Next Article in Journal
Some Properties of the C. elegans Multicopper Oxidase F21D5.3, an Ortholog of Human Ceruloplasmin
Next Article in Special Issue
Anti-Obesity Effects of Rosa rugosa Thunb. Flower Bud Extracts on Lipid Metabolism Regulation in 3T3-L1 Adipocytes and Sprague Dawley Rats
Previous Article in Journal
Targeting the IL-23 Receptor Gene: A Promising Approach in Inflammatory Bowel Disease Treatment
Previous Article in Special Issue
Cholesterol and SREBP2 Dynamics During Spermatogenesis Stages in Rabbits: Effects of High-Fat Diet and Protective Role of Extra Virgin Olive Oil
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

High-Fat Diet and Metabolic Diseases: A Comparative Analysis of Sex-Dependent Responses and Mechanisms

School of Sports, Shenzhen University, Shenzhen 518060, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(10), 4777; https://doi.org/10.3390/ijms26104777
Submission received: 11 April 2025 / Revised: 10 May 2025 / Accepted: 14 May 2025 / Published: 16 May 2025
(This article belongs to the Special Issue High Fat Diet Metabolism and Diseases)

Abstract

Sex differences in metabolic disorders and susceptibility to chronic diseases induced by a high-fat diet (HFD) exhibit significant dimorphic characteristics. A long-standing male-centric bias in medical research and healthcare, predominantly focused on male physiological traits, has hindered the precise treatment of metabolic diseases in female patients. A comprehensive understanding of sex differences in metabolic health and their underlying mechanisms is crucial for advancing personalized health promotion and precision medicine. This review systematically elucidates sex-specific manifestations in high-fat diet-associated metabolic disorders: males predominantly develop visceral adiposity, insulin resistance, and dyslipidemia, accompanied by a significantly elevated risk of cardiovascular and metabolic syndromes. Premenopausal females maintain metabolic homeostasis through the estrogen-mediated optimization of glucose and lipid metabolism and oxidative stress buffering mechanisms, whereas postmenopausal-phase females experience dramatic metabolic vulnerability due to z loss of protective barriers. Furthermore, we emphasize multidimensional mechanistic interpretations of metabolic sexual dimorphism from perspectives including sex chromosome complement, sex hormone signaling pathways, epigenetic regulation, gut microbiota composition, and neuroendocrine dimorphism. This work provides critical theoretical foundations for rectifying unisex research paradigms and optimizing sex-specific early warning systems and precision therapeutic strategies for metabolic disorders.

1. Introduction

Global economic expansion and aggressive food marketing strategies have precipitated profound shifts in dietary patterns worldwide. Notably, the consumption of energy-dense foods rich in saturated fatty acids, trans-fatty acids, and ultra-processed ingredients has surged across developed nations and rapidly urbanizing low-to-middle-income countries [1,2,3,4]. Chronic HFD exposure, particularly involving saturated fatty acids and trans-fatty acids, has been mechanistically linked to escalating epidemics of obesity [5], cardiometabolic diseases, diabetes, and other many metabolic diseases, which constitutes a paramount global public health challenge [6]. Mechanistically, excessive fat intake dysregulates lipid homeostasis by elevating serum low-density lipoprotein cholesterol (LDL-C) while suppressing high-density lipoprotein cholesterol (HDL-C), thereby accelerating atherogenesis and cardiovascular events [7]. In parallel, HFD induces pancreatic β-cell dysfunction and systemic insulin resistance (IR), establishing a causal pathway to type 2 diabetes mellitus (T2DM) [2]. Furthermore, HFD potentiates inflammation through NF-κB (nuclear factor-kappa B) pathway activation and reactive oxygen species (ROS) production [8,9].
Emerging evidence reveals striking sexual dimorphism in HFD-associated metabolic responses. Preclinical models demonstrate male-predominant susceptibility, with accelerated adiposity gain, adipose tissue inflammation, macrophage infiltration [10], and IR [11], contrasting with relative protection in females during reproductive age [12,13]. Clinical observations, though limited, corroborate this sex-divergent vulnerability: males exhibit higher metabolic syndrome prevalence, while premenopausal females maintain protection through estrogen-mediated mechanisms. Postmenopausal estrogen decline abolishes this advantage, rendering females vulnerable to cardiovascular dysfunction [14] and IR [15]. Sexual dichotomy extends to IR subtypes—impaired glucose tolerance (postprandial phase) predominates in females versus impaired fasting glucose in males [16,17]. Alarmingly, the persistent male-centric bias in biomedical research compromises clinical practice. Current diagnostic frameworks and therapeutic protocols predominantly derive from male-predominant studies [18], exemplified by the U.S. Government Accountability Office report identifying 80% of withdrawn drugs (1997–2000) as posing elevated risks for females [19]. This underscores the imperative to integrate sex as a critical biological variable in research design.
This review synthesizes current evidence on HFD-induced metabolic dysregulation through a sex-specific perspective, elucidating underlying mechanisms spanning molecular pathways to clinical manifestations. By bridging this knowledge gap, we aim to catalyze sex-informed research paradigms, advance personalized therapeutic strategies, and propel precision medicine for metabolic disease management.

2. Sex Differences in Metabolic Responses Associated with HFD and the Development of Metabolic Diseases

Chronic HFD consumption serves as a critical etiological factor driving pathological alterations in body composition, metabolic homeostasis, inflammatory cascades, and oxidative stress profiles, which collectively establish the pathophysiological foundation for diverse metabolic disorders. This review systematically examines the sexually dimorphic responses to HFD exposure and differential progression trajectories of obesity, diabetes, and cardiovascular diseases between sexes through multidimensional analysis encompassing these pathological alterations (Figure 1). To achieve this goal, a systematic literature search was conducted across PubMed, Web of Science, and Science Direct using MeSH-based keywords (e.g., “high-fat diet”, “sex differences”, “metabolic diseases”, “gut microbiota”) combined with Boolean operators (e.g., “HFD AND sex differences AND mechanisms”). Inclusion criteria encompassed English-language preclinical and clinical studies exploring sex-dependent mechanisms. The exclusion criteria applied were (i) studies with an off-topic or non-mechanistic focus and (ii) gray literature (Ph.D. dissertations, conference proceedings) or unpublished data. Reference lists of key articles were manually screened to ensure comprehensive coverage of sex-specific molecular pathways and therapeutic interventions.
Prior to mechanistic dissection, conceptual clarification remains imperative. The term “sex differences” denotes biologically determined disparities rooted in chromosomal complement, gonadal hormone profiles, and reproductive system architecture. In contrast, “gender differences” encompass sociocultural constructs shaped by psychosocial roles, behavioral norms, and identity expressions within specific cultural contexts [20]. While acknowledging potential gender-related influences, this review focuses explicitly on biological sex-specific metabolic adaptations, particularly those governed by physiological determinants.

2.1. Sex Differences in Body Composition Abnormalities and Obesity Development

Sexual dimorphism in body composition and adipose tissue distribution is well documented, with males typically exhibiting greater lean mass, higher basal metabolic rates, and lower overall adiposity compared to females [21,22]. This divergence extends to fat deposition patterns: males predominantly accumulate visceral fat within abdominal compartments, whereas females demonstrate preferential subcutaneous fat storage in gluteofemoral regions. Such anatomical partitioning aligns with sex-specific energy allocation strategies. Premenopausal females preferentially utilize carbohydrates and lipids for energy production, a metabolic orientation that promotes subcutaneous fat sequestration while mitigating visceral and ectopic lipid deposition [20]. At the cellular level, male visceral adipocytes exhibit heightened lipid storage capacity under HFD conditions [23] and demonstrate elevated secretion of proinflammatory cytokines [24]. These inflammatory mediators disrupt insulin signaling cascades [25], establishing a mechanistic link to the increased prevalence of metabolic syndrome (MetS) observed in males. MetS is a cluster of metabolic risk factors that include central obesity, hypertension, insulin resistance, and atherogenic dyslipidemia [26]. It serves as a critical precursor to life-threatening comorbidities, elevating risks for cardiovascular diseases and T2DM [22]. Conversely, female subcutaneous adipose tissue displays distinct metabolic properties, including enhanced insulin sensitivity [20,27]. Clinical evidence reveals accelerated weight gain trajectories in male versus female subjects under HFD conditions, a pattern consistently replicated in rodent models [28]. These findings collectively support the clinical consensus that males exhibit greater susceptibility to central obesity-associated metabolic disorders compared to premenopausal females.
However, the decline in estrogen levels during menopause drives a masculinized shift in fat distribution, characterized by preferential visceral adipose tissue accumulation, which increases the risk of obesity and other metabolic diseases. A recent clinical study on 3.5 million Chinese older adults reported that the prevalence of central obesity was higher in older females than in males (37.4% vs. 25.2%) [22]. This suggests that estrogen plays a critical role in regulating lipolysis metabolism in females [29]. For instance, estrogen is regulated by the expression of aromatase genes, which inhibits the hypertrophy of visceral fat depots mainly through the activation of estrogen receptor α (ERα) [29]. Additionally, the calcium-sensing receptor in adipocytes may mediate sex differences in fat distribution. HFD-fed females have lower visceral white adipose tissue mass than males, but this sex difference disappears in calcium-sensing receptor-deficient mice [30].

2.2. Sex Differences in Disorders of Glucose Metabolism and Development of T2DM

T2DM and its complications represent a critical global health burden, with prevalence projected to reach 12.2% by 2045 due to aging populations and urbanization [31]. The disease pathogenesis centers on two interconnected defects: IR in peripheral tissues and progressive pancreatic β-cell failure. IR refers to a defect in the control of glucose metabolism mediated by insulin, which manifests as impaired glucose uptake in the skeletal muscle, adipose tissue, and liver, coupled with enhanced hepatic gluconeogenesis, collectively elevating systemic glucose levels. Compensatory hyperinsulinemia initially mitigates hyperglycemia but ultimately drives β-cell exhaustion through endoplasmic reticulum stress and apoptotic pathways, culminating in irreversible insulin secretion defects [32]. Both preclinical and clinical evidence confirms that HFD exposure accelerates diabetes progression through coordinated mechanisms, including insulin signaling disruption [33,34], gut microbiota dysbiosis [35], ectopic lipid deposition [36], and neuroendocrine dysregulation. HFD-induced norepinephrine surges trigger uncontrolled lipolysis, resulting in systemic fatty acid overflow in rodents. This lipotoxic environment directly exacerbates IR while promoting β-cell dysfunction, establishing a self-reinforcing cycle of metabolic deterioration [37].
Sexual dimorphism in HFD-induced metabolic dysregulation is particularly evident across glycemic parameters. Clinical evidence indicates that males exhibit heightened susceptibility to fasting hyperglycemia [20], exaggerated postprandial glucose excursions [38], earlier onset of IR, and 30% lower insulin sensitivity than age-matched premenopausal females [39]. Enhanced insulin secretion capacity and delayed gastric emptying in females collectively buffer postprandial glycemic spikes under HFD conditions [40]. Longitudinal studies reveal sex-divergent HbA1c trajectories: males show greater HFD-associated HbA1c elevation, strongly correlated with oxidative stress severity [41], while premenopausal females maintain lower HbA1c through estrogen-mediated protection. This advantage dissipates after menopause, with declining estrogen levels precipitating parallel increases in fasting glucose, postprandial hyperglycemia, and HbA1c [42,43]. Compared with women of normal menopausal age, women with early menopause or late menopause may experience a large or moderate increase in the risk of T2DM, respectively [44]. Clinical observations in hyperandrogenic states like polycystic ovary syndrome (PCOS) further validate the vital role of sex hormones, as androgen excess induces male-pattern IR in females [45].
This sexual divergence stems from fundamental biological distinctions in body composition, adipose biology, and hormonal regulation. Visceral adiposity in males drives metabolic inflammation through the elevated secretion of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), which directly impair insulin receptor signaling [46]. Greater muscle mass paradoxically exacerbates the systemic impact of skeletal muscle IR in young men [47]. Preclinical evidence reveals that at the pancreatic level, chronic HFD accelerates β-cell failure in males via unresolved endoplasmic reticulum stress and apoptosis [48,49,50], whereas estrogen preserves β-cell function in females through ERα-mediated suppression of cellular stress pathways [51]. Beyond β-cell protection, estrogen enhances insulin sensitivity via the activation of insulin signaling effectors and mitochondrial bioenergetics, facilitating peripheral glucose utilization in female mice [52,53]. These mechanistic insights underscore the necessity for sex-specific diabetes prevention strategies, prioritizing early intervention in males and tailored approaches for postmenopausal females to optimize precision medicine outcomes.

2.3. Sex Differences in HFD-Induced Lipid Metabolism Disorders and Cardiovascular Disease Development

Extensive preclinical and clinical evidence demonstrates that chronic HFD exposure disrupts systemic lipid homeostasis, manifesting as ectopic lipid deposition; elevated total cholesterol (TC), triglycerides (TG), and LDL; and reduced HDL levels [54,55]. These metabolic perturbations exhibit pronounced sexual dimorphism. Males predominantly develop visceral adiposity with heightened lipolytic activity and proinflammatory cytokine secretion, predisposing them to hepatic lipid accumulation and free fatty acid spillover [42]. Clinical data reveal a male predisposition to hypertriglyceridemia and hypoalphalipoproteinemia under HFD conditions [56], whereas premenopausal females maintain favorable lipid profiles characterized by elevated HDL and reduced TC/LDL levels [57]. Preclinical studies also reveal 2–3-fold higher hepatic TG levels and severe mitochondrial dysfunction in HFD-fed male mice versus female counterparts [58]. Mechanistically, female mice exhibit attenuated upregulation of hepatic and adipose G-protein-coupled receptors under HFD treatment, mitigating lipid metabolism impairment and dyslipidemia risk [59]. Estrogen mediates this protection by enhancing adipose lipoprotein lipase (LPL) activity to promote TG hydrolysis and lipoprotein clearance [60,61], while HFD suppresses adipose LPL activity in males, exacerbating hypertriglyceridemia [62]. Postmenopausal estrogen decline abolishes these benefits, inducing male-pattern dyslipidemia [63,64,65], while estrogen replacement therapy restores lipid homeostasis [65]. However, there is controversy regarding the use of hormone replacement therapy by postmenopausal women to reduce the risk of cardiovascular diseases. Some studies suggest that hormone replacement therapy may bring benefits, while others indicate that it might increase risks of stroke and venous thromboembolism [66]. The therapy duration and timing may explain this inconsistency [67].
These HFD-induced dyslipidemias directly fuel cardiovascular pathogenesis through atherogenic mechanisms. Abundant LDL penetrates the damaged vascular endothelium, undergoing oxidation to form oxidized LDL, which is internalized by macrophages to generate foam cells—the hallmark of early atherosclerotic lesions [68,69]. Progressive lipid accumulation forms fatty streaks, while inflammatory mediators drive smooth muscle proliferation and fibrous cap formation. Plaque rupture exposes thrombogenic components, precipitating acute coronary events [70]. Sexual dimorphism permeates this pathogenic cascade: males exhibit a higher atherosclerosis risk due to elevated LDL, reduced HDL, and impaired endothelial progenitor cell function compared to premenopausal females [71,72]. Estrogen confers cardioprotection via dual mechanisms: enhancing nitric oxide-mediated endothelial function and HDL-dependent reverse cholesterol transport while suppressing LDL oxidation and vascular inflammation [73]. Clinical evidence shows that males develop unstable, rupture-prone plaques in contrast to stabilized lesions in premenopausal females [74]. The postmenopausal female loss of estrogen elevates atherosclerosis risk in females to male-equivalent levels [74]. Epidemiological analyses confirm sex-divergent outcomes in diet-related cardiovascular diseases, with males showing higher ischemic heart disease mortality from 1990 to 2021 [75].
Emerging evidence underscores the critical role of sex-specific factors in modulating heart–brain axis interactions that drive pathophysiology in cardiovascular and cerebrovascular diseases, with shared risk factors (hypertension, diabetes, dyslipidemia) and common pathways, including systemic inflammation, neuroendocrine dysfunction, and accelerated atherosclerosis [76]. Notably, diabetes and hypertension confer higher cardiovascular risks in women. Unique female reproductive events (e.g., pregnancy complications, menopause) substantially elevate stroke vulnerability, with post-stroke outcomes showing higher disability rates and depression risks compared to men. While middle-aged men demonstrate greater stroke incidence associated with premature atherosclerosis and ST elevation myocardial infarction patterns, women face diagnostic delays, therapeutic inequities, and stress-mediated heart–brain axis activation exacerbating ischemic risk. Addressing these disparities requires gender-optimized clinical trial designs and precision interventions to advance equitable cardiovascular and cerebrovascular care [76,77].
Collectively, compelling evidence reveals fundamental sex differences in HFD-induced lipid dysregulation and consequent cardiovascular and/or cerebrovascular pathologies. Men exhibit heightened susceptibility to atherogenic dyslipidemia-driven plaque progression, whereas premenopausal women retain estrogen-mediated atheroprotection until menopause yet demonstrate elevated postmenopausal stroke risk compared to age-matched males. These findings underscore the necessity for sex-stratified approaches in cardiovascular research and clinical management. Future investigations should delineate molecular drivers of sexual dimorphism to develop precision therapies countering HFD-related metabolic and cardiovascular risks.

