Next Article in Journal
Decoding Fear: Analysis and Prognosis of Preoperatory Stress Level Through Advanced Statistical Modelling—A Prospective Study Across Multiple Surgical Specialties
Previous Article in Journal
Pediatric Medication Prescribing Across Urgent Care Visits: An Epidemiologic View from a Primary Care Setting in the Kingdom of Saudi Arabia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Metabolic Reprogramming Through Polyphenol Networks: A Systems Approach to Metabolic Inflammation and Insulin Resistance

by
Shakila Jahan Shimu
1,
Jawad Ul Karim Mahir
2,
Fardin Al Fahad Shakib
2,
Arafath Amin Ridoy
2,
Ratin Al Samir
2,
Nadia Jahan
2,
Md Fahim Hasan
2,
Sadman Sazzad
3,
Shamima Akter
4,
Mohammad Sarif Mohiuddin
5,
Md Jalal Ahmed Shawon
6,
Mohammad Hossain Shariare
2,
Mohammad Mohabbulla Mohib
2,7,* and
Mohammad Borhan Uddin
2,*
1
Department of Health Informatics, Harrisburg University of Science and Technology, 326 Market St., Harrisburg, PA 17101, USA
2
Department of Pharmaceutical Sciences, North South University, Dhaka 1229, Bangladesh
3
Department of Biomedical Engineering and Informatics, Luddy School of Informatics, Computing and Engineering, Indiana University Indianapolis, 420 University Blvd, Indianapolis, IN 46202, USA
4
Department of Endocrinology, Diabetes & Metabolism, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, 705 Maple Road, Williamsville, NY 14221, USA
5
Department of Foundations of Medicine, NYU Grossman Long Island School of Medicine, 101 Mineola Blvd, Mineola, NY 11501, USA
6
Department of Applied Biosciences and Process Engineering, Anhalt University of Applied Sciences, Bernburger Strasse 55, 06366 Köthen, Germany
7
Julius Bernstein Institute of Physiology, Medical School, Martin Luther University of Halle-Wittenberg, Magdeburger Str. 6, 06112 Halle, Germany
*
Authors to whom correspondence should be addressed.
Med. Sci. 2025, 13(3), 180; https://doi.org/10.3390/medsci13030180
Submission received: 7 August 2025 / Revised: 30 August 2025 / Accepted: 1 September 2025 / Published: 5 September 2025
(This article belongs to the Section Endocrinology and Metabolic Diseases)

Abstract

Obesity-induced insulin resistance and type 2 diabetes mellitus (T2DM) represent complex systemic disorders marked by chronic inflammation, oxidative stress, mitochondrial dysfunction, and endoplasmic reticulum (ER) stress. These pathophysiological processes disrupt insulin signaling and β-cell function, leading to impaired glucose homeostasis across multiple organs. Conventional therapies often target isolated pathways, overlooking the intricate molecular crosstalk and organelle-level disturbances driving disease progression. Citrus-derived polyphenols—including hesperidin, naringenin, nobiletin, and tangeretin—have emerged as promising agents capable of orchestrating a multi-targeted “metabolic reprogramming.” These compounds modulate key signaling pathways, including AMPK, PI3K/Akt, NF-κB, and Nrf2, thereby enhancing insulin sensitivity, reducing pro-inflammatory cytokine expression, and restoring redox balance. Furthermore, they improve mitochondrial biogenesis, stabilize membrane potential, and alleviate ER stress by modulating the unfolded protein response (UPR), thus supporting cellular energy homeostasis and protein folding capacity. Evidence from preclinical studies and select clinical trials suggests that citrus polyphenols can significantly improve glycemic control, reduce oxidative and inflammatory markers, and preserve β-cell function. Their pleiotropic actions across molecular and organ-level targets position them as integrative metabolic modulators. This review presents a systems-level synthesis of how citrus polyphenols rewire metabolic signaling networks and organelle resilience, offering a holistic therapeutic strategy to mitigate the root causes of obesity-induced insulin resistance.

1. Introduction

It begins quietly, an expanding waistline, a subtle shift in blood sugar levels, a creeping fatigue. But behind these seemingly mundane signs brews a global biological crisis: the unstoppable rise in obesity-linked type 2 diabetes mellitus (T2DM). This twin epidemic, now termed “diabesity,” has grown into a full-blown metabolic pandemic [1,2]. As of 2023, more than 650 million adults are classified as obese, and over 500 million people live with diabetes, with projections pointing toward 700 million cases by 2045 [3,4,5,6]. Numbers, yes, but behind them lies a silent molecular war.
At the heart of this storm is a cascade of metabolic miscommunications. As adipose tissue expands, it not only stores excess energy but also morphs into a pro-inflammatory endocrine organ. Hypertrophied adipocytes release a torrent of cytokines such as TNF-α, IL-6, and MCP-1—recruiting macrophages and igniting a chronic inflammatory loop [7,8,9,10]. The result? The activation of NF-κB, JNK, and SOCS3, key molecular saboteurs that degrade insulin signaling, disrupt glucose uptake, and sow the seeds of insulin resistance across the liver, muscle, and pancreas [10,11,12,13]. But inflammation is only the beginning. With each calorie surplus, cells are flooded with free fatty acids, triggering lipotoxicity, ceramide accumulation, and a collapse in mitochondrial efficiency [14,15]. Overloaded mitochondria generate reactive oxygen species (ROS), feeding a cycle of oxidative damage that mutates proteins, disrupts redox balance, and disables insulin receptors [16,17]. Meanwhile, the endoplasmic reticulum (ER) tasked with protein folding buckles under metabolic pressure. UPR sensors, such as PERK, IRE1, and ATF6, are activated, prompting β-cells to undergo apoptosis and further exacerbating metabolic dysfunction in the body [18,19].
Despite our arsenal of pharmacological tools such as metformin, insulin, and GLP-1 agonists, there remains no cure for diabesity, only containment. These agents address symptoms but leave the root networks of dysfunction largely untouched [20]. This limitation has propelled research toward integrative, multi-targeted interventions derived from nature’s pharmacopeia.
Among these, citrus polyphenols have emerged as compelling candidates. In this review we introduce a unique framework—systemic metabolic reprogramming—which emphasizes how citrus polyphenols simultaneously recalibrate mitochondrial–ER crosstalk, immune–metabolic interactions, and organ-level adaptation [21]. These compounds work through AMPK, NF-κB, and Nrf2., and restore insulin sensitivity in the liver, adipose tissue, and skeletal muscle [21,22,23,24,25]. They modulate SIRT1–PGC1α pathways, stabilize mitochondrial membrane potential, and reduce ER stress through unfolded protein response signaling [21]. In doing so, they mitigate hyperglycemia, improve lipid metabolism, and prevent β-cell failure in preclinical models [25].
Previous reviews have largely focused on the antioxidant or single-pathway actions of citrus polyphenols. However, the concept of citrus flavonoids as systemic metabolic reprogrammers—simultaneously recalibrating mitochondrial–ER crosstalk, immune–metabolic signaling, and multi-organ adaptation—has not yet been synthesized. This review uniquely frames citrus polyphenols as network-level modulators within diabesity.
This review unpacks how citrus polyphenols intervene at key metabolic junctions, recalibrating molecular circuits, rejuvenating organelle function, and restoring systemic homeostasis. As our understanding deepens, these nature-derived compounds may offer more than dietary support; they may serve as network-based therapeutics in the fight against diabesity.

2. Pathophysiology of Obesity-Induced Diabetes

2.1. Lipotoxicity and Free Fatty Acid-Mediated Insulin Resistance

As the storm of chronic inflammation rages through expanding adipose tissue, another silent disruptor spreads through the bloodstream, free fatty acids (FFAs). In the obese state, basal lipolysis is persistently elevated, turning fat depots into active endocrine disruptors. These circulating FFAs are eagerly taken up by insulin-sensitive tissues, including the liver and skeletal muscle, where they undergo incomplete oxidation or are converted into lipotoxic intermediates such as diacylglycerols (DAGs) and ceramides [26,27].
These molecules act not merely as metabolic byproducts, but as intracellular saboteurs. By activating serine/threonine kinases such as c-Jun N-terminal kinase (JNK) and protein kinase C (PKC), they interfere with insulin signaling at its core, through the inhibitory serine phosphorylation of insulin receptor substrates (IRS1/2). The downstream result is twofold: in skeletal muscle, glucose uptake is impaired; in the liver, glucose production remains inappropriately elevated [28,29,30,31]. These defects form the biochemical bedrock of systemic insulin resistance. Yet, this mechanism is not universally expressed. A paradox remains: not all individuals with obesity develop T2DM. Beneath this variability lies a complex interplay of mitochondrial oxidative capacity, lipid partitioning, and intracellular buffering efficiency. Some individuals appear to be equipped with metabolic machinery capable of neutralizing lipotoxicity [32,33,34,35]. This variability suggests the urgent need for stratified approaches, where mitochondrial phenotype, not body mass index, guides therapeutic strategy.
In the liver, lipotoxicity takes on another form: hepatic insulin resistance. This defect impairs the suppression of gluconeogenic enzymes under insulin stimulation, resulting in fasting and postprandial hyperglycemia. Lipid-laden hepatocytes, struggling under the weight of FFA influx, experience impaired AKT activation and PKC-mediated insulin receptor dysfunction [36,37,38]. Meanwhile, Kupffer cells, the liver’s resident macrophages, release inflammatory cytokines that compound the damage and amplify insulin resistance [39]. Still, one critical question lingers: is hepatic steatosis the cause or merely a companion of insulin resistance? Although these conditions often coexist, emerging evidence suggests that hepatic inflammation and lipotoxicity, not simple steatosis, are the more potent drivers of metabolic impairment [40,41,42]. As such, targeting inflammatory and lipid-signaling pathways within the liver may offer greater therapeutic promise than attempts to merely de-fat the organ.

