstrated broad cardiometabolic benefits extending to adipose tissue function and cardiac health [96,97]. Within adipose tissue, DASH reduces visceral adiposity and attenuates chronic low-grade inflammation by decreasing pro-inflammatory immune cell infiltration and suppressing NF-κB-mediated cytokine production. Clinical trials report significant reductions in circulating C-reactive protein, LDL-cholesterol, and markers of oxidative stress, accompanied by improvements in insulin sensitivity and adipokine profiles [98,99]. The diet’s high potassium, magnesium, and fiber content promotes vasodilation, enhances endothelial function, and favorably modulates the renin–angiotensin–aldosterone system. Cardiac benefits include cumulative reductions in subclinical myocardial injury (hs-cTnI) and systemic inflammation over sustained adherence periods, with meta-analyses demonstrating 6–11 mmHg reductions in systolic blood pressure independent of weight loss [100]. DASH shares anti-inflammatory characteristics with the Mediterranean diet but places greater emphasis on sodium restriction and dairy inclusion, making it particularly suitable for hypertensive populations at elevated cardiovascular risk.
3.6. Plant-Based Diets
Plant-based diets, encompassing vegetarian, vegan, and whole-food plant-based patterns, exert favorable effects on adipose tissue biology and cardiac function through multiple mechanisms [101,102]. High intake of dietary fiber, polyphenols, and antioxidants reduces oxidative stress and reactive oxygen species (ROS) generation in adipose tissue, thereby attenuating adipocyte hypertrophy and inflammatory adipokine secretion. Plant-based patterns consistently lower circulating leptin while increasing adiponectin, reflecting improved adipose tissue insulin sensitivity and reduced visceral fat accumulation [103]. The absence of dietary cholesterol and reduced saturated fat intake decrease hepatic VLDL secretion and improve lipid profiles, with vegan diets showing the most pronounced reductions in total cholesterol, LDL-cholesterol, and apolipoprotein B [104]. Cardiac benefits extend beyond risk factor modification: interventional studies in heart failure patients report improvements in ejection fraction, stroke volume, and cardiac remodeling following adoption of plant-based diets [105,106]. Mechanistically, reduced TMAO production (due to altered gut microbiota composition lacking TMA-producing bacteria), lower advanced glycation end-products (AGEs), and enhanced nitric oxide bioavailability contribute to improved endothelial function and reduced myocardial fibrosis. Plant-based diets also lower blood pressure through mechanisms similar to DASH, including increased potassium intake and favorable effects on vascular tone [107]. Potential limitations include the need for careful planning to ensure adequate intake of vitamin B12, iron, zinc, and omega-3 fatty acids, particularly in strictly vegan patterns.
3.7. Direct Inter-Diet Comparisons and Personalization Strategies
Direct comparisons of dietary patterns reveal distinct macronutrient compositions, caloric densities, adipose tissue effects, and cardiac outcomes. The MedDiet offers broad cardiometabolic benefits with high sustainability; ketogenic diets provide potent short-term metabolic switching at the cost of long-term adherence and variable lipid responses; IF/TRE protocols optimize circadian alignment and cellular repair, and CR delivers robust improvements in insulin sensitivity and mitochondrial function but is challenging to maintain chronically [76,85,108]. Personalization strategies should account for adipose phenotype (visceral vs. subcutaneous predominance, inflammatory state, BAT activity), metabolic status, comorbidities, and individual lifestyle factors. Nutrigenomics further refines these approaches by identifying genetic polymorphisms that modulate responses to dietary interventions like TRE or CR. For example, APOE variants influence lipid responses to Mediterranean diet patterns, with ε4 carriers showing greater cholesterol reductions from monounsaturated fat emphasis, while FTO rs9939609 polymorphism affects satiety signaling and body weight loss efficacy during intermittent fasting, particularly in obesity-prone individuals. MTHFR polymorphisms also impact metabolic responses to nutrient intake, underscoring genotype-specific diet optimization. Beyond genetic determinants, substantial inter-individual heterogeneity in dietary response arises from age, biological sex, baseline metabolic phenotype, gut microbiome composition, and physical activity level [109,110]. Older adults exhibit reduced metabolic flexibility and anabolic resistance that may blunt intervention efficacy; women and men differ in fat distribution and substrate oxidation, leading to sexually dimorphic responses; and individuals with greater baseline insulin resistance or adipose inflammation often show larger absolute improvements but may require more intensive protocols [111]. These multidimensional sources of variability underscore that optimal dietary prescription requires integration of genetic, phenotypic, demographic, and lifestyle factors toward truly personalized nutrition approaches. Synergistic approaches, such as combining MedDiet with TRE or cycling mild CR periods, may maximize metabolic flexibility and cardioprotection while maintaining adherence.
In summary, dietary patterns modulate the adipose-cardiac axis through distinct but overlapping mechanisms: the Mediterranean diet enhances adiponectin and PPAR-γ signaling; ketogenic diets shift fuel utilization toward ketone bodies and activate BAT; intermittent fasting promotes AMPK-driven autophagy and circadian alignment; and caloric restriction activates the SIRT1–PGC-1α axis. Selecting among these approaches should be guided by individual metabolic phenotype, comorbidities, and long-term adherence potential.
4. Role of Micronutrients in Adipose and Cardiac Metabolism
While the preceding sections have addressed macronutrients and dietary patterns, micronutrients serve as essential cofactors and regulators that intersect with the same molecular nodes—AMPK, SIRT1, NF-κB, and mitochondrial signaling pathways—that govern adipose-cardiac crosstalk [112,113]. Rather than functioning as isolated factors, micronutrients modulate the activity and expression of nutrient-sensing cascades discussed earlier, thereby amplifying or mitigating macronutrient-driven metabolic dysfunction. For example, vitamin D insufficiency compromises AMPK/SIRT1 activation in adipose tissue, whereas magnesium deficiency impairs mitochondrial ATP synthesis and promotes oxidative stress through shared pathways affected by caloric excess [114,115]. This mechanistic overlap underscores the importance of adequate micronutrient status in preserving the metabolic flexibility and anti-inflammatory balance that protect cardiac function.
Micronutrient status profoundly influences metabolic and inflammatory pathways in both adipose tissue and the heart. Deficiencies or excesses of certain vitamins and minerals can perturb adipokine secretion, insulin sensitivity, and mitochondrial function, thereby contributing to cardiometabolic diseases such as obesity, type 2 diabetes, and heart failure [116,117].
Vitamin D insufficiency is common in obesity and metabolic syndrome, and restoring vitamin D may ameliorate adipose dysfunction. Mechanistically, the active form (1,25-dihydroxyvitamin D3) acts via the vitamin D receptor (VDR) in adipocytes to reduce inflammation and modulate adipokine release [118,119]. Vitamin D suppresses pro-inflammatory cytokines (e.g., IL-6, IL-1β, IL-8, MCP-1) and even leptin in human adipocytes, while potentially boosting adiponectin, an insulin-sensitizing adipokine [118,120,121]. These anti-inflammatory and pro-metabolic effects can improve insulin sensitivity in adipose tissue. In the heart, vitamin D signaling is emerging as an important regulator of cardiac metabolism and remodeling. Cardiomyocytes and other cardiac cells express VDR, and vitamin D exerts cardioprotective effects: it dampens inflammatory cell infiltration in myocardium, reduces cardiomyocyte apoptosis, and modulates autophagy [122,123]. Vitamin D also helps preserve mitochondrial function in the heart; it promotes mitochondrial homeostasis and limits oxidative damage to proteins and lipids [124]. Consistently, vitamin D deficiency has been linked to myocardial dysfunction, whereas sufficient vitamin D may support normal contractility and prevent pathological hypertrophy [125].
Magnesium is a cofactor in hundreds of enzymes crucial for energy metabolism, including those governing glucose uptake and ATP synthesis [126]. In adipose tissue, magnesium deficiency triggers a pro-inflammatory, insulin-resistant state. Hypomagnesemia in obesity is associated with excessive reactive oxygen species (ROS) generation, mitochondrial dysfunction, and reduced ATP production in adipocytes [127]. Clinically, low magnesium correlates with greater insulin resistance, whereas higher magnesium intake is strongly associated with improved insulin sensitivity, especially in overweight individuals [128]. Magnesium repletion can enhance adipocyte glucose uptake and adipokine profiles, thereby improving metabolic homeostasis. In the cardiovascular system, magnesium plays a similarly vital role. Magnesium deficiency is known to suppress cardiac mitochondrial function and increase oxidative stress, which can impair myocardial energetics [129]. Indeed, in heart failure models, magnesium restoration improved mitochondrial ATP production and reduced oxidative damage, translating into better diastolic function [130]. Adequate magnesium thus helps maintain normal cardiac bioenergetics and electrical stability, whereas deficiency has been linked to arrhythmias, hypertrophy, and worsened outcomes in heart failure [129].
Zinc is an essential trace element that modulates antioxidant defenses and metabolic signaling. In adipose tissue, low zinc status exacerbates metabolic inflammation. Obese individuals often show suboptimal zinc levels, which are associated with aggravated insulin resistance, chronic inflammation, and heightened oxidative stress in adipose depots [131,132]. Mechanistically, zinc is required to activate antioxidant enzymes (such as Cu/Zn-superoxide dismutase) that scavenge ROS and to regulate cytokine expression [133]. Zinc sufficiency restrains adipose tissue inflammation and helps maintain healthy adipokine secretion; for example, zinc deficiency can directly contribute to an inflammatory adipose phenotype and alter leptin production in obesity [134,135]. Conversely, zinc supplementation in at-risk populations has been shown to reduce systemic inflammatory markers and improve metabolic control (e.g., glucose and lipid levels) [136,137]. In the heart, zinc is crucial for redox homeostasis and cellular survival. Zinc deficiency in cardiac tissue leads to oxidative stress, inflammation, and cell death, factors that promote cardiomyopathy and heart failure [138]. A lack of zinc can impair cardiomyocyte function by disrupting calcium handling and increasing apoptosis, whereas optimal zinc levels protect the myocardium [139]. Indeed, zinc supplementation has demonstrated protective effects against myocardial ischemia–reperfusion injury and oxidative damage [140]. Thus, maintaining zinc homeostasis is important for preventing cardiac oxidative injury and unfavorable remodeling.
Iron has a dual and context-dependent role in cardiometabolic health, as both iron overload and iron deficiency can be detrimental. In adipose tissue, excess iron accumulation is pathogenic [141]. Obesity is often characterized by iron sequestration in adipose, particularly within macrophages, which creates a pro-inflammatory environment [142]. Iron overload in adipocytes and adipose macrophages promotes local oxidative stress and triggers the release of inflammatory cytokines, contributing to insulin resistance. High iron levels in fat are also linked to adipokine dysregulation—notably, adiponectin expression is suppressed when adipose iron content is elevated [143]. This combination of elevated inflammation and lower insulin-sensitizing adipokines can exacerbate metabolic syndrome. On the other hand, in the heart, iron deficiency is a prevalent problem that impairs cardiac energy metabolism [144]. Iron is required for oxygen transport (hemoglobin/myoglobin) and for mitochondrial enzymes of oxidative phosphorylation. When iron is lacking, cardiomyocytes exhibit reduced mitochondrial respiration and ATP production, leading to diminished contractile performance [145]. Approximately 50% of patients with chronic heart failure have iron deficiency, which is associated with worse exercise capacity and outcomes [146]. Iron deficiency in the failing heart correlates with mitochondrial dysfunction, greater oxidative stress, and adverse remodeling [147]. Encouragingly, treating iron deficiency (e.g., with intravenous iron) in heart failure improves symptoms and functional status, underlining the importance of iron for cardiac metabolism [148]. Yet iron excess is equally harmful in the heart: in iron-overload conditions (such as hemochromatosis), surplus iron deposits in cardiac myocytes and mitochondria, catalyzing ROS generation and lipid peroxidation that lead to fibrosis and cardiomyopathy [149]. Importantly, micronutrient deficiencies frequently coexist in patients with metabolic syndrome and obesity, creating a state of “high-calorie malnutrition” in which energy excess paradoxically accompanies inadequate vitamin and mineral status [150,151]. Deficiencies in vitamin D, magnesium, zinc, and B-vitamins cluster together and may exert synergistic deleterious effects: concurrent vitamin D insufficiency and magnesium depletion amplify NF-κB-driven adipose inflammation, while combined zinc and selenium deficits compromise antioxidant defenses and impair mitochondrial function in both adipocytes and cardiomyocytes [152,153]. This clustering suggests that single-nutrient interventions may yield limited benefit, whereas comprehensive micronutrient repletion strategies targeting multiple deficiencies simultaneously could more effectively restore adipose-cardiac axis homeostasis and interrupt the inflammatory-metabolic feed-forward loops characteristic of cardiometabolic disease [154]. A detailed summary of the key micronutrients and their roles in regulating adipose inflammation, adipokine secretion, insulin sensitivity, and cardiac mitochondrial function is presented in Table 1.
Table 1.
Overview of Key Micronutrients (Vitamin D, Magnesium, Zinc, and Iron) and Their Roles in Regulating Adipose Inflammation, Adipokine Secretion, Insulin Sensitivity, and Cardiac Mitochondrial Function.
In summary, micronutrients act as essential cofactors and modulators of adipose-cardiac communication. Vitamin D and magnesium support mitochondrial function and insulin sensitivity, while zinc maintains redox balance and adipokine homeostasis. Iron requires careful balance, as both deficiency and excess impair cardiac energetics. Addressing micronutrient status represents an accessible and often overlooked strategy for optimizing cardiometabolic health.
5. Macronutrients Effects on Adipose-Cardiac Metabolic Flexibility
Metabolic flexibility is the capacity to rapidly switch between carbohydrate and fat oxidation as feeding or fasting states change [155]. A healthy adipose-cardiac axis buffers nutrient flux, storing fat after meals and releasing fatty acids during fasting, while the heart adjusts its fuel use accordingly. In obesity and diabetes, this coordination breaks down: insulin-resistant adipose tissue releases fatty acids even when fed, and the heart becomes fat-reliant, impairing metabolic flexibility [156,157]. Eventually, adipose storage capacity is overwhelmed, and surplus lipids accumulate in non-adipose sites like myocardium, promoting lipotoxicity and cardiac dysfunction [158,159].