2.4. Sex Differences in HFD-Induced Inflammation, Oxidative Stress, and Associated Disease Development

HFD exposure induces systemic inflammation and oxidative stress through preferential visceral adipose deposition. This metabolic–inflammatory axis drives pathological processes including myocardial injury, cardiac functional impairment, and IR via coordinated mechanisms [78].

2.4.1. Inflammation

HFD exposure induces systemic low-grade inflammation through adipose tissue-derived cytokine overproduction (e.g., TNF-α, IL-6), which exacerbates metabolic dysregulation, including IR [79,80]. This inflammatory cascade is mechanistically linked to adipose macrophage dynamics: proinflammatory M1 macrophages amplify inflammation via TNF-α and IL-1β secretion, while anti-inflammatory M2 macrophages counteract through IL-10 release [81,82,83]. Visceral adipose-derived mediators propagate systemic inflammation and oxidative damage by elevating mitochondrial ROS, accelerating DNA/protein/lipid oxidation, and promoting atherosclerotic progression [9,84]. For instance, myocardial pathological hypertrophy is frequently accompanied by intensified inflammatory responses that exacerbate cardiac injury [85].
Sexual dimorphism in HFD-induced inflammatory responses is well established. It has been demonstrated in preclinical studies that reveal elevated circulating IL-6 and TNF-α levels with predominant M1 macrophage polarization under HFD conditions in male mice [11,86], whereas females demonstrate M2-dominant macrophage profiles [81,87]. Estrogen mediates this protection by suppressing M1 polarization, enhancing IL-10 production [88,89,90], and inhibiting NF-κB nuclear translocation. Although testosterone exhibits anti-inflammatory effects through NF-κB suppression, its protective capacity is attenuated by male-predominant visceral adiposity [91,92,93,94], compounded by androgen-mediated toll-like receptor 4 (TLR4)/myeloid differentiation marker 88 (MyD88) pathway activation that potentiates inflammatory signaling [95].
Cardiovascular susceptibility further demonstrates sex-specific patterns. Female mice maintain superior aortic compliance compared to males, which develop vascular stiffness and endothelial dysfunction under HFD [96]. Estrogen confers cardiovascular protection through multi-mechanistic actions: dampening inflammation and oxidative stress, promoting angiogenesis, and preserving vasodilatory capacity [97].

2.4.2. Oxidative Stress

Evidence from both preclinical and clinical studies indicates that oxidative stress arises from an imbalance between ROS production (e.g., superoxide anion, hydrogen peroxide, hydroxyl radicals) and cellular antioxidant defenses, leading to macromolecular damage that drives inflammatory cascades, tissue dysfunction [98], IR [99], neurodegeneration [100], and cardiovascular pathologies [101]. Physiological redox homeostasis is maintained through coordinated enzymatic, including superoxide dismutase (SOD), glutathione peroxidase (GPx), catalase (CAT), and non-enzymatic antioxidant systems (e.g., vitamins E/C, glutathione) that neutralize ROS [102].
Chronic HFD exposure disrupts this equilibrium, with sexually dimorphic manifestations of oxidative damage. Female organisms exhibit enhanced antioxidant capacity, effectively mitigating ROS accumulation and associated tissue injury compared to males [103]. This sex-specific protection stems from elevated baseline antioxidant enzyme activities in females, mediated principally by estrogen’s redox-modulatory effects [104]. Estrogen enhances antioxidant defenses through two synergistic mechanisms: (1) upregulating cystathionine gamma-lyase expression and activating extracellular regulated kinase 1/2-NF-κB signaling to boost SOD/GPx/CAT activity [105,106,107] and (2) preserving mitochondrial integrity to suppress ROS generation [97]. Conversely, male rodents fed an HFD demonstrate elevated plasma malondialdehyde and total oxidative status, alongside reduced total antioxidant status, glutathione levels, and GPx/CAT activity [108,109]. Androgen signaling exacerbates this vulnerability by transcriptionally repressing antioxidant enzymes. For instance, testosterone downregulates SOD, CAT, and GPx expression in human endothelial cells, impairing ROS clearance [110,111,112].
In summary, chronic HFD exposure promotes cardiovascular pathogenesis through sexually dimorphic inflammatory and oxidative stress pathways. Males exhibit heightened susceptibility to ROS-driven atherosclerosis and myocardial injury compared to premenopausal females, a disparity rooted in sex hormone regulation of redox homeostasis. Estrogen serves as a critical modulator of antioxidant defenses, while androgens paradoxically amplify oxidative vulnerability despite partial anti-inflammatory activity. These findings underscore the necessity for sex-specific therapeutic strategies targeting oxidative stress in metabolic disease management.

3. Mechanisms Behind the Sex Differences in the Development of Metabolic Diseases

HFD exposure drives the pathogenesis of metabolic disorders, including obesity, T2DM, and atherosclerosis, by disrupting glucose and lipid homeostasis, amplifying inflammatory cascades, and compromising oxidative stress regulation. These metabolic perturbations exhibit pronounced sex-specific manifestations rooted in fundamental biological divergences. Emerging research has revealed five interconnected mechanistic determinants of sexual dimorphism: (1) sex chromosome complement-driven transcriptional regulation, (2) sex hormone receptor signaling dynamics, (3) genetic and epigenetic regulatory networks, (4) gut microbiota–host metabolic crosstalk, and (5) neuroendocrine pathway dimorphisms (Figure 2).

3.1. Dosage Effects and Compensatory Mechanisms of Sex Chromosome Genes

The impact of biological sex on metabolic disease susceptibility extends beyond sex hormone variations to encompass sex chromosome dosage effects and their compensatory mechanisms [113]. The inherent imbalance in X-linked gene expression between XX (female) and XY (male) configurations is partially resolved in females through random X-chromosome inactivation (XCI). However, about 15% of X-linked genes (e.g., histone demethylase KDM5C, apoptosis inhibitor XIAP) escape inactivation, exhibiting biallelic expression that drives sex-specific metabolic regulation [114,115]. Murine four-core genotype models reveal that XX chromosomal complement, independent of gonadal hormones, promotes subcutaneous adipogenesis, whereas XY predisposes individuals to visceral obesity [116]. Clinically, Klinefelter syndrome (47,XXY) is a sex chromosome aneuploidy characterized by an additional X chromosome in males. While clinical features typically include hypogonadism, infertility, gynecomastia, and learning difficulties, phenotypic expression shows marked interindividual variability. Diagnosis is confirmed through karyotype analysis [117]. The prevalence of MetS is significantly higher in patients with Klinefelter syndrome than that in the general population [118]. Clinical evidence shows that patients with Klinefelter syndrome display central adiposity, IR, and atherogenic dyslipidemia (elevated triglyceride–glucose index, reduced HDL-C) linked to X-chromosome overdosage [119], while individuals with Turner syndrome (45,XO) exhibit heightened visceral adiposity, fasting hyperglycemia, and atherosclerosis risk due to X monosomy [120].
XCI-escape genes critically mediate metabolic sexual dimorphism through adipocyte differentiation and mitochondrial bioenergetics. Lysine-specific histone demethylase 5C (KDM5C), an X-linked escape gene overexpressed in females, serves as a key obesity determinant [121]. The KDM5 histone demethylase family (KDM5A/B/C/D) exhibits sex-divergent expression, with KDM5C (X-linked) and KDM5D (Y-linked) showing reciprocal sexual dimorphism in humans and mice [122,123,124]. Female-enriched KDM5C regulates adipogenesis via histone H3K4 methylation dynamics: in white adipose tissue, it suppresses delta-like homolog 1 to promote preadipocyte maturation [125,126]; in brown fat, it activates uncoupling protein 1 (UCP1) and mitochondrial respiratory genes to enhance thermogenesis. These effects are abolished by KDM5 inhibitors like C70 [126]. Similarly, biallelic XIAP expression may protect pancreatic β-cell function, potentially attenuating diabetes progression [127,128,129], though its sex-specific roles require validation.
Y-chromosome genes equally shape metabolic disparities. Males with Y-chromosome deletions exhibit impaired insulin sensitivity [130], while preclinical and clinical evidence shows that hepatic sex-determining region Y gene upregulation in fibrotic livers activates stellate cells via platelet-derived growth factor receptor α and high-mobility group box-1 protein signaling, exacerbating lipid metabolism-associated liver pathology in mice [131]. Despite progress, critical challenges persist: the functional divergence of XCI-escape genes between humans and mice, as well as undefined sex chromosome–autosome interactions. Integrating single-cell multi-omics with sex-stratified cellular models will elucidate dosage–effect regulatory networks in adipocytes, hepatocytes, and other metabolically active cells, advancing targeted interventions for sex-specific metabolic disorders.

3.2. Sex Hormone Receptor Signaling Dynamics

Sex hormones are mainly secreted by the gonads. The ovaries of females mainly secrete estrogens and progesterone, while the testes of males mainly secrete androgens primarily composed of testosterone. These steroid hormones share a tetracyclic hydrocarbon backbone and are mainly derived from cholesterol. In detail, estrogen is characterized by an aromatic A-ring with a hydroxyl group at C3. Its synthesis begins with cholesterol conversion to pregnenolone via CYP11A1, followed by oxidation to progesterone. Progesterone undergoes sequential modifications by CYP17A1 to form androstenedione, which is reduced to testosterone. Testosterone is aromatized by aromatase, removing the C19 methyl group and inducing A-ring aromatization to yield estradiol. Estrogens and androgens, classically recognized for their roles in sexual differentiation and reproductive function, exert profound regulatory effects on systemic metabolic homeostasis. Accumulating evidence reveals striking sexual dimorphism in metabolic disease susceptibility mediated by these hormones. Epidemiological analyses demonstrate that premenopausal females exhibit preferential subcutaneous fat distribution and enhanced insulin sensitivity relative to males, conferring protection against visceral adiposity and metabolic dysfunction. This protective phenotype dissipates after menopause, with declining estrogen levels precipitating central obesity, cardiovascular pathologies, IR, and hypertension at rates paralleling those in male counterparts [27]. Conversely, clinical evidence suggests that hypogonadal males with reduced testosterone levels display elevated risks of abdominal obesity and T2DM, establishing a bidirectional relationship between androgen status and metabolic health [132,133,134]. These clinical observations, corroborated by preclinical studies, position sex hormones as pivotal modulators of HFD-induced metabolic adaptations and key determinants of sex-specific disease trajectories [135].

3.2.1. Androgens/Androgen Receptor

Androgens, predominantly testosterone synthesized by testicular Leydig cells with minor adrenal contributions, regulate metabolic homeostasis through genomic and non-genomic mechanisms mediated by androgen receptors (ARs). The genomic pathway involves ligand-dependent AR nuclear translocation, subsequent binding to androgen response elements (AREs), and transcriptional regulation via coregulator recruitment [136]. Non-genomic actions rapidly modulate glucose and lipid metabolism through membrane-initiated signaling cascades. Preclinical models reveal that androgen deprivation or AR antagonism induces lipid dysregulation and IR [137], whereas AR activation (e.g., ostarine administration) counteracts metabolic perturbations in mice [138] Clinically, male hypogonadism strongly correlates with obesity, IR, and dyslipidemia, and these metabolic abnormalities are reversible through testosterone replacement therapy [139,140,141,142,143]. These findings collectively establish androgens/AR signaling as a central determinant of sex-specific metabolic responses.
The androgen–AR axis exerts dual regulatory control through central and peripheral pathways. Central modulation occurs via hypothalamic AR-mediated suppression of NF-κB-dependent protein tyrosine phosphatase 1B (PTP1B) expression, enhancing insulin sensitivity under HFD conditions [144]. Concurrently, AR signaling regulates energy balance by modulating pro-opiomelanocortin/neuropeptide Y (POMC/NPY) neuronal activity and attenuates neuroinflammation through TLR4/NF-κB/mitogen-activated protein kinase (MAPK) pathway inhibition, elevating anti-inflammatory cytokines IL-10/IL-13 [145]. In peripheral tissues, AR activation promotes skeletal muscle glucose uptake via glucose transporter 4 (GLUT4) induction [146], stimulates adipose lipolysis through hormone-sensitive lipase and LPL activation while suppressing lipogenesis via peroxisome proliferator-activated receptor gamma/stearoyl-CoA desaturase 1/acetyl-CoA carboxylase-1 downregulation, and enhances brown fat thermogenesis via UCP1 transcriptional activation [147]. Hepatic AR signaling improves lipid metabolism by increasing bile acid synthesis and taurine production via cysteine sulfinic acid decarboxylase regulation [148,149], whereas pancreatic AR activity enhances β-cell function through glucagon-like peptide-1 (GLP-1) receptor upregulation and focal adhesion kinase/phosphatidylinositol 3-kinase/mammalian target of rapamycin complex 2-mediated insulin secretion potentiation [150,151].
While females maintain lower circulating androgen levels, primarily through adrenal synthesis and peripheral aromatization, these hormones critically regulate reproductive development, bone homeostasis, and metabolic physiology. Pathologically, hyperandrogenism, a hallmark of polycystic ovary syndrome, exacerbates IR and T2DM risk in females [152,153]. Preclinical studies demonstrate that HFD-fed female rats with dihydrotestosterone supplementation develop profound IR, which can be reversed through hypothalamic-specific AR deletion [154]. Mechanistically, supraphysiological androgens disrupt metabolic homeostasis via dual pathways: centrally, they impair hypothalamic function by suppressing kisspeptin/gonadotropin-releasing hormone and neuronal activity while stimulating agouti-related peptide (AgRP) neurons to enhance feeding behavior, concurrently inducing leptin resistance and brown adipose tissue dysfunction [155,156]. Peripherally, androgen excess promotes visceral adiposity through adipocyte hypertrophy, macrophage infiltration, and oxidative stress while simultaneously inducing pancreatic β-cell damage and gut microbiota dysbiosis [154,157]. Notably, hepatic AR ablation in female mice attenuates HFD-induced metabolic derangements by preserving insulin signaling and mitigating weight gain [158].
Overall, the androgen–AR axis exhibits sexually dimorphic metabolic regulation. In males, physiological AR signaling maintains metabolic homeostasis through hypothalamic suppression of appetite via POMC/NPY neuronal modulation and NF-κB/PTP1B pathway inhibition, coupled with peripheral enhancements in skeletal muscle GLUT4-mediated glucose uptake, brown adipose UCP1-driven thermogenesis, and hepatic suppression of gluconeogenic enzymes. In contrast, female hyperandrogenism drives metabolic dysfunction via central appetite stimulation (AgRP upregulation/POMC suppression) and leptin resistance, compounded by peripheral visceral adipocyte inflammation, oxidative stress, and gut microbiota alterations that collectively exacerbate β-cell failure. These divergent pathways underscore the necessity for sex-specific therapeutic strategies targeting androgen/AR signaling in metabolic disease management.