2.2. Oxidative Stress, Mitochondrial Dysfunction, and ER Disruption

As lipotoxic intermediates sabotage insulin signaling from the surface, a deeper disturbance brews within the cell—oxidative stress. In obesity, the ectopic deposition of lipids in liver and skeletal muscle overburdens cellular respiration, particularly within the mitochondria. The result is an overproduction of reactive oxygen species (ROS), driven not only by mitochondrial electron transport chain leakage but also by upregulated NADPH oxidase (NOX) activity [43,44,45] [Table 1].
These ROS act as rogue messengers, damaging proteins, lipids, and DNA, and activating redox-sensitive inflammatory pathways, such as NF-κB and JNK, which further disrupt insulin receptor signaling [74,75]. Moreover, elevated oxidative stress impairs mitochondrial ATP production, undermines metabolic flexibility, and sets the stage for mitochondrial dysfunction—a central node in the pathogenesis of insulin resistance [76,77]. Yet, ROS are not universally destructive. At physiological levels, they function as essential signaling molecules, fine-tuning insulin sensitivity and energy flux. It is only when their production surpasses the buffering capacity of antioxidant systems that they become pathological. This duality makes ROS modulation a therapeutic tightrope—too much suppression may blunt necessary signaling, while too little invites metabolic collapse [77,78].
But the damage does not end at the mitochondria. Just beyond lies another vulnerable organelle, the endoplasmic reticulum (ER), which is tasked with protein folding, calcium storage, and lipid biosynthesis. In obesity, ER homeostasis is frequently disrupted, as excess nutrients and oxidative signals trigger the unfolded protein response (UPR). Key sensors, such as PERK, IRE1, and ATF6, become chronically activated, leading to maladaptive signaling, impaired insulin receptor trafficking, and even β-cell apoptosis [78,79,80].
Compounding the crisis, ER stress and mitochondrial dysfunction do not act in isolation. Instead, they engage in bidirectional cross-talk, where ROS produced by mitochondrial overload exacerbate ER stress, and ER stress in turn destabilizes mitochondrial dynamics and calcium homeostasis [81,82]. This self-reinforcing loop amplifies cellular distress, propelling the insulin-resistant phenotype forward.
Thus, in the obese insulin-resistant state, the combined dysfunction of mitochondria and ER forms a molecular sinkhole, collapsing the delicate architecture of intracellular communication and energy balance. Targeting this axis, mitochondrial redox tuning, and restoration of ER homeostasis may be essential for effective metabolic reprogramming.

2.3. β-Cell Compensation and Failure

As insulin resistance deepens in peripheral tissues, the metabolic burden shifts to the pancreas. For a time, the β-cells rise to the occasion. In early obesity, these endocrine sentinels respond to rising glycemic demand with compensatory hypersecretion of insulin, working overtime to preserve normoglycemia [83,84].
But this heroism comes at a cost. With chronic exposure to hyperglycemia and elevated FFAs, a biochemical storm known as glucolipotoxicity, β-cells begin to falter. Initially, their function deteriorates: insulin granule synthesis declines, secretory machinery loses fidelity, and glucose-stimulated insulin secretion becomes blunted [85,86].
Over time, this dysfunction becomes irreversible. A convergence of molecular insults, oxidative stress, ER stress, amyloid deposition, and islet-localized inflammation drives β-cells toward apoptosis and loss of mass [87]. Mitochondrial inefficiency further impairs insulin granule exocytosis, while pro-inflammatory cytokines such as IL-1β and IFN-γ infiltrate islets and exacerbate cell death [88]. Eventually, the tipping point is reached: the remaining β-cell pool is no longer sufficient to compensate. Glucose levels rise unchecked, and what began as a compensatory adaptation gives way to overt T2DM [89]. Yet the fate of the β-cell may not be entirely sealed. Some dysfunction appears reversible, at least in early stages, raising hope for therapeutic intervention. GLP-1 receptor agonists, for example, have been shown to enhance β-cell function, promote insulin gene expression, and even reduce apoptosis. However, restoring β-cell mass, not just improving function, remains a challenging task [90,91].
To this end, regenerative strategies such as β-cell neogenesis, trans-differentiation, and reprogramming of other pancreatic cells are under active exploration. While preclinical data offer tantalizing glimpses of success, translation into durable human therapies is still in its infancy [92]. Thus, the story of the β-cell in diabesity is one of early heroism, gradual unraveling, and an uncertain future. Preserving its function and perhaps, one day, restoring its mass may hold the key to altering the trajectory of metabolic disease.

2.4. Adipose Tissue Dysfunction and Immune Activation

As systemic insulin resistance develops, one of its earliest instigators lies buried in the expansion of fat depots. In the setting of chronic overnutrition, adipocytes enlarge through hypertrophy, outpacing their oxygen supply and creating localized hypoxia. This cellular stress activates hypoxia-inducible factors (HIFs) and initiates an inflammatory cascade [93,94].
Stressed adipocytes begin secreting danger signals, including chemokines such as monocyte chemoattractant protein-1 (MCP-1), which draw immune cells into the adipose microenvironment. There, monocytes differentiate into classically activated (M1) macrophages, forming crown-like structures around dying adipocytes and releasing a storm of pro-inflammatory cytokines: TNF-α, IL-6, and IL-1β [93]. These cytokines interfere with insulin receptor signaling by inhibiting the serine phosphorylation of IRS1, ultimately contributing to systemic insulin resistance [95]. At the same time, levels of adiponectin decline—an insulin-sensitizing adipokine with potent anti-inflammatory and vascular-protective properties. This drop further tips the balance toward a pro-inflammatory adipose phenotype, perpetuating metabolic dysfunction [96] [Figure 1].
But this immune–metabolic dialog is far from uniform. Depot-specific differences—between visceral, subcutaneous, and brown adipose tissue—shape the inflammatory landscape, as do the plastic and dynamic phenotypes of resident immune cells. In some depots, regulatory T cells, eosinophils, and alternatively activated (M2) macrophages attempt to counterbalance inflammation, though their roles remain incompletely understood [93]. As this immunological remodeling unfolds, translational researchers face a challenge: current animal models and cell lines oversimplify the dynamic complexity of human adipose tissue. The precise orchestration of immune-adipocyte interactions during obesity progression and weight loss remains a frontier of investigation, requiring refined model systems and longitudinal tissue profiling [97,98,99].
Thus, the immune–adipocyte axis stands as a central pillar of metabolic inflammation, a battlefield where energy sensing, immune signaling, and hormonal control collide.

3. Citrus Polyphenols in Metabolic Reprogramming

3.1. Sour Yet Sweet Salvation: How Citrus Polyphenols Rewire Diabetic Metabolism

Until now, the story has unfolded as a progressive unraveling of metabolic order, characterized by lipotoxicity, oxidative stress, mitochondrial dysfunction, inflammatory infiltration, and β-cell failure [15,100,101]. But nature, too, offers its counterstrike. Quietly nestled among citrus fruits, a class of polyphenolic compounds has shown a surprising capacity to engage the metabolic network, not as blunt tools, but as precise modulators capable of reprogramming disordered pathways [21].
In addition to functional outcomes, the structural features of citrus polyphenols are critical in explaining their biological activity. Figure 2 presents the chemical structures of the major flavonoids discussed in this review, including hesperidin, naringenin, nobiletin, tangeretin, eriocitrin, quercetin, and neohesperidin. These compounds share a flavanone or flavone backbone, with variation in hydroxylation, glycosylation, and methoxylation patterns. Such structural differences underlie their capacity to modulate redox balance and metabolic pathways.
Hesperidin, a glycosylated flavanone, possesses hydroxyl groups at the 5- and 7-positions of the A-ring and initiates its metabolic rescue by activating AMPK the cell’s energy sensor. This activation restores PI3K/Akt signaling, enabling the translocation of GLUT4 in skeletal muscle and adipose tissue, thereby reviving insulin responsiveness [21,102,103,104]. Simultaneously, hesperidin attenuates ROS accumulation and boosts endogenous antioxidants like SOD, GPx, and catalase, relieving oxidative stress and restoring mitochondrial integrity key to breaking the self-perpetuating cycle of insulin resistance [103]. Quercetin, abundant in citrus and other plants, reinforces this network-level modulation. It not only improves glycemic control via PI3K/Akt and IRS1 restoration, but also blocks NF-κB-mediated inflammation, decreasing expression of TNF-α and IL-6—two upstream drivers of adipose and hepatic insulin resistance [104,105]. Quercetin and eriocitrin, with multiple catechol groups, provide potent metal-chelating and antioxidant properties. These structural insights highlight common factors—redox modulation, lipid solubility, and receptor interactions—that enable citrus flavonoids to reverse metabolic dysfunction [106,107].
Then comes naringenin, the aglycone of naringin, which enters the liver and shifts metabolic flux toward fatty acid oxidation. It activates PPARα, downregulates SREBP-1c, and inhibits JNK signaling, thereby reducing lipogenesis, inflammation, and insulin resistance in hepatocytes [21,108,109]. In skeletal muscle, it reinforces AMPK signaling, improving glucose uptake and restoring oxidative capacity [66,110].
Tangeretin, sourced from citrus peels, contributes by modulating adipokine profiles—notably restoring adiponectin levels, which enhances insulin sensitivity through both AMPK activation and Pparγ modulation [111,112]. It also suppresses oxidative stress markers in insulin-sensitive tissues, reinforcing mitochondrial health and preserving metabolic signaling fidelity [113]. Kaempferol, while less abundant, wields a unique role in suppressing hepatic gluconeogenesis. By activating AMPK, it downregulates PEPCK and G6Pase, two key enzymes responsible for excess hepatic glucose output in insulin-resistant states [114,115,116,117]. Through this mechanism, it reprograms hepatic energy flow and corrects fasting hyperglycemia [116,117]. Rutin, structurally similar to quercetin, amplifies this ensemble response by restoring mitochondrial biogenesis, enhancing SIRT1–PGC1α signaling, and mitigating oxidative stress, particularly in the pancreas and liver [116].
Together, these compounds do not simply oppose insulin resistance—they rewire the system. Through synergistic targeting of AMPK, PI3K/Akt, NF-κB, PPARs, SIRT1, and UPR components, citrus polyphenols restore cellular energy sensing, reduce inflammatory signaling, and regenerate insulin sensitivity across the liver, muscle, adipose, and pancreas tissues. Their strength lies not in singular action, but in their ability to act in concert, modulating interlinked metabolic pathways at the core of diabesity. Thus, citrus polyphenols represent more than nutraceuticals. They are biochemical architects, reshaping the very systems that obesity and hypernutrition seek to dismantle.