Beyond macronutrient quantity, food source quality critically influences the adipose-cardiac axis through ultra-processed foods (UPFs). Even balanced macronutrient profiles from UPF impair metabolic flexibility, as emulsifiers and additives disrupt gut microbiota composition and function. These perturbations elevate microbial production of trimethylamine N-oxide (TMAO), a pro-atherogenic metabolite linked to endothelial dysfunction, adipose inflammation, and cardiac lipotoxicity independent of caloric intake. Prioritizing whole-food sources thus preserves microbiota integrity and optimal nutrient signaling to adipose and cardiac tissues, enhancing overall metabolic resilience [160,161].
Carbohydrate intake acutely increases insulin, which suppresses adipose lipolysis and promotes cardiac glucose uptake, a hallmark of metabolic flexibility. In healthy individuals, a high-carbohydrate meal shifts the heart toward glucose utilization as intended [162]. However, chronic excess of refined sugars can drive insulin resistance and ectopic fat deposition that impair this flexibility. In a 24-week rat study, a fructose-enriched diet (60% of calories) induced prediabetes with early diastolic dysfunction and significant molecular remodeling of the heart [163]. Despite only mild hyperglycemia, fructose-fed rats showed mitochondrial lipid perturbations, including a wholesale rearrangement of the cardiac lipidome, defective cardiolipin remodeling, and changes in 75 cardiac proteins (many related to mitochondrial function, oxidative stress, and apoptosis) [163]. Notably, these profound changes occurred before overt oxidative damage, indicating that high sugar intake can remodel cardiac metabolism and impair function at early stages of metabolic disease [163,164]. Chronic high-carbohydrate diets, especially those rich in fructose, can overload adipose stores and spill excess fuel to the heart, impairing mitochondrial function and substrate flexibility. In contrast, reducing carbohydrate intake lowers insulin, enhances adipose lipolysis, and promotes fat oxidation. Clinical trials show that replacing carbs with protein improves insulin sensitivity and glycemic control in obese, insulin-resistant individuals [165]. Together, these findings highlight that quality and quantity of carbohydrates modulate the adipose-cardiac axis: chronic sugar excess inflicts inflexibility via adipose overload and mitochondrial tweaks in the heart, whereas controlled or lower-glycemic carb intake helps maintain insulin-sensitive fuel switching [166,167].
The heart relies heavily on fatty acids for ATP, but it remains metabolically flexible—increasing glucose oxidation when glucose is abundant. High-fat diets can undermine this flexibility by chronically elevating fatty acid supply. In insulin-sensitive individuals, a short-term isocaloric high-fat feeding can paradoxically improve insulin responsiveness (likely by lowering postprandial glycemia/insulinemia) [168]. Over the long term, however, excess dietary fat often leads to adipose expansion and systemic insulin resistance, tilting the heart into a fat-dependent, glucose-underutilizing state [157]. In diabetes, elevated circulating free fatty acids drive myocardial uptake beyond energetic needs, so the heart oxidizes more fat and less glucose, with excess fuel shunted into storage as triglycerides and harmful intermediates (e.g., diacylglycerols and ceramides) [169]. Lipid accumulation drives oxidative stress, apoptosis, and contractile dysfunction, hallmarks of lipotoxic cardiomyopathy in metabolic disease. Encouragingly, enhancing adipose fat handling can protect the heart; in an HFpEF mouse model, adipose-specific MitoNEET overexpression reduced cardiac damage despite high-fat feeding [170]. These mice, which gained weight without insulin resistance, had less cardiac fibrosis and higher myocardial SIRT3 and mitochondrial dynamism proteins compared to equally obese insulin-resistant mice [170]. Efficient adipose lipid storage prevents ectopic overflow and supports adaptive cardiac mitochondrial remodeling. Early in high-fat feeding, the heart may compensate with increased glucose oxidation, but prolonged excess leads to inflexibility and energy deficits [171]. Targeting adipose-cardiac lipid handling shows promise; in diabetic mice, the adiponectin agonist AdipoRon reduced myocardial lipid accumulation and improved insulin sensitivity and cardiac function [14]. In diabetic mice, the adiponectin receptor agonist AdipoRon reduced myocardial fatty acid, triglyceride, and ceramide accumulation while improving insulin sensitivity, inflammation, and cardiac function [169]. Mechanistically, AdipoRon stimulated AMP-activated protein kinase (AMPK) and PPARα/PGC-1α pathways in the heart, enhancing mitochondrial fatty acid oxidation and clearance of toxic lipids [169,172].
Dietary protein can indirectly modulate the adipose-heart metabolic axis through its effects on body composition, satiety, and insulin dynamics [173]. Higher-protein diets often facilitate fat loss and lean mass retention, which improves whole-body insulin sensitivity and eases the burden on both adipose tissue and the heart [174]. In clinical studies, isocaloric high-protein diets have outperformed higher-carbohydrate diets in reducing insulin resistance and glycemic variability in at-risk populations [174]. By alleviating hyperinsulinemia and aiding weight reduction, increased protein intake allows adipose tissue to shrink and become more insulin-responsive, thus lowering circulating fatty acids and toxic lipid byproducts that would otherwise strain the myocardium [175]. However, not all aspects of protein are beneficial: certain amino acids, notably branched-chain amino acids (BCAAs), are elevated in obesity and have been linked to insulin resistance when chronically high. In insulin-resistant states, BCAA breakdown in adipose tissue and liver is blunted, potentially shifting metabolic stress to the heart and muscles [176]. Overall, dietary protein supports metabolic flexibility, especially when replacing refined carbs or excess fat [177]. Protein-induced weight loss lowers leptin and often raises adiponectin, a favorable shift that enhances cardiac fuel flexibility via AMPK activation, boosting glucose use and fat oxidation while reducing apoptosis [169]. Chronic protein deficiency or muscle loss can impair metabolic health, even with controlled fat intake, highlighting the need for balanced macronutrients. Protein primarily supports the adipose-cardiac axis by enhancing insulin sensitivity and adipokine signalling, promoting efficient cardiac fuel-switching [172]. Synthesizing these macronutrient-specific effects reveals a unifying principle: the adipose-cardiac axis functions optimally when nutrient supply matches metabolic demand, and dysfunction arises when this balance is chronically disrupted. Carbohydrate excess promotes hyperinsulinemia and adipose lipogenesis; fat overconsumption saturates adipose buffering, leading to ectopic lipid deposition; and protein, while generally supportive, can contribute to dysfunction when BCAAs accumulate in insulin-resistant states. Clinically, these insights support individualized dietary approaches based on metabolic phenotype rather than rigid macronutrient prescriptions. The convergence of all three macronutrients on shared endpoints, including AMPK activity, mitochondrial function, and adipokine balance, highlights the potential for synergistic dietary interventions [155,169]. An overview of how carbohydrates, fats, and proteins differentially modulate adipose tissue function, cardiac fuel selection, and overall metabolic flexibility within the adipose–cardiac axis is provided in Table 2.
Table 2.
Summary of the Distinct Metabolic and Functional Effects of Carbohydrates, Fats, and Proteins on Adipose Tissue Function, Cardiac Fuel Selection, and Overall Adipose–Cardiac Metabolic Flexibility.
In summary, all three macronutrients influence the adipose-cardiac axis through effects on insulin signaling, lipid handling, and mitochondrial function. Carbohydrate and fat excess overwhelm adipose storage capacity, promoting ectopic lipid deposition and cardiac lipotoxicity, whereas adequate protein intake supports metabolic flexibility. Optimal macronutrient balance should be tailored to individual metabolic status to preserve adipose-cardiac coordination.
6. Inter-Organ Communication Beyond Adipokines: Exosomes, miRNAs, and Gut Microbiota
Inter-organ signalling from adipose tissue to the heart extends well beyond classical adipokines. Small extracellular vesicles (sEVs, often referred to as exosomes) and their non-coding RNA cargo, together with gut-microbiota-derived metabolites, form a highly dynamic communication network that transduces nutrient and inflammatory states in adipose depots into cardiac metabolic and structural responses [47,178]. Exosomes are 40–150 nm vesicles formed within multivesicular bodies and secreted by most cell types, including mature adipocytes, stromal vascular cells, and adipose-derived stem cells (ASCs). Adipose-derived sEVs carry lipids, proteins, metabolites, and a characteristic set of non-coding RNAs (miRNAs, circRNAs), whose composition changes with depot (visceral vs. subcutaneous), nutrient state (fed vs. fasted), and adipose inflammation.
Several recent studies show that visceral adipose tissue releases sEVs enriched in miRNAs that impair insulin signalling in peripheral organs and modulate inflammation; conversely, sEVs from healthy or “browned” adipose are enriched in miRNAs and proteins that promote mitochondrial biogenesis and anti-inflammatory phenotypes [47]. Adipose sEVs are taken up by cardiomyocytes, cardiac fibroblasts, and endothelial cells via endocytosis or receptor-mediated uptake; once internalized, vesicular miRNAs regulate gene networks controlling fatty acid oxidation, mitochondrial quality control, autophagy, and extracellular matrix remodelling [178].
Preclinical models implicate specific miRNAs, for example, adipose-derived miR-132/212 and miR-223, in promoting endothelial dysfunction, cardiomyocyte apoptosis, or macrophage polarization within the heart, thereby linking adipose inflammation to atherosclerosis progression and myocardial remodelling. Conversely, ASC-derived exosomes carrying cardioprotective miRNAs (e.g., miR-93, miR-22 in some models) reduce apoptosis and stimulate angiogenesis after ischemic injury. These data position sEVs as both mediators and potential therapeutic vectors [179,180].
Nutritional states reshape exosomal output: high-fat diets and obesity upregulate pro-inflammatory miRNA signatures in adipose sEVs (promoters of NF-κB and TLR4 pathways), while caloric restriction, exercise, or Mediterranean-type diets shift cargo toward mitochondrial-supportive and anti-fibrotic miRNAs [180]. Importantly, visceral adipose sEVs are often more pathogenic than subcutaneous depot sEVs in animal studies, aligning with clinical observations that visceral adiposity more strongly predicts cardiometabolic risk [181]. The gut microbiome modulates cardiac physiology indirectly through multiple metabolites, short-chain fatty acids (SCFAs), secondary bile acids, and trimethylamine-N-oxide (TMAO) being chief examples, and through effects on adipose inflammation and exosome secretion [182]. SCFAs produced by fiber fermentation improve adipose insulin sensitivity and can promote thermogenesis, whereas microbiota that favor TMAO production (from dietary choline/carnitine) correlate with adverse cardiac outcomes. There is growing evidence of crosstalk: microbiota-derived metabolites alter adipose sEV cargo and secretion rate, and adipose inflammation alters gut barrier integrity and microbial composition, creating feed-forward loops that modulate cardiac risk [183].
Mechanistically, gut microbiota modulate host exosome production through several interconnected pathways. Microbial metabolites, particularly SCFAs such as butyrate and propionate, act on adipocytes via G-protein-coupled receptors (GPR41/GPR43) and histone deacetylase inhibition, altering intracellular signaling cascades that regulate exosome biogenesis and cargo loading [184,185]. Lipopolysaccharide (LPS) from dysbiotic microbiota activates TLR4-NF-κB signaling in adipocytes, promoting release of pro-inflammatory sEVs enriched in miRNAs such as miR-34a and miR-155 that target anti-inflammatory pathways [186,187]. Dietary inputs thus exert simultaneous effects across this network: high-fiber intake promotes SCFA-producing bacteria (Bifidobacterium, Faecalibacterium), which enhance adiponectin secretion, reduce inflammatory adipokine output, and shift adipose sEV miRNA profiles toward anti-inflammatory signatures (increased miR-22, miR-146a; decreased miR-34a) [188]. Conversely, Western diets rich in saturated fat and L-carnitine favor TMAO-producing taxa and LPS translocation, suppressing adiponectin while elevating resistin, IL-6, and pro-fibrotic exosomal miRNAs [183]. These integrated signals converge on cardiomyocytes: SCFA-conditioned adipose sEVs deliver miRNAs that support mitochondrial biogenesis (via AMPK/PGC-1α) and reduce oxidative stress, whereas dysbiosis-associated sEVs carrying miR-34a inhibit SIRT1 and promote cardiomyocyte apoptosis and fibroblast activation [186,189]. TMAO itself synergizes with pro-inflammatory exosomal cargo to activate TGF-β/Smad pathways in cardiac fibroblasts, accelerating fibrosis and diastolic dysfunction [190]. This tripartite gut–adipose–heart communication network demonstrates how nutrient inputs shape microbial ecology, adipokine profiles, and sEV-mediated miRNA transfer in an integrated manner that ultimately determines cardiac metabolic flexibility and resilience.
Adipose sEVs and their miRNA cargo are attractive biomarkers (circulating vesicles can reflect depot-specific pathology) and promising therapeutic modalities [191]. Two translational strategies are emerging: inhibit pathogenic sEV production or uptake (targeting the ESCRT machinery, neutral sphingomyelinase, or blocking key surface ligands); or engineer sEVs or synthetic nanoparticles to deliver cardioprotective miRNAs or metabolic regulators to the heart. Parallel microbiome interventions (dietary fiber, targeted pre-/probiotics, and inhibitors of microbial TMA production) aim to shift metabolite profiles and, indirectly, reprogram adipose signalling. Early preclinical successes have been reported, but human proof-of-concept trials remain limited [192]. This multi-layered adipose–cardiac communication network, integrating classical adipokines, sEV-derived miRNAs, and gut microbiota metabolites, is summarized in Figure 1.
Figure 1.