3.2.2. Estrogen and Estrogen Receptors

Estrogens, the predominant female sex hormone synthesized primarily by ovarian follicles and the corpus luteum with minor adrenal contributions, exerts critical regulatory effects on glucose and lipid metabolism through estrogen receptor α/β (ERα/ERβ)-mediated mechanisms. This signaling axis mitigates HFD-induced metabolic disturbances and protects against obesity-related pathologies [159,160]. Evidence from preclinical and clinical studies suggests that postmenopausal estrogen decline precipitates visceral adiposity, IR, atherogenic dyslipidemia, and cardiovascular risk elevation. These metabolic derangements are reversed through estrogen replacement therapy [161,162,163,164]. The estrogen/ER system orchestrates metabolic homeostasis via dual regulatory tiers: centrally, hypothalamic ERα engages glutamate/aspartate-rich carboxy-terminal domain-containing protein 1 (Cited1) in arcuate nucleus neurons to potentiate leptin sensitivity and suppress hyperphagia [165]. Peripherally, ER activation enhances skeletal muscle/adipose glucose uptake through PI3K–Akt–GLUT4 axis stimulation while preserving β-cell mass via anti-apoptotic mechanisms [52]. Concurrently, ERα modulates lipid metabolism by upregulating LDL receptor and scavenger receptor class B type I to accelerate cholesterol clearance [159,166,167,168] while suppressing visceral adipogenesis through AMP-activated protein kinase/peroxisome proliferator-activated receptor-α (AMPK/PPARα) pathway activation and adipose tissue inflammation resolution [29,169]. These coordinated actions establish estrogen signaling as a cornerstone of female metabolic health across the lifespan, with its age-related decline constituting a key driver of postmenopausal MetS susceptibility.
While males maintain lower physiological estrogen levels via adrenal synthesis and testicular aromatase-mediated androgen conversion, this hormone critically regulates bone homeostasis, reproductive function, and metabolic health through ERα/ERβ signaling. Estrogen modulates hypothalamic–pituitary–gonadal axis activity via negative feedback on luteinizing hormone/follicle-stimulating hormone secretion, while ERα activation suppresses visceral adiposity and enhances insulin sensitivity through adipocyte differentiation inhibition [170]. Clinical evidence from aromatase-deficient males reveals an estrogen-dependent MetS phenotype characterized by dyslipidemia, ectopic fat deposition, and glucose intolerance, which is reversed by estrogen therapy [170]. Preclinical studies further demonstrate that estrogen confers hepatic protection in male mice through formyl peptide receptor 2 induction, attenuating HFD-induced hepatocyte damage [171]. In summary, these findings underscore the conserved metabolic regulatory role of estrogen across sexes, with sex-specific circulating levels explaining divergent disease susceptibilities.

3.3. Genetic and Epigenetic Regulatory Networks

Epigenetic regulation dynamically modulates energy metabolism through DNA methylation and histone modifications at metabolic loci. The hypermethylation of the peroxisome proliferator-activated receptor gamma coactivator 1 α (PGC-1α) promoter suppresses mitochondrial function in cardiomyocytes, while HFD-induced tissue-specific alterations, including adipose PPARα hypomethylation and hepatic ERα histone hyperacetylation, disrupt lipid homeostasis and insulin signaling [172]. Pathological remodeling in metabolic diseases features β-cell insulin promoter hypermethylation and adipocyte PPARG hypermethylation, exacerbating glucose intolerance through impaired secretion and differentiation [173]. Concurrently, hepatic nuclear factor 4α methylation and enhancer of zeste homolog/tripartite motif-containing 14-mediated histone modifications drive diabetes susceptibility and inflammatory dysregulation [174,175].
Sexual dimorphism in epigenetic regulation profoundly shapes metabolic phenotypes. Genome-wide association studies reveal sex-biased obesity-risk loci: SAFB-like transcription modulator (SLTM) variants selectively increase male adiposity, whereas rare death inducer obliterator 1 (DIDO1) and SLC12A5 mutations exhibit female-specific associations with elevated body mass index (>80% penetrance in females). Notably, clinical evidence indicated that DIDO1 variants correlate with hyperandrogenism and central adiposity in females, while solute carrier family 12 member 5 (SLC12A5) mutations heighten T2DM risk in this population [176]. X-chromosome epigenetic mechanisms further drive metabolic divergence. Xist-mediated chromatin remodeling calibrates metabolic plasticity in female mice [177], while male-specific hypermethylation of the X-linked angiotensin-converting enzyme 2 (ACE2) gene reduces pancreatic islet expression compared to that in females [178]. Key metabolic genes exhibit sex-divergent epigenetic controls: MC4R variants promote a male-biased preference for hypercaloric diets through dimorphic enhancer methylation [179], whereas fat mass and obesity-associated protein (FTO) polymorphisms differentially regulate iroquois homeobox 3/5 expression via AT-rich interaction domain 5B-dependent histone modifications, altering adipogenesis and thermogenic capacity [180].
These findings establish an integrative paradigm wherein sex-specific epigenetic networks encompassing X-chromosome reprogramming, allele-specific DNA methylation, and histone modifier activity sculpt metabolic dimorphism. The dynamic interplay between sex hormones and chromatin-modifying enzymes reinforces divergent regulatory trajectories, with androgen/estrogen signaling modulating epigenetic writer/eraser recruitment and X-linked modifiers like XIST fine-tuning metabolic adaptation. Future investigations should prioritize deciphering the molecular crosstalk between steroid receptors and epigenetic remodelers, elucidating X-chromosome dosage compensation mechanisms in metabolic regulation, and developing sex-stratified epigenetic therapies targeting MC4R, FTO, or ACE2 pathways. Such advances will catalyze precision medicine strategies for metabolic disorders through gender-informed chromatin modulation.

3.4. Gut Microbiota–Host Metabolic Crosstalk

The gut microbiota serves as a pivotal regulator of host metabolic homeostasis, with dysbiosis strongly implicated in obesity, IR, and MetS pathogenesis [181]. Preclinical evidence indicates that HFD induces microbial imbalance, characterized by an increased Firmicutes/Bacteroidetes (F/B) ratio, through the selective enrichment of Firmicutes over Bacteroidetes. This shift reduces populations of short-chain fatty acid (SCFA)-producing taxa (e.g., Lachnospiraceae, Ruminococcus), diminishing butyrate levels that normally reinforce intestinal barrier integrity via G-protein-coupled receptor 41/43 activation and histone deacetylase inhibition. The resultant barrier dysfunction facilitates systemic lipopolysaccharide translocation, driving chronic inflammation [182,183]. Concurrently, preclinical and clinical evidence shows that HFD-induced alterations in secondary bile acid metabolism exacerbate hepatic lipid accumulation and suppress GLP-1 secretion through farnesoid X receptor (FXR) signaling activation [184,185].
Sexual dimorphism profoundly influences HFD-induced microbiota remodeling. Preclinical evidence indicates that male mice develop adipose inflammation and IR associated with Bacteroidaceae/Clostridia expansion alongside Lactobacillus and Desulfovibrionaceae enrichment [11,186,187]. In contrast, female counterparts exhibit more severe intestinal barrier disruption, evidenced by tight junction protein dysregulation and colonic proinflammatory cytokine elevation, correlating with Actinomycetota/Bacillota depletion [11,183,187]. Baseline microbial differences underlie this divergence: males harbor lower α-diversity with Prevotella/Veillonella predominance, while females maintain richer communities enriched in SCFA producers [10,188,189,190,191,192,193,194]. These sex-specific communities differentially modulate host metabolism through SCFA-mediated histone deacetylase inhibition/G-protein-coupled receptor signaling and bile acid–FXR interactions that regulate hepatic lipid synthesis genes (e.g., SREBP-1c). Emerging evidence further indicates the promise of microbiota–gut–brain axis signaling, where SCFAs suppress hypothalamic NPY via vagal afferents to reduce hyperphagia [184].
These findings establish gut microbiota sexual dimorphism as a central determinant of HFD-induced metabolic disparities. While male susceptibility stems from low-diversity baseline flora predisposing individuals to proinflammatory taxon shifts, female pathophysiology involves α-diversity loss compromising barrier-protective SCFA producers. Mechanistically, sex-specific microbiota metabolites engage complementary pathways. For instance, SCFAs enhance insulin sensitivity through epigenetic and receptor-mediated mechanisms, while bile acids modulate lipid homeostasis via FXR. Critical knowledge gaps persist regarding sex hormone–microbiota crosstalk and translational applicability to human populations. Future investigations should integrate multi-omics profiling with gnotobiotic models to decipher dynamic microbiota–host interactions, enabling the development of sex-stratified therapeutic strategies targeting microbial networks for the precision management of metabolic disorders.

3.5. Neuroendocrine Pathway Dimorphisms

Hypothalamic glial cells play pivotal roles in energy homeostasis and metabolic regulation. In HFD-fed mice, hypothalamic microglia exhibit rapid activation marked by morphological changes and elevated proinflammatory cytokine production. These microglia undergo immunometabolic reprogramming, shifting toward fatty acid oxidation and oxidative phosphorylation with suppressed glycolysis, while displaying mixed M1/M2-like polarization [195]. HFD further induces astrocyte proliferation and NF-κB-dependent IL-6 secretion [195], with notable sex-specific responses: male mice develop hypothalamic inflammation under HFD, whereas females remain resistant [196]. This protection may be attributed to the function of Cited1, which is enriched in hypothalamic POMC neurons and governs appetite and energy balance by facilitating crosstalk between ERα and leptin-activated Stat3 [165]. Preclinical models demonstrate that POMC neuron-specific cited1 deletion causes diet-induced obesity exclusively in females [165].
Collectively, sex-specific neuroendocrine pathways engage distinct molecular hubs: serotonin 2C receptor helps maintain metabolic stability in males, while Cited1 orchestrates hormone–neuron interactions in females. Future research directions include further investigations into the differences in the responses of other hypothalamic neuron populations to HFD exposure across sexes. Combining the multidimensional information of patients’ genetic backgrounds, lifestyles, and disease status, we can design a personalized treatment plan to precisely regulate the neuroendocrine pathway in order to improve metabolic status.
Interestingly, increasing evidence suggests that maternal HFD adherence leads to obesity, IR, impaired glucose tolerance, and increased cardiovascular disease risk in offspring. The sex-specific remodeling of hypothalamic energy homeostasis networks represents a critical neural mechanism underlying this detrimental impact of maternal HFD consumption on offspring. Preclinical studies demonstrate that maternal HFD exposure sex-dependently reprograms arcuate nucleus circuitry: Male offspring exhibit amplified orexigenic signaling through increased AgRP neuron populations, elevated AgRP-to-POMC neuron ratios, and expanded arginine vasopressin/retinoic acid receptor-related orphan receptor β (Avp/Rorb) neuronal clusters, collectively driving hyperphagia and insulin dysregulation that predisposes them to obesity [177]. Neuron–glial interaction analyses reveal the male-specific enhancement of AgRP neuron–astrocyte crosstalk, coupled with the activation of neurotrophic regulatory networks involving the obesity-associated gene neuronal growth regulator 1. In contrast, female offspring show compensatory upregulation of the X-chromosome-encoded long noncoding RNA Xist in hypothalamic tissue, suggesting sex chromosome-linked epigenetic mechanisms may mitigate metabolic perturbations caused by maternal overnutrition [177]. Additionally, maternal HFD leads to metabolic disorder in offspring through the upregulation of cytokines, neutrophil infiltration, damage to the intestinal barrier, and the induction of systemic inflammation. Moreover, maternal HFD impairs pancreatic β-cell function in offspring; significantly increases lipid content, blocks the AMPK/SIRT1/PGC-1α signaling pathway; and thereby promotes the occurrence of obesity, hyperglycemia, and cardiovascular diseases [197]. These findings establish a molecular framework wherein maternal metabolic stress induces sexually dimorphic hypothalamic rewiring through divergent neuron–glia interaction patterns and neuroendocrine pathway activation, ultimately shaping offspring susceptibility to energy imbalance.

4. Dietary and Pharmacotherapeutic Interventions Targeting Sex-Specific HFD-Induced Metabolic Diseases

Sexual dimorphism profoundly influences dietary regulation and pharmacological responses through divergent nutritional requirements and metabolic adaptations. Females benefit from high-fiber diets that optimize gut microbiota and modulate estrogen levels to reduce breast cancer risk [198], whereas males require restricted refined carbohydrate intake to mitigate MetS predisposition [199]. While protein consumption enhances energy metabolism in both sexes, females exhibit superior muscle anabolic responses [200,201]. Lipid regulation strategies also diverge: clinical data show that omega-3 and monounsaturated fatty acids preferentially support female cardiovascular health [202], while males require focused monounsaturated fat intake for adiposity control [203]. The Mediterranean diet exerts stronger cardioprotective effects on males via anti-inflammatory mechanisms [204], whereas ketogenic diets may compromise female gut barrier integrity despite short-term weight loss [205]. Micronutrient antioxidants (e.g., vitamins C/E, selenium, zinc) alleviate HFD-induced metabolic stress by scavenging reactive oxygen species and stabilizing hormonal balance [206,207], with iron supplementation being particularly critical for female physiology [208].
Pharmacological interventions exhibit sex-biased efficacy and safety profiles. Clinical data reveal that reduced cytochrome P450 activity predisposes females to elevated drug concentrations and adverse effects [209]. Glucose-lowering agents demonstrate sex-divergent responses: males show heightened sulfonylurea sensitivity [210], while GLP-1 receptor agonists (e.g., liraglutide) elicit stronger weight-modulatory effects on females [211]. Fluctuating estrogen levels during menstrual cycles and menopause alter metformin’s cardioprotective efficacy [212,213,214], whereas males exhibit superior metabolic responses to sodium-glucose co-transporter 2 inhibitors and statins [215]. Hormone replacement strategies further underscore sexual dimorphism: clinical studies show that estrogen therapy enhances insulin sensitivity in postmenopausal females [216], while testosterone supplementation reduces visceral adiposity and improves IR in males [217,218,219]. However, long-term safety profiles require rigorous sex-specific evaluation [97].
These findings underscore the necessity for sex-tailored therapeutic approaches. Nutritional strategies should prioritize female gut microbiota optimization through fiber-rich diets and targeted micronutrients, while males require carbohydrate restriction and lipid profile modulation. Pharmacological regimens must account for sex-specific drug metabolism patterns and hormonal interactions, with individualized adjustments based on metabolic risk profiles and therapeutic objectives.

5. Sex Differences in Exercise-Related Exercise Capacity and Metabolic Health

Sexual dimorphism in exercise-mediated metabolic regulation arises from fundamental differences in skeletal muscle architecture and energy flux pathways. Males exhibit higher lean mass and glycolytic type II fiber predominance, favoring anaerobic performance but exhibiting lower basal energy expenditure compared to females [200,220]. Conversely, female muscle physiology prioritizes oxidative metabolism through type I fiber dominance and estrogen–AMPK axis-driven lipid mobilization, particularly enhancing fatty acid oxidation during endurance activities [221,222,223]. This metabolic divergence manifests as male superiority in power/strength tasks versus female advantages in fatigue resistance and fat utilization efficiency [222,224]. Hormonal regulation further stratifies these responses: males display greater exercise-induced AMPK activation [225], while females optimize intramuscular lipid storage and hypoxic metabolic flexibility via 17β-estradiol signaling [223,226].
Exercise intervention outcomes demonstrate sex-biased cardiometabolic benefits. High-intensity interval training preferentially enhances male glycolytic capacity and VO2max, albeit with paradoxical reductions in insulin sensitivity [200,227,228]. Females achieve superior cardioprotection and all-cause mortality reduction through moderate-intensity regimens [229]. Sexual dimorphism extends to HFD-induced metabolic rescue: preclinical studies have shown that male mice develop exercise-aggravated cardiac lipotoxicity under high-intensity interval training [230], while females improve insulin sensitivity via endurance training through IRS-1 serine dephosphorylation and mitochondrial biogenesis [200]. Menstrual cycle dynamics introduce additional complexity: luteal-phase progesterone elevation suppresses lipid oxidation while enhancing glucose utilization, necessitating hormone-phase-adapted exercise prescriptions [200]. Notably, sex-specific intergenerational metabolic programming emerges where maternal preconception/prenatal exercise preferentially potentiates male offspring metabolic fitness [231].
These findings establish a paradigm wherein sex-specific exercise adaptations are orchestrated through intersecting hormonal, cellular, and metabolic networks. While male physiology capitalizes on anabolic capacity and acute AMPK responsiveness, female metabolic resilience stems from estrogen-optimized oxidative efficiency and lipid flexibility. Critical knowledge gaps persist regarding sex hormone–exercise crosstalk at a molecular resolution and the clinical translation of sex-stratified exercise therapies. Future research must employ multi-omics approaches to decode the dynamic interactome of exercise–sex hormone–metabolism networks, enabling precision exercise regimens tailored to hormonal status and metabolic phenotypes.