3.2. Citrus Polyphenols and Inflammatory Reprogramming in Diabesity

In the evolving landscape of diabesity, inflammation emerges not as a byproduct but as a driving force—a molecular amplifier that sustains insulin resistance and disrupts metabolic homeostasis. The epicenter of this inflammatory storm lies within adipose tissue, where immune–metabolic crosstalk distorts insulin signaling at every level.
TNF-α, first identified as a pivotal factor in obese adipose tissue by Hotamisligil et al. [118], sets this cascade in motion [119,120]. As previously described in Section 3.1, citrus polyphenols suppress NF-κB nuclear translocation and normalize IRS1–Akt signaling [121,122,123]. Simultaneously, NF-κB, a master inflammatory transcription factor, translocates to the nucleus, driving the expression of cytokines such as IL-6, MCP-1, and iNOS, which reinforce metabolic dysfunction and immune infiltration [7,124,125]. Amid this inflammatory gridlock, citrus polyphenols have emerged not merely as suppressors of inflammation but as metabolic circuit breakers, capable of disconnecting inflammatory feedback loops and restoring intracellular communication [21].
Naringin, hesperidin, and quercetin have all been shown to suppress NF-κB nuclear translocation, thereby silencing pro-inflammatory transcriptional programs at their source. In murine and cellular models, these polyphenols reduce the expression of COX-2, iNOS, and inflammatory cytokines, thereby reshaping the cytokine environment from a pro-inflammatory to a homeostatic one [126,127]. Further upstream, they interfere with the AGE-RAGE signaling pathway, a critical amplifier of chronic inflammation and oxidative stress in metabolic tissues. Naringin and hesperidin downregulate RAGE expression and reduce AGE accumulation, blunting ROS generation and downstream cytokine bursts [128,129,130]. Importantly, these molecular insights are not confined to the bench. Clinical data reveal that citrus bioflavonoid complexes significantly lower plasma levels of IL-6 and TNF-α, as well as biomarkers of DNA oxidative damage, such as 8-OHdG, suggesting a systemic reprogramming of inflammatory tone in human subjects [131]. This anti-inflammatory reprogramming does not merely reduce cytokines; it restores the integrity of insulin signaling. With NF-κB silenced and JNK/IKKβ activity reduced, IRS1 phosphorylation normalizes, Akt activation is restored, and glucose uptake resumes. This is metabolic recalibration—not via single-point intervention, but through network-level remodeling of the immune–metabolic interface.
Thus, citrus polyphenols act as molecular disruptors of inflammatory inertia, targeting not only the messengers of inflammation but the architecture that sustains it. Their ability to realign immune and metabolic signaling pathways elevates them from anti-inflammatory agents to true agents of metabolic reprogramming.

3.3. Role of Citrus in Mitochondrial Health and Endoplasmic Reticulum Stress: Restoring Protein Homeostasis

In the battle against metabolic disease, restoring energy balance and cellular homeostasis is no longer just a downstream goal; it is a frontline strategy. As insulin signaling falters and inflammation mounts, the cell’s two central organelles for metabolic coordination—the mitochondria and the endoplasmic reticulum (ER) emerge as critical nodes of dysfunction [21]. Their deterioration underlies the systemic collapse in energy sensing, protein folding, and glucose regulation seen in obesity-induced T2DM.
At the heart of this collapse lies mitochondrial dysfunction, driven by nutrient overload, lipotoxicity, and oxidative stress. In metabolically active tissues—especially liver, adipose, and muscle—excess free fatty acids impair mitochondrial respiration, increase reactive oxygen species (ROS), and destabilize membrane potential, culminating in reduced ATP synthesis and suppression of insulin-stimulated glucose uptake [132,133].
Here, citrus polyphenols—particularly naringenin, hesperidin, and tangeretin—intervene as metabolic stabilizers. Their actions go beyond antioxidant defense. By activating the Nrf2–ARE pathway, they enhance endogenous antioxidant enzyme expression (SOD, catalase, GPx) and limit mitochondrial ROS accumulation [134,135]. More importantly, these polyphenols stimulate PGC-1α, NRF1, and TFAM, transcriptional regulators of mitochondrial biogenesis and energy renewal, reprogramming the cell toward oxidative resilience [134].
Naringenin, in particular, has been shown to preserve mitochondrial membrane potential, boost ATP output, and regulate PINK1/Parkin-mediated mitophagy, thereby eliminating dysfunctional mitochondria and sustaining metabolic flux in hepatocytes and myocytes under lipotoxic conditions [136,137,138]. Meanwhile, hesperidin enhances mitochondrial efficiency and prevents apoptotic signaling by reducing cytochrome c release and caspase activation, thereby preserving cell viability in the face of metabolic overload [139].
Yet mitochondria are not isolated actors. Their function is tightly intertwined with that of the endoplasmic reticulum (ER)—an organelle responsible for protein folding, lipid biosynthesis, and calcium signaling [140]. Under chronic nutrient stress, the ER accumulates misfolded proteins, triggering unfolded protein response (UPR) pathways. If unresolved, this stress spills over into apoptosis, fueling insulin resistance and β-cell failure [99,141,142,143].
Once again, citrus flavonoids offer a path to restoration. Compounds like naringenin and hesperidin selectively attenuate the PERK–eIF2α, IRE1–XBP1, and ATF6 arms of the UPR, preventing the maladaptive amplification of ER stress. In high-fat-diet models, naringenin reduces CHOP expression, a pro-apoptotic UPR marker, and restores adaptive ER signaling, thereby rescuing β-cell and hepatocyte function [141,144,145].
These polyphenols also enhance GRP78/BiP expression, boosting chaperone capacity and facilitating correct protein folding under stress conditions [140,146]. Additionally, by modulating ER-mitochondrial crosstalk, citrus compounds support coordinated calcium transfer, optimize oxidative metabolism, and prevent apoptosis—a level of control that aligns precisely with systemic metabolic reprogramming [147,148,149].
Thus, citrus polyphenols do not merely suppress damage; they restore the intracellular architecture upon which metabolic homeostasis depends. By recalibrating mitochondrial bioenergetics, enhancing mitophagy, stabilizing ER folding capacity, and reinforcing cross-organelle signaling, they initiate a cellular response that rewires the failing metabolic circuitry of diabesity from within.

3.4. Free Radicals, Oxidative Stress, and Citrus Polyphenols: A Natural Line of Defense

As the metabolic storm deepens, a new biochemical disruptor takes center stage: oxidative stress. While reactive oxygen species (ROS) serve as transient messengers under physiological conditions, chronic metabolic overload transforms them into agents of cellular sabotage, eroding the signaling pathways that maintain metabolic equilibrium.
In obesity-induced diabetes, the overproduction of ROS—particularly superoxide (O2), hydrogen peroxide (H2O2), and hydroxyl radicals (•OH)—disrupts insulin receptor integrity, impairs glucose uptake, and accelerates β-cell dysfunction [45,150,151]. These free radicals are not isolated threats; they are embedded within a vicious cycle where lipotoxicity, inflammation, and mitochondrial inefficiency reinforce oxidative damage, amplifying insulin resistance across tissues [152] [Figure 3].
The collapse of redox homeostasis is especially destructive in insulin-sensitive cells like hepatocytes, myocytes, and adipocytes, where antioxidant defense systems are often overwhelmed [67,153]. The antioxidant enzymatic triad—superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx)—is essential for neutralizing ROS. However, in metabolic disease, these systems are either underexpressed or dysfunctional, allowing oxidative stress to accumulate unchecked [150,151].
At this critical juncture, citrus polyphenols emerge as a dual-action metabolic defense—scavengers of free radicals and regulators of redox-responsive gene expression. Unlike synthetic antioxidants that act in isolation, these natural compounds integrate into the metabolic network, restoring antioxidant capacity from within [154,155]. For example, supplementation with Citrus unshiu peel extract has been shown to significantly increase hepatic levels of SOD, CAT, GPx, and glutathione (GSH) in high-fat-diet-fed mice, leading to reductions in lipid peroxidation and improvement in insulin sensitivity [156]. This restoration of redox balance helps normalize insulin signaling cascades and suppress stress-induced kinase activation, such as JNK and IKKβ, which are known inhibitors of insulin receptor substrates [154,157].
The effect is not compound-specific but rather synergistic. Flavonoids such as naringenin, hesperidin, and quercetin, abundant in citrus species like Citrus paradisi, Citrus sinensis, and Citrus maxima, not only scavenge ROS directly but also activate the Nrf2–ARE pathway—a master regulator of endogenous antioxidant gene expression [53,155,158]. Activation of Nrf2 leads to increased transcription of SOD, GPx, and CAT, reinforcing intracellular defenses at the transcriptional level [158].
Notably, extracts of red pummelo (Citrus maxima) demonstrated 20–40% higher free radical scavenging capacity than vitamin C or synthetic antioxidants like BHA, underlining the superior efficacy of these phytochemical complexes in redox modulation [159]. Thus, oxidative stress is not merely a consequence of metabolic dysfunction; it is a pathogenic amplifier of insulin resistance and systemic energy imbalance. By disrupting this oxidative-inflammation loop, citrus polyphenols do more than clean up molecular debris; they restore the redox environment required for metabolic precision. In doing so, they enable cells to re-establish insulin responsiveness, protect β-cell integrity, and suppress the molecular triggers of inflammation. This metabolic recalibration through antioxidant programming firmly positions citrus-derived polyphenols as frontline defenders in the systems-level war against diabesity (Figure 4).