Multi-Level Adipose–Cardiac Axis Communication: Integrating Classical and Emerging Signaling Pathways. The figure illustrates the multilayered communication network between adipose tissue (left) and the heart (right). Adipose-to-heart signaling (efferent arm): Adipose tissue releases classical adipokines (adiponectin, leptin, resistin) that modulate myocardial metabolism, insulin sensitivity, and inflammatory tone. Extracellular vesicles (sEVs) carrying miRNAs (e.g., miR-34a, miR-122, miR-146a) transfer post-transcriptional regulatory signals that influence cardiomyocyte gene expression, mitochondrial function, and fibrotic pathways. Lipid mediators, including free fatty acids and ceramides, serve as metabolic substrates but in excess contribute to cardiac lipotoxicity and metabolic inflexibility. Heart-to-adipose signaling (afferent arm): The failing or stressed heart releases inflammatory cytokines (TNF-α, IL-6, IL-1β) and natriuretic peptides that act on adipose tissue to alter lipolysis, adipokine secretion, and thermogenic capacity. These cardiac-derived signals contribute to systemic inflammation and can exacerbate adipose dysfunction, creating pathological feed-forward loops. Gut microbiota as a modulatory axis: Microbiota-derived metabolites, including short-chain fatty acids (SCFAs), trimethylamine N-oxide (TMAO), and secondary bile acids, independently influence both adipose tissue function and cardiac responses, representing an environmental layer that intersects with host-derived signaling. Central convergence on adipose dysfunction: The intersection of these signaling layers at adipose tissue dysfunction (center) illustrates how metabolic, inflammatory, and microbiota-derived inputs converge to drive cardiac metabolic inflexibility and lipotoxicity. Overall, the figure depicts an integrated system in which classical hormones, vesicle-bound non-coding RNAs, lipid substrates, inflammatory mediators, and microbiota-derived metabolites form distinct but interconnected communication layers modulating the adipose–cardiac axis.
In summary, inter-organ communication extends beyond classical adipokines to include adipose-derived extracellular vesicles carrying miRNAs and gut microbiota-derived metabolites such as SCFAs and TMAO. These emerging mediators translate nutritional and inflammatory states in adipose tissue into cardiac metabolic and structural responses, offering novel biomarker and therapeutic opportunities for cardiometabolic disease.
7. Biomarkers and Translational Therapeutic Targets in the Adipose-Cardiac Axis
Accurate biomarkers that capture adipose health, inter-organ signalling, and cardiac metabolic flexibility are central for translating mechanistic insights into patient care. Biomarkers relevant to the adipose-cardiac axis can be categorized according to their clinical utility: diagnostic biomarkers identify the presence or severity of adipose dysfunction and its cardiac consequences; prognostic biomarkers predict disease trajectory, adverse events, or mortality risk; and therapeutic-response biomarkers track the efficacy of nutritional, pharmacological, or lifestyle interventions. Several molecules serve dual or triple roles depending on clinical context, underscoring the integrated nature of adipose-cardiac communication. Complementing biomarkers, several translational targets have matured to the point of clinical testing or near-term clinical translation [193].
Diagnostic biomarkers enable identification and phenotyping of adipose-cardiac dysfunction. Circulating adiponectin (particularly the high-molecular-weight fraction) remains a robust inverse marker of adipose dysfunction and a predictor of improved cardiac metabolic profile; low adiponectin associates with impaired myocardial FAO and greater lipotoxicity [194]. Ceramides (distinct species, e.g., C16:0/C18:0) have emerged as reproducible lipid biomarkers of lipotoxic stress and cardiovascular risk that integrate adipose lipolytic activity and hepatic lipid handling; ceramide panels have predictive value beyond traditional lipids in multiple cohorts. Advanced imaging (cardiac MR spectroscopy for myocardial steatosis, FDG-PET or dedicated tracers for brown adipose quantification, CT/MRI-derived visceral adipose radiodensity) adds tissue-level phenotyping to circulating biomarkers and enhances the prediction of who will benefit from targeted metabolic interventions [195,196].
Prognostic biomarkers predict disease trajectory and cardiovascular outcomes. Acylcarnitine profiles, ketone body levels, and circulating mtDNA/mitochondria-derived peptides provide readouts of mitochondrial substrate flux and damage and are promising markers of cardiac metabolic flexibility [197,198]. Circulating adipose-derived sEVs carry depot-specific miRNA signatures that may serve as early indicators of pathogenic adipose signalling before overt clinical disease. Several groups have described EV-miRNA panels (including miR-34a, miR-122, and miR-192) associated with fibrosis, insulin resistance, and atherosclerotic progression. EVs have practical advantages for stability and tissue specificity, but methodological standardization and large prospective validation are outstanding needs [191]. Elevated TMAO levels independently predict major adverse cardiovascular events and heart failure progression, linking gut microbiota composition to long-term cardiac risk. Echocardiography and circulating NT-proBNP/hs-troponin remain essential for clinical endpoints [47,199].
Therapeutic-response biomarkers track the efficacy of interventions targeting the adipose-cardiac axis. Changes in adiponectin levels, HOMA-IR, and ceramide profiles following dietary interventions (Mediterranean diet, caloric restriction) or pharmacotherapy (GLP-1 agonists, SGLT2 inhibitors) provide mechanistic confirmation of treatment effects. Shifts in circulating sEV-miRNA signatures and reductions in TMAO with dietary fiber or microbiome-targeted therapies offer additional metrics for monitoring metabolic reprogramming. These biomarkers complement traditional endpoints when assessing dietary or metabolic therapies [47,199]. Several mechanistic targets have direct translational lines. Adiponectin pathway—adiponectin receptor agonists and agents increasing adiponectin expression are under preclinical/early clinical evaluation to restore cardiac mitochondrial function and reduce fibrosis [194]. Lipotoxicity axis, inhibitors of ceramide synthesis and modulators of fatty-acid oxidation (CPT1 modulators, ATGL regulators) aim to rebalance myocardial substrate handling; ceramide-lowering shows promising associations with improved outcomes in early human studies [198]. SIRT1/AMPK activation—NAD+ precursors, SIRT1 activators (resveratrol analogues), and AMPK activators remain attractive for enhancing mitochondrial biogenesis and metabolic flexibility; formulation and target specificity are active development challenges [200,201]. EV therapeutics, ASC-derived exosome products designed to deliver cardioprotective miRNAs or proteins, are in translational pipelines; scalability and safety (immunogenicity, off-target effects) remain to be resolved [202]. Microbiome-targeted approaches—dietary fiber interventions to raise SCFAs, inhibitors of microbial TMA formation, and targeted probiotic consortia are being evaluated for their capacity to lower systemic inflammation, reprogram adipose function, and reduce cardiac risk [192].
GLP-1 receptor agonists and SGLT2 inhibitors represent emerging therapeutic modalities that impact the adipose-cardiac axis beyond glycemic control. GLP-1 agonists modulate appetite and energy intake and activate key metabolic pathways, such as AMPK and mTOR, that overlap with mechanisms of calorie restriction and healthy diets. Clinically, they reduce visceral and epicardial adipose tissue volumes, thereby improving cardiac metabolism and reducing inflammation. SGLT2 inhibitors also support cardiac remodeling by inducing mild ketosis, enhancing myocardial ketone utilization, and improving metabolic flexibility, similar to ketogenic diets. These agents complement biomarker-driven strategies by targeting pathological adipose signals and promoting mitochondria [203,204,205].
A translational pathway requires combining accessible biomarkers (serum ceramides, adiponectin, EV-miRNA panels) with imaging phenotyping and randomized intervention trials that use biomarker changes as mechanistic endpoints. Critical gaps include the standardization of EV assays, prospective validation of ceramide panels across diverse populations, and well-powered human trials to determine whether biomarker-guided interventions (e.g., ceramide lowering, adiponectin agonism, or microbiome modulation) can achieve durable reductions in primary cardiovascular outcomes [198]. In summary, biomarkers reflecting adipose dysfunction (adiponectin, ceramides, sEV-miRNAs) and imaging modalities (cardiac MR spectroscopy, FDG-PET) can stratify patients for targeted interventions. Emerging therapies, including adiponectin receptor agonists, AMPK/SIRT1 activators, GLP-1 receptor agonists, SGLT2 inhibitors, and microbiome-targeted approaches, aim to restore adipose-cardiac coordination and improve cardiometabolic outcomes. Key biomarkers and emerging therapeutic targets that reflect adipose dysfunction, inter-organ crosstalk, and cardiac metabolic flexibility across obesity, diabetes, and heart failure are summarised in Table 3.
Table 3.
Principal Biomarkers and Emerging Therapeutic Targets Reflecting Adipose Dysfunction, Inter-Organ Crosstalk, and Cardiac Metabolic Flexibility in Obesity, Diabetes, and Heart Failure.
8. Conclusions and Future Directions
Metabolic and signaling crosstalk between adipose tissue and the heart is profoundly influenced by nutrient status. This review illustrates that macronutrients and key micronutrients can remodel the adipose-cardiac axis, promoting metabolic flexibility or driving maladaptation in obesity, diabetes, and heart failure. However, despite growing insights into how carbohydrates, fats, proteins, and vitamins/minerals (e.g., vitamin D, magnesium, zinc, selenium, and iron) affect adipokine signaling and cardiac fuel use, significant knowledge gaps remain. The interplay among multiple nutrients and the long-term consequences of dietary patterns on the adipose-heart axis remain poorly elucidated, underscoring the need for more comprehensive mechanistic studies and longitudinal clinical research [116,126,138,145,147].
A primary future direction is to decipher the complex, context-dependent effects of nutrients on inter-organ communication. For instance, how combined macronutrient excess (or restriction) and micronutrient imbalances jointly shape adipose inflammation, adipokine profiles, and cardiac substrate preference remains unclear. Advanced in vivo models and multi-omics approaches could help unravel nutrient–gene interactions and identify new mediators of adipose-cardiac signaling [117,155,156]. Emerging evidence indicates that adipose-derived exosomal microRNAs and gut microbiota metabolites convey nutritional signals to the heart, warranting further investigation [158,162,163]. Standardization of assays (e.g., for extracellular vesicle biomarkers) and large-scale validation studies are critical next steps. Several specific unmet clinical needs warrant prioritization: standardized EV-miRNA panels require consensus protocols for vesicle isolation, miRNA quantification, and normalization to enable cross-study comparisons and clinical translation [206]; longitudinal dietary intervention trials simultaneously tracking nutritional biomarkers (adiponectin, ceramides, TMAO, sEV-miRNAs) and cardiac remodeling parameters (echocardiographic indices, MR-derived steatosis and fibrosis markers) are needed to clarify whether biomarker changes precede or follow structural cardiac adaptations [207]; and head-to-head comparisons of dietary patterns using hard cardiometabolic endpoints would strengthen evidence-based prescription for specific patient phenotypes. Likewise, well-powered human trials are needed to test whether modulating these pathways—through diets or pharmaceuticals—can meaningfully improve cardiac outcomes [148,149,159,166].
From a clinical standpoint, these findings carry promising therapeutic implications. Correcting common nutrient deficiencies (such as vitamin D or iron) is an attractive strategy, given their links to myocardial dysfunction and heart failure outcomes [159,174,178,180]. Novel interventions targeting adipose-derived signals are also on the horizon; for example, adiponectin receptor agonists aim to restore cardioprotective adipokine activity, and GLP-1 receptor agonists and SGLT2 inhibitors modulate adipose tissue inflammation and enhance cardiac metabolic flexibility by reducing visceral fat and lipotoxicity while improving energy substrate utilization [169,194]. Microbiome-targeted diets or probiotics seek to reduce pro-inflammatory signaling [47,172]. Such approaches could enhance cardiac metabolic flexibility and reduce lipotoxic damage. Importantly, translating these advances into practice will require rigorous clinical trials and biomarker-guided strategies to confirm improved patient outcomes [169,193]. Bridging these gaps through future research will pave the way for nutrient-based interventions that bolster the adipose-cardiac axis and ultimately combat cardiometabolic disease.
Author Contributions
Conceptualization, N.P., P.T., M.M., M.K. and J.B.; methodology, N.P.; investigation, N.P. and P.T.; resources, J.B.; writing—original draft preparation, N.P., P.T., M.M., M.K. and J.B.; writing—review and editing, N.P. and J.B.; visualization, N.P.; supervision, J.B.; project administration, J.B. All authors have read and agreed to the published version of the manuscript.
Funding
This research received no external funding.
Institutional Review Board Statement
Not applicable.
Informed Consent Statement
Not applicable.
Data Availability Statement
Not applicable.
Acknowledgments
The authors have reviewed and edited the output and take full responsibility for the content of this publication.
Conflicts of Interest
The authors declare no conflicts of interest.