6. Conclusions and Perspectives

The pervasive sexual dimorphism in HFD-induced metabolic disorders underscores an urgent need to recalibrate biomedical research paradigms historically skewed toward male physiology. This synthesis reveals that sex-specific metabolic vulnerabilities emerge through an intricate interplay of sex chromosome-mediated transcriptional programs, steroid hormone-regulated energy partitioning, dynamic epigenetic remodeling, microbiota–metabolite crosstalk, and hypothalamic nutrient-sensing plasticity. Crucially, the sexually divergent therapeutic responses observed in exercise adaptation and drug metabolism underscore the limitations of one-size-fits-all interventions, advocating for clinical strategies that harmonize sex-specific molecular signatures with physiological traits. In addition, other variables, such as age or ethnicity, may also play a role in the sex-dependent impact of HFD on metabolic diseases, which warrants further studies in the future.
Looking ahead, the field must prioritize elucidating the spatiotemporal choreography of sex hormone fluctuations across developmental milestones and their crosstalk with epigenetic–microbiome networks. Advanced multi-omics integration spanning metabolomic, epigenomic, and proteomic landscapes will be instrumental in building predictive models that map sex-specific molecular cascades to clinical phenotypes. Parallel efforts should focus on translating these insights into precision medicine through two synergistic pathways: developing therapies targeting conserved nodes of sexually dimorphic regulation and implementing personalized prevention frameworks that account for hormonal status, genetic predisposition, and environmental exposures. By bridging these frontiers, we can catalyze a transformative shift toward sex-stratified therapeutic regimens, ultimately enabling tailored nutritional and pharmacological strategies to combat HFD-associated cardiometabolic diseases with precision.

Author Contributions

All authors conceived the manuscript. Q.M. drafted the initial version of the manuscript; X.D. systematically conducted comprehensive literature investigations; Z.Z. developed an intellectual framework by offering valuable methodological insights; L.Y. critically revised and contributed to the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This study was sponsored by Shenzhen Research Start-up Funding for High-level Talents (827-00097826) and the Start-up Research Grant of Shenzhen University (868-000001032054).