3.5. Translating Mechanisms to Humans: Clinical Evidence of Citrus Polyphenol-Driven Metabolic Reprogramming

Having followed citrus polyphenols through cellular landscapes—where they extinguish oxidative stress, rewire mitochondrial signaling, restore protein-folding homeostasis, and silence inflammatory circuits—we arrive at the final test: do these molecular shifts translate to human physiology?
Clinical trials provide preliminary but encouraging signals that citrus polyphenols may recalibrate metabolic function. However, most studies remain limited by small sample sizes, short durations, and population heterogeneity. Therefore, current evidence should be regarded as exploratory, highlighting the need for multicenter RCTs and large longitudinal studies to establish efficacy [58,159] [Table 2].
In a double-blind, randomized clinical trial, 500 mg/day of hesperidin administered over three weeks significantly reduced TNF-α and VCAM-1, thereby restoring endothelial function—a proxy for systemic inflammatory tone and insulin sensitivity [22]. Importantly, these effects are not isolated to the vasculature but echo upstream changes in NF-κB signaling and insulin receptor substrate activity, consistent with earlier mechanistic studies.
While current clinical studies remain limited in scale and duration, ongoing large-scale randomized controlled trials are investigating citrus flavonoid supplementation in populations with metabolic syndrome and NAFLD. These trials are expected to provide more definitive evidence on efficacy, dose–response relationships, and long-term safety. Such future research will be essential for translating the promising mechanistic and preclinical findings into clinical practice.
Naringin, too, has moved from bench to bedside. Oral supplementation in patients with metabolic syndrome resulted in reductions in total cholesterol, LDL-C, and triglycerides, while increasing HDL-C, an indirect yet essential component of insulin signaling restoration [160,161]. These lipid profile improvements align with its known PPARγ activation and inhibition of JNK signaling, bridging preclinical mechanisms with clinical outcome [25,162].
In a dietary intervention study, individuals with T2DM who followed a Mediterranean diet enriched with citrus fruits experienced a reduction in HbA1c from 7.1% to 6.8% over 12 weeks, indicating an improvement in long-term glycemic control through enhanced insulin sensitivity and restored glucose uptake mechanisms [163].
Perhaps most compelling is the clinical validation of bergamot polyphenolic fraction (BPF)—a mixture rich in neoeriocitrin, naringin, and neohesperidin. In hyperlipidemic and prediabetic subjects, BPF not only lowered LDL-C and improved glucose metabolism, but achieved these results with efficacy comparable to statin therapy, yet without pharmacological side effects [164,165].
These studies converge on a single, powerful theme: citrus polyphenols act at the systems level, engaging molecular targets already mapped in preclinical models—AMPK, NF-κB, PI3K/Akt, PPARs—to bring about clinically meaningful metabolic restoration. Their ability to reduce inflammatory biomarkers, improve insulin action, recalibrate lipid metabolism, and preserve vascular function confirms their role not just as dietary supplements, but as translational agents in the therapeutic modulation of diabesity. And so, from the molecular to the metabolic, from mitochondria to macrosystems, citrus polyphenols close the loop: a natural, multifaceted, clinically validated reprogramming of the diseased metabolic network.
Table 2. Clinical studies of the effect of citrus polyphenols on metabolic reprogramming. This table summarizes randomized clinical trials, dietary interventions, or meta-analyses evaluating hesperidin, naringin, neoeriocitrin, and other citrus-derived polyphenols. Reported outcomes include changes in lipid profile, glycemic markers, inflammatory cytokines, liver fat, and endothelial function. Study size, intervention dose, and duration are also included.
Table 2. Clinical studies of the effect of citrus polyphenols on metabolic reprogramming. This table summarizes randomized clinical trials, dietary interventions, or meta-analyses evaluating hesperidin, naringin, neoeriocitrin, and other citrus-derived polyphenols. Reported outcomes include changes in lipid profile, glycemic markers, inflammatory cytokines, liver fat, and endothelial function. Study size, intervention dose, and duration are also included.
Compound(s)Study Design and PopulationIntervention Details (Dose and Duration)Reported OutcomesMechanism of ActionSource
Neoeriocitrin, Naringin, NeohesperidinRCT, overweight adults with MASLD (n = 80)Bergamot extract (500–1000 mg/day, 12 weeks)↓ Liver fat content (−18%), ↓ body weight (−5% vs. placebo)Enhances bile flow; antioxidant activity reduces oxidative stress[166]
Hesperidin, Naringin, NeohesperidinRCT, metabolic syndrome (n = 95)Mixed citrus extracts (500 mg/day, 8 weeks)↑ Endothelial function (FMD ↑12%), improved vascular toneAntioxidant effects improve vascular inflammation and nitric oxide availability[167]
Hesperidin → Hesperetin; SCFAsClinical trial, healthy volunteers (n = 40)Citrus fruit extract (500 mg/day, 4 weeks)Gut microbiota modulation: ↑ Bifidobacterium, ↑ SCFA production; ↓ systemic inflammationHesperidin metabolized to hesperetin → SCFA-mediated endothelial protection and anti-inflammatory response[168]
Flavones, Flavanones, OleuropeinRCT, high-CV-risk adults (n = 120)Citrus + olive polyphenol mix (500 mg/day, 12 weeks)↓ Cardiovascular risk biomarkers; ↓ hs-CRP (−20%); improved metabolic-inflammatory profileAntioxidant activity; NF-κB inhibition[169]
Hesperidin, Naringin, OleuropeinRCT, adults with dyslipidemia (n = 72)Citrus + olive leaf extracts (500 mg/day, 10 weeks)↓ LDL oxidation (−12%), ↓ TNF-α (−18%), ↓ IL-6 (−15%)Free radical scavenging; cytokine modulation[170]
HesperidinRCT, obese adults (n = 64)Orange juice (Citrus sinensis, ~500 mL/day, 12 weeks)↓ BMI (−1.2 kg/m2), ↓ waist circumference (−3.4 cm), ↓ IL-1β, IL-6, TNF-αInhibits pro-inflammatory cytokine release; antioxidant endothelial protection[171]
HesperidinRCT, NAFLD patients (n = 82)Hesperidin 1 g/day + lifestyle changes (12 weeks)↓ Liver fat (−22%), ↓ ALT (−30%), ↓ TG (−18%), ↓ weight (−4 kg)NF-κB inhibition; ↓ TNF-α, ↓ hs-CRP[172]
Hesperidin (meta-analysis)Meta-analysis of RCTs (n = 525 metabolic subjects)Hesperidin (500–1000 mg/day, 4–12 weeks)↓ TG, ↓ TC, ↓ LDL (especially in BMI >30); ↓ TNF-α, ↓ IL-6 at higher dosesAnti-inflammatory; lipid-lowering[173]
Orange juice (flavonoids)4-week RCT, MASLD patients (n = 62)Orange juice (500 mL/day)↓ Liver steatosis (by FibroScan), ↓ GGT (−10%)Antioxidant effect; modest anti-inflammatory[174]
Flavonoid-enriched orange juiceRCT, metabolic syndrome patients (n = 48)Enriched OJ (500 mL/day, 6 weeks)↑ Antioxidant status (TAC ↑15%), improved glycemic trend↓ CRP, ↓ endothelial inflammation[175]
HesperidinRCT, vascular function study (n = 24 metabolic syndrome patients)Hesperidin 500 mg/day, 3 weeks↑ FMD (+12%), ↓ IL-6 (−15%), ↓ TNF-α (−12%)↑ NO bioavailability; ↓ inflammatory cytokines[22]
Eriomin® (Eriocitrin)Crossover RCT, prediabetes patients (n = 103)Eriomin® 200–500 mg/day, 12 weeks↓ FBG (−5.5 mg/dL), ↓ HOMA-IR (−18%), ↑ GLP-1 (+15%), ↑ adiponectin (+20%)↓ IL-6, TNF-α, hs-CRP[176]
Polyphenols incl. NaringeninMeta-analysis in NAFLD patients (12 RCTs, n ≈ 950)Various flavonoids (6–12 weeks)↓ BMI, ↓ ALT (−12%), ↓ AST (−10%), ↓ TG (−18%), ↓ TNF-α (−14%)Anti-inflammatory; metabolic reprogramming[177]

4. Role of Lipoproteins in Diabetes and the Impact of Citrus Polyphenols

Lipoproteins play a central role in the pathophysiology of insulin resistance and type 2 diabetes [178]. In diabetic states, apoB-containing lipoproteins such as very-low-density lipoprotein (VLDL) and low-density lipoprotein (LDL) are elevated, driving lipid overload in the liver and peripheral tissues. These particles are highly enriched in triglyceride-derived lipid hydroperoxides, which amplify oxidative stress and vascular inflammation [179]. In contrast, levels of high-density lipoprotein (HDL) are reduced, and HDL function is often impaired. Dysfunctional HDL loses its antioxidant and anti-inflammatory properties, limiting its capacity to neutralize lipid peroxides and further exacerbating the pro-inflammatory state [180].
Citrus polyphenols have been reported to beneficially modulate lipoprotein metabolism. Hesperidin supplementation in humans reduces plasma LDL and triglycerides while modestly increasing HDL levels [23]. Bergamot polyphenolic fraction (rich in neoeriocitrin, naringin, and neohesperidin) lowers apoB-containing lipoproteins and improves HDL function [181]. Animal studies demonstrate that naringin and nobiletin suppress hepatic VLDL secretion and enhance reverse cholesterol transport. These actions collectively reduce circulating lipid hydroperoxides and restore lipoprotein redox balance [182].
By targeting both lipoprotein levels and lipoprotein functionality, citrus polyphenols address an under-recognized contributor to metabolic dysfunction in diabetes [183]. Their ability to restore HDL’s antioxidant capacity and attenuate VLDL-derived oxidative stress provides a mechanistic link between polyphenol chemistry, lipid metabolism, and vascular protection. This integration expands the translational relevance of citrus polyphenols beyond glucose homeostasis to the broader spectrum of cardiometabolic risk.