References
- Isomaa, B.; Almgren, P.; Tuomi, T.; Forsén, B.; Lahti, K.; Nissén, M.; Taskinen, M.R.; Groop, L. Cardiovascular morbidity and mortality associated with the metabolic syndrome. Diabetes Care 2001, 24, 683–689. [Google Scholar] [CrossRef] [PubMed]
- Al Suwaidi, J.; Zubaid, M.; El-Menyar, A.A.; Singh, R.; Rashed, W.; Ridha, M.; Shehab, A.; Al-Lawati, J.; Amin, H.; Al-Mottareb, A. Prevalence of the metabolic syndrome in patients with acute coronary syndrome in six middle eastern countries. J. Clin. Hypertens. 2010, 12, 890–899. [Google Scholar] [CrossRef] [PubMed]
- Milionis, H.J.; Kalantzi, K.J.; Papathanasiou, A.J.; Kosovitsas, A.A.; Doumas, M.T.; Goudevenos, J.A. Metabolic syndrome and risk of acute coronary syndromes in patients younger than 45 years of age. Coron. Artery Dis. 2007, 18, 247–252. [Google Scholar] [CrossRef] [PubMed]
- Tune, J.D.; Goodwill, A.G.; Sassoon, D.J.; Mather, K.J. Cardiovascular consequences of metabolic syndrome. Transl. Res. 2017, 183, 57–70. [Google Scholar] [CrossRef]
- Lavie, C.J.; Milani, R.V.; Artham, S.M.; Patel, D.A.; Ventura, H.O. The obesity paradox, weight loss, and coronary disease. Am. J. Med. 2009, 122, 1106–1114. [Google Scholar] [CrossRef]
- Thakker, J.; Khaliq, I.; Ardeshna, N.S.; Lavie, C.J.; Oktay, A.A. The Obesity Paradox of Cardiovascular Outcomes in Patients with Diabetes Mellitus. Curr. Diabetes Rep. 2025, 25, 35. [Google Scholar] [CrossRef]
- Diab, A.; Dastmalchi, L.N.; Gulati, M.; Michos, E.D. A Heart-Healthy Diet for Cardiovascular Disease Prevention: Where Are We Now? Vasc. Health Risk Manag. 2023, 19, 237–253. [Google Scholar] [CrossRef]
- Le Goff, D.; Aerts, N.; Odorico, M.; Guillou-Landreat, M.; Perraud, G.; Bastiaens, H.; Musinguzi, G.; Le Reste, J.Y.; Barais, M. Practical dietary interventions to prevent cardiovascular disease suitable for implementation in primary care: An ADAPTE-guided systematic review of international clinical guidelines. Int. J. Behav. Nutr. Phys. Act. 2023, 20, 93. [Google Scholar] [CrossRef]
- Ha, E.E.; Bauer, R.C. Emerging Roles for Adipose Tissue in Cardiovascular Disease. Arter. Thromb. Vasc. Biol. 2018, 38, e137–e144. [Google Scholar] [CrossRef]
- Oikonomou, E.K.; Antoniades, C. The role of adipose tissue in cardiovascular health and disease. Nat. Rev. Cardiol. 2019, 16, 83–99. [Google Scholar] [CrossRef]
- Alotaibi, M.M.; Alrashdi, N.Z.; Almutairi, M.K.; Alqahtani, M.M.; Almutairi, A.B.; Alqahtani, S.M.; Alajel, H.M.; Bajunayd, A.K. Association of adipose tissue infiltration with cardiac function: Scoping review. Adipocyte 2025, 14, 2489467. [Google Scholar] [CrossRef]
- Bou Matar, D.; Zhra, M.; Nassar, W.K.; Altemyatt, H.; Naureen, A.; Abotouk, N.; Elahi, M.A.; Aljada, A. Adipose tissue dysfunction disrupts metabolic homeostasis: Mechanisms linking fat dysregulation to disease. Front. Endocrinol. 2025, 16, 1592683. [Google Scholar] [CrossRef] [PubMed]
- Giralt, M.; Villarroya, F. White, brown, beige/brite: Different adipose cells for different functions? Endocrinology 2013, 154, 2992–3000. [Google Scholar] [CrossRef]
- Walkiewicz, K.W.; Dzięgielewska-Gęsiak, S.; Myrcik, D.; Bednarczyk, M.; Muc-Wierzgoń, M. Eating behavior patterns in relation to obesity phenotypes and beige adipose tissue content with a focus on young women; a narrative review. Front. Nutr. 2025, 12, 1692944. [Google Scholar] [CrossRef] [PubMed]
- Wiszniewski, K.; Grudniewska, A.; Szabłowska-Gadomska, I.; Pilichowska-Paszkiet, E.; Zaborska, B.; Zgliczyński, W.; Dudek, P.; Bik, W.; Sota, M.; Mrozikiewicz-Rakowska, B. Epicardial Adipose Tissue-A Novel Therapeutic Target in Obesity Cardiomyopathy. Int. J. Mol. Sci. 2025, 26, 7963. [Google Scholar] [CrossRef] [PubMed]
- Wen, X.; Zhang, B.; Wu, B.; Xiao, H.; Li, Z.; Li, R.; Xu, X.; Li, T. Signaling pathways in obesity: Mechanisms and therapeutic interventions. Signal Transduct. Target. Ther. 2022, 7, 298, Erratum in Signal Transduct. Target Ther. 2022, 7, 369. https://doi.org/10.1038/s41392-022-01188-4. [Google Scholar] [CrossRef]
- Munoz, M.D.; Zamudio, A.; McCann, M.; Gil, V.; Xu, P.; Liew, C.W. Activation of brown adipose tissue by a low-protein diet ameliorates hyperglycemia in a diabetic lipodystrophy mouse model. Sci. Rep. 2023, 13, 11808. [Google Scholar] [CrossRef]
- Littlejohn, P.T.; Bar-Yoseph, H.; Edwards, K.; Li, H.; Ramirez-Contreras, C.Y.; Holani, R.; Metcalfe-Roach, A.; Fan, Y.M.; Yang, T.M.; Radisavljevic, N.; et al. Multiple micronutrient deficiencies alter energy metabolism in host and gut microbiome in an early-life murine model. Front. Nutr. 2023, 10, 1151670. [Google Scholar] [CrossRef]
- Liu, H.; Wang, S.; Wang, J.; Guo, X.; Song, Y.; Fu, K.; Gao, Z.; Liu, D.; He, W.; Yang, L.L. Energy metabolism in health and diseases. Signal Transduct. Target. Ther. 2025, 10, 69. [Google Scholar] [CrossRef]
- Oldham, S. Obesity and nutrient sensing TOR pathway in flies and vertebrates: Functional conservation of genetic mechanisms. Trends Endocrinol. Metab. 2011, 22, 45–52. [Google Scholar] [CrossRef]
- Choe, S.S.; Huh, J.Y.; Hwang, I.J.; Kim, J.I.; Kim, J.B. Adipose Tissue Remodeling: Its Role in Energy Metabolism and Metabolic Disorders. Front. Endocrinol. 2016, 7, 30. [Google Scholar] [CrossRef] [PubMed]
- 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] [CrossRef]
- Tseng, Y.H. Adipose tissue in communication: Within and without. Nat. Rev. Endocrinol. 2023, 19, 70–71. [Google Scholar] [CrossRef] [PubMed]
- Ruan, H.; Dong, L.Q. Adiponectin signaling and function in insulin target tissues. J. Mol. Cell Biol. 2016, 8, 101–109. [Google Scholar] [CrossRef]
- Hivert, M.F.; Manning, A.K.; McAteer, J.B.; Florez, J.C.; Dupuis, J.; Fox, C.S.; O’Donnell, C.J.; Cupples, L.A.; Meigs, J.B. Common variants in the adiponectin gene (ADIPOQ) associated with plasma adiponectin levels, type 2 diabetes, and diabetes-related quantitative traits: The Framingham Offspring Study. Diabetes 2008, 57, 3353–3359. [Google Scholar] [CrossRef]
- Adler, E.M. The Adiponectin-Mitochondria Connection. Sci. Signal. 2010, 3, ec136. [Google Scholar] [CrossRef]
- Qiao, L.; Kinney, B.; Yoo, H.S.; Lee, B.; Schaack, J.; Shao, J. Adiponectin increases skeletal muscle mitochondrial biogenesis by suppressing mitogen-activated protein kinase phosphatase-1. Diabetes 2012, 61, 1463–1470. [Google Scholar] [CrossRef]
- Choi, H.M.; Doss, H.M.; Kim, K.S. Multifaceted Physiological Roles of Adiponectin in Inflammation and Diseases. Int. J. Mol. Sci. 2020, 21, 1219. [Google Scholar] [CrossRef]
- Ouchi, N.; Walsh, K. A novel role for adiponectin in the regulation of inflammation. Arter. Thromb. Vasc. Biol. 2008, 28, 1219–1221. [Google Scholar] [CrossRef]
- Clemente-Suárez, V.J.; Redondo-Flórez, L.; Beltrán-Velasco, A.I.; Martín-Rodríguez, A.; Martínez-Guardado, I.; Navarro-Jiménez, E.; Laborde-Cárdenas, C.C.; Tornero-Aguilera, J.F. The Role of Adipokines in Health and Disease. Biomedicines 2023, 11, 1290. [Google Scholar] [CrossRef]
- Kawai, T.; Autieri, M.V.; Scalia, R. Adipose tissue inflammation and metabolic dysfunction in obesity. Am. J. Physiol. Cell Physiol. 2021, 320, C375–C391. [Google Scholar] [CrossRef]
- Menzaghi, C.; Marucci, A.; Antonucci, A.; De Bonis, C.; Ortega Moreno, L.; Salvemini, L.; Copetti, M.; Trischitta, V.; Di Paola, R. Suggestive evidence of a multi-cytokine resistin pathway in humans and its role on cardiovascular events in high-risk individuals. Sci. Rep. 2017, 7, 44337. [Google Scholar] [CrossRef] [PubMed]
- Ghanem, S.E.; Abdel-Samiee, M.; Torky, M.H.; Gaafar, A.; Mohamed, S.M.; Salah Eldin, G.M.M.; Awad, S.M.; Diab, K.A.; Elsabaawy, D.M.; Yehia, S.A.; et al. Role of resistin, IL-6 and NH2-terminal portion proBNP in the pathogenesis of cardiac disease in type 2 diabetes mellitus. BMJ Open Diabetes Res. Care 2020, 8, e001206. [Google Scholar] [CrossRef] [PubMed]
- Lopez-Garcia, E.; Hu, F.B. Nutrition and the endothelium. Curr. Diabetes Rep. 2004, 4, 253–259. [Google Scholar] [CrossRef] [PubMed]
- Dehghani, F.; Hajhashemy, Z.; Keshteli, A.H.; Yazdannik, A.; Falahi, E.; Saneei, P.; Esmaillzadeh, A. Nutrient patterns in relation to insulin resistance and endothelial dysfunction in Iranian women. Sci. Rep. 2024, 14, 2857. [Google Scholar] [CrossRef]
- Hall, M.E.; Harmancey, R.; Stec, D.E. Lean heart: Role of leptin in cardiac hypertrophy and metabolism. World J. Cardiol. 2015, 7, 511–524. [Google Scholar] [CrossRef]
- Kamareddine, L.; Ghantous, C.M.; Allouch, S.; Al-Ashmar, S.A.; Anlar, G.; Kannan, S.; Djouhri, L.; Korashy, H.M.; Agouni, A.; Zeidan, A. Between Inflammation and Autophagy: The Role of Leptin-Adiponectin Axis in Cardiac Remodeling. J. Inflamm. Res. 2021, 14, 5349–5365. [Google Scholar] [CrossRef]
- Dong, F.; Zhang, X.; Ren, J. Leptin regulates cardiomyocyte contractile function through endothelin-1 receptor-NADPH oxidase pathway. Hypertension 2006, 47, 222–229. [Google Scholar] [CrossRef]
- Gruzdeva, O.; Borodkina, D.; Uchasova, E.; Dyleva, Y.; Barbarash, O. Leptin resistance: Underlying mechanisms and diagnosis. Diabetes, Metab. Syndr. Obesity 2019, 12, 191–198. [Google Scholar] [CrossRef]
- Martin, S.S.; Qasim, A.; Reilly, M.P. Leptin resistance: A possible interface of inflammation and metabolism in obesity-related cardiovascular disease. J. Am. Coll. Cardiol. 2008, 52, 1201–1210. [Google Scholar] [CrossRef]
- Li, C.; Sun, X.N.; Zhao, S.; Scherer, P.E. Crosstalk Between Adipose Tissue and the Heart: An Update. J. Transl. Intern. Med. 2022, 10, 219–226. [Google Scholar] [CrossRef]
- Berezin, A.E.; Berezin, A.A.; Lichtenauer, M. Emerging Role of Adipocyte Dysfunction in Inducing Heart Failure Among Obese Patients with Prediabetes and Known Diabetes Mellitus. Front. Cardiovasc. Med. 2020, 7, 583175. [Google Scholar] [CrossRef]
- Zhang, K.; Yuan, Z.; Wang, S.; Zhao, S.; Cui, H.; Lai, Y. The abnormalities of free fatty acid metabolism in patients with hypertrophic cardiomyopathy, a single-center retrospective observational study. BMC Cardiovasc. Disord. 2024, 24, 312. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Yang, S.; Chen, J.; Su, Z. Unraveling the Regulation of Hepatic Gluconeogenesis. Front. Endocrinol. 2019, 9, 802. [Google Scholar] [CrossRef] [PubMed]
- Aubert, G.; Martin, O.J.; Horton, J.L.; Lai, L.; Vega, R.B.; Leone, T.C.; Koves, T.; Gardell, S.J.; Krüger, M.; Hoppel, C.L.; et al. The Failing Heart Relies on Ketone Bodies as a Fuel. Circulation 2016, 133, 698–705. [Google Scholar] [CrossRef]