Acknowledgments

We would like to thank Servier Medical Art for providing free, professional-grade medical elements for the creation of figures in this manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. WHO. Guidelines Approved by the Guidelines Review Committee. In Total Fat Intake for the Prevention of Unhealthy Weight Gain in Adults and Children: WHO Guideline; World Health Organization: Geneva, Switzerland, 2023. [Google Scholar]
  2. Cyr-Scully, A.; Howard, A.G.; Sanzone, E.; Meyer, K.A.; Du, S.; Zhang, B.; Wang, H.; Gordon-Larsen, P. Characterizing the urban diet: Development of an urbanized diet index. Nutr. J. 2022, 21, 55. [Google Scholar] [CrossRef]
  3. Shan, Z.; Rehm, C.D.; Rogers, G.; Ruan, M.; Wang, D.D.; Hu, F.B.; Mozaffarian, D.; Zhang, F.F.; Bhupathiraju, S.N. Trends in Dietary Carbohydrate, Protein, and Fat Intake and Diet Quality Among US Adults, 1999–2016. JAMA 2019, 322, 1178–1187. [Google Scholar] [CrossRef] [PubMed]
  4. Retterstøl, K.; Rosqvist, F. Fat and fatty acids—A scoping review for Nordic Nutrition Recommendations 2023. Food Nutr. Res. 2024, 68, 9980. [Google Scholar] [CrossRef] [PubMed]
  5. Koliaki, C.; Dalamaga, M.; Liatis, S. Update on the Obesity Epidemic: After the Sudden Rise, Is the Upward Trajectory Beginning to Flatten? Curr. Obes. Rep. 2023, 12, 514–527. [Google Scholar] [CrossRef]
  6. Talukdar, J.R.; Steen, J.P.; Goldenberg, J.Z.; Zhang, Q.; Vernooij, R.W.M.; Ge, L.; Zeraatkar, D.; Bała, M.M.; Ball, G.D.C.; Thabane, L.; et al. Saturated fat, the estimated absolute risk and certainty of risk for mortality and major cancer and cardiometabolic outcomes: An overview of systematic reviews. Syst. Rev. 2023, 12, 179. [Google Scholar] [CrossRef] [PubMed]
  7. Maki, K.C.; Dicklin, M.R.; Kirkpatrick, C.F. Saturated fats and cardiovascular health: Current evidence and controversies. J. Clin. Lipidol. 2021, 15, 765–772. [Google Scholar] [CrossRef]
  8. Mei, J.; Qian, M.; Hou, Y.; Liang, M.; Chen, Y.; Wang, C.; Zhang, J. Association of saturated fatty acids with cancer risk: A systematic review and meta-analysis. Lipids Health Dis. 2024, 23, 32. [Google Scholar] [CrossRef]
  9. Tan, B.L.; Norhaizan, M.E. Effect of High-Fat Diets on Oxidative Stress, Cellular Inflammatory Response and Cognitive Function. Nutrients 2019, 11, 2579. [Google Scholar] [CrossRef]
  10. Hases, L.; Stepanauskaite, L.; Birgersson, M.; Brusselaers, N.; Schuppe-Koistinen, I.; Archer, A.; Engstrand, L.; Williams, C. High-fat diet and estrogen modulate the gut microbiota in a sex-dependent manner in mice. Commun. Biol. 2023, 6, 20. [Google Scholar] [CrossRef]
  11. Lefebvre, C.; Tiffay, A.; Breemeersch, C.E.; Dreux, V.; Bôle-Feysot, C.; Guérin, C.; Breton, J.; Maximin, E.; Monnoye, M.; Déchelotte, P.; et al. Sex-dependent effects of a high fat diet on metabolic disorders, intestinal barrier function and gut microbiota in mouse. Sci. Rep. 2024, 14, 19835. [Google Scholar] [CrossRef]
  12. Tran, V.; Brettle, H.; Diep, H.; Dinh, Q.N.; O’Keeffe, M.; Fanson, K.V.; Sobey, C.G.; Lim, K.; Drummond, G.R.; Vinh, A.; et al. Sex-specific effects of a high fat diet on aortic inflammation and dysfunction. Sci. Rep. 2023, 13, 21644. [Google Scholar] [CrossRef] [PubMed]
  13. Medrikova, D.; Jilkova, Z.M.; Bardova, K.; Janovska, P.; Rossmeisl, M.; Kopecky, J. Sex differences during the course of diet-induced obesity in mice: Adipose tissue expandability and glycemic control. Int. J. Obes. 2012, 36, 262–272. [Google Scholar] [CrossRef] [PubMed]
  14. El Khoudary, S.R.; Aggarwal, B.; Beckie, T.M.; Hodis, H.N.; Johnson, A.E.; Langer, R.D.; Limacher, M.C.; Manson, J.E.; Stefanick, M.L.; Allison, M.A. Menopause Transition and Cardiovascular Disease Risk: Implications for Timing of Early Prevention: A Scientific Statement From the American Heart Association. Circulation 2020, 142, e506–e532. [Google Scholar] [CrossRef]
  15. Oya, J.; Nakagami, T.; Yamamoto, Y.; Fukushima, S.; Takeda, M.; Endo, Y.; Uchigata, Y. Effects of age on insulin resistance and secretion in subjects without diabetes. Intern. Med. 2014, 53, 941–947. [Google Scholar] [CrossRef] [PubMed]
  16. Mauvais-Jarvis, F. Gender differences in glucose homeostasis and diabetes. Physiol. Behav. 2018, 187, 20–23. [Google Scholar] [CrossRef]
  17. Kautzky-Willer, A.; Harreiter, J.; Pacini, G. Sex and Gender Differences in Risk, Pathophysiology and Complications of Type 2 Diabetes Mellitus. Endocr. Rev. 2016, 37, 278–316. [Google Scholar] [CrossRef]
  18. Clayton, J.A. Studying both sexes: A guiding principle for biomedicine. FASEB J. 2016, 30, 519–524. [Google Scholar] [CrossRef]
  19. U.S. Government Accountability Office. Drug Safety: Most Drugs Withdrawn in Recent Years Had Greater Health R isks for Women. 19 January 2001. Available online: https://www.gao.gov/products/gao-01-286r (accessed on 9 February 2001).
  20. Tramunt, B.; Smati, S.; Grandgeorge, N.; Lenfant, F.; Arnal, J.F.; Montagner, A.; Gourdy, P. Sex differences in metabolic regulation and diabetes susceptibility. Diabetologia 2020, 63, 453–461. [Google Scholar] [CrossRef]
  21. Goossens, G.H.; Jocken, J.W.E.; Blaak, E.E. Sexual dimorphism in cardiometabolic health: The role of adipose tissue, muscle and liver. Nat. Rev. Endocrinol. 2021, 17, 47–66. [Google Scholar] [CrossRef]
  22. Jiang, X.; Zhao, Y.; Yang, Q.; Wang, W.; Lin, T.; Qiu, Y. Gender differences in associations between obesity and hypertension, diabetes, dyslipidemia: Evidence from electronic health records of 3.5 million Chinese senior population. BMC Public Health 2025, 25, 405. [Google Scholar] [CrossRef]
  23. Nauli, A.M.; Matin, S. Why Do Men Accumulate Abdominal Visceral Fat? Front. Physiol. 2019, 10, 1486. [Google Scholar] [CrossRef]
  24. Kolb, H. Obese visceral fat tissue inflammation: From protective to detrimental? BMC Med. 2022, 20, 494. [Google Scholar] [CrossRef] [PubMed]
  25. McArdle, M.A.; Finucane, O.M.; Connaughton, R.M.; McMorrow, A.M.; Roche, H.M. Mechanisms of obesity-induced inflammation and insulin resistance: Insights into the emerging role of nutritional strategies. Front. Endocrinol. 2013, 4, 52. [Google Scholar] [CrossRef] [PubMed]
  26. Bovolini, A.; Garcia, J.; Andrade, M.A.; Duarte, J.A. Metabolic Syndrome Pathophysiology and Predisposing Factors. Int. J. Sports Med. 2021, 42, 199–214. [Google Scholar] [CrossRef]
  27. Karastergiou, K.; Smith, S.R.; Greenberg, A.S.; Fried, S.K. Sex differences in human adipose tissues—The biology of pear shape. Biol. Sex. Differ. 2012, 3, 13. [Google Scholar] [CrossRef] [PubMed]
  28. Shetty, S.; Duesman, S.J.; Patel, S.; Huynh, P.; Toh, P.; Shroff, S.; Das, A.; Chowhan, D.; Keller, B.; Alvarez, J.; et al. Sex-specific role of high-fat diet and stress on behavior, energy metabolism, and the ventromedial hypothalamus. Biol. Sex. Differ. 2024, 15, 55. [Google Scholar] [CrossRef]
  29. Bracht, J.R.; Vieira-Potter, V.J.; De Souza Santos, R.; Öz, O.K.; Palmer, B.F.; Clegg, D.J. The role of estrogens in the adipose tissue milieu. Ann. N. Y. Acad. Sci. 2020, 1461, 127–143. [Google Scholar] [CrossRef]
  30. Adam, S.; Maas, S.L.; Huchzermeier, R.; Rakateli, L.; Abschlag, K.; Hohl, M.; Liao, L.; Bartneck, M.; Teunissen, M.; Wouters, K.; et al. The calcium-sensing-receptor (CaSR) in adipocytes contributes to sex-differences in the susceptibility to high fat diet induced obesity and atherosclerosis. EBioMedicine 2024, 107, 105293. [Google Scholar] [CrossRef] [PubMed]
  31. Sun, H.; Saeedi, P.; Karuranga, S.; Pinkepank, M.; Ogurtsova, K.; Duncan, B.B.; Stein, C.; Basit, A.; Chan, J.C.N.; Mbanya, J.C.; et al. IDF Diabetes Atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res. Clin. Pract. 2022, 183, 109119. [Google Scholar] [CrossRef]
  32. James, D.E.; Stöckli, J.; Birnbaum, M.J. The aetiology and molecular landscape of insulin resistance. Nat. Rev. Mol. Cell Biol. 2021, 22, 751–771. [Google Scholar] [CrossRef]
  33. Du, W.; Jiang, S.; Yin, S.; Wang, R.; Zhang, C.; Yin, B.C.; Li, J.; Li, L.; Qi, N.; Zhou, Y.; et al. The microbiota-dependent tryptophan metabolite alleviates high-fat diet-induced insulin resistance through the hepatic AhR/TSC2/mTORC1 axis. Proc. Natl. Acad. Sci. USA 2024, 121, e2400385121. [Google Scholar] [CrossRef] [PubMed]
  34. Elkanawati, R.Y.; Sumiwi, S.A.; Levita, J. Impact of Lipids on Insulin Resistance: Insights from Human and Animal Studies. Drug Des. Dev. Ther. 2024, 18, 3337–3360. [Google Scholar] [CrossRef] [PubMed]
  35. Zeng, N.; Wu, F.; Lu, J.; Li, X.; Lin, S.; Zhou, L.; Wang, Z.; Wu, G.; Huang, Q.; Zheng, D.; et al. High-fat diet impairs gut barrier through intestinal microbiota-derived reactive oxygen species. Sci. China Life Sci. 2024, 67, 879–891. [Google Scholar] [CrossRef]
  36. Li, M.; Chi, X.; Wang, Y.; Setrerrahmane, S.; Xie, W.; Xu, H. Trends in insulin resistance: Insights into mechanisms and therapeutic strategy. Signal Transduct. Target. Ther. 2022, 7, 216. [Google Scholar] [CrossRef]
  37. Sakamoto, K.; Butera, M.A.; Zhou, C.; Maurizi, G.; Chen, B.; Ling, L.; Shawkat, A.; Patlolla, L.; Thakker, K.; Calle, V.; et al. Overnutrition causes insulin resistance and metabolic disorder through increased sympathetic nervous system activity. Cell Metab. 2025, 37, 121–137.E6. [Google Scholar] [CrossRef] [PubMed]
  38. Abdou, M.; Hafez, M.H.; Anwar, G.M.; Fahmy, W.A.; Abd Alfattah, N.M.; Salem, R.I.; Arafa, N. Effect of high protein and fat diet on postprandial blood glucose levels in children and adolescents with type 1 diabetes in Cairo, Egypt. Diabetes Metab. Syndr. 2021, 15, 7–12. [Google Scholar] [CrossRef]
  39. Smith, J.A.; Soares, R.N.; McMillan, N.J.; Jurrissen, T.J.; Martinez-Lemus, L.A.; Padilla, J.; Manrique-Acevedo, C. Young Women Are Protected Against Vascular Insulin Resistance Induced by Adoption of an Obesogenic Lifestyle. Endocrinology 2022, 163, bqac137. [Google Scholar] [CrossRef]
  40. Varlamov, O.; Bethea, C.L.; Roberts, C.T., Jr. Sex-specific differences in lipid and glucose metabolism. Front. Endocrinol. 2014, 5, 241. [Google Scholar] [CrossRef]
  41. Churuangsuk, C.; Lean, M.E.J.; Combet, E. Lower carbohydrate and higher fat intakes are associated with higher hemoglobin A1c: Findings from the UK National Diet and Nutrition Survey 2008–2016. Eur. J. Nutr. 2020, 59, 2771–2782. [Google Scholar] [CrossRef]
  42. Palmer, B.F.; Clegg, D.J. The sexual dimorphism of obesity. Mol. Cell Endocrinol. 2015, 402, 113–119. [Google Scholar] [CrossRef]
  43. Speksnijder, E.M.; Ten Noever de Brauw, G.V.; Malekzadeh, A.; Bisschop, P.H.; Stenvers, D.J.; Siegelaar, S.E. Effect of Postmenopausal Hormone Therapy on Glucose Regulation in Women With Type 1 or Type 2 Diabetes: A Systematic Review and Meta-analysis. Diabetes Care 2023, 46, 1866–1875. [Google Scholar] [CrossRef] [PubMed]
  44. Yazdkhasti, M.; Jafarabady, K.; Shafiee, A.; Omran, S.P.; Mahmoodi, Z.; Esmaeilzadeh, S.; Babaheidari, T.B.; Kabir, K.; Peisepar, M.; Bakhtiyari, M. The association between age of menopause and type 2 diabetes: A systematic review and meta-analysis. Nutr. Metab. 2024, 21, 87. [Google Scholar] [CrossRef] [PubMed]
  45. Lizcano, F.; Guzmán, G. Estrogen Deficiency and the Origin of Obesity during Menopause. Biomed. Res. Int. 2014, 2014, 757461. [Google Scholar] [CrossRef] [PubMed]
  46. Szukiewicz, D. Molecular Mechanisms for the Vicious Cycle between Insulin Resistance and the Inflammatory Response in Obesity. Int. J. Mol. Sci. 2023, 24, 9818. [Google Scholar] [CrossRef]
  47. Sjøberg, K.A.; Frøsig, C.; Kjøbsted, R.; Sylow, L.; Kleinert, M.; Betik, A.C.; Shaw, C.S.; Kiens, B.; Wojtaszewski, J.F.P.; Rattigan, S.; et al. Exercise Increases Human Skeletal Muscle Insulin Sensitivity via Coordinated Increases in Microvascular Perfusion and Molecular Signaling. Diabetes 2017, 66, 1501–1510. [Google Scholar] [CrossRef]
  48. Gupta, D.; Jetton, T.L.; LaRock, K.; Monga, N.; Satish, B.; Lausier, J.; Peshavaria, M.; Leahy, J.L. Temporal characterization of β cell-adaptive and -maladaptive mechanisms during chronic high-fat feeding in C57BL/6NTac mice. J. Biol. Chem. 2017, 292, 12449–12459. [Google Scholar] [CrossRef]
  49. Volpe, C.M.O.; Villar-Delfino, P.H.; Dos Anjos, P.M.F.; Nogueira-Machado, J.A. Cellular death, reactive oxygen species (ROS) and diabetic complications. Cell Death Dis. 2018, 9, 119. [Google Scholar] [CrossRef]
  50. Eguchi, N.; Vaziri, N.D.; Dafoe, D.C.; Ichii, H. The Role of Oxidative Stress in Pancreatic β Cell Dysfunction in Diabetes. Int. J. Mol. Sci. 2021, 22, 1509. [Google Scholar] [CrossRef]
  51. Zhou, Z.; Ribas, V.; Rajbhandari, P.; Drew, B.G.; Moore, T.M.; Fluitt, A.H.; Reddish, B.R.; Whitney, K.A.; Georgia, S.; Vergnes, L.; et al. Estrogen receptor α protects pancreatic β-cells from apoptosis by preserving mitochondrial function and suppressing endoplasmic reticulum stress. J. Biol. Chem. 2018, 293, 4735–4751. [Google Scholar] [CrossRef]
  52. De Paoli, M.; Zakharia, A.; Werstuck, G.H. The Role of Estrogen in Insulin Resistance: A Review of Clinical and Preclinical Data. Am. J. Pathol. 2021, 191, 1490–1498. [Google Scholar] [CrossRef]
  53. Ribas, V.; Drew, B.G.; Zhou, Z.; Phun, J.; Kalajian, N.Y.; Soleymani, T.; Daraei, P.; Widjaja, K.; Wanagat, J.; de Aguiar Vallim, T.Q.; et al. Skeletal muscle action of estrogen receptor α is critical for the maintenance of mitochondrial function and metabolic homeostasis in females. Sci. Transl. Med. 2016, 8, 334ra54. [Google Scholar] [CrossRef] [PubMed]
  54. Moholdt, T.; Parr, E.B.; Devlin, B.L.; Giskeødegård, G.F.; Hawley, J.A. Effect of high-fat diet and morning or evening exercise on lipoprotein subfraction profiles: Secondary analysis of a randomised trial. Sci. Rep. 2023, 13, 4008. [Google Scholar] [CrossRef] [PubMed]
  55. Shinohata, R.; Shibakura, M.; Arao, Y.; Watanabe, S.; Hirohata, S.; Usui, S. A high-fat/high-cholesterol diet, but not high-cholesterol alone, increases free cholesterol and apoE-rich HDL serum levels in rats and upregulates hepatic ABCA1 expression. Biochimie 2022, 197, 49–58. [Google Scholar] [CrossRef] [PubMed]
  56. Ye, Y.; Zhou, Q.; Dai, W.; Peng, H.; Zhou, S.; Tian, H.; Shen, L.; Han, H. Gender differences in metabolic syndrome and its components in southern china using a healthy lifestyle index: A cross-sectional study. BMC Public Health 2023, 23, 686. [Google Scholar] [CrossRef]
  57. Ambikairajah, A.; Walsh, E.; Cherbuin, N. Lipid profile differences during menopause: A review with meta-analysis. Menopause 2019, 26, 1327–1333. [Google Scholar] [CrossRef]
  58. McCoin, C.S.; Von Schulze, A.; Allen, J.; Fuller, K.N.Z.; Xia, Q.; Koestler, D.C.; Houchen, C.J.; Maurer, A.; Dorn, G.W., 2nd; Shankar, K.; et al. Sex modulates hepatic mitochondrial adaptations to high-fat diet and physical activity. Am. J. Physiol. Endocrinol. Metab. 2019, 317, E298–E311. [Google Scholar] [CrossRef]
  59. Arcones, A.C.; Cruces-Sande, M.; Ramos, P.; Mayor, F., Jr.; Murga, C. Sex Differences in High Fat Diet-Induced Metabolic Alterations Correlate with Changes in the Modulation of GRK2 Levels. Cells 2019, 8, 1464. [Google Scholar] [CrossRef]
  60. Ryczkowska, K.; Adach, W.; Janikowski, K.; Banach, M.; Bielecka-Dabrowa, A. Menopause and women’s cardiovascular health: Is it really an obvious relationship? Arch. Med. Sci. 2023, 19, 458–466. [Google Scholar] [CrossRef]
  61. Ko, S.H.; Jung, Y. Energy Metabolism Changes and Dysregulated Lipid Metabolism in Postmenopausal Women. Nutrients 2021, 13, 4556. [Google Scholar] [CrossRef]
  62. Spitler, K.M.; Davies, B.S.J. Aging and plasma triglyceride metabolism. J. Lipid Res. 2020, 61, 1161–1167. [Google Scholar] [CrossRef]