5. Conclusions and Future Direction

In the orchestra of cellular life, metabolic reprogramming is the conductor’s baton, guiding the tempo of nutrient sensing, mitochondrial energy flow, insulin action, and inflammatory resolution. But in obesity-induced diabetes, the rhythm descends into disarray. The once-synchronized network of metabolic signals becomes a cacophony of oxidative stress, unfolded proteins, inflammatory feedback, and lipotoxic derailment.
Yet, amid this dysfunction, a new score emerges, one not composed in laboratories but grown in groves of citrus, where nature has refined its symphony of healing.
Throughout this review, we followed citrus-derived polyphenols as they stepped into this metabolic dissonance, not as singular antioxidants or hypoglycemic agents, but as systemic modulators orchestrating a return to cellular harmony. From naringenin’s mitochondrial recalibration to hesperidin’s anti-inflammatory tuning, these flavonoids reactivated dormant pathways, rewired dysfunctional circuits, and restored homeostatic balance at every biological level—liver, adipose, muscle, pancreas, and beyond.
This review integrates evidence across molecular, organelle, and clinical dimensions to present citrus polyphenols as systemic reprogramming agents. This framework underscores their role not merely as antioxidants but as network-based therapeutic candidates.
Their strength lies not in pharmacological force, but in biological fluency. Citrus polyphenols do not attack the system—they speak its language, modulating master nodes like AMPK, PPARγ, Nrf2, and NF-κB, enabling tissues to heal from within. This systems-level approach is what places them at the forefront of a new therapeutic paradigm: one where nutritional networks, not isolated molecules, reshape disease trajectories.
Clinical studies now echo these findings. From reductions in HbA1c and inflammatory markers to improvements in lipid profiles and insulin sensitivity, the early human evidence is promising. These are not miracle cures—but metabolic nudges that push the body back toward its innate balance, especially when paired with lifestyle realignment.
As we close this chapter, we leave not with finality but with purpose. The story of citrus polyphenols in metabolic reprogramming is still unfolding. While this review focused on their core impact on insulin resistance, oxidative stress, mitochondrial-ER integrity, and inflammatory remodeling, much remains to be explored.
For now, we end where we began: with complexity, with nature, and with a hopeful question. If food is information, and polyphenols are its syntax, then perhaps within the citrus grove lies not just nutrition, but a code for reprogramming metabolism itself. And we have only just started decoding it.

Author Contributions

S.J.S. (Conceptualization: Writing—Original Draft: Lead; Writing—Review and Editing), M.S.M.; J.U.K.M.; M.H.S.; (Writing—Review and Editing: Equal), S.A.; F.A.F.S. and M.J.A.S.; (Writing—Review and Editing: Equal), A.A.R. and M.F.H. (Writing—Review and Editing: Equal), S.S.; R.A.S. and N.J. (Conceptualization: Equal), M.M.M. (Supervision: Equal), M.B.U. (Supervision: Lead; Project Administration: Lead). All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We utilized AI-based tools, to assist with grammar and language refinement in this manuscript. All the figures of this manuscript were created with licensed version of BioRender.com. We want to acknowledge Naivedya Choubey and Umayrah Mohiuddin for their help.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

AMPKAMP-activated protein kinase
ALTAlanine aminotransferase
ASTAspartate aminotransferase
ATF3Activating transcription factor 3
BMIBody mass index
CPT1Carnitine palmitoyltransferase 1
CRP/hs-CRPC-reactive protein/high-sensitivity C-reactive protein
EREndoplasmic reticulum
FAFatty acid
FASFatty acid synthase
FMDFlow-mediated dilation
FOXO1Forkhead box protein O1
GGTGamma-glutamyl transferase
GLP-1Glucagon-like peptide-1
GLUT4Glucose transporter type 4
HbA1cGlycated hemoglobin A1c
HDLHigh-density lipoprotein
HFDHigh-fat diet
HMGCR3-hydroxy-3-methylglutaryl-CoA reductase
HOMA-IRHomeostatic model assessment of insulin resistance
IL-1β, IL-6Interleukin-1 beta, Interleukin-6
IRS1Insulin receptor substrate 1
JNKc-Jun N-terminal kinase
LDLLow-density lipoprotein
MAPKMitogen-activated protein kinase
MASLDMetabolic dysfunction–associated steatotic liver disease
MCDMethionine-choline deficient diet
NF-κBNuclear factor kappa-light-chain-enhancer of activated B cells
NLRP3NOD-, LRR- and pyrin domain-containing protein 3 inflammasome
NONitric oxide
Nrf2Nuclear factor erythroid 2–related factor 2
OXPHOSOxidative phosphorylation
PERKPKR-like ER kinase
IRE1Inositol-requiring enzyme 1
ATF6Activating transcription factor 6
PGC-1αPeroxisome proliferator-activated receptor gamma coactivator 1-alpha
PI3K/AKTPhosphoinositide 3-kinase/protein kinase B pathway
PPARγPeroxisome proliferator-activated receptor gamma
ROSReactive oxygen species
SCFAShort-chain fatty acid
SODSuperoxide dismutase
SREBP1cSterol regulatory element-binding protein 1c
TFEBTranscription factor EB
TG/TCTriglycerides/Total cholesterol
TNF-αTumor necrosis factor-alpha
T2DMType 2 diabetes mellitus
UPRUnfolded protein response
UCP2Uncoupling protein 2
VLDLVery-low-density lipoprotein