- Matsuura, T.R.; Puchalska, P.; Crawford, P.A.; Kelly, D.P. Ketones and the Heart: Metabolic Principles and Therapeutic Implications. Circ. Res. 2023, 132, 882–898. [Google Scholar] [CrossRef] [PubMed]
- Michel, L.Y.M. Extracellular Vesicles in Adipose Tissue Communication with the Healthy and Pathological Heart. Int. J. Mol. Sci. 2023, 24, 7745. [Google Scholar] [CrossRef]
- Duncan, J.G.; Finck, B.N. The PPARalpha-PGC-1alpha Axis Controls Cardiac Energy Metabolism in Healthy and Diseased Myocardium. PPAR Res. 2008, 2008, 253817. [Google Scholar] [CrossRef]
- Parodi-Rullán, R.M.; Chapa-Dubocq, X.R.; Javadov, S. Acetylation of Mitochondrial Proteins in the Heart: The Role of SIRT3. Front. Physiol. 2018, 9, 1094. [Google Scholar] [CrossRef]
- Pillai, V.B.; Sundaresan, N.R.; Jeevanandam, V.; Gupta, M.P. Mitochondrial SIRT3 and heart disease. Cardiovasc. Res. 2010, 88, 250–256. [Google Scholar] [CrossRef]
- Tang, S.; Li, R.; Ma, W.; Lian, L.; Gao, J.; Cao, Y.; Gan, L. Cardiac-to-adipose axis in metabolic homeostasis and diseases: Special instructions from the heart. Cell Biosci. 2023, 13, 161. [Google Scholar] [CrossRef] [PubMed]
- Yan, Y.; Wang, L.; Zhong, N.; Wen, D.; Liu, L. Multifaceted Roles of Adipokines in Endothelial Cell Function. Front. Endocrinol. 2024, 15, 1490143. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Han, X.; Song, J.; Dong, M.; Xie, T. Mechanism of Action and Risk Prediction of Adiponectin in Cardiovascular Diseases. Front. Biosci. 2024, 29, 286. [Google Scholar] [CrossRef] [PubMed]
- Osorio-Conles, Ó.; Olbeyra, R.; Moizé, V.; Ibarzabal, A.; Giró, O.; Viaplana, J.; Jiménez, A.; Vidal, J.; de Hollanda, A. Positive Effects of a Mediterranean Diet Supplemented with Almonds on Female Adipose Tissue Biology in Severe Obesity. Nutrients 2022, 14, 2617. [Google Scholar] [CrossRef]
- Grahovac, M.; Kumric, M.; Vilovic, M.; Supe-Domic, D.; Pavlovic, N.; Bukic, J.; Ticinovic Kurir, T.; Bozic, J. Endocannabinoid and AGE Interactions in Prediabetes: The Role of Mediterranean Diet Adherence. Nutrients 2025, 17, 2517. [Google Scholar] [CrossRef]
- Martinovic, D.; Tokic, D.; Martinovic, L.; Kumric, M.; Vilovic, M.; Rusic, D.; Vrdoljak, J.; Males, I.; Ticinovic Kurir, T.; Lupi-Ferandin, S.; et al. Adherence to the Mediterranean Diet and Its Association with the Level of Physical Activity in Fitness Center Users: Croatian-Based Study. Nutrients 2021, 13, 4038. [Google Scholar] [CrossRef]
- Hsiao, G.; Chapman, J.; Ofrecio, J.M.; Wilkes, J.; Resnik, J.L.; Thapar, D.; Subramaniam, S.; Sears, D.D. Multi-tissue, selective PPARγ modulation of insulin sensitivity and metabolic pathways in obese rats. Am. J. Physiol. Endocrinol. Metab. 2011, 300, E164–E174. [Google Scholar] [CrossRef]
- Florkowski, M.; Abiona, E.; Frank, K.M.; Brichacek, A.L. Obesity-associated inflammation countered by a Mediterranean diet: The role of gut-derived metabolites. Front. Nutr. 2024, 11, 1392666. [Google Scholar] [CrossRef]
- Iwabu, M.; Yamauchi, T.; Okada-Iwabu, M.; Sato, K.; Nakagawa, T.; Funata, M.; Yamaguchi, M.; Namiki, S.; Nakayama, R.; Tabata, M.; et al. Adiponectin and AdipoR1 regulate PGC-1alpha and mitochondria by Ca2+ and AMPK/SIRT1. Nature 2010, 464, 1313–1319. [Google Scholar] [CrossRef]
- Rius-Pérez, S.; Torres-Cuevas, I.; Millán, I.; Ortega, Á.L.; Pérez, S. PGC-1α, Inflammation, and Oxidative Stress: An Integrative View in Metabolism. Oxid. Med. Cell. Longev. 2020, 2020, 1452696. [Google Scholar] [CrossRef]
- Hareer, L.W.; Lau, Y.Y.; Mole, F.; Reidlinger, D.P.; O’Neill, H.M.; Mayr, H.L.; Greenwood, H.; Albarqouni, L. The effectiveness of the Mediterranean Diet for primary and secondary prevention of cardiovascular disease: An umbrella review. Nutr. Diet. 2025, 82, 8–41. [Google Scholar] [CrossRef] [PubMed]
- Otręba, M.; Kośmider, L.; Stojko, J.; Rzepecka-Stojko, A. Cardioprotective Activity of Selected Polyphenols Based on Epithelial and Aortic Cell Lines. A Review. Molecules 2020, 25, 5343. [Google Scholar] [CrossRef] [PubMed]
- Hedayati, N.; Yaghoobi, A.; Salami, M.; Gholinezhad, Y.; Aghadavood, F.; Eshraghi, R.; Aarabi, M.H.; Homayoonfal, M.; Asemi, Z.; Mirzaei, H.; et al. Impact of polyphenols on heart failure and cardiac hypertrophy: Clinical effects and molecular mechanisms. Front. Cardiovasc. Med. 2023, 10, 1174816. [Google Scholar] [CrossRef] [PubMed]
- Grosso, G.; Marventano, S.; Yang, J.; Micek, A.; Pajak, A.; Scalfi, L.; Galvano, F.; Kales, S.N. A comprehensive meta-analysis on evidence of Mediterranean diet and cardiovascular disease: Are individual components equal? Crit. Rev. Food Sci. Nutr. 2017, 57, 3218–3232. [Google Scholar] [CrossRef]
- Andriantsitohaina, R.; Auger, C.; Chataigneau, T.; Étienne-Selloum, N.; Li, H.; Martínez, M.C.; Schini-Kerth, V.B.; Laher, I. Molecular mechanisms of the cardiovascular protective effects of polyphenols. Br. J. Nutr. 2012, 108, 1532–1549. [Google Scholar] [CrossRef]
- Akiyama, M.; Akiyama, T.; Saigusa, D.; Hishinuma, E.; Matsukawa, N.; Shibata, T.; Tsuchiya, H.; Mori, A.; Fujii, Y.; Mogami, Y.; et al. Comprehensive study of metabolic changes induced by a ketogenic diet therapy using GC/MS- and LC/MS-based metabolomics. Seizure 2023, 107, 52–59. [Google Scholar] [CrossRef]
- Biesiekierska, M.; Strigini, M.; Śliwińska, A.; Pirola, L.; Balcerczyk, A. The Impact of Ketogenic Nutrition on Obesity and Metabolic Health: Mechanisms and Clinical Implications. Nutr. Rev. 2025, 83, 1957–1972, Erratum in Nutr. Rev. 2025, 83, 1385. https://doi.org/10.1093/nutrit/nuaf058. [Google Scholar] [CrossRef]
- Li, J.; He, W.; Wu, Q.; Qin, Y.; Luo, C.; Dai, Z.; Long, Y.; Yan, P.; Huang, W.; Cao, L. Ketogenic diets and β-hydroxybutyrate in the prevention and treatment of diabetic kidney disease: Current progress and future perspectives. BMC Nephrol. 2025, 26, 127. [Google Scholar] [CrossRef]
- Llorente-Folch, I.; Düssmann, H.; Watters, O.; Connolly, N.M.C.; Prehn, J.H.M. Ketone body β-hydroxybutyrate (BHB) preserves mitochondrial bioenergetics. Sci. Rep. 2023, 13, 19664. [Google Scholar] [CrossRef]
- Da Eira, D.; Jani, S.; Stefanovic, M.; Ceddia, R.B. The ketogenic diet promotes triacylglycerol recycling in white adipose tissue and uncoupled fat oxidation in brown adipose tissue, but does not reduce adiposity in rats. J. Nutr. Biochem. 2023, 120, 109412. [Google Scholar] [CrossRef]
- Ahmad, Y.; Seo, D.S.; Jang, Y. Metabolic Effects of Ketogenic Diets: Exploring Whole-Body Metabolism in Connection with Adipose Tissue and Other Metabolic Organs. Int. J. Mol. Sci. 2024, 25, 7076. [Google Scholar] [CrossRef] [PubMed]
- Xu, S.; Tao, H.; Cao, W.; Cao, L.; Lin, Y.; Zhao, S.M.; Xu, W.; Cao, J.; Zhao, J.Y. Ketogenic diets inhibit mitochondrial biogenesis and induce cardiac fibrosis. Signal Transduct. Target. Ther. 2021, 6, 54. [Google Scholar] [CrossRef] [PubMed]
- Schmidt, T.; Harmon, D.M.; Kludtke, E.; Mickow, A.; Simha, V.; Kopecky, S. Dramatic elevation of LDL cholesterol from ketogenic-dieting: A Case Series. Am. J. Prev. Cardiol. 2023, 14, 100495. [Google Scholar] [CrossRef] [PubMed]
- Li, S.; Du, Y.; Meireles, C.; Sharma, K.; Qi, L.; Castillo, A.; Wang, J. Adherence to ketogenic diet in lifestyle interventions in adults with overweight or obesity and type 2 diabetes: A scoping review. Nutr. Diabetes 2023, 13, 16. [Google Scholar] [CrossRef]
- Crosby, L.; Davis, B.; Joshi, S.; Jardine, M.; Paul, J.; Neola, M.; Barnard, N.D. Ketogenic Diets and Chronic Disease: Weighing the Benefits Against the Risks. Front. Nutr. 2021, 8, 702802. [Google Scholar] [CrossRef]
- Zambuzzi, W.F.; Ferreira, M.R.; Wang, Z.; Peppelenbosch, M.P. A Biochemical View on Intermittent Fasting’s Effects on Human Physiology-Not Always a Beneficial Strategy. Biology 2025, 14, 669. [Google Scholar] [CrossRef]
- Kazmirczak, F.; Hartweck, L.M.; Vogel, N.T.; Mendelson, J.B.; Park, A.K.; Raveendran, R.M.; O-Uchi, J.; Jhun, B.S.; Prisco, S.Z.; Prins, K.W. Intermittent Fasting Activates AMP-Kinase to Restructure Right Ventricular Lipid Metabolism and Microtubules. JACC Basic Transl. Sci. 2023, 8, 239–254. [Google Scholar] [CrossRef]
- Liu, B.; Page, A.J.; Hutchison, A.T.; Wittert, G.A.; Heilbronn, L.K. Intermittent fasting increases energy expenditure and promotes adipose tissue browning in mice. Nutrition 2019, 66, 38–43. [Google Scholar] [CrossRef]
- Kim, K.H.; Kim, Y.H.; Son, J.E.; Lee, J.H.; Kim, S.; Choe, M.S.; Moon, J.H.; Zhong, J.; Fu, K.; Lenglin, F.; et al. Intermittent fasting promotes adipose thermogenesis and metabolic homeostasis via VEGF-mediated alternative activation of macrophage. Cell Res. 2017, 27, 1309–1326. [Google Scholar] [CrossRef]
- Sebastian, S.A.; Shah, Y.; Arsene, C. Intermittent fasting and cardiovascular disease: A scoping review of the evidence. Dis. Mon. 2024, 70, 101778. [Google Scholar] [CrossRef]
- Kahraman, S.; Dogan, A. Ventricular arrhythmia linked to long intermittent fasting. J. Electrocardiol. 2020, 58, 125–127. [Google Scholar] [CrossRef]
- Eliopoulos, A.G.; Gkouskou, K.K.; Tsioufis, K.; Sanoudou, D. A perspective on intermittent fasting and cardiovascular risk in the era of obesity pharmacotherapy. Front. Nutr. 2025, 12, 1524125. [Google Scholar] [CrossRef] [PubMed]
- Sun, M.L.; Yao, W.; Wang, X.Y.; Gao, S.; Varady, K.A.; Forslund, S.K.; Zhang, M.; Shi, Z.Y.; Cao, F.; Zou, B.J.; et al. Intermittent fasting and health outcomes: An umbrella review of systematic reviews and meta-analyses of randomised controlled trials. EClinicalMedicine 2024, 70, 102519. [Google Scholar] [CrossRef] [PubMed]
- Xiaoyu, W.; Yuxin, X.; Li, L. The effects of different intermittent fasting regimens in people with type 2 diabetes: A network meta-analysis. Front. Nutr. 2024, 11, 1325894. [Google Scholar] [CrossRef] [PubMed]
- Most, J.; Redman, L.M. Impact of calorie restriction on energy metabolism in humans. Exp. Gerontol. 2020, 133, 110875. [Google Scholar] [CrossRef]
- Browning, J.D.; Weis, B.; Davis, J.; Satapati, S.; Merritt, M.; Malloy, C.R.; Burgess, S.C. Alterations in hepatic glucose and energy metabolism as a result of calorie and carbohydrate restriction. Hepatology 2008, 48, 1487–1496. [Google Scholar] [CrossRef]
- Kim, K.E.; Jung, Y.; Min, S.; Nam, M.; Heo, R.W.; Jeon, B.T.; Song, D.H.; Yi, C.O.; Jeong, E.A.; Kim, H.; et al. Caloric restriction of db/db mice reverts hepatic steatosis and body weight with divergent hepatic metabolism. Sci. Rep. 2016, 6, 30111. [Google Scholar] [CrossRef]