  63. Nie, G.; Yang, X.; Wang, Y.; Liang, W.; Li, X.; Luo, Q.; Yang, H.; Liu, J.; Wang, J.; Guo, Q.; et al. The Effects of Menopause Hormone Therapy on Lipid Profile in Postmenopausal Women: A Systematic Review and Meta-Analysis. Front. Pharmacol. 2022, 13, 850815. [Google Scholar] [CrossRef] [PubMed]
  64. Ko, S.H.; Kim, H.S. Menopause-Associated Lipid Metabolic Disorders and Foods Beneficial for Postmenopausal Women. Nutrients 2020, 12, 202. [Google Scholar] [CrossRef] [PubMed]
  65. Szudzik, M.; Hutsch, T.; Chabowski, D.; Zajdel, M.; Ufnal, M. Normal caloric intake with high-fat diet induces metabolic dysfunction-associated steatotic liver disease and dyslipidemia without obesity in rats. Sci. Rep. 2024, 14, 22796. [Google Scholar] [CrossRef]
  66. Gu, Y.; Han, F.; Xue, M.; Wang, M.; Huang, Y. The benefits and risks of menopause hormone therapy for the cardiovascular system in postmenopausal women: A systematic review and meta-analysis. BMC Womens Health 2024, 24, 60. [Google Scholar] [CrossRef] [PubMed]
  67. Hodis, H.N.; Mack, W.J. Menopausal Hormone Replacement Therapy and Reduction of All-Cause Mortality and Cardiovascular Disease: It Is About Time and Timing. Cancer J. 2022, 28, 208–223. [Google Scholar] [CrossRef]
  68. Jiang, H.; Zhou, Y.; Nabavi, S.M.; Sahebkar, A.; Little, P.J.; Xu, S.; Weng, J.; Ge, J. Mechanisms of Oxidized LDL-Mediated Endothelial Dysfunction and Its Consequences for the Development of Atherosclerosis. Front. Cardiovasc. Med. 2022, 9, 925923. [Google Scholar] [CrossRef]
  69. Gui, Y.; Zheng, H.; Cao, R.Y. Foam Cells in Atherosclerosis: Novel Insights Into Its Origins, Consequences, and Molecular Mechanisms. Front. Cardiovasc. Med. 2022, 9, 845942. [Google Scholar] [CrossRef]
  70. Libby, P.; Buring, J.E.; Badimon, L.; Hansson, G.K.; Deanfield, J.; Bittencourt, M.S.; Tokgözoğlu, L.; Lewis, E.F. Atherosclerosis. Nat. Rev. Dis. Primers 2019, 5, 56. [Google Scholar] [CrossRef]
  71. Yerly, A.; van der Vorst, E.P.C.; Baumgartner, I.; Bernhard, S.M.; Schindewolf, M.; Döring, Y. Sex-specific and hormone-related differences in vascular remodelling in atherosclerosis. Eur. J. Clin. Investig. 2023, 53, e13885. [Google Scholar] [CrossRef]
  72. Ren, Z.; Guo, J.; Xiao, X.; Huang, J.; Li, M.; Cai, R.; Zeng, H. The Effect of Sex Differences on Endothelial Function and Circulating Endothelial Progenitor Cells in Hypertriglyceridemia. Cardiol. Res. Pract. 2020, 2020, 2132918. [Google Scholar] [CrossRef]
  73. Fairweather, D. Sex differences in inflammation during atherosclerosis. Clin. Med. Insights Cardiol. 2014, 8 (Suppl. S3), 49–59. [Google Scholar] [CrossRef] [PubMed]
  74. Vakhtangadze, T.; Singh Tak, R.; Singh, U.; Baig, M.S.; Bezsonov, E. Gender Differences in Atherosclerotic Vascular Disease: From Lipids to Clinical Outcomes. Front. Cardiovasc. Med. 2021, 8, 707889. [Google Scholar] [CrossRef] [PubMed]
  75. Wu, X.; Zhang, Y.; Yao, C.; Zhao, X.; Xiong, W.; Rang, W. Trend and Prediction of Changes in the Disease Burden of Diet-related Ischemic Heart Disease in China, 1990–2021. Chin. Gen. Pract. 2025, 28, 305–312. (In Chinese) [Google Scholar]
  76. Regitz-Zagrosek, V.; Gebhard, C. Gender medicine: Effects of sex and gender on cardiovascular disease manifestation and outcomes. Nat. Rev. Cardiol. 2023, 20, 236–247. [Google Scholar] [CrossRef]
  77. Rexrode, K.M.; Madsen, T.E.; Yu, A.Y.X.; Carcel, C.; Lichtman, J.H.; Miller, E.C. The Impact of Sex and Gender on Stroke. Circ. Res. 2022, 130, 512–528. [Google Scholar] [CrossRef]
  78. Jia, Z.; Wang, Z.; Pan, H.; Zhang, J.; Wang, Q.; Zhou, C.; Liu, J. Crosstalk between fat tissue and muscle, brain, liver, and heart in obesity: Cellular and molecular perspectives. Eur. J. Med. Res. 2024, 29, 637. [Google Scholar] [CrossRef]
  79. Funcke, J.B.; Scherer, P.E. Beyond adiponectin and leptin: Adipose tissue-derived mediators of inter-organ communication. J. Lipid Res. 2019, 60, 1648–1684. [Google Scholar]
  80. Moghbeli, M.; Khedmatgozar, H.; Yadegari, M.; Avan, A.; Ferns, G.A.; Ghayour Mobarhan, M. Cytokines and the immune response in obesity-related disorders. Adv. Clin. Chem. 2021, 101, 135–168. [Google Scholar] [PubMed]
  81. Strizova, Z.; Benesova, I.; Bartolini, R.; Novysedlak, R.; Cecrdlova, E.; Foley, L.K.; Striz, I. M1/M2 macrophages and their overlaps—Myth or reality? Clin. Sci. 2023, 137, 1067–1093. [Google Scholar] [CrossRef]
  82. Li, Y.; Dong, X.; He, W.; Quan, H.; Chen, K.; Cen, C.; Wei, W. Ube2L6 Promotes M1 Macrophage Polarization in High-Fat Diet-Fed Obese Mice via ISGylation of STAT1 to Trigger STAT1 Activation. Obes. Facts 2024, 17, 24–36. [Google Scholar] [CrossRef]
  83. Duan, Z.; Luo, Y. Targeting macrophages in cancer immunotherapy. Signal Transduct. Target. Ther. 2021, 6, 127. [Google Scholar] [CrossRef] [PubMed]
  84. Alfaddagh, A.; Martin, S.S.; Leucker, T.M.; Michos, E.D.; Blaha, M.J.; Lowenstein, C.J.; Jones, S.R.; Toth, P.P. Inflammation and cardiovascular disease: From mechanisms to therapeutics. Am. J. Prev. Cardiol. 2020, 4, 100130. [Google Scholar] [CrossRef] [PubMed]
  85. Liu, Y.; Chen, X.; Zhang, H.G. Editorial: Cardiac Hypertrophy: From Compensation to Decompensation and Pharmacological Interventions. Front. Pharmacol. 2021, 12, 665936. [Google Scholar] [CrossRef]
  86. Braga Tibaes, J.R.; Azarcoya-Barrera, J.; Wollin, B.; Veida-Silva, H.; Makarowski, A.; Vine, D.; Tsai, S.; Jacobs, R.; Richard, C. Sex Differences Distinctly Impact High-Fat Diet-Induced Immune Dysfunction in Wistar Rats. J. Nutr. 2022, 152, 1347–1357. [Google Scholar] [CrossRef]
  87. Peng, Y.; Zhou, M.; Yang, H.; Qu, R.; Qiu, Y.; Hao, J.; Bi, H.; Guo, D. Regulatory Mechanism of M1/M2 Macrophage Polarization in the Development of Autoimmune Diseases. Mediators Inflamm. 2023, 2023, 8821610. [Google Scholar] [CrossRef] [PubMed]
  88. Shu, Z.; Zhang, G.; Zhu, X.; Xiong, W. Estrogen receptor α mediated M1/M2 macrophages polarization plays a critical role in NASH of female mice. Biochem. Biophys. Res. Commun. 2022, 596, 63–70. [Google Scholar] [CrossRef]
  89. Liu, H.; Liu, K.; Bodenner, D.L. Estrogen receptor inhibits interleukin-6 gene expression by disruption of nuclear factor kappaB transactivation. Cytokine 2005, 31, 251–257. [Google Scholar] [CrossRef]
  90. Kurmann, L.; Azzarito, G.; Leeners, B.; Rosselli, M.; Dubey, R.K. 17β-Estradiol Abrogates TNF-α-Induced Human Brain Vascular Pericyte Migration by Downregulating miR-638 via ER-β. Int. J. Mol. Sci. 2024, 25, 11416. [Google Scholar] [CrossRef]
  91. Bandala, C.; Cárdenas-Rodríguez, N.; Reyes-Long, S.; Cortés-Algara, A.; Contreras-García, I.J.; Cruz-Hernández, T.R.; Alfaro-Rodriguez, A.; Cortes-Altamirano, J.L.; Perez-Santos, M.; Anaya-Ruiz, M.; et al. Estrogens as a Possible Therapeutic Strategy for the Management of Neuroinflammation and Neuroprotection in COVID-19. Curr. Neuropharmacol. 2023, 21, 2110–2125. [Google Scholar] [CrossRef]
  92. Ketchem, J.M.; Bowman, E.J.; Isales, C.M. Male sex hormones, aging, and inflammation. Biogerontology 2023, 24, 1–25. [Google Scholar] [CrossRef]
  93. Koning, T.; Calaf, G.M. Association of Inflammation and Immune Cell Infiltration with Estrogen Receptor Alpha in an Estrogen and Ionizing Radiation-Induced Breast Cancer Model. Int. J. Mol. Sci. 2024, 25, 8604. [Google Scholar] [CrossRef] [PubMed]
  94. Zhu, B.T.; Liao, Q.Q.; Tian, H.Y.; Yu, D.J.; Xie, T.; Sun, X.L.; Zhou, X.M.; Han, Y.X.; Zhao, Y.J.; El-Kassas, M.; et al. Estrogen: The forgotten player in metaflammation. Front. Pharmacol. 2024, 15, 1478819. [Google Scholar] [CrossRef] [PubMed]
  95. Kovats, S. Estrogen receptors regulate innate immune cells and signaling pathways. Cell Immunol. 2015, 294, 63–69. [Google Scholar] [CrossRef] [PubMed]
  96. Steppan, J.; Jandu, S.; Wang, H.; Kang, S.; Savage, W.; Narayanan, R.; Nandakumar, K.; Santhanam, L. Commonly used mouse strains have distinct vascular properties. Hypertens. Res. 2020, 43, 1175–1181. [Google Scholar] [CrossRef]
  97. Iorga, A.; Cunningham, C.M.; Moazeni, S.; Ruffenach, G.; Umar, S.; Eghbali, M. The protective role of estrogen and estrogen receptors in cardiovascular disease and the controversial use of estrogen therapy. Biol. Sex. Differ. 2017, 8, 33. [Google Scholar] [CrossRef]
  98. Schieber, M.; Chandel, N.S. ROS function in redox signaling and oxidative stress. Curr. Biol. 2014, 24, R453–R462. [Google Scholar] [CrossRef]
  99. Marseglia, L.; Manti, S.; D’Angelo, G.; Nicotera, A.; Parisi, E.; Di Rosa, G.; Gitto, E.; Arrigo, T. Oxidative stress in obesity: A critical component in human diseases. Int. J. Mol. Sci. 2014, 16, 378–400. [Google Scholar] [CrossRef]
  100. Bej, E.; Cesare, P.; Volpe, A.R.; d’Angelo, M.; Castelli, V. Oxidative Stress and Neurodegeneration: Insights and Therapeutic Strategies for Parkinson’s Disease. Neurol. Int. 2024, 16, 502–517. [Google Scholar] [CrossRef]
  101. Jin, S.; Kang, P.M. A Systematic Review on Advances in Management of Oxidative Stress-Associated Cardiovascular Diseases. Antioxidants 2024, 13, 923. [Google Scholar] [CrossRef]
  102. Chaudhary, P.; Janmeda, P.; Docea, A.O.; Yeskaliyeva, B.; Abdull Razis, A.F.; Modu, B.; Calina, D.; Sharifi-Rad, J. Oxidative stress, free radicals and antioxidants: Potential crosstalk in the pathophysiology of human diseases. Front. Chem. 2023, 11, 1158198. [Google Scholar] [CrossRef]
  103. Tiberi, J.; Cesarini, V.; Stefanelli, R.; Canterini, S.; Fiorenza, M.T.; La Rosa, P. Sex differences in antioxidant defence and the regulation of redox homeostasis in physiology and pathology. Mech. Ageing Dev. 2023, 211, 111802. [Google Scholar] [CrossRef] [PubMed]
  104. Diaz-Castro, J.; Pulido-Moran, M.; Moreno-Fernandez, J.; Kajarabille, N.; de Paco, C.; Garrido-Sanchez, M.; Prados, S.; Ochoa, J.J. Gender specific differences in oxidative stress and inflammatory signaling in healthy term neonates and their mothers. Pediatr. Res. 2016, 80, 595–601. [Google Scholar] [CrossRef] [PubMed]
  105. Xiang, D.; Liu, Y.; Zhou, S.; Zhou, E.; Wang, Y. Protective Effects of Estrogen on Cardiovascular Disease Mediated by Oxidative Stress. Oxid. Med. Cell. Longev. 2021, 2021, 5523516. [Google Scholar] [CrossRef] [PubMed]
  106. Borrás, C.; Ferrando, M.; Inglés, M.; Gambini, J.; Lopez-Grueso, R.; Edo, R.; Mas-Bargues, C.; Pellicer, A.; Viña, J. Estrogen Replacement Therapy Induces Antioxidant and Longevity-Related Genes in Women after Medically Induced Menopause. Oxid. Med. Cell. Longev. 2021, 2021, 8101615. [Google Scholar] [CrossRef]
  107. Sahoo, D.K.; Samanta, L.; Kesari, K.K.; Mukherjee, S. Editorial: Hormonal imbalance-associated oxidative stress and protective benefits of nutritional antioxidants. Front. Endocrinol. 2024, 15, 1368580. [Google Scholar] [CrossRef]
  108. Ayala, A.; Muñoz, M.F.; Argüelles, S. Lipid peroxidation: Production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxid. Med. Cell. Longev. 2014, 2014, 360438. [Google Scholar] [CrossRef]
  109. Maithili Karpaga Selvi, N.; Sridhar, M.G.; Swaminathan, R.P.; Sripradha, R. Curcumin Attenuates Oxidative Stress and Activation of Redox-Sensitive Kinases in High Fructose- and High-Fat-Fed Male Wistar Rats. Sci. Pharm. 2015, 83, 159–175. [Google Scholar] [CrossRef]
  110. Hong, Y.; Boiti, A.; Vallone, D.; Foulkes, N.S. Reactive Oxygen Species Signaling and Oxidative Stress: Transcriptional Regulation and Evolution. Antioxidants 2024, 13, 312. [Google Scholar] [CrossRef]
  111. Darbandi, M.; Darbandi, S.; Agarwal, A.; Sengupta, P.; Durairajanayagam, D.; Henkel, R.; Sadeghi, M.R. Reactive oxygen species and male reproductive hormones. Reprod. Biol. Endocrinol. 2018, 16, 87. [Google Scholar] [CrossRef]
  112. Tostes, R.C.; Carneiro, F.S.; Carvalho, M.H.; Reckelhoff, J.F. Reactive oxygen species: Players in the cardiovascular effects of testosterone. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2016, 310, R1–R14. [Google Scholar] [CrossRef]
  113. Lavorando, E.; Owens, M.C.; Liu, K.F. Comparing the roles of sex chromosome-encoded protein homologs in gene regulation. Genes. Dev. 2024, 38, 585–596. [Google Scholar] [CrossRef] [PubMed]
  114. Tukiainen, T.; Villani, A.C.; Yen, A.; Rivas, M.A.; Marshall, J.L.; Satija, R.; Aguirre, M.; Gauthier, L.; Fleharty, M.; Kirby, A.; et al. Landscape of X chromosome inactivation across human tissues. Nature 2017, 550, 244–248. [Google Scholar] [CrossRef] [PubMed]
  115. Reue, K. Sex differences in obesity: X chromosome dosage as a risk factor for increased food intake, adiposity and co-morbidities. Physiol. Behav. 2017, 176, 174–182. [Google Scholar] [CrossRef] [PubMed]
  116. Chen, X.; McClusky, R.; Itoh, Y.; Reue, K.; Arnold, A.P. X and Y chromosome complement influence adiposity and metabolism in mice. Endocrinology 2013, 154, 1092–1104. [Google Scholar] [CrossRef]
  117. White, M.; Zacharin, M.R.; Fawcett, S.; McGillivray, G. Klinefelter Syndrome: What should we tell prospective parents? Prenat. Diagn. 2023, 43, 240–249. [Google Scholar] [CrossRef]
  118. Granato, S.; Barbaro, G.; Di Giorgio, M.R.; Rossi, F.M.; Marzano, C.; Impronta, F.; Spaziani, M.; Anzuini, A.; Lenzi, A.; Radicioni, A.F. Epicardial fat: The role of testosterone and lipid metabolism in a cohort of patients with Klinefelter syndrome. Metabolism 2019, 95, 21–26. [Google Scholar] [CrossRef]
  119. Demirci, I.; Haymana, C.; Candemir, B.; Yuksel, B.; Eser, M.; Meric, C.; Akin, S.; Gulcelik, N.E.; Sonmez, A. Triglyceride-glucose index levels in patients with Klinefelter syndrome and its relationship with endothelial dysfunction and insulin resistance: A cross-sectional observational study. Arch. Endocrinol. Metab. 2023, 67, 378–384. [Google Scholar] [CrossRef]
  120. Krzyścin, M.; Sowińska-Przepiera, E.; Gruca-Stryjak, K.; Soszka-Przepiera, E.; Syrenicz, I.; Przepiera, A.; Bumbulienė, Ž.; Syrenicz, A. Are Young People with Turner Syndrome Who Have Undergone Treatment with Growth and Sex Hormones at Higher Risk of Metabolic Syndrome and Its Complications? Biomedicines 2024, 12, 1034. [Google Scholar] [CrossRef]
  121. Link, J.C.; Wiese, C.B.; Chen, X.; Avetisyan, R.; Ronquillo, E.; Ma, F.; Guo, X.; Yao, J.; Allison, M.; Chen, Y.I.; et al. X chromosome dosage of histone demethylase KDM5C determines sex differences in adiposity. J. Clin. Investig. 2020, 130, 5688–5702. [Google Scholar] [CrossRef]
  122. Qian, S.; Wang, Y.; Ma, H.; Zhang, L. Expansion and Functional Divergence of Jumonji C-Containing Histone Demethylases: Significance of Duplications in Ancestral Angiosperms and Vertebrates. Plant Physiol. 2015, 168, 1321–1337. [Google Scholar] [CrossRef]
  123. Godfrey, A.K.; Naqvi, S.; Chmátal, L.; Chick, J.M.; Mitchell, R.N.; Gygi, S.P.; Skaletsky, H.; Page, D.C. Quantitative analysis of Y-Chromosome gene expression across 36 human tissues. Genome Res. 2020, 30, 860–873. [Google Scholar] [CrossRef] [PubMed]
  124. Cortez, D.; Marin, R.; Toledo-Flores, D.; Froidevaux, L.; Liechti, A.; Waters, P.D.; Grützner, F.; Kaessmann, H. Origins and functional evolution of Y chromosomes across mammals. Nature 2014, 508, 488–493. [Google Scholar] [CrossRef] [PubMed]
  125. Mortensen, S.B.; Jensen, C.H.; Schneider, M.; Thomassen, M.; Kruse, T.A.; Laborda, J.; Sheikh, S.P.; Andersen, D.C. Membrane-tethered delta-like 1 homolog (DLK1) restricts adipose tissue size by inhibiting preadipocyte proliferation. Diabetes 2012, 61, 2814–2822. [Google Scholar] [CrossRef] [PubMed]
  126. Vergnes, L.; Wiese, C.B.; Zore, T.; Riestenberg, C.; Avetisyan, R.; Reue, K. Gene Regulation and Mitochondrial Activity During White and Brown Adipogenesis Are Modulated by KDM5 Histone Demethylase. J. Endocr. Soc. 2024, 8, bvae029. [Google Scholar] [CrossRef]