References

  1. Allocca, S.; Monda, A.; Messina, A.; Casillo, M.; Sapuppo, W.; Monda, V.; Polito, R.; Di Maio, G.; Monda, M.; La Marra, M. Endocrine and Metabolic Mechanisms Linking Obesity to Type 2 Diabetes: Implications for Targeted Therapy. Healthcare 2025, 13, 1437. [Google Scholar] [CrossRef]
  2. Kyrou, I.; Randeva, H.S.; Tsigos, C.; Kaltsas, G.; Weickert, M.O. Clinical Problems Caused by Obesity; MD Text.com, Inc.: South Dartmouth, MA, USA, 2015. [Google Scholar]
  3. Saeedi, P.; Petersohn, I.; Salpea, P.; Malanda, B.; Karuranga, S.; Unwin, N.; Colagiuri, S.; Guariguata, L.; Motala, A.A.; Ogurtsova, K.; et al. Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: Results from the International Diabetes Federation Diabetes Atlas, 9(th) edition. Diabetes Res. Clin. Pract. 2019, 157, 107843. [Google Scholar] [PubMed]
  4. Ong, K.L.; Stafford, L.K.; McLaughlin, S.A.; Boyko, E.J.; Vollset, S.E.; Smith, A.E.; Dalton, B.E.; Duprey, J.; Cruz, J.A.; Hagins, H. Global, regional, and national burden of diabetes from 1990 to 2021, with projections of prevalence to 2050: A systematic analysis for the Global Burden of Disease Study 2021. Lancet 2023, 402, 203–234. [Google Scholar] [CrossRef]
  5. Kocatepe, D.; Büyükkol, D.C.; Hinislioğlu, K.N. Obesity Prevalence in World and Türkiye. North. J. Health Sci. 2025, 1, 26–32. [Google Scholar]
  6. Shimu, S.J. A 10-Year Retrospective Quantitative Analysis of The CDC Database: Examining the Prevalence of Depression in the Us Adult Urban Population. Front. Health Inform. 2024, 13, 2676–7104. [Google Scholar]
  7. Zatterale, F.; Longo, M.; Naderi, J.; Raciti, G.A.; Desiderio, A.; Miele, C.; Beguinot, F. Chronic adipose tissue inflammation linking obesity to insulin resistance and type 2 diabetes. Front. Physiol. 2020, 10, 1607. [Google Scholar] [CrossRef]
  8. Makki, K.; Froguel, P.; Wolowczuk, I. Adipose tissue in obesity-related inflammation and insulin resistance: Cells, cytokines, and chemokines. Int. Sch. Res. Not. 2013, 2013, 139239. [Google Scholar]
  9. Li, X.; Ren, Y.; Chang, K.; Wu, W.; Griffiths, H.R.; Lu, S.; Gao, D. Adipose tissue macrophages as potential targets for obesity and metabolic diseases. Front. Immunol. 2023, 14, 1153915. [Google Scholar] [CrossRef] [PubMed]
  10. Gkrinia, E.M.M.; Belančić, A. The Mechanisms of Chronic Inflammation in Obesity and Potential Therapeutic Strategies: A Narrative Review. Curr. Issues Mol. Biol. 2025, 47, 357. [Google Scholar] [CrossRef]
  11. Gudise, V.; Chowdhury, B. Molecular mechanisms and the vital roles of resistin, TLR 4, and NF-κB in treating type 2 diabetic complications. Beni-Suef Univ. J. Basic. Appl. Sci. 2020, 9, 54. [Google Scholar] [CrossRef]
  12. Kim, J.K. Endothelial nuclear factor κB in obesity and aging: Is endothelial nuclear factor κB a master regulator of inflammation and insulin resistance? Circulation 2012, 125, 1081–1083. [Google Scholar] [CrossRef]
  13. Chen, L.; Chen, R.; Wang, H.; Liang, F. Mechanisms linking inflammation to insulin resistance. Int. J. Endocrinol. 2015, 2015, 508409. [Google Scholar] [CrossRef]
  14. Sergi, D.; Zauli, E.; Celeghini, C.; Previati, M.; Zauli, G. Ceramides as the molecular link between impaired lipid metabolism, saturated fatty acid intake and insulin resistance: Are all saturated fatty acids to be blamed for ceramide-mediated lipotoxicity? Nutr. Res. Rev. 2024, 38, 256–266. [Google Scholar] [CrossRef]
  15. Römer, A.; Linn, T.; Petry, S.F. Lipotoxic impairment of mitochondrial function in β-cells: A review. Antioxidants 2021, 10, 293. [Google Scholar] [CrossRef]
  16. Corkey, B.E. Reactive oxygen species: Role in obesity and mitochondrial energy efficiency. Philos. Trans. R. Soc. B 2023, 378, 20220210. [Google Scholar] [CrossRef] [PubMed]
  17. Apostolova, N.; Vezza, T.; Muntane, J.; Rocha, M.; Victor, V.M. Mitochondrial dysfunction and mitophagy in type 2 diabetes: Pathophysiology and therapeutic targets. Antioxid. Redox Signal. 2023, 39, 278–320. [Google Scholar] [CrossRef] [PubMed]
  18. Patel, S.; Majumdar, A. Endoplasmic Reticulum Stress and Unfolded Protein Response in Metabolic Syndrome. In Biochemical Mechanisms for Metabolic Syndrome; Springer: Berlin/Heidelberg, Germany, 2024; pp. 203–222. [Google Scholar]
  19. Ghosh, R.; Colon-Negron, K.; Papa, F.R. Endoplasmic reticulum stress, degeneration of pancreatic islet β-cells, and therapeutic modulation of the unfolded protein response in diabetes. Mol. Metab. 2019, 27, S60–S68. [Google Scholar] [CrossRef]
  20. Aufi, S.S.A.; Hasnin, K. Diabesity An Emerging Epidemic and Advances in Treatment. Barind Med. Coll. J. 2024, 10, 29–34. [Google Scholar] [CrossRef]
  21. Mohib, M.; Afnan, K.; Paran, T.Z.; Khan, S.; Sarker, J.; Hasan, N.; Hasan, I.; Sagor, A.T. Beneficial role of citrus fruit polyphenols against hepatic dysfunctions: A review. J. Diet. Suppl. 2018, 15, 223–250. [Google Scholar] [CrossRef] [PubMed]
  22. Rizza, S.; Muniyappa, R.; Iantorno, M.; Kim, J.-A.; Chen, H.; Pullikotil, P.; Senese, N.; Tesauro, M.; Lauro, D.; Cardillo, C. Citrus polyphenol hesperidin stimulates production of nitric oxide in endothelial cells while improving endothelial function and reducing inflammatory markers in patients with metabolic syndrome. J. Clin. Endocrinol. Metab. 2011, 96, E782–E792. [Google Scholar] [CrossRef]
  23. Mahmoud, A.M.; Hernandez Bautista, R.J.; Sandhu, M.A.; Hussein, O.E. Beneficial Effects of Citrus Flavonoids on Cardiovascular and Metabolic Health. Oxidative Med. Cell. Longev. 2019, 2019, 5484138. [Google Scholar] [CrossRef]
  24. Xue, Y.; Huang, Z.; Chen, X.; Jia, G.; Zhao, H.; Liu, G. Naringin induces skeletal muscle fiber type transformation via AMPK/PGC-1α signaling pathway in mice and C2C12 myotubes. Nutr. Res. 2021, 92, 99–108. [Google Scholar] [CrossRef]
  25. Den Hartogh, D.J.; Tsiani, E. Antidiabetic properties of naringenin: A citrus fruit polyphenol. Biomolecules 2019, 9, 99. [Google Scholar] [CrossRef]
  26. Hammerschmidt, P.; Brüning, J.C. Contribution of specific ceramides to obesity-associated metabolic diseases. Cell. Mol. Life Sci. 2022, 79, 395. [Google Scholar] [CrossRef]
  27. Sears, B.; Perry, M. The role of fatty acids in insulin resistance. Lipids Health Dis. 2015, 14, 121. [Google Scholar] [CrossRef] [PubMed]
  28. Yung, J.H.M.; Giacca, A. Role of c-Jun N-terminal kinase (JNK) in obesity and type 2 diabetes. Cells 2020, 9, 706. [Google Scholar] [CrossRef] [PubMed]
  29. Kim, G.-T.; Kim, S.-J.; Park, S.-H.; Lee, D.; Park, T.-S. Hepatic expression of the serine palmitoyltransferase subunit Sptlc2 reduces lipid droplets in the liver by activating VLDL secretion. J. Lipid Atheroscler. 2020, 9, 291. [Google Scholar] [CrossRef]
  30. 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]
  31. Sokolowska, E.; Blachnio-Zabielska, A. The role of ceramides in insulin resistance. Front. Endocrinol. 2019, 10, 577. [Google Scholar] [CrossRef]
  32. Schrauwen, P.; Hesselink, M.K. Oxidative capacity, lipotoxicity, and mitochondrial damage in type 2 diabetes. Diabetes 2004, 53, 1412–1417. [Google Scholar] [CrossRef] [PubMed]
  33. Dubé, J.J.; Amati, F.; Stefanovic-Racic, M.; Toledo, F.G.; Sauers, S.E.; Goodpaster, B.H. Exercise-induced alterations in intramyocellular lipids and insulin resistance: The athlete’s paradox revisited. Am. J. Physiol.-Endocrinol. Metab. 2008, 294, E882–E888. [Google Scholar] [CrossRef]
  34. Goodpaster, B.H.; Sparks, L.M. Metabolic flexibility in health and disease. Cell Metab. 2017, 25, 1027–1036. [Google Scholar] [CrossRef]
  35. Mohib, M.M.; Rabe, S.; Nolze, A.; Rooney, M.; Ain, Q.; Zipprich, A.; Gekle, M.; Schreier, B. Eplerenone, a mineralocorticoid receptor inhibitor, reduces cirrhosis associated changes of hepatocyte glucose and lipid metabolism. Cell Commun. Signal. 2024, 22, 614. [Google Scholar] [CrossRef] [PubMed]
  36. Cusi, K. Role of obesity and lipotoxicity in the development of nonalcoholic steatohepatitis: Pathophysiology and clinical implications. Gastroenterology 2012, 142, 711–725.e6. [Google Scholar] [CrossRef] [PubMed]
  37. Qin, W.; Weng, J. Hepatocyte NLRP3 interacts with PKCε to drive hepatic insulin resistance and steatosis. Sci. Bull. 2023, 68, 1413–1429. [Google Scholar] [CrossRef] [PubMed]