- Derous, D.; Mitchell, S.E.; Wang, L.; Green, C.L.; Wang, Y.; Chen, L.; Han, J.J.; Promislow, D.E.L.; Lusseau, D.; Douglas, A.; et al. The effects of graded levels of calorie restriction: XI. Evaluation of the main hypotheses underpinning the life extension effects of CR using the hepatic transcriptome. Aging 2017, 9, 1770–1824. [Google Scholar] [CrossRef]
- Rahmani, S.; Roohbakhsh, A.; Pourbarkhordar, V.; Karimi, G. The Cardiovascular Protective Function of Natural Compounds Through AMPK/SIRT1/PGC-1α Signaling Pathway. Food Sci. Nutr. 2024, 12, 9998–10009. [Google Scholar] [CrossRef]
- Chen, W.K.; Tsai, Y.L.; Shibu, M.A.; Shen, C.Y.; Chang-Lee, S.N.; Chen, R.J.; Yao, C.H.; Ban, B.; Kuo, W.W.; Huang, C.Y. Exercise training augments Sirt1-signaling and attenuates cardiac inflammation in D-galactose induced-aging rats. Aging 2018, 10, 4166–4174. [Google Scholar] [CrossRef]
- Walker, A.E.; Henson, G.D.; Reihl, K.D.; Nielson, E.I.; Morgan, R.G.; Lesniewski, L.A.; Donato, A.J. Beneficial effects of lifelong caloric restriction on endothelial function are greater in conduit arteries compared to cerebral resistance arteries. Age 2014, 36, 559–569. [Google Scholar] [CrossRef] [PubMed]
- Di Daniele, N.; Marrone, G.; Di Lauro, M.; Di Daniele, F.; Palazzetti, D.; Guerriero, C.; Noce, A. Effects of Caloric Restriction Diet on Arterial Hypertension and Endothelial Dysfunction. Nutrients 2021, 13, 274. [Google Scholar] [CrossRef]
- Ungvari, Z.; Parrado-Fernandez, C.; Csiszar, A.; de Cabo, R. Mechanisms underlying caloric restriction and lifespan regulation: Implications for vascular aging. Circ. Res. 2008, 102, 519–528. [Google Scholar] [CrossRef] [PubMed]
- Simsek, B.; Yanar, K.; Kansu, A.D.; Belce, A.; Aydin, S.; Çakatay, U. Caloric restriction improves the redox homeostasis in the aging male rat heart even when started in middle-adulthood and when the body weight is stable. Biogerontology 2019, 20, 127–140. [Google Scholar] [CrossRef] [PubMed]
- Das, J.K.; Banskota, N.; Candia, J.; Griswold, M.E.; Orenduff, M.; de Cabo, R.; Corcoran, D.L.; Das, S.K.; De, S.; Huffman, K.M.; et al. Calorie restriction modulates the transcription of genes related to stress response and longevity in human muscle: The CALERIE study. Aging Cell 2023, 22, e13963. [Google Scholar] [CrossRef]
- Sacks, F.M.; Svetkey, L.P.; Vollmer, W.M.; Appel, L.J.; Bray, G.A.; Harsha, D.; Obarzanek, E.; Conlin, P.R.; Miller, E.R.; Simons-Morton, D.G.; et al. Effects on blood pressure of reduced dietary sodium and the Dietary Approaches to Stop Hypertension (DASH) diet. N. Engl. J. Med. 2001, 344, 3–10. [Google Scholar] [CrossRef]
- Juraschek, S.P.; Miller, E.R., 3rd; Chang, A.R.; Anderson, C.A.M.; Hall, J.E.; Appel, L.J. Effects of the Dietary Approaches to Stop Hypertension Diet on Change in Cardiac Biomarkers Over Time: Results From the DASH-Sodium Trial. J. Am. Heart Assoc. 2022, 12, e026684. [Google Scholar] [CrossRef]
- Azadbakht, L.; Surkan, P.J.; Esmaillzadeh, A.; Willett, W.C. The Dietary Approaches to Stop Hypertension eating plan affects C-reactive protein, coagulation abnormalities, and hepatic function tests among type 2 diabetic patients. J. Nutr. 2011, 141, 1083–1088. [Google Scholar] [CrossRef]
- Soltani, S.; Chitsazi, M.J.; Salehi-Abargouei, A. The effect of dietary approaches to stop hypertension (DASH) on serum inflammatory markers: A systematic review and meta-analysis of randomized trials. Clin. Nutr. 2018, 37, 542–550. [Google Scholar] [CrossRef]
- Appel, L.J.; Moore, T.J.; Obarzanek, E.; Vollmer, W.M.; Svetkey, L.P.; Sacks, F.M.; Bray, G.A.; Vogt, T.M.; Cutler, J.A.; Windhauser, M.M.; et al. A clinical trial of the effects of dietary patterns on blood pressure. N. Engl. J. Med. 1997, 336, 1117–1124. [Google Scholar] [CrossRef]
- Kim, H.; Caulfield, L.E.; Garcia-Larsen, V.; Steffen, L.M.; Coresh, J.; Rebholz, C.M. Plant-Based Diets Are Associated With a Lower Risk of Incident Cardiovascular Disease, Cardiovascular Disease Mortality, and All-Cause Mortality in a General Population of Middle-Aged Adults. J. Am. Heart Assoc. 2019, 8, e012865. [Google Scholar] [CrossRef]
- Kahleova, H.; Levin, S.; Barnard, N.D. Plant-Based Diets for Cardiovascular Risk Reduction: A Review of Observational and Interventional Studies. Nutrients 2017, 9, 848. [Google Scholar] [CrossRef]
- Kahleova, H.; Tura, A.; Hill, M.; Holubkov, R.; Barnard, N.D. A Plant-Based Dietary Intervention Improves Beta-Cell Function and Insulin Resistance in Overweight Adults: A 16-Week Randomized Clinical Trial. Nutrients 2018, 10, 189. [Google Scholar] [CrossRef] [PubMed]
- Yokoyama, Y.; Levin, S.M.; Barnard, N.D. Association between plant-based diets and plasma lipids: A systematic review and meta-analysis. Nutr. Rev. 2017, 75, 683–698. [Google Scholar] [CrossRef] [PubMed]
- Choi, E.Y.; Allen, K.; McDonnough, M.; Massera, D.; Ostfeld, R.J. A plant-based diet and heart failure: Case report and literature review. J. Geriatr. Cardiol. 2017, 14, 375–378. [Google Scholar] [CrossRef]
- Najjar, R.S.; Montgomery, B.D. A defined, plant-based diet as a potential therapeutic approach in the treatment of heart failure: A clinical case series. Complement. Ther. Med. 2019, 45, 211–214. [Google Scholar] [CrossRef]
- Appleby, P.N.; Davey, G.K.; Key, T.J. Hypertension and blood pressure among meat eaters, fish eaters, vegetarians and vegans in EPIC-Oxford. Public Health Nutr. 2002, 5, 645–654. [Google Scholar] [CrossRef]
- Sebastian, S.A.; Padda, I.; Johal, G. Long-term impact of mediterranean diet on cardiovascular disease prevention: A systematic review and meta-analysis of randomized controlled trials. Curr. Probl. Cardiol. 2024, 49, 102509. [Google Scholar] [CrossRef]
- Ordovas, J.M.; Ferguson, L.R.; Tai, E.S.; Mathers, J.C. Personalised nutrition and health. BMJ 2018, 361, bmj.k2173. [Google Scholar] [CrossRef]
- Berry, S.E.; Valdes, A.M.; Drew, D.A.; Asnicar, F.; Mazidi, M.; Wolf, J.; Capdevila, J.; Hadjigeorgiou, G.; Davies, R.; Al Khatib, H.; et al. Human postprandial responses to food and potential for precision nutrition. Nat. Med. 2020, 26, 964–973. [Google Scholar] [CrossRef]
- van de Rest, O.; Schutte, B.A.M.; Deelen, J.; Stassen, S.A.M.; van den Akker, E.B.; van Heemst, D.; Dibbets-Schneider, P.; van Dipten-van der Veen, R.A.; Kelderman, M.; Hankemeier, T.; et al. Metabolic effects of a 13-weeks lifestyle intervention in older adults: The Growing Old Together Study. Aging 2016, 8, 111–126. [Google Scholar] [CrossRef] [PubMed]
- Chang, E.; Kim, Y. Vitamin D Insufficiency Exacerbates Adipose Tissue Macrophage Infiltration and Decreases AMPK/SIRT1 Activity in Obese Rats. Nutrients 2017, 9, 338. [Google Scholar] [CrossRef] [PubMed]
- Ruderman, N.B.; Xu, X.J.; Nelson, L.; Cacicedo, J.M.; Saha, A.K.; Lan, F.; Ido, Y. AMPK and SIRT1: A long-standing partnership? Am. J. Physiol. Endocrinol. Metab. 2010, 298, E751–E760. [Google Scholar] [CrossRef] [PubMed]
- Cazzola, R.; Della Porta, M.; Piuri, G.; Maier, J.A. Magnesium: A Defense Line to Mitigate Inflammation and Oxidative Stress in Adipose Tissue. Antioxidants 2024, 13, 893. [Google Scholar] [CrossRef]
- Manna, P.; Achari, A.E.; Jain, S.K. Vitamin D supplementation inhibits oxidative stress and upregulate SIRT1/AMPK/GLUT4 cascade in high glucose-treated 3T3L1 adipocytes and in adipose tissue of high fat diet-fed diabetic mice. Arch. Biochem. Biophys. 2017, 615, 22–34. [Google Scholar] [CrossRef]
- Vijver, M.A.T.; Bomer, N.; Verdonk, R.C.; van der Meer, P.; van Veldhuisen, D.J.; Dams, O.C. Micronutrient Deficiencies in Heart Failure and Relationship with Exocrine Pancreatic Insufficiency. Nutrients 2024, 17, 56. [Google Scholar] [CrossRef]
- van den Broek, T.J.; Kremer, B.H.A.; Marcondes Rezende, M.; Hoevenaars, F.P.M.; Weber, P.; Hoeller, U.; van Ommen, B.; Wopereis, S. The Impact of Micronutrient Status on Health: Correlation Network Analysis to Understand the Role of Micronutrients in Metabolic-Inflammatory Processes Regulating Homeostasis and Phenotypic Flexibility. Genes Nutr. 2017, 12, 5. [Google Scholar] [CrossRef]
- Marcotorchino, J.; Gouranton, E.; Romier, B.; Tourniaire, F.; Astier, J.; Malezet, C.; Amiot, M.; Landrier, J. Vitamin D Reduces the Inflammatory Response and Restores Glucose Uptake in Adipocytes. Mol. Nutr. Food Res. 2012, 56, 1771–1782. [Google Scholar] [CrossRef]
- Manna, P.; Jain, S.K. Vitamin D Up-Regulates Glucose Transporter 4 (GLUT4) Translocation and Glucose Utilization Mediated by Cystathionine-γ-Lyase (CSE) Activation and H2S Formation in 3T3L1 Adipocytes. J. Biol. Chem. 2012, 287, 42324–42332. [Google Scholar] [CrossRef]
- Lorente-Cebrián, S.; Eriksson, A.; Dunlop, T.; Mejhert, N.; Dahlman, I.; Åström, G.; Sjölin, E.; Wåhlén, K.; Carlberg, C.; Laurencikiene, J.; et al. Differential Effects of 1α,25-Dihydroxycholecalciferol on MCP-1 and Adiponectin Production in Human White Adipocytes. Eur. J. Nutr. 2012, 51, 335–342. [Google Scholar] [CrossRef]
- Ding, C.; Wilding, J.P.H.; Bing, C. 1,25-Dihydroxyvitamin D3 Protects against Macrophage-Induced Activation of NFκB and MAPK Signalling and Chemokine Release in Human Adipocytes. PLoS ONE 2013, 8, e61707. [Google Scholar] [CrossRef] [PubMed]
- Hu, F.; Yan, L.; Lu, S.; Ma, W.; Wang, Y.; Wei, Y.; Yan, X.; Zhao, X.; Chen, Z.; Wang, Z.; et al. Effects of 1, 25-Dihydroxyvitamin D3 on Experimental Autoimmune Myocarditis in Mice. Cell. Physiol. Biochem. 2016, 38, 2219–2229. [Google Scholar] [CrossRef] [PubMed]
- Lontchi-Yimagou, E.; Kang, S.; Goyal, A.; Zhang, K.; You, J.Y.; Carey, M.; Jain, S.; Bhansali, S.; Kehlenbrink, S.; Guo, P.; et al. Insulin-Sensitizing Effects of Vitamin D Repletion Mediated by Adipocyte Vitamin D Receptor: Studies in Humans and Mice. Mol. Metab. 2020, 42, 101095. [Google Scholar] [CrossRef] [PubMed]
- Jeffery, L.E.; Qureshi, O.S.; Gardner, D.; Hou, T.Z.; Briggs, Z.; Soskic, B.; Baker, J.; Raza, K.; Sansom, D.M. Vitamin D Antagonises the Suppressive Effect of Inflammatory Cytokines on CTLA-4 Expression and Regulatory Function. PLoS ONE 2015, 10, e0131539. [Google Scholar] [CrossRef]
- Ozer, P.; Emet, S.; Karaayvaz, E.; Elitok, A.; Bilge, A.; Adalet, K.; Oncul, A. Silent Myocardial Dysfunction in Vitamin D Deficiency. Arch. Med Sci.—Atheroscler. Dis. 2020, 5, 153–162. [Google Scholar] [CrossRef]
- Hosseini Dastgerdi, A.; Ghanbari Rad, M.; Soltani, N. The Therapeutic Effects of Magnesium in Insulin Secretion and Insulin Resistance. Adv. Biomed. Res. 2022, 11, 54. [Google Scholar] [CrossRef]
- Oost, L.J.; Kurstjens, S.; Ma, C.; Hoenderop, J.G.J.; Tack, C.J.; de Baaij, J.H.F. Magnesium Increases Insulin-Dependent Glucose Uptake in Adipocytes. Front. Endocrinol. 2022, 13, 986616. [Google Scholar] [CrossRef]