  127. Long, J.; Su, Y.X.; Deng, H.C. Lipoapoptosis pathways in pancreatic β-cells and the anti-apoptosis mechanisms of adiponectin. Horm. Metab. Res. 2014, 46, 722–727. [Google Scholar] [CrossRef]
  128. Zhang, Y.; Huang, F.; Luo, Q.; Wu, X.; Liu, Z.; Chen, H.; Huang, Y. Inhibition of XIAP increases carboplatin sensitivity in ovarian cancer. OncoTargets Ther. 2018, 11, 8751–8759. [Google Scholar] [CrossRef]
  129. Jiang, C.; Yi, X.P.; Shen, H.; Li, Y.X. Targeting X-linked inhibitor of apoptosis protein inhibits pancreatic cancer cell growth through p-Akt depletion. World J. Gastroenterol. 2012, 18, 2956–2965. [Google Scholar] [CrossRef]
  130. Zhao, Y.; Stankovic, S.; Koprulu, M.; Wheeler, E.; Day, F.R.; Lango Allen, H.; Kerrison, N.D.; Pietzner, M.; Loh, P.R.; Wareham, N.J.; et al. GIGYF1 loss of function is associated with clonal mosaicism and adverse metabolic health. Nat. Commun. 2021, 12, 4178. [Google Scholar] [CrossRef]
  131. Wu, X.N.; Wang, M.Z.; Zhang, N.; Zhang, W.; Dong, J.; Ke, M.Y.; Xiang, J.X.; Ma, F.; Xue, F.; Hou, J.J.; et al. Sex-determining region Y gene promotes liver fibrosis and accounts for sexual dimorphism in its pathophysiology. J. Hepatol. 2024, 80, 928–940. [Google Scholar] [CrossRef]
  132. Wang, P.; Li, Q.; Wu, L.; Yu, X.; Zheng, Y.; Liu, J.; Yao, J.; Liu, Z.; Fan, S.; Li, Y. Association between the weight-adjusted-waist index and testosterone deficiency in adult males: A cross-sectional study. Sci. Rep. 2024, 14, 25574. [Google Scholar] [CrossRef]
  133. Santosa, S.; Bush, N.C.; Jensen, M.D. Acute Testosterone Deficiency Alters Adipose Tissue Fatty Acid Storage. J. Clin. Endocrinol. Metab. 2017, 102, 3056–3064. [Google Scholar] [CrossRef] [PubMed]
  134. Miller, C.; Madden-Doyle, L.; Jayasena, C.; McIlroy, M.; Sherlock, M.; O’Reilly, M.W. Mechanisms in endocrinology: Hypogonadism and metabolic health in men-novel insights into pathophysiology. Eur. J. Endocrinol. 2024, 191, R1–R17. [Google Scholar] [CrossRef] [PubMed]
  135. Mauvais-Jarvis, F.; Bairey Merz, N.; Barnes, P.J.; Brinton, R.D.; Carrero, J.J.; DeMeo, D.L.; De Vries, G.J.; Epperson, C.N.; Govindan, R.; Klein, S.L.; et al. Sex and gender: Modifiers of health, disease, and medicine. Lancet 2020, 396, 565–582. [Google Scholar] [CrossRef]
  136. Naamneh Elzenaty, R.; du Toit, T.; Flück, C.E. Basics of androgen synthesis and action. Best. Pract. Res. Clin. Endocrinol. Metab. 2022, 36, 101665. [Google Scholar] [CrossRef]
  137. Apaiajai, N.; Chunchai, T.; Jaiwongkam, T.; Kerdphoo, S.; Chattipakorn, S.C.; Chattipakorn, N. Testosterone Deprivation Aggravates Left-Ventricular Dysfunction in Male Obese Insulin-Resistant Rats via Impairing Cardiac Mitochondrial Function and Dynamics Proteins. Gerontology 2018, 64, 333–343. [Google Scholar] [CrossRef]
  138. Leciejewska, N.; Pruszynska-Oszmalek, E.; Bien, J.; Nogowski, L.; Kolodziejski, P.A. Effect of ostarine (enobosarm/GTX024), a selective androgen receptor modulator, on adipocyte metabolism in Wistar rats. J. Physiol. Pharmacol. 2019, 70, 525–533. [Google Scholar]
  139. Raza, M.T.; Sharif, S.; Khan, Z.A.; Naz, S.; Mushtaq, S.; Umer, A. Frequency of Hypogonadism in Type 2 Diabetes Mellitus Patients with and without Coronary Artery Disease. Cureus 2019, 11, e6500. [Google Scholar] [CrossRef]
  140. Ruth, K.S.; Day, F.R.; Tyrrell, J.; Thompson, D.J.; Wood, A.R.; Mahajan, A.; Beaumont, R.N.; Wittemans, L.; Martin, S.; Busch, A.S.; et al. Using human genetics to understand the disease impacts of testosterone in men and women. Nat. Med. 2020, 26, 252–258. [Google Scholar] [CrossRef]
  141. Dhindsa, S.; Ghanim, H.; Batra, M.; Kuhadiya, N.D.; Abuaysheh, S.; Sandhu, S.; Green, K.; Makdissi, A.; Hejna, J.; Chaudhuri, A.; et al. Insulin Resistance and Inflammation in Hypogonadotropic Hypogonadism and Their Reduction After Testosterone Replacement in Men with Type 2 Diabetes. Diabetes Care 2016, 39, 82–91. [Google Scholar] [CrossRef]
  142. Yassin, A.; Haider, A.; Haider, K.S.; Caliber, M.; Doros, G.; Saad, F.; Garvey, W.T. Testosterone Therapy in Men With Hypogonadism Prevents Progression from Prediabetes to Type 2 Diabetes: Eight-Year Data From a Registry Study. Diabetes Care 2019, 42, 1104–1111. [Google Scholar] [CrossRef]
  143. Saad, F.; Yassin, A.; Doros, G.; Haider, A. Effects of long-term treatment with testosterone on weight and waist size in 411 hypogonadal men with obesity classes I-III: Observational data from two registry studies. Int. J. Obes. 2016, 40, 162–170. [Google Scholar] [CrossRef] [PubMed]
  144. Yu, I.C.; Lin, H.Y.; Liu, N.C.; Sparks, J.D.; Yeh, S.; Fang, L.Y.; Chen, L.; Chang, C. Neuronal androgen receptor regulates insulin sensitivity via suppression of hypothalamic NF-κB-mediated PTP1B expression. Diabetes 2013, 62, 411–423. [Google Scholar] [CrossRef]
  145. Yang, L.; Zhou, R.; Tong, Y.; Chen, P.; Shen, Y.; Miao, S.; Liu, X. Neuroprotection by dihydrotestosterone in LPS-induced neuroinflammation. Neurobiol. Dis. 2020, 140, 104814. [Google Scholar] [CrossRef] [PubMed]
  146. Antinozzi, C.; Marampon, F.; Corinaldesi, C.; Vicini, E.; Sgrò, P.; Vannelli, G.B.; Lenzi, A.; Crescioli, C.; Di Luigi, L. Testosterone insulin-like effects: An in vitro study on the short-term metabolic effects of testosterone in human skeletal muscle cells. J. Endocrinol. Investig. 2017, 40, 1133–1143. [Google Scholar] [CrossRef] [PubMed]
  147. Moreira-Pais, A.; Ferreira, R.; Neves, J.S.; Vitorino, R.; Moreira-Gonçalves, D.; Nogueira-Ferreira, R. Sex differences on adipose tissue remodeling: From molecular mechanisms to therapeutic interventions. J. Mol. Med. 2020, 98, 483–493. [Google Scholar] [CrossRef]
  148. Zhang, D.; Fan, J.; Liu, H.; Qiu, G.; Cui, S. Testosterone enhances taurine synthesis by upregulating androgen receptor and cysteine sulfinic acid decarboxylase expressions in male mouse liver. Am. J. Physiol. Gastrointest. Liver Physiol. 2023, 324, G295–G304. [Google Scholar] [CrossRef]
  149. Wei, W.; Lyu, X.; Markhard, A.L.; Fu, S.; Mardjuki, R.E.; Cavanagh, P.E.; Zeng, X.; Rajniak, J.; Lu, N.; Xiao, S.; et al. PTER is a N-acetyltaurine hydrolase that regulates feeding and obesity. Nature 2024, 633, 182–188. [Google Scholar] [CrossRef]
  150. Zhu, L.; Zhou, J.; Pan, Y.; Lv, J.; Liu, Y.; Yu, S.; Zhang, Y. Glucagon-like peptide-1 receptor expression and its functions are regulated by androgen. Biomed. Pharmacother. 2019, 120, 109555. [Google Scholar] [CrossRef]
  151. Xu, W.; Qadir, M.M.F.; Nasteska, D.; Mota de Sa, P.; Gorvin, C.M.; Blandino-Rosano, M.; Evans, C.R.; Ho, T.; Potapenko, E.; Veluthakal, R.; et al. Architecture of androgen receptor pathways amplifying glucagon-like peptide-1 insulinotropic action in male pancreatic β cells. Cell Rep. 2023, 42, 112529. [Google Scholar] [CrossRef]
  152. Sanchez-Garrido, M.A.; Tena-Sempere, M. Metabolic dysfunction in polycystic ovary syndrome: Pathogenic role of androgen excess and potential therapeutic strategies. Mol. Metab. 2020, 35, 100937. [Google Scholar] [CrossRef]
  153. Liu, L.; Liu, S.; Song, Q.; Luo, D.; Su, Y.; Qi, X.; Wang, Q.; Ning, J.; Lv, Y.; Guan, Q. Association of Metabolic Obesity Phenotypes and Total Testosterone in Chinese Male Population. Diabetes Metab. Syndr. Obes. 2021, 14, 399–408. [Google Scholar] [CrossRef] [PubMed]
  154. Navarro, G.; Allard, C.; Morford, J.J.; Xu, W.; Liu, S.; Molinas, A.J.; Butcher, S.M.; Fine, N.H.; Blandino-Rosano, M.; Sure, V.N.; et al. Androgen excess in pancreatic β cells and neurons predisposes female mice to type 2 diabetes. JCI Insight 2018, 3, e98607. [Google Scholar] [CrossRef]
  155. Brown, R.E.; Wilkinson, D.A.; Imran, S.A.; Caraty, A.; Wilkinson, M. Hypothalamic kiss1 mRNA and kisspeptin immunoreactivity are reduced in a rat model of polycystic ovary syndrome (PCOS). Brain Res. 2012, 1467, 1–9. [Google Scholar] [CrossRef] [PubMed]
  156. Nohara, K.; Laque, A.; Allard, C.; Münzberg, H.; Mauvais-Jarvis, F. Central mechanisms of adiposity in adult female mice with androgen excess. Obesity 2014, 22, 1477–1484. [Google Scholar] [CrossRef]
  157. Han, Q.; Wang, J.; Li, W.; Chen, Z.J.; Du, Y. Androgen-induced gut dysbiosis disrupts glucolipid metabolism and endocrinal functions in polycystic ovary syndrome. Microbiome 2021, 9, 101. [Google Scholar] [CrossRef]
  158. Osei-Ntansah, A.; Oliver, T.; Lofton, T.; Falzarano, C.; Carr, K.; Huang, R.; Wilson, A.; Damaser, E.; Harvey, G.; Rahman, M.A.; et al. Liver Androgen Receptor Knockout Improved High-fat Diet Induced Glucose Dysregulation in Female Mice but Not Male Mice. J. Endocr. Soc. 2024, 8, bvae021. [Google Scholar] [CrossRef] [PubMed]
  159. Palmisano, B.T.; Zhu, L.; Stafford, J.M. Role of Estrogens in the Regulation of Liver Lipid Metabolism. Adv. Exp. Med. Biol. 2017, 1043, 227–256. [Google Scholar]
  160. Alemany, M. Estrogens and the regulation of glucose metabolism. World J. Diabetes 2021, 12, 1622–1654. [Google Scholar] [CrossRef]
  161. Nappi, R.E.; Chedraui, P.; Lambrinoudaki, I.; Simoncini, T. Menopause: A cardiometabolic transition. Lancet Diabetes Endocrinol. 2022, 10, 442–456. [Google Scholar] [CrossRef]
  162. Reue, K.; Wiese, C.B. Illuminating the Mechanisms Underlying Sex Differences in Cardiovascular Disease. Circ. Res. 2022, 130, 1747–1762. [Google Scholar] [CrossRef]
  163. de Oliveira, M.C.; Gilglioni, E.H.; de Boer, B.A.; Runge, J.H.; de Waart, D.R.; Salgueiro, C.L.; Ishii-Iwamoto, E.L.; Oude Elferink, R.P.; Gaemers, I.C. Bile acid receptor agonists INT747 and INT777 decrease oestrogen deficiency-related postmenopausal obesity and hepatic steatosis in mice. Biochim. Biophys. Acta 2016, 1862, 2054–2062. [Google Scholar] [CrossRef] [PubMed]
  164. Papadakis, G.E.; Hans, D.; Gonzalez Rodriguez, E.; Vollenweider, P.; Waeber, G.; Marques-Vidal, P.; Lamy, O. Menopausal Hormone Therapy Is Associated with Reduced Total and Visceral Adiposity: The OsteoLaus Cohort. J. Clin. Endocrinol. Metab. 2018, 103, 1948–1957. [Google Scholar] [CrossRef] [PubMed]
  165. González-García, I.; García-Clavé, E.; Cebrian-Serrano, A.; Le Thuc, O.; Contreras, R.E.; Xu, Y.; Gruber, T.; Schriever, S.C.; Legutko, B.; Lintelmann, J.; et al. Estradiol regulates leptin sensitivity to control feeding via hypothalamic Cited1. Cell Metab. 2023, 35, 438–455.E7. [Google Scholar] [CrossRef] [PubMed]
  166. Villa, A.; Della Torre, S.; Stell, A.; Cook, J.; Brown, M.; Maggi, A. Tetradian oscillation of estrogen receptor α is necessary to prevent liver lipid deposition. Proc. Natl. Acad. Sci. USA 2012, 109, 11806–11811. [Google Scholar] [CrossRef]
  167. Fukata, Y.; Yu, X.; Imachi, H.; Nishiuchi, T.; Lyu, J.; Seo, K.; Takeuchi, A.; Iwama, H.; Masugata, H.; Hoshikawa, H.; et al. 17β-Estradiol regulates scavenger receptor class BI gene expression via protein kinase C in vascular endothelial cells. Endocrine 2014, 46, 644–650. [Google Scholar] [CrossRef]
  168. Zhu, L.; Shi, J.; Luu, T.N.; Neuman, J.C.; Trefts, E.; Yu, S.; Palmisano, B.T.; Wasserman, D.H.; Linton, M.F.; Stafford, J.M. Hepatocyte estrogen receptor alpha mediates estrogen action to promote reverse cholesterol transport during Western-type diet feeding. Mol. Metab. 2018, 8, 106–116. [Google Scholar] [CrossRef]
  169. Sutjarit, N.; Sueajai, J.; Boonmuen, N.; Sornkaew, N.; Suksamrarn, A.; Tuchinda, P.; Zhu, W.; Weerachayaphorn, J.; Piyachaturawat, P. Curcuma comosa reduces visceral adipose tissue and improves dyslipidemia in ovariectomized rats. J. Ethnopharmacol. 2018, 215, 167–175. [Google Scholar] [CrossRef]
  170. Hammes, S.R.; Levin, E.R. Impact of estrogens in males and androgens in females. J. Clin. Investig. 2019, 129, 1818–1826. [Google Scholar] [CrossRef]
  171. Lee, C.; Kim, J.; Han, J.; Oh, D.; Kim, M.; Jeong, H.; Kim, T.J.; Kim, S.W.; Kim, J.N.; Seo, Y.S.; et al. Formyl peptide receptor 2 determines sex-specific differences in the progression of nonalcoholic fatty liver disease and steatohepatitis. Nat. Commun. 2022, 13, 578. [Google Scholar] [CrossRef]
  172. Axsom, J.E.; Schmidt, H.D.; Matura, L.A.; Libonati, J.R. The Influence of Epigenetic Modifications on Metabolic Changes in White Adipose Tissue and Liver and Their Potential Impact in Exercise. Front. Physiol. 2021, 12, 686270. [Google Scholar] [CrossRef]
  173. Mannar, V.; Boro, H.; Patel, D.; Agstam, S.; Dalvi, M.; Bundela, V. Epigenetics of the Pathogenesis and Complications of Type 2 Diabetes Mellitus. touchREVIEWS Endocrinol. 2023, 19, 46–53. [Google Scholar] [CrossRef] [PubMed]
  174. Wu, Y.L.; Lin, Z.J.; Li, C.C.; Lin, X.; Shan, S.K.; Guo, B.; Zheng, M.H.; Li, F.; Yuan, L.Q.; Li, Z.H. Epigenetic regulation in metabolic diseases: Mechanisms and advances in clinical study. Signal Transduct. Target. Ther. 2023, 8, 98. [Google Scholar] [CrossRef] [PubMed]
  175. Li, B.; Xia, Y.; Mei, S.; Ye, Z.; Song, B.; Yan, X.; Lin, F.; Rao, T.; Yu, W.; Mei, C.; et al. Histone H3K27 methyltransferase EZH2 regulates apoptotic and inflammatory responses in sepsis-induced AKI. Theranostics 2023, 13, 1860–1875. [Google Scholar] [CrossRef] [PubMed]
  176. Kaisinger, L.R.; Kentistou, K.A.; Stankovic, S.; Gardner, E.J.; Day, F.R.; Zhao, Y.; Mörseburg, A.; Carnie, C.J.; Zagnoli-Vieira, G.; Puddu, F.; et al. Large-scale exome sequence analysis identifies sex- and age-specific determinants of obesity. Cell Genom. 2023, 3, 100362. [Google Scholar] [CrossRef]
  177. Huang, Y.; Wang, A.; Zhou, W.; Li, B.; Zhang, L.; Rudolf, A.M.; Jin, Z.; Hambly, C.; Wang, G.; Speakman, J.R. Maternal dietary fat during lactation shapes single nucleus transcriptomic profile of postnatal offspring hypothalamus in a sexually dimorphic manner in mice. Nat. Commun. 2024, 15, 2382. [Google Scholar] [CrossRef]
  178. Shi, Y.; Ma, J.; Li, S.; Liu, C.; Liu, Y.; Chen, J.; Liu, N.; Liu, S.; Huang, H. Sex difference in human diseases: Mechanistic insights and clinical implications. Signal Transduct. Target. Ther. 2024, 9, 238. [Google Scholar] [CrossRef]
  179. Horstmann, A.; Kovacs, P.; Kabisch, S.; Boettcher, Y.; Schloegl, H.; Tönjes, A.; Stumvoll, M.; Pleger, B.; Villringer, A. Common genetic variation near MC4R has a sex-specific impact on human brain structure and eating behavior. PLoS ONE 2013, 8, e74362. [Google Scholar] [CrossRef]
  180. Claussnitzer, M.; Dankel, S.N.; Kim, K.H.; Quon, G.; Meuleman, W.; Haugen, C.; Glunk, V.; Sousa, I.S.; Beaudry, J.L.; Puviindran, V.; et al. FTO Obesity Variant Circuitry and Adipocyte Browning in Humans. N. Engl. J. Med. 2015, 373, 895–907. [Google Scholar] [CrossRef]
  181. Adak, A.; Khan, M.R. An insight into gut microbiota and its functionalities. Cell Mol. Life Sci. 2019, 76, 473–493. [Google Scholar] [CrossRef]
  182. Lemons, J.M.S.; Liu, L. Chewing the Fat with Microbes: Lipid Crosstalk in the Gut. Nutrients 2022, 14, 573. [Google Scholar] [CrossRef]
  183. Fan, S.; Chen, S.; Lin, L. Research progress of gut microbiota and obesity caused by high-fat diet. Front. Cell Infect. Microbiol. 2023, 13, 1139800. [Google Scholar] [CrossRef]
  184. Krautkramer, K.A.; Fan, J.; Bäckhed, F. Gut microbial metabolites as multi-kingdom intermediates. Nat. Rev. Microbiol. 2021, 19, 77–94. [Google Scholar] [CrossRef]
  185. Friedman, E.S.; Li, Y.; Shen, T.D.; Jiang, J.; Chau, L.; Adorini, L.; Babakhani, F.; Edwards, J.; Shapiro, D.; Zhao, C.; et al. FXR-Dependent Modulation of the Human Small Intestinal Microbiome by the Bile Acid Derivative Obeticholic Acid. Gastroenterology 2018, 155, 1741–1752.e5. [Google Scholar] [CrossRef] [PubMed]