  38. Mota, M.; Banini, B.A.; Cazanave, S.C.; Sanyal, A.J. Molecular mechanisms of lipotoxicity and glucotoxicity in nonalcoholic fatty liver disease. Metabolism 2016, 65, 1049–1061. [Google Scholar] [CrossRef]
  39. Huang, W.; Metlakunta, A.; Dedousis, N.; Zhang, P.; Sipula, I.; Dube, J.J.; Scott, D.K.; O’Doherty, R.M. Depletion of liver Kupffer cells prevents the development of diet-induced hepatic steatosis and insulin resistance. Diabetes 2010, 59, 347–357. [Google Scholar] [CrossRef]
  40. Ziolkowska, S.; Binienda, A.; Jabłkowski, M.; Szemraj, J.; Czarny, P. The Interplay between Insulin Resistance, Inflammation, Oxidative Stress, Base Excision Repair and Metabolic Syndrome in Nonalcoholic Fatty Liver Disease. Int. J. Mol. Sci. 2021, 22, 11128. [Google Scholar] [CrossRef]
  41. Gruben, N.; Shiri-Sverdlov, R.; Koonen, D.P.; Hofker, M.H. Nonalcoholic fatty liver disease: A main driver of insulin resistance or a dangerous liaison? Biochim. Biophys. Acta 2014, 1842, 2329–2343. [Google Scholar] [CrossRef]
  42. Chen, Z.; Yu, R.; Xiong, Y.; Du, F.; Zhu, S. A vicious circle between insulin resistance and inflammation in nonalcoholic fatty liver disease. Lipids Health Dis. 2017, 16, 203. [Google Scholar] [CrossRef]
  43. Ly, L.D.; Xu, S.; Choi, S.K.; Ha, C.M.; Thoudam, T.; Cha, S.K.; Wiederkehr, A.; Wollheim, C.B.; Lee, I.K.; Park, K.S. Oxidative stress and calcium dysregulation by palmitate in type 2 diabetes. Exp. Mol. Med. 2017, 49, e291. [Google Scholar] [CrossRef]
  44. Morawietz, H.; Brendel, H.; Diaba-Nuhoho, P.; Catar, R.; Perakakis, N.; Wolfrum, C.; Bornstein, S.R. Cross-Talk of NADPH Oxidases and Inflammation in Obesity. Antioxidants 2023, 12, 1589. [Google Scholar] [CrossRef]
  45. Vilas-Boas, E.A.; Almeida, D.C.; Roma, L.P.; Ortis, F.; Carpinelli, A.R. Lipotoxicity and β-Cell Failure in Type 2 Diabetes: Oxidative Stress Linked to NADPH Oxidase and ER Stress. Cells 2021, 10, 3328. [Google Scholar] [CrossRef]
  46. Wang, S.W.; Sheng, H.; Bai, Y.-F.; Weng, Y.-Y.; Fan, X.-Y.; Lou, L.-J.; Zhang, F. Neohesperidin enhances PGC-1α-mediated mitochondrial biogenesis and alleviates hepatic steatosis in high fat diet fed mice. Nutr. Diabetes 2020, 10, 27. [Google Scholar] [CrossRef]
  47. Morrow, N.M.; Burke, A.C.; Samsoondar, J.P.; Seigel, K.E.; Wang, A.; Telford, D.E.; Sutherland, B.G.; O’Dwyer, C.; Steinberg, G.R.; Fullerton, M.D.; et al. The citrus flavonoid nobiletin confers protection from metabolic dysregulation in high-fat-fed mice independent of AMPK. J. Lipid Res. 2020, 61, 387–402. [Google Scholar] [CrossRef] [PubMed]
  48. Lu, Z.; Li, X.; Wang, M.; Zhang, X.; Zhuang, R.; Wu, F.; Li, W.; Zhu, W.; Zhang, B. Liver-Specific Bmal1 Depletion Reverses the Beneficial Effects of Nobiletin on Liver Cholesterol Homeostasis in Mice Fed with High-Fat Diet. Nutrients 2023, 15, 2547. [Google Scholar] [CrossRef] [PubMed]
  49. Lee, Y.S.; Cha, B.-Y.; Saito, K.; Yamakawa, H.; Choi, S.-S.; Yamaguchi, K.; Yonezawa, T.; Teruya, T.; Nagai, K.; Woo, J.-T. Nobiletin improves hyperglycemia and insulin resistance in obese diabetic ob/ob mice. Biochem. Pharmacol. 2010, 79, 1674–1683. [Google Scholar] [CrossRef] [PubMed]
  50. Yang, Y.; Wu, Y.; Zou, J.; Wang, Y.-H.; Xu, M.-X.; Huang, W.; Yu, D.-J.; Zhang, L.; Zhang, Y.-Y.; Sun, X.-D. Naringenin Attenuates Non-Alcoholic Fatty Liver Disease by Enhancing Energy Expenditure and Regulating Autophagy via AMPK. Front. Pharmacol. 2021, 12, 687095. [Google Scholar] [CrossRef]
  51. Liu, X.; Wang, N.; Fan, S.; Zheng, X.; Yang, Y.; Zhu, Y.; Lu, Y.; Chen, Q.; Zhou, H.; Zheng, J. The citrus flavonoid naringenin confers protection in a murine endotoxaemia model through AMPK-ATF3-dependent negative regulation of the TLR4 signalling pathway. Sci. Rep. 2016, 6, 39735. [Google Scholar] [CrossRef]
  52. Sarkar, S.; Ghosh, S.; Biswas, M. Naringin ameliorates high-fat diet-induced hepatotoxicity and dyslipidemia in experimental rat model via modulation of anti-oxidant enzymes, AMPK and SERBP-1c signaling pathways. Toxicol. Rep. 2025, 14, 102062. [Google Scholar] [CrossRef]
  53. Pengnet, S.; Sumarithum, P.; Phongnu, N.; Prommaouan, S.; Kantip, N.; Phoungpetchara, I.; Malakul, W. Naringin attenuates fructose-induced NAFLD progression in rats through reducing endogenous triglyceride synthesis and activating the Nrf2/HO-1 pathway. Front. Pharmacol. 2022, 13, 1049818. [Google Scholar] [CrossRef]
  54. Guan, L.; Guo, L.; Zhang, H.; Liu, H.; Zhou, W.; Zhai, Y.; Yan, X.; Men, X.; Peng, L. Naringin Protects against Non-Alcoholic Fatty Liver Disease by Promoting Autophagic Flux and Lipophagy. Mol. Nutr. Food Res. 2024, 68, e2200812. [Google Scholar] [CrossRef]
  55. Tian, M.; Han, Y.-B.; Zhao, C.-C.; Liu, L.; Zhang, F.-L. Hesperidin alleviates insulin resistance by improving HG-induced oxidative stress and mitochondrial dysfunction by restoring miR-149. Diabetol. Metab. Syndr. 2021, 13, 50. [Google Scholar] [CrossRef]
  56. Estruel-Amades, S.; Massot-Cladera, M.; Garcia-Cerdà, P.; Pérez-Cano, F.J.; Franch, À.; Castell, M.; Camps-Bossacoma, M. Protective Effect of Hesperidin on the Oxidative Stress Induced by an Exhausting Exercise in Intensively Trained Rats. Nutrients 2019, 11, 783. [Google Scholar] [CrossRef] [PubMed]
  57. Morshedzadeh, N.; Ahmadi, A.R.; Behrouz, V.; Mir, E. A narrative review on the role of hesperidin on metabolic parameters, liver enzymes, and inflammatory markers in nonalcoholic fatty liver disease. Food Sci. Nutr. 2023, 11, 7523–7533. [Google Scholar] [CrossRef]
  58. Yeh, C.H.; Shen, Z.Q.; Wang, T.W.; Kao, C.H.; Teng, Y.C.; Yeh, T.K.; Lu, C.K.; Tsai, T.F. Hesperetin promotes longevity and delays aging via activation of Cisd2 in naturally aged mice. J. Biomed. Sci. 2022, 29, 53. [Google Scholar] [CrossRef]
  59. Durço, A.O.; de Souza, D.S.; Heimfarth, L.; Miguel-Dos-Santos, R.; Rabelo, T.K.; Barreto, T.d.O.; Rhana, P.; Santana, M.N.S.; Braga, W.F.; Cruz, J.d.S.; et al. d-Limonene Ameliorates Myocardial Infarction Injury by Reducing Reactive Oxygen Species and Cell Apoptosis in a Murine Model. J. Nat. Prod. 2019, 82, 3010–3019. [Google Scholar] [CrossRef] [PubMed]
  60. Valerii, M.C.; Turroni, S.; Ferreri, C.; Zaro, M.; Sansone, A.; Dalpiaz, A.; Botti, G.; Ferraro, L.; Spigarelli, R.; Bellocchio, I.; et al. Effect of a Fiber D-Limonene-Enriched Food Supplement on Intestinal Microbiota and Metabolic Parameters of Mice on a High-Fat Diet. Pharmaceutics 2021, 13, 1753. [Google Scholar] [CrossRef]
  61. Hiramitsu, M.; Shimada, Y.; Kuroyanagi, J.; Inoue, T.; Katagiri, T.; Zang, L.; Nishimura, Y.; Nishimura, N.; Tanaka, T. Eriocitrin ameliorates diet-induced hepatic steatosis with activation of mitochondrial biogenesis. Sci. Rep. 2014, 4, 3708. [Google Scholar] [CrossRef]
  62. Tsutsumi, R.; Yoshida, T.; Nii, Y.; Okahisa, N.; Iwata, S.; Tsukayama, M.; Hashimoto, R.; Taniguchi, Y.; Sakaue, H.; Hosaka, T.; et al. Sudachitin, a polymethoxylated flavone, improves glucose and lipid metabolism by increasing mitochondrial biogenesis in skeletal muscle. Nutr. Metab. 2014, 11, 32. [Google Scholar] [CrossRef]
  63. Sundaram, R.; Shanthi, P.; Sachdanandam, P. Effect of tangeretin, a polymethoxylated flavone on glucose metabolism in streptozotocin-induced diabetic rats. Phytomedicine 2014, 21, 793–799. [Google Scholar] [CrossRef]
  64. Wang, Q.; Ou, Y.; Hu, G.; Wen, C.; Yue, S.; Chen, C.; Xu, L.; Xie, J.; Dai, H.; Xiao, H.; et al. Naringenin attenuates non-alcoholic fatty liver disease by down-regulating the NLRP3/NF-κB pathway in mice. Br. J. Pharmacol. 2020, 177, 1806–1821. [Google Scholar]
  65. Wu, L.; Niu, Y.; Ren, B.; Wang, S.; Song, Y.; Wang, X.; Zhao, K.; Yue, Z.; Li, Y.; Gao, J. Naringenin Promotes Gastrointestinal Motility in Mice by Impacting the SCF/c-Kit Pathway and Gut Microbiota. Foods 2024, 13, 2520. [Google Scholar] [PubMed]
  66. Zygmunt, K.; Faubert, B.; MacNeil, J.; Tsiani, E. Naringenin, a citrus flavonoid, increases muscle cell glucose uptake via AMPK. Biochem. Biophys. Res. Commun. 2010, 398, 178–183. [Google Scholar] [CrossRef] [PubMed]
  67. Pu, P.; Gao, D.M.; Mohamed, S.; Chen, J.; Zhang, J.; Zhou, X.Y.; Zhou, N.J.; Xie, J.; Jiang, H. Naringin ameliorates metabolic syndrome by activating AMP-activated protein kinase in mice fed a high-fat diet. Arch. Biochem. Biophys. 2012, 518, 61–70. [Google Scholar] [CrossRef]