- Cahill, F.; Shahidi, M.; Shea, J.; Wadden, D.; Gulliver, W.; Randell, E.; Vasdev, S.; Sun, G. High Dietary Magnesium Intake Is Associated with Low Insulin Resistance in the Newfoundland Population. PLoS ONE 2013, 8, e58278. [Google Scholar] [CrossRef]
- Liu, M.; Dudley, S.C. Magnesium, Oxidative Stress, Inflammation, and Cardiovascular Disease. Antioxidants 2020, 9, 907. [Google Scholar] [CrossRef]
- Liu, M.; Liu, H.; Feng, F.; Xie, A.; Kang, G.; Zhao, Y.; Hou, C.R.; Zhou, X.; Dudley, S.C. Magnesium Deficiency Causes a Reversible, Metabolic, Diastolic Cardiomyopathy. J. Am. Heart Assoc. 2021, 10, e020205. [Google Scholar] [CrossRef]
- Ahn, B.-I.; Kim, M.J.; Koo, H.S.; Seo, N.; Joo, N.-S.; Kim, Y.-S. Serum Zinc Concentration Is Inversely Associated with Insulin Resistance but Not Related with Metabolic Syndrome in Nondiabetic Korean Adults. Biol. Trace Element Res. 2014, 160, 169–175. [Google Scholar] [CrossRef] [PubMed]
- de Luis, D.A.; Pacheco, D.; Izaola, O.; Terroba, M.C.; Cuellar, L.; Cabezas, G. Micronutrient Status in Morbidly Obese Women before Bariatric Surgery. Surg. Obes. Relat. Dis. 2013, 9, 323–327. [Google Scholar] [CrossRef] [PubMed]
- Li, H.-T.; Jiao, M.; Chen, J.; Liang, Y. Roles of Zinc and Copper in Modulating the Oxidative Refolding of Bovine Copper, Zinc Superoxide Dismutase. Acta Biochim. Biophys. Sin. 2010, 42, 183–194. [Google Scholar] [CrossRef] [PubMed]
- Franco, C.; Canzoniero, L.M.T. Zinc Homeostasis and Redox Alterations in Obesity. Front. Endocrinol. 2024, 14, 1273177. [Google Scholar] [CrossRef]
- Liu, M.-J.; Bao, S.; Bolin, E.R.; Burris, D.L.; Xu, X.; Sun, Q.; Killilea, D.W.; Shen, Q.; Ziouzenkova, O.; Belury, M.A.; et al. Zinc Deficiency Augments Leptin Production and Exacerbates Macrophage Infiltration into Adipose Tissue in Mice Fed a High-Fat Diet1–3. J. Nutr. 2013, 143, 1036–1045. [Google Scholar] [CrossRef]
- Nazari, M.; Nikbaf-Shandiz, M.; Pashayee-Khamene, F.; Bagheri, R.; Goudarzi, K.; Hosseinnia, N.V.; Dolatshahi, S.; Omran, H.S.; Amirani, N.; Ashtary-larky, D.; et al. Zinc Supplementation in Individuals with Prediabetes and Type 2 Diabetes: A GRADE-Assessed Systematic Review and Dose-Response Meta-Analysis. Biol. Trace Elem. Res. 2024, 202, 2966–2990. [Google Scholar] [CrossRef]
- Rezaei, S.M.A.; Mohammadi, F.; Eftekhari, M.H.; Ejtehadi, F.; Ghaem, H.; Mohammadipoor, N. The Effects of Zinc Supplementation on the Metabolic Factors in Patients with Non-Alcoholic Fatty Liver Disease: A Randomized, Double-Blinded, Placebo-Controlled Clinical Trial. BMC Nutr. 2023, 9, 138. [Google Scholar] [CrossRef]
- Rosenblum, H.; Bikdeli, B.; Wessler, J.; Gupta, A.; Jacoby, D.L. Zinc Deficiency as a Reversible Cause of Heart Failure. Tex. Heart Inst. J. 2020, 47, 152–154. [Google Scholar] [CrossRef]
- Foster, M.; Samman, S. Zinc and Redox Signaling: Perturbations Associated with Cardiovascular Disease and Diabetes Mellitus. Antioxid. Redox Signal. 2010, 13, 1549–1573. [Google Scholar] [CrossRef]
- Ozyıldırım, S.; Baltaci, A.K.; Sahna, E.; Mogulkoc, R. Effects of Chronic and Acute Zinc Supplementation on Myocardial Ischemia-Reperfusion Injury in Rats. Biol. Trace Elem. Res. 2017, 178, 64–70. [Google Scholar] [CrossRef]
- Zimiao, C.; Dongdong, L.; Shuoping, C.; Peng, Z.; Fan, Z.; Rujun, C.; Xiaohua, G. Correlations Between Iron Status and Body Composition in Patients with Type 2 Diabetes Mellitus. Front. Nutr. 2022, 9, 911860. [Google Scholar] [CrossRef]
- Ameka, M.K.; Beavers, W.N.; Shaver, C.M.; Ware, L.B.; Kerchberger, V.E.; Schoenfelt, K.Q.; Sun, L.; Koyama, T.; Skaar, E.P.; Becker, L.; et al. An Iron Refractory Phenotype in Obese Adipose Tissue Macrophages Leads to Adipocyte Iron Overload. Int. J. Mol. Sci. 2022, 23, 7417. [Google Scholar] [CrossRef] [PubMed]
- Pihan-Le Bars, F.; Bonnet, F.; Loréal, O.; Le Loupp, A.-G.; Ropert, M.; Letessier, E.; Prieur, X.; Bach, K.; Deugnier, Y.; Fromenty, B.; et al. Indicators of Iron Status Are Correlated with Adiponectin Expression in Adipose Tissue of Patients with Morbid Obesity. Diabetes Metab. 2016, 42, 105–111. [Google Scholar] [CrossRef] [PubMed]
- Dongiovanni, P.; Ruscica, M.; Rametta, R.; Recalcati, S.; Steffani, L.; Gatti, S.; Girelli, D.; Cairo, G.; Magni, P.; Fargion, S.; et al. Dietary Iron Overload Induces Visceral Adipose Tissue Insulin Resistance. Am. J. Pathol. 2013, 182, 2254–2263. [Google Scholar] [CrossRef] [PubMed]
- Hoes, M.F.; Grote Beverborg, N.; Kijlstra, J.D.; Kuipers, J.; Swinkels, D.W.; Giepmans, B.N.G.; Rodenburg, R.J.; van Veldhuisen, D.J.; de Boer, R.A.; van der Meer, P. Iron Deficiency Impairs Contractility of Human Cardiomyocytes through Decreased Mitochondrial Function. Eur. J. Heart Fail. 2018, 20, 910–919. [Google Scholar] [CrossRef]
- Beavers, C.J.; Ambrosy, A.P.; Butler, J.; Davidson, B.T.; Gale, S.E.; PIÑA, I.L.; Mastoris, I.; Reza, N.; Mentz, R.J.; Lewis, G.D. Iron Deficiency in Heart Failure: A Scientific Statement from the Heart Failure Society of America. J. Card. Fail. 2023, 29, 1059–1077. [Google Scholar] [CrossRef]
- Zhang, H.; Jamieson, K.L.; Grenier, J.; Nikhanj, A.; Tang, Z.; Wang, F.; Wang, S.; Seidman, J.G.; Seidman, C.E.; Thompson, R.; et al. Myocardial Iron Deficiency and Mitochondrial Dysfunction in Advanced Heart Failure in Humans. J. Am. Heart Assoc. 2022, 11, e022853. [Google Scholar] [CrossRef]
- Mentz, R.J.; Garg, J.; Rockhold, F.W.; Butler, J.; De Pasquale, C.G.; Ezekowitz, J.A.; Lewis, G.D.; O’Meara, E.; Ponikowski, P.; Troughton, R.W.; et al. Ferric Carboxymaltose in Heart Failure with Iron Deficiency. N. Engl. J. Med. 2023, 389, 975–986. [Google Scholar] [CrossRef]
- Yan, X.; Xie, Y.; Liu, H.; Huang, M.; Yang, Z.; An, D.; Jiang, G. Iron Accumulation and Lipid Peroxidation: Implication of Ferroptosis in Diabetic Cardiomyopathy. Diabetol. Metab. Syndr. 2023, 15, 161. [Google Scholar] [CrossRef]
- García, O.P.; Long, K.Z.; Rosado, J.L. Impact of micronutrient deficiencies on obesity. Nutr. Rev. 2009, 67, 559–572. [Google Scholar] [CrossRef]
- Via, M. The malnutrition of obesity: Micronutrient deficiencies that promote diabetes. ISRN Endocrinol. 2012, 2012, 103472. [Google Scholar] [CrossRef] [PubMed]
- DiNicolantonio, J.J.; O’Keefe, J.H.; Wilson, W. Subclinical magnesium deficiency: A principal driver of cardiovascular disease and a public health crisis. Open Heart 2018, 5, e000668. [Google Scholar] [CrossRef] [PubMed]
- Cheung, M.M.; Dall, R.D.; Shewokis, P.A.; Altasan, A.; Volpe, S.L.; Amori, R.; Singh, H.; Sukumar, D. The effect of combined magnesium and vitamin D supplementation on vitamin D status, systemic inflammation, and blood pressure: A randomized double-blinded controlled trial. Nutrition 2022, 99–100, 111674. [Google Scholar] [CrossRef] [PubMed]
- Calcaterra, V.; Verduci, E.; Milanta, C.; Agostinelli, M.; Todisco, C.F.; Bona, F.; Dolor, J.; La Mendola, A.; Tosi, M.; Zuccotti, G. Micronutrient Deficiency in Children and Adolescents with Obesity-A Narrative Review. Children 2023, 10, 695. [Google Scholar] [CrossRef]
- Muoio, D.M. Metabolic Inflexibility: When Mitochondrial Indecision Leads to Metabolic Gridlock. Cell 2014, 159, 1253–1262. [Google Scholar] [CrossRef]
- Goldberg, I.J.; Trent, C.M.; Schulze, P.C. Lipid Metabolism and Toxicity in the Heart. Cell Metab. 2012, 15, 805–812. [Google Scholar] [CrossRef]
- Banerjee, S.; Peterson, L.R. Myocardial Metabolism and Cardiac Performance in Obesity and Insulin Resistance. Curr. Cardiol. Rep. 2007, 9, 143–149. [Google Scholar] [CrossRef]
- Oneglia, A.P.; Szczepaniak, L.S.; Jaffery, M.F.; Cipher, D.J.; McDonald, J.G.; Haykowsky, M.J.; Moreau, K.L.; Clegg, D.J.; Zaha, V.; Nelson, M.D. Myocardial Steatosis Impairs Left Ventricular Diastolic–Systolic Coupling in Healthy Humans. J. Physiol. 2023, 601, 1371–1382. [Google Scholar] [CrossRef]
- Geng, J.; Zhang, X.; Guo, Y.; Wen, H.; Guo, D.; Liang, Q.; Pu, S.; Wang, Y.; Liu, M.; Li, Z.; et al. Moderate-Intensity Interval Exercise Exacerbates Cardiac Lipotoxicity in High-Fat, High-Calories Diet-Fed Mice. Nat. Commun. 2025, 16, 613. [Google Scholar] [CrossRef]
- Geladari, E.V.; Kounatidis, D.; Christodoulatos, G.S.; Psallida, S.; Pavlou, A.; Geladari, C.V.; Sevastianos, V.; Dalamaga, M.; Vallianou, N.G. Ultra-Processed Foods and Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): What Is the Evidence So Far? Nutrients 2025, 17, 2098. [Google Scholar] [CrossRef]
- Bauer, J.; Ayala, F.O.; Marcadenti, A.; Machado, R.H.V.; Cristina Bersch-Ferreira, Â.; Moreira, M.F.S.; Beretta, M.V.; Feoli, A.M.P.; Busnello, F.M. Consumption of Ultra-Processed Foods and Metabolic Parameters in Type 2 Diabetes Mellitus: A Cross-Sectional Study. Int. J. Environ. Res. Public Health 2025, 22, 1275. [Google Scholar] [CrossRef] [PubMed]
- Choi, Y.; Shin, H.; Tang, Z.; Yeh, Y.; Ma, Y.; Kadegowda, A.K.G.; Wang, H.; Jiang, L.; Arya, R.K.; Chen, L.; et al. Adipose Lipolysis Regulates Cardiac Glucose Uptake and Function in Mice under Cold Stress. Int. J. Mol. Sci. 2021, 22, 13361. [Google Scholar] [CrossRef] [PubMed]
- Szűcs, G.; Sója, A.; Péter, M.; Sárközy, M.; Bruszel, B.; Siska, A.; Földesi, I.; Szabó, Z.; Janáky, T.; Vígh, L.; et al. Prediabetes Induced by Fructose-Enriched Diet Influences Cardiac Lipidome and Proteome and Leads to Deterioration of Cardiac Function Prior to the Development of Excessive Oxidative Stress and Cell Damage. Oxidative Med. Cell. Longev. 2019, 2019, 1–21. [Google Scholar] [CrossRef] [PubMed]
- Velagic, A.; Li, M.; Deo, M.; Li, J.C.; Kiriazis, H.; Donner, D.G.; Anderson, D.; De Blasio, M.J.; Woodman, O.L.; Kemp-Harper, B.K.; et al. A High-Sucrose Diet Exacerbates the Left Ventricular Phenotype in a High Fat-Fed Streptozotocin Rat Model of Diabetic Cardiomyopathy. Am. J. Physiol. Circ. Physiol. 2023, 324, H241–H257. [Google Scholar] [CrossRef]
- Tettamanzi, F.; Bagnardi, V.; Louca, P.; Nogal, A.; Monti, G.S.; Mambrini, S.P.; Lucchetti, E.; Maestrini, S.; Mazza, S.; Rodriguez-Mateos, A.; et al. A High Protein Diet Is More Effective in Improving Insulin Resistance and Glycemic Variability Compared to a Mediterranean Diet—A Cross-Over Controlled Inpatient Dietary Study. Nutrients 2021, 13, 4380. [Google Scholar] [CrossRef]
- Kotzé-Hörstmann, L.; Cois, A.; Johnson, R.; Mabasa, L.; Shabalala, S.; Van Jaarsveld, P.J.; Sadie-Van Gijsen, H. Characterization and Comparison of the Divergent Metabolic Consequences of High-Sugar and High-Fat Diets in Male Wistar Rats. Front. Physiol. 2022, 13, 904366. [Google Scholar] [CrossRef]