  186. Santos-Marcos, J.A.; Mora-Ortiz, M.; Tena-Sempere, M.; Lopez-Miranda, J.; Camargo, A. Interaction between gut microbiota and sex hormones and their relation to sexual dimorphism in metabolic diseases. Biol. Sex. Differ. 2023, 14, 4. [Google Scholar] [CrossRef] [PubMed]
  187. Bolnick, D.I.; Snowberg, L.K.; Hirsch, P.E.; Lauber, C.L.; Org, E.; Parks, B.; Lusis, A.J.; Knight, R.; Caporaso, J.G.; Svanbäck, R. Individual diet has sex-dependent effects on vertebrate gut microbiota. Nat. Commun. 2014, 5, 4500. [Google Scholar] [CrossRef] [PubMed]
  188. Org, E.; Mehrabian, M.; Parks, B.W.; Shipkova, P.; Liu, X.; Drake, T.A.; Lusis, A.J. Sex differences and hormonal effects on gut microbiota composition in mice. Gut Microbes 2016, 7, 313–322. [Google Scholar] [CrossRef]
  189. Fransen, F.; van Beek, A.A.; Borghuis, T.; Meijer, B.; Hugenholtz, F.; van der Gaast-de Jongh, C.; Savelkoul, H.F.; de Jonge, M.I.; Faas, M.M.; Boekschoten, M.V.; et al. The Impact of Gut Microbiota on Gender-Specific Differences in Immunity. Front. Immunol. 2017, 8, 754. [Google Scholar] [CrossRef]
  190. Elderman, M.; Hugenholtz, F.; Belzer, C.; Boekschoten, M.; van Beek, A.; de Haan, B.; Savelkoul, H.; de Vos, P.; Faas, M. Sex and strain dependent differences in mucosal immunology and microbiota composition in mice. Biol. Sex. Differ. 2018, 9, 26. [Google Scholar] [CrossRef]
  191. Sinha, T.; Vich Vila, A.; Garmaeva, S.; Jankipersadsing, S.A.; Imhann, F.; Collij, V.; Bonder, M.J.; Jiang, X.; Gurry, T.; Alm, E.J.; et al. Analysis of 1135 gut metagenomes identifies sex-specific resistome profiles. Gut Microbes 2019, 10, 358–366. [Google Scholar] [CrossRef]
  192. Liu, D.S.; Wang, X.S.; Zhong, X.H.; Cao, H.; Zhang, F. Sexual dimorphism in the gut microbiota and sexual dimorphism in chronic diseases: Association or causation? J. Steroid Biochem. Mol. Biol. 2024, 237, 106451. [Google Scholar] [CrossRef]
  193. Ma, Z.S.; Li, W. How and Why Men and Women Differ in Their Microbiomes: Medical Ecology and Network Analyses of the Microgenderome. Adv. Sci. 2019, 6, 1902054. [Google Scholar] [CrossRef] [PubMed]
  194. Ding, T.; Schloss, P.D. Dynamics and associations of microbial community types across the human body. Nature 2014, 509, 357–360. [Google Scholar] [CrossRef] [PubMed]
  195. Liu, T.; Xu, Y.; Yi, C.X.; Tong, Q.; Cai, D. The hypothalamus for whole-body physiology: From metabolism to aging. Protein Cell 2022, 13, 394–421. [Google Scholar] [CrossRef] [PubMed]
  196. Chowen, J.A.; Argente-Arizón, P.; Freire-Regatillo, A.; Argente, J. Sex differences in the neuroendocrine control of metabolism and the implication of astrocytes. Front. Neuroendocrinol. 2018, 48, 3–12. [Google Scholar] [CrossRef]
  197. Tang, C.; Wang, Y.; Xu, Z.; Chen, D.; Xu, J.; Yang, D.; Zhang, L.; Liu, J.; Kan, J. The relationships between high-fat diet and metabolic syndrome: Potential mechanisms. Food Biosci. 2024, 59, 104261. [Google Scholar] [CrossRef]
  198. Mann, E.R.; Lam, Y.K.; Uhlig, H.H. Short-chain fatty acids: Linking diet, the microbiome and immunity. Nat. Rev. Immunol. 2024, 24, 577–595. [Google Scholar] [CrossRef]
  199. Kwon, Y.J.; Lee, H.S.; Lee, J.W. Association of carbohydrate and fat intake with metabolic syndrome. Clin. Nutr. 2018, 37, 746–751. [Google Scholar] [CrossRef]
  200. MacGregor, K.; Ellefsen, S.; Pillon, N.J.; Hammarström, D.; Krook, A. Sex differences in skeletal muscle metabolism in exercise and type 2 diabetes mellitus. Nat. Rev. Endocrinol. 2025, 21, 166–179. [Google Scholar] [CrossRef]
  201. Tomé, D.; Benoit, S.; Azzout-Marniche, D. Protein metabolism and related body function: Mechanistic approaches and health consequences. Proc. Nutr. Soc. 2021, 80, 243–251. [Google Scholar] [CrossRef]
  202. Bailey, R.L.; Dog, T.L.; Smith-Ryan, A.E.; Das, S.K.; Baker, F.C.; Madak-Erdogan, Z.; Hammond, B.R.; Sesso, H.D.; Eapen, A.; Mitmesser, S.H.; et al. Sex Differences Across the Life Course: A Focus On Unique Nutritional and Health Considerations among Women. J. Nutr. 2022, 152, 1597–1610. [Google Scholar] [CrossRef]
  203. DiNicolantonio, J.J.; O’Keefe, J.H. Monounsaturated Fat vs Saturated Fat: Effects on Cardio-Metabolic Health and Obesity. Mo. Med. 2022, 119, 69–73. [Google Scholar] [PubMed]
  204. Delgado-Lista, J.; Alcala-Diaz, J.F.; Torres-Peña, J.D.; Quintana-Navarro, G.M.; Fuentes, F.; Garcia-Rios, A.; Ortiz-Morales, A.M.; Gonzalez-Requero, A.I.; Perez-Caballero, A.I.; Yubero-Serrano, E.M.; et al. Long-term secondary prevention of cardiovascular disease with a Mediterranean diet and a low-fat diet (CORDIOPREV): A randomised controlled trial. Lancet 2022, 399, 1876–1885. [Google Scholar] [CrossRef]
  205. Güzey Akansel, M.; Baş, M.; Gençalp, C.; Kahrıman, M.; Şahin, E.; Öztürk, H.; Gür, G.; Gür, C. Effects of the Ketogenic Diet on Microbiota Composition and Short-Chain Fatty Acids in Women with Overweight/Obesity. Nutrients 2024, 16, 4374. [Google Scholar] [CrossRef] [PubMed]
  206. Didier, A.J.; Stiene, J.; Fang, L.; Watkins, D.; Dworkin, L.D.; Creeden, J.F. Antioxidant and Anti-Tumor Effects of Dietary Vitamins A, C, and E. Antioxidants 2023, 12, 632. [Google Scholar] [CrossRef]
  207. Di Berardino, C.; Barceviciute, U.; Camerano Spelta Rapini, C.; Peserico, A.; Capacchietti, G.; Bernabò, N.; Russo, V.; Gatta, V.; Konstantinidou, F.; Donato, M.; et al. High-fat diet-negative impact on female fertility: From mechanisms to protective actions of antioxidant matrices. Front. Nutr. 2024, 11, 1415455. [Google Scholar] [CrossRef] [PubMed]
  208. Tang, G.H.; Sholzberg, M. Iron deficiency anemia among women: An issue of health equity. Blood Rev. 2024, 64, 101159. [Google Scholar] [CrossRef]
  209. Zucker, I.; Prendergast, B.J. Sex differences in pharmacokinetics predict adverse drug reactions in women. Biol. Sex. Differ. 2020, 11, 32. [Google Scholar] [CrossRef]
  210. Kautzky-Willer, A.; Leutner, M.; Harreiter, J. Sex differences in type 2 diabetes. Diabetologia 2023, 66, 986–1002. [Google Scholar] [CrossRef]
  211. Wilding, J.P.; Overgaard, R.V.; Jacobsen, L.V.; Jensen, C.B.; le Roux, C.W. Exposure-response analyses of liraglutide 3.0 mg for weight management. Diabetes Obes. Metab. 2016, 18, 491–499. [Google Scholar] [CrossRef]
  212. Moyer, A.M.; Matey, E.T.; Miller, V.M. Individualized medicine: Sex, hormones, genetics, and adverse drug reactions. Pharmacol. Res. Perspect. 2019, 7, e00541. [Google Scholar] [CrossRef]
  213. Aroda, V.R.; Christophi, C.A.; Edelstein, S.L.; Zhang, P.; Herman, W.H.; Barrett-Connor, E.; Delahanty, L.M.; Montez, M.G.; Ackermann, R.T.; Zhuo, X.; et al. The effect of lifestyle intervention and metformin on preventing or delaying diabetes among women with and without gestational diabetes: The Diabetes Prevention Program outcomes study 10-year follow-up. J. Clin. Endocrinol. Metab. 2015, 100, 1646–1653. [Google Scholar] [CrossRef] [PubMed]
  214. Raparelli, V.; Elharram, M.; Moura, C.S.; Abrahamowicz, M.; Bernatsky, S.; Behlouli, H.; Pilote, L. Sex Differences in Cardiovascular Effectiveness of Newer Glucose-Lowering Drugs Added to Metformin in Type 2 Diabetes Mellitus. J. Am. Heart Assoc. 2020, 9, e012940. [Google Scholar] [CrossRef] [PubMed]
  215. Funck, K.L.; Bjerg, L.; Isaksen, A.A.; Sandbæk, A.; Grove, E.L. Gender disparities in time-to-initiation of cardioprotective glucose-lowering drugs in patients with type 2 diabetes and cardiovascular disease: A Danish nationwide cohort study. Cardiovasc. Diabetol. 2022, 21, 279. [Google Scholar] [CrossRef]
  216. Christakis, M.K.; Hasan, H.; De Souza, L.R.; Shirreff, L. The effect of menopause on metabolic syndrome: Cross-sectional results from the Canadian Longitudinal Study on Aging. Menopause 2020, 27, 999–1009. [Google Scholar] [CrossRef]
  217. Tishova, Y.; Kalinchenko, S.; Mskhalaya, G.; Hackett, G.; Livingston, M.; König, C.; Strange, R.; Zitzmann, M.; Mann, A.; Maarouf, A.; et al. Testosterone therapy reduces insulin resistance in men with adult-onset testosterone deficiency and metabolic syndrome. Results from the Moscow Study, a randomized controlled trial with an open-label phase. Diabetes Obes. Metab. 2024, 26, 2147–2157. [Google Scholar] [CrossRef] [PubMed]
  218. Kanakis, G.A.; Pofi, R.; Goulis, D.G.; Isidori, A.M.; Armeni, E.; Erel, C.T.; Fistonić, I.; Hillard, T.; Hirschberg, A.L.; Meczekalski, B.; et al. EMAS position statement: Testosterone replacement therapy in older men. Maturitas 2023, 178, 107854. [Google Scholar] [CrossRef]
  219. Maseroli, E.; Comeglio, P.; Corno, C.; Cellai, I.; Filippi, S.; Mello, T.; Galli, A.; Rapizzi, E.; Presenti, L.; Truglia, M.C.; et al. Testosterone treatment is associated with reduced adipose tissue dysfunction and nonalcoholic fatty liver disease in obese hypogonadal men. J. Endocrinol. Investig. 2021, 44, 819–842. [Google Scholar] [CrossRef]
  220. Archacki, D.; Zieliński, J.; Ciekot-Sołtysiak, M.; Zarębska, E.A.; Kusy, K. Sex Differences in the Energy System Contribution during Sprint Exercise in Speed-Power and Endurance Athletes. J. Clin. Med. 2024, 13, 4812. [Google Scholar] [CrossRef]
  221. Mauvais-Jarvis, F.; Clegg, D.J.; Hevener, A.L. The role of estrogens in control of energy balance and glucose homeostasis. Endocr. Rev. 2013, 34, 309–338. [Google Scholar] [CrossRef]
  222. Landen, S.; Hiam, D.; Voisin, S.; Jacques, M.; Lamon, S.; Eynon, N. Physiological and molecular sex differences in human skeletal muscle in response to exercise training. J. Physiol. 2023, 601, 419–434. [Google Scholar] [CrossRef]
  223. Tarnopolsky, M.A. Sex differences in exercise metabolism and the role of 17-beta estradiol. Med. Sci. Sports Exerc. 2008, 40, 648–654. [Google Scholar] [CrossRef] [PubMed]
  224. Hunter, S.K.; Angadi, S.S.; Bhargava, A.; Harper, J.; Hirschberg, A.L.; Levine, B.D.; Moreau, K.L.; Nokoff, N.J.; Stachenfeld, N.S.; Bermon, S. The Biological Basis of Sex Differences in Athletic Performance: Consensus Statement for the American College of Sports Medicine. Med. Sci. Sports Exerc. 2023, 55, 2328–2360. [Google Scholar] [CrossRef]
  225. Brown, K.D.; Waggy, E.D.; Nair, S.; Robinson, T.J.; Schmitt, E.E.; Bruns, D.R.; Thomas, D.P. Sex Differences in Cardiac AMP-Activated Protein Kinase Following Exhaustive Exercise. Sports Med. Int. Open 2020, 4, E13–E18. [Google Scholar] [CrossRef] [PubMed]
  226. Mauvais-Jarvis, F. Sex differences in energy metabolism: Natural selection, mechanisms and consequences. Nat. Rev. Nephrol. 2024, 20, 56–69. [Google Scholar] [CrossRef] [PubMed]
  227. Ansdell, P.; Thomas, K.; Hicks, K.M.; Hunter, S.K.; Howatson, G.; Goodall, S. Physiological sex differences affect the integrative response to exercise: Acute and chronic implications. Exp. Physiol. 2020, 105, 2007–2021. [Google Scholar] [CrossRef]
  228. Blüher, S.; Käpplinger, J.; Herget, S.; Reichardt, S.; Böttcher, Y.; Grimm, A.; Kratzsch, J.; Petroff, D. Cardiometabolic risk markers, adipocyte fatty acid binding protein (aFABP) and the impact of high-intensity interval training (HIIT) in obese adolescents. Metabolism 2017, 68, 77–87. [Google Scholar] [CrossRef]
  229. Ji, H.; Gulati, M.; Huang, T.Y.; Kwan, A.C.; Ouyang, D.; Ebinger, J.E.; Casaletto, K.; Moreau, K.L.; Skali, H.; Cheng, S. Sex Differences in Association of Physical Activity With All-Cause and Cardiovascular Mortality. J. Am. Coll. Cardiol. 2024, 83, 783–793. [Google Scholar] [CrossRef]
  230. 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]
  231. Kusuyama, J.; Alves-Wagner, A.B.; Makarewicz, N.S.; Goodyear, L.J. Effects of maternal and paternal exercise on offspring metabolism. Nat. Metab. 2020, 2, 858–872. [Google Scholar] [CrossRef]
Figure 1. Sex differences in metabolic responses associated with HFD and the development of metabolic diseases. Some elements of this figure are from Servier Medical Art (https://smart.servier.com/, accessed on 15 of March 2025).
Figure 1. Sex differences in metabolic responses associated with HFD and the development of metabolic diseases. Some elements of this figure are from Servier Medical Art (https://smart.servier.com/, accessed on 15 of March 2025).
Ijms 26 04777 g001
Figure 2. Mechanisms behind sex differences in the development of metabolic diseases. Notes: Some elements in this figure are from Servier Medical Art (https://smart.servier.com/, accessed on 15 of March 2025). KDM5C: lysine demethylase 5C; UCP1: uncoupling protein 1; SRY: sex-determining region Y; ERs: estrogen receptors; PDGFRα: platelet-derived growth factor receptor α; HMGB1: high-mobility group box-1 protein; Cited1: glutamate/aspartate-rich carboxy-terminal domain-containing protein 1; PIK3: phosphatidylinositol 3-kinase; Akt: protein kinase b; GLUT4: glucose transporter type 4; LDLR: low-density lipoprotein receptor; SR-B1: scavenger receptor class B type 1; AMPK: adenosine 5′-monophosphate (AMP)-activated protein kinase; PPARα: peroxisome proliferator-activated receptor α; AR: androgen receptor; GnRH: gonadotropin-releasing hormone; AgRP: agouti-related protein; TLR4: toll-like receptor 4; NF-κB: nuclear factor-kappa B; MAPK: mitogen-activated protein kinase; HSL: hormone-sensitive triglyceride lipase; LPL: lipoprotein lipase; PPARG: peroxisome proliferator-activated receptor gamma; SCD1: stearoyl-CoA desaturase 1; ACC1: acetyl-CoA carboxylases-1; NEGR1: neuronal growth regulator 1.
Figure 2. Mechanisms behind sex differences in the development of metabolic diseases. Notes: Some elements in this figure are from Servier Medical Art (https://smart.servier.com/, accessed on 15 of March 2025). KDM5C: lysine demethylase 5C; UCP1: uncoupling protein 1; SRY: sex-determining region Y; ERs: estrogen receptors; PDGFRα: platelet-derived growth factor receptor α; HMGB1: high-mobility group box-1 protein; Cited1: glutamate/aspartate-rich carboxy-terminal domain-containing protein 1; PIK3: phosphatidylinositol 3-kinase; Akt: protein kinase b; GLUT4: glucose transporter type 4; LDLR: low-density lipoprotein receptor; SR-B1: scavenger receptor class B type 1; AMPK: adenosine 5′-monophosphate (AMP)-activated protein kinase; PPARα: peroxisome proliferator-activated receptor α; AR: androgen receptor; GnRH: gonadotropin-releasing hormone; AgRP: agouti-related protein; TLR4: toll-like receptor 4; NF-κB: nuclear factor-kappa B; MAPK: mitogen-activated protein kinase; HSL: hormone-sensitive triglyceride lipase; LPL: lipoprotein lipase; PPARG: peroxisome proliferator-activated receptor gamma; SCD1: stearoyl-CoA desaturase 1; ACC1: acetyl-CoA carboxylases-1; NEGR1: neuronal growth regulator 1.
Ijms 26 04777 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mo, Q.; Deng, X.; Zhou, Z.; Yin, L. High-Fat Diet and Metabolic Diseases: A Comparative Analysis of Sex-Dependent Responses and Mechanisms. Int. J. Mol. Sci. 2025, 26, 4777. https://doi.org/10.3390/ijms26104777

AMA Style

Mo Q, Deng X, Zhou Z, Yin L. High-Fat Diet and Metabolic Diseases: A Comparative Analysis of Sex-Dependent Responses and Mechanisms. International Journal of Molecular Sciences. 2025; 26(10):4777. https://doi.org/10.3390/ijms26104777

Chicago/Turabian Style

Mo, Qiaoling, Xinquan Deng, Ziyu Zhou, and Lijun Yin. 2025. "High-Fat Diet and Metabolic Diseases: A Comparative Analysis of Sex-Dependent Responses and Mechanisms" International Journal of Molecular Sciences 26, no. 10: 4777. https://doi.org/10.3390/ijms26104777

APA Style

Mo, Q., Deng, X., Zhou, Z., & Yin, L. (2025). High-Fat Diet and Metabolic Diseases: A Comparative Analysis of Sex-Dependent Responses and Mechanisms. International Journal of Molecular Sciences, 26(10), 4777. https://doi.org/10.3390/ijms26104777

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

Article Metrics

Back to TopTop