- Victorio, J.A.; Guizoni, D.M.; Freitas, I.N.; Araujo, T.R.; Davel, A.P. Effects of High-Fat and High-Fat/High-Sucrose Diet-Induced Obesity on PVAT Modulation of Vascular Function in Male and Female Mice. Front. Pharmacol. 2021, 12, 720224. [Google Scholar] [CrossRef]
- Pointke, M.; Strenge, F.; Piotrowski, D.; Matteikat, A.; Meyhöfer, S.; Meyhöfer, S.M.; Chamorro, R.; Wilms, B. Short-Term High-Fat and High-Carb Diet Effects on Glucose Metabolism and Hedonic Regulation in Young Healthy Men. Front. Nutr. 2024, 11, 1469230. [Google Scholar] [CrossRef]
- Kim, Y.; Lim, J.H.; Kim, E.N.; Hong, Y.A.; Park, H.-J.; Chung, S.; Choi, B.S.; Kim, Y.-S.; Park, J.Y.; Kim, H.W.; et al. Adiponectin Receptor Agonist Ameliorates Cardiac Lipotoxicity via Enhancing Ceramide Metabolism in Type 2 Diabetic Mice. Cell Death Dis. 2022, 13, 282. [Google Scholar] [CrossRef]
- Valero-Muñoz, M.; Cooper, H.L.; Li, S.; Saw, E.L.; Wilson, R.M.; Kusminski, C.M.; Scherer, P.E.; Sam, F. Metabolic Dysregulation in the Heart in Obesity-Associated HFpEF. Front. Cardiovasc. Med. 2025, 12, 1678992. [Google Scholar] [CrossRef]
- Chung, Y.-H.; Lu, K.-Y.; Chiu, S.-C.; Lo, C.-J.; Hung, L.-M.; Huang, J.-P.; Cheng, M.-L.; Wang, C.-H.; Tsai, C.-K.; Lin, Y.-C.; et al. Early Imaging Biomarker of Myocardial Glucose Adaptations in High-Fat-Diet-Induced Insulin Resistance Model by Using 18 F-FDG PET and [U-13C]Glucose Nuclear Magnetic Resonance Tracer. Contrast Media Mol. Imaging 2018, 2018, 8751267. [Google Scholar] [CrossRef] [PubMed]
- Minato-Inokawa, S.; Tsuboi-Kaji, A.; Honda, M.; Takeuchi, M.; Kitaoka, K.; Kurata, M.; Wu, B.; Kazumi, T.; Fukuo, K. Low Muscle Mass Is Associated with Low Insulin Sensitivity, Impaired Pancreatic β Cell Function, and High Glucose Excursion in Nondiabetic Nonobese Japanese Women. Metab. Open 2024, 23, 100306. [Google Scholar] [CrossRef] [PubMed]
- Fuglsang-Nielsen, R.; Rakvaag, E.; Langdahl, B.; Knudsen, K.E.B.; Hartmann, B.; Holst, J.J.; Hermansen, K.; Gregersen, S. Effects of Whey Protein and Dietary Fiber Intake on Insulin Sensitivity, Body Composition, Energy Expenditure, Blood Pressure, and Appetite in Subjects with Abdominal Obesity. Eur. J. Clin. Nutr. 2021, 75, 611–619. [Google Scholar] [CrossRef] [PubMed]
- Chen, C.-N.; Hsu, K.-J.; Chien, K.-Y.; Chen, J.-J. Effects of Combined High-Protein Diet and Exercise Intervention on Cardiometabolic Health in Middle-Aged Obese Adults: A Randomized Controlled Trial. Front. Cardiovasc. Med. 2021, 8, 705282. [Google Scholar] [CrossRef]
- Zhang, J.; Pivovarova-Ramich, O.; Kabisch, S.; Markova, M.; Hornemann, S.; Sucher, S.; Rohn, S.; Machann, J.; Pfeiffer, A.F.H. High Protein Diets Improve Liver Fat and Insulin Sensitivity by Prandial but Not Fasting Glucagon Secretion in Type 2 Diabetes. Front. Nutr. 2022, 9, 808346. [Google Scholar] [CrossRef]
- Choi, B.H.; Hyun, S.; Koo, S.-H. The Role of BCAA Metabolism in Metabolic Health and Disease. Exp. Mol. Med. 2024, 56, 1552–1559. [Google Scholar] [CrossRef]
- Oh, A.; Okazaki, R.; Sam, F.; Valero-Muñoz, M. Heart Failure with Preserved Ejection Fraction and Adipose Tissue: A Story of Two Tales. Front. Cardiovasc. Med. 2019, 6, 110. [Google Scholar] [CrossRef]
- Kita, S.; Maeda, N.; Shimomura, I. Interorgan Communication by Exosomes, Adipose Tissue, and Adiponectin in Metabolic Syndrome. J. Clin. Investig. 2019, 129, 4041–4049. [Google Scholar] [CrossRef]
- Wang, L.; Zhang, J.-J.; Wang, S.-S.; Li, L. Mechanism of Adipose-Derived Mesenchymal Stem Cell Exosomes in the Treatment of Heart Failure. World J. Stem Cells 2023, 15, 897–907. [Google Scholar] [CrossRef]
- Guo, B.; Zhuang, T.-T.; Li, C.-C.; Li, F.; Shan, S.-K.; Zheng, M.-H.; Xu, Q.-S.; Wang, Y.; Lei, L.-M.; Tang, K.-X.; et al. MiRNA-132/212 Encapsulated by Adipose Tissue-Derived Exosomes Worsen Atherosclerosis Progression. Cardiovasc. Diabetol. 2024, 23, 331. [Google Scholar] [CrossRef]
- Chen, L.; Amraee, F.; Sadegh-Nejadi, S.; Saberian, M.; Ghahari, S.A.; Miao, X.; Lisco, G.; Afrisham, R. Molecular Mechanisms Linking Adipose Tissue-Derived Small Extracellular Vesicles/Exosomes to the Development or Amelioration of Obesity, Insulin Resistance, and Diabetes-Related Complications. Eur. J. Med Res. 2025, 30, 1049. [Google Scholar] [CrossRef]
- Hatamnejad, M.R.; Medzikovic, L.; Dehghanitafti, A.; Rahman, B.; Vadgama, A.; Eghbali, M. Role of Gut Microbial Metabolites in Ischemic and Non-Ischemic Heart Failure. Int. J. Mol. Sci. 2025, 26, 2242. [Google Scholar] [CrossRef] [PubMed]
- Snelson, M.; Muralitharan, R.R.; Liu, C.-F.; Markó, L.; Forslund, S.K.; Marques, F.Z.; Tang, W.H.W. Gut-Heart Axis: The Role of Gut Microbiota and Metabolites in Heart Failure. Circ. Res. 2025, 136, 1382–1406. [Google Scholar] [CrossRef] [PubMed]
- Cani, P.D.; Van Hul, M. Gut microbiota in overweight and obesity: Crosstalk with adipose tissue. Nat. Rev. Gastroenterol. Hepatol. 2024, 21, 164–183. [Google Scholar] [CrossRef] [PubMed]
- Mauney, E.E.; Wibowo, M.C.; Tseng, Y.H.; Kostic, A.D. Adipose tissue-gut microbiome crosstalk in inflammation and thermogenesis. Trends Endocrinol. Metab. 2025, 36, 721–732. [Google Scholar] [CrossRef]
- Pan, Y.; Liu, Y.; Gao, Y.; Xing, Y.; Si, Y.; Liu, F.; Jiang, X. Adipocyte-secreted exosomal microRNA-34a inhibits M2 macrophage polarization to promote obesity-induced adipose inflammation. J. Clin. Investig. 2019, 129, 834–849. [Google Scholar] [CrossRef]
- Thomou, T.; Mori, M.A.; Dreyfuss, J.M.; Konishi, M.; Sakaguchi, M.; Wolfrum, C.; Rao, T.N.; Winnay, J.N.; Garcia-Martin, R.; Grinspoon, S.K.; et al. Adipose-derived circulating miRNAs regulate gene expression in other tissues. Nature 2017, 542, 450–455. [Google Scholar] [CrossRef]
- Virtue, A.T.; McCright, S.J.; Wright, J.M.; Jimenez, M.T.; Mowel, W.K.; Kotzin, J.J.; Joannas, L.; Basavappa, M.G.; Spencer, S.P.; Clark, M.L.; et al. The gut microbiota regulates white adipose tissue inflammation and obesity via a family of microRNAs. Sci. Transl. Med. 2019, 11, eaav1892. [Google Scholar] [CrossRef]
- Gan, L.; Xie, D.; Liu, J.; Bond Lau, W.; Christopher, T.A.; Lopez, B.; Zhang, L.; Gao, E.; Koch, W.J.; Ma, X.L.; et al. Small extracellular microvesicles mediated pathological communications between dysfunctional adipocytes and cardiomyocytes as a novel mechanism exacerbating ischemia/reperfusion injury in diabetic mice. Circulation 2020, 141, 968–983. [Google Scholar] [CrossRef]
- Li, Z.; Wu, Z.; Yan, J.; Liu, H.; Liu, Q.; Deng, Y.; Ou, C.; Chen, M. Gut microbe-derived metabolite trimethylamine N-oxide induces cardiac hypertrophy and fibrosis. Lab. Investig. 2019, 99, 346–357. [Google Scholar] [CrossRef]
- Diez-Roda, P.; Perez-Navarro, E.; Garcia-Martin, R. Adipose Tissue as a Major Launch Spot for Circulating Extracellular Vesicle-Carried MicroRNAs Coordinating Tissue and Systemic Metabolism. Int. J. Mol. Sci. 2024, 25, 13488. [Google Scholar] [CrossRef] [PubMed]
- Gan, L.; Guo, X.; Dong, S.; Sun, C. The Biology of Exosomes and Exosomal Non-Coding RNAs in Cardiovascular Diseases. Front. Pharmacol. 2025, 16, 1529375. [Google Scholar] [CrossRef] [PubMed]
- Antoniades, C.; Tousoulis, D.; Vavlukis, M.; Fleming, I.; Duncker, D.J.; Eringa, E.; Manfrini, O.; Antonopoulos, A.S.; Oikonomou, E.; Padró, T.; et al. Perivascular Adipose Tissue as a Source of Therapeutic Targets and Clinical Biomarkers. Eur. Heart J. 2023, 44, 3827–3844. [Google Scholar] [CrossRef] [PubMed]
- Lei, X.; Qiu, S.; Yang, G.; Wu, Q. Adiponectin and Metabolic Cardiovascular Diseases: Therapeutic Opportunities and Challenges. Genes Dis. 2023, 10, 1525–1536. [Google Scholar] [CrossRef]
- Morselli, F.; Pierce, I.; Webber, M.; Falconer, D.; Hughes, A.D.; Orini, M.; Lambiase, P.D.; Moon, J.C.; Captur, G. Imaging Cardiac Fat by Cardiovascular Magnetic Resonance—A State-of-the Art Review. Magn. Reson. Imaging 2025, 124, 110529. [Google Scholar] [CrossRef]
- Frankl, J.; Sherwood, A.; Clegg, D.J.; Scherer, P.E.; Öz, O.K. Imaging Metabolically Active Fat: A Literature Review and Mechanistic Insights. Int. J. Mol. Sci. 2019, 20, 5509. [Google Scholar] [CrossRef]
- Yang, S.; Wu, Y. Ceramide Metabolism and Cardiovascular Risk Factors: Insights into Therapeutic Strategies. Front. Cardiovasc. Med. 2025, 12, 1656113. [Google Scholar] [CrossRef]
- Delcheva, G.; Stefanova, K.; Stankova, T. Ceramides—Emerging Biomarkers of Lipotoxicity in Obesity, Diabetes, Cardiovascular Diseases, and Inflammation. Diseases 2024, 12, 195. [Google Scholar] [CrossRef]
- Xie, H.; Zhang, B.; Xie, M.; Li, T. Circulating Metabolic Signatures of Heart Failure in Precision Cardiology. Precis. Clin. Med. 2023, 6, pbad005. [Google Scholar] [CrossRef]
- Xu, W.; Luo, Y.; Yin, J.; Huang, M.; Luo, F. Targeting AMPK Signaling by Polyphenols: A Novel Strategy for Tackling Aging. Food Funct. 2023, 14, 56–73. [Google Scholar] [CrossRef]
- Sahoo, S.; Losordo, D.W. Exosomes and Cardiac Repair After Myocardial Infarction. Circ. Res. 2014, 114, 333–344. [Google Scholar] [CrossRef]
- Zhou, F.; Li, K.; Yang, K. Adipose-Derived Stem Cell Exosomes and Related MicroRNAs in Atherosclerotic Cardiovascular Disease. J. Cardiovasc. Transl. Res. 2023, 16, 453–462. [Google Scholar] [CrossRef]
- Bakkar, N.Z.; AlZaim, I.; El-Yazbi, A.F. Depot-specific adipose tissue modulation by SGLT2 inhibitors and GLP1 agonists mediates their cardioprotective effects in metabolic disease. Clin. Sci. 2022, 136, 1631–1651. [Google Scholar] [CrossRef]
- Bao, Y.; Hu, Y.; Shi, M.; Zhao, Z. SGLT2 inhibitors reduce epicardial adipose tissue more than GLP-1 agonists or exercise interventions in patients with type 2 diabetes mellitus and/or obesity: A systematic review and network meta-analysis. Diabetes Obes. Metab. 2025, 27, 1096–1112. [Google Scholar] [CrossRef]
- Morciano, C.; Gugliandolo, S.; Capece, U.; Di Giuseppe, G.; Mezza, T.; Ciccarelli, G.; Soldovieri, L.; Brunetti, M.; Avolio, A.; Splendore, A.; et al. SGLT2 inhibition and adipose tissue metabolism: Current outlook and perspectives. Cardiovasc. Diabetol. 2024, 23, 449. [Google Scholar] [CrossRef]
- Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef]
- Szczepaniak, L.S.; Victor, R.G.; Orber, L.; Peshock, R.M.; Grundy, S.M. Myocardial triglycerides and systolic function in humans: In vivo evaluation by localized proton spectroscopy and cardiac imaging. Magn. Reson. Med. 2003, 49, 417–423. [Google Scholar] [CrossRef]
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. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).