  68. Dong, L.; Lou, W.; Xu, C.; Wang, J. Naringenin cationic lipid-modified nanoparticles mitigate MASLD progression by modulating lipid homeostasis and gut microbiota. J. Nanobiotechnology 2025, 23, 168. [Google Scholar] [CrossRef] [PubMed]
  69. Lv, Z.; Meng, J.; Yao, S.; Xiao, F.; Li, S.; Shi, H.; Cui, C.; Chen, K.; Luo, X.; Ye, Y.; et al. Naringenin improves muscle endurance via activation of the Sp1-ERRγ transcriptional axis. Cell Rep. 2023, 42, 113288. [Google Scholar] [CrossRef]
  70. Jia, S.; Hu, Y.; Zhang, W.; Zhao, X.; Chen, Y.; Sun, C.; Li, X.; Chen, K. Hypoglycemic and hypolipidemic effects of neohesperidin derived from Citrus aurantium L. in diabetic KK-A(y) mice. Food Funct. 2015, 6, 878–886. [Google Scholar]
  71. Wu, H.; Liu, Y.; Chen, X.; Zhu, D.; Ma, J.; Yan, Y.; Si, M.; Li, X.; Sun, C.; Yang, B.; et al. Neohesperidin Exerts Lipid-Regulating Effects in vitro and in vivo via Fibroblast Growth Factor 21 and AMP-Activated Protein Kinase/Sirtuin Type 1/Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1α Signaling Axis. Pharmacology 2017, 100, 115–126. [Google Scholar]
  72. Jamal, A.; Brettle, H.; Jamil, D.A.; Tran, V.; Diep, H.; Bobik, A.; van der Poel, C.; Vinh, A.; Drummond, G.R.; Thomas, C.J.; et al. Reduced Insulin Resistance and Oxidative Stress in a Mouse Model of Metabolic Syndrome following Twelve Weeks of Citrus Bioflavonoid Hesperidin Supplementation: A Dose-Response Study. Biomolecules 2024, 14, 637. [Google Scholar]
  73. Yuk, T.; Kim, Y.; Yang, J.; Sung, J.; Jeong, H.S.; Lee, J.; Dini, L. Nobiletin Inhibits Hepatic Lipogenesis via Activation of AMP-Activated Protein Kinase. Evid. Based Complement. Altern. Med. 2018, 2018, 7420265. [Google Scholar] [CrossRef]
  74. Rains, J.L.; Jain, S.K. Oxidative stress, insulin signaling, and diabetes. Free Radic. Biol. Med. 2011, 50, 567–575. [Google Scholar] [CrossRef]
  75. Manna, P.; Jain, S.K. Obesity, Oxidative Stress, Adipose Tissue Dysfunction, and the Associated Health Risks: Causes and Therapeutic Strategies. Metab. Syndr. Relat. Disord. 2015, 13, 423–444. [Google Scholar] [CrossRef]
  76. Bhatti, J.S.; Bhatti, G.K.; Reddy, P.H. Mitochondrial dysfunction and oxidative stress in metabolic disorders—A step towards mitochondria based therapeutic strategies. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 1066–1077. [Google Scholar] [CrossRef] [PubMed]
  77. Cojocaru, K.A.; Luchian, I.; Goriuc, A.; Antoci, L.M.; Ciobanu, C.G.; Popescu, R.; Vlad, C.E.; Blaj, M.; Foia, L.G. Mitochondrial Dysfunction, Oxidative Stress, and Therapeutic Strategies in Diabetes, Obesity, and Cardiovascular Disease. Antioxidants 2023, 12, 658. [Google Scholar] [CrossRef]
  78. Argaev-Frenkel, L.; Rosenzweig, T. Redox Balance in Type 2 Diabetes: Therapeutic Potential and the Challenge of Antioxidant-Based Therapy. Antioxidants 2023, 12, 994. [Google Scholar] [CrossRef] [PubMed]
  79. Shrestha, N.; De Franco, E.; Arvan, P.; Cnop, M. Pathological β-Cell Endoplasmic Reticulum Stress in Type 2 Diabetes: Current Evidence. Front. Endocrinol. 2021, 12, 650158. [Google Scholar] [CrossRef] [PubMed]
  80. Kim, G.; Lee, J.; Ha, J.; Kang, I.; Choe, W. Endoplasmic Reticulum Stress and Its Impact on Adipogenesis: Molecular Mechanisms Implicated. Nutrients 2023, 15, 5082. [Google Scholar] [CrossRef]
  81. Bhattarai, K.R.; Riaz, T.A.; Kim, H.R.; Chae, H.J. The aftermath of the interplay between the endoplasmic reticulum stress response and redox signaling. Exp. Mol. Med. 2021, 53, 151–167. [Google Scholar] [CrossRef]
  82. Veluthakal, R.; Esparza, D.; Hoolachan, J.M.; Balakrishnan, R.; Ahn, M.; Oh, E.; Jayasena, C.S.; Thurmond, D.C. Mitochondrial Dysfunction, Oxidative Stress, and Inter-Organ Miscommunications in T2D Progression. Int. J. Mol. Sci. 2024, 25, 1504. [Google Scholar]
  83. Benito-Vicente, A.; Jebari-Benslaiman, S.; Galicia-Garcia, U.; Larrea-Sebal, A.; Uribe, K.B.; Martin, C. Molecular mechanisms of lipotoxicity-induced pancreatic β-cell dysfunction. Int. Rev. Cell Mol. Biol. 2021, 359, 357–402. [Google Scholar]
  84. Biondi, G.; Marrano, N.; Borrelli, A.; Rella, M.; Palma, G.; Calderoni, I.; Siciliano, E.; Lops, P.; Giorgino, F.; Natalicchio, A. Adipose Tissue Secretion Pattern Influences β-Cell Wellness in the Transition from Obesity to Type 2 Diabetes. Int. J. Mol. Sci. 2022, 23, 5522. [Google Scholar] [CrossRef]
  85. Ježek, P.; Jabůrek, M.; Holendová, B.; Plecitá-Hlavatá, L. Fatty Acid-Stimulated Insulin Secretion vs. Lipotoxicity. Molecules 2018, 23, 1483. [Google Scholar]
  86. Kim, J.W.; Yoon, K.H. Glucolipotoxicity in Pancreatic β-Cells. Diabetes Metab. J. 2011, 35, 444–450. [Google Scholar] [CrossRef] [PubMed]
  87. Dalle, S.; Abderrahmani, A.; Renard, E. Pharmacological inhibitors of β-cell dysfunction and death as therapeutics for diabetes. Front. Endocrinol. 2023, 14, 1076343. [Google Scholar] [CrossRef]
  88. Brozzi, F.; Eizirik, D.L. ER stress and the decline and fall of pancreatic beta cells in type 1 diabetes. Ups. J. Med. Sci. 2016, 121, 133–139. [Google Scholar] [CrossRef] [PubMed]
  89. Diane, A.; Allouch, A.; Mu-U-Min, R.B.A.; Al-Siddiqi, H.H. Endoplasmic reticulum stress in pancreatic β-cell dysfunctionality and diabetes mellitus: A promising target for generation of functional hPSC-derived β-cells in vitro. Front. Endocrinol. 2024, 15, 1386471. [Google Scholar] [CrossRef] [PubMed]
  90. Yang, L.; Yao, D.; Yang, H.; Wei, Y.; Peng, Y.; Ding, Y.; Shu, L. Puerarin Protects Pancreatic β-Cells in Obese Diabetic Mice via Activation of GLP-1R Signaling. Mol. Endocrinol. 2016, 30, 361–371. [Google Scholar]
  91. Yusta, B.; Baggio, L.L.; Estall, J.L.; Koehler, J.A.; Holland, D.P.; Li, H.; Pipeleers, D.; Ling, Z.; Drucker, D.J. GLP-1 receptor activation improves beta cell function and survival following induction of endoplasmic reticulum stress. Cell Metab. 2006, 4, 391–406. [Google Scholar]
  92. Li, Y.; Zhu, J.; Yue, C.; Song, S.; Tian, L.; Wang, Y. Recent advances in pancreatic α-cell transdifferentiation for diabetes therapy. Front. Immunol. 2025, 16, 1551372. [Google Scholar] [CrossRef]
  93. Choe, S.S.; Huh, J.Y.; Hwang, I.J.; Kim, J.I.; Kim, J.B. Adipose Tissue Remodeling: Its Role in Energy Metabolism and Metabolic Disorders. Front. Endocrinol. 2016, 7, 30. [Google Scholar] [CrossRef] [PubMed]
  94. Huynh, P.M.; Wang, F.; An, Y.A. Hypoxia signaling in the adipose tissue. J. Mol. Cell Biol. 2025, 16, mjae039. [Google Scholar] [CrossRef]
  95. Mirabelli, M.; Misiti, R.; Sicilia, L.; Brunetti, F.S.; Chiefari, E.; Brunetti, A.; Foti, D.P. Hypoxia in Human Obesity: New Insights from Inflammation towards Insulin Resistance-A Narrative Review. Int. J. Mol. Sci. 2024, 25, 9802. [Google Scholar]
  96. Begum, M.; Choubey, M.; Tirumalasetty, M.B.; Arbee, S.; Mohib, M.M.; Wahiduzzaman, M.; Mamun, M.A.; Uddin, M.B.; Mohiuddin, M.S. Adiponectin: A promising target for the treatment of diabetes and its complications. Life 2023, 13, 2213. [Google Scholar] [CrossRef]
  97. Corvera, S. Cellular Heterogeneity in Adipose Tissues. Annu. Rev. Physiol. 2021, 83, 257–278. [Google Scholar] [CrossRef]
  98. Massier, L.; Jalkanen, J.; Elmastas, M.; Zhong, J.; Wang, T.; Nono Nankam, P.A.; Frendo-Cumbo, S.; Bäckdahl, J.; Subramanian, N.; Sekine, T.; et al. An integrated single cell and spatial transcriptomic map of human white adipose tissue. Nat. Commun. 2023, 14, 1438. [Google Scholar] [CrossRef]
  99. Sultana, A.; Jahan Shimu, S.; Jakanta Faika, M.; Islam, T.; Ferdous, N.; Nessa, A. Bangladeshi parents’ knowledge and awareness about cervical cancer and willingness to vaccinate female family members against human papilloma virus: A cross sectional study. Int. J. Community Med. Public. Health 2023, 10, 3446–3453. [Google Scholar] [CrossRef]
  100. Khin, P.P.; Lee, J.H.; Jun, H.-S. Pancreatic beta-cell dysfunction in type 2 diabetes. Eur. J. Inflamm. 2023, 21. [Google Scholar] [CrossRef]
  101. Chen, B.; Li, T.; Wu, Y.; Song, L.; Wang, Y.; Bian, Y.; Qiu, Y.; Yang, Z. Lipotoxicity: A New Perspective in Type 2 Diabetes Mellitus. Diabetes Metab. Syndr. Obes. 2025, 18, 1223–1237. [Google Scholar] [CrossRef] [PubMed]
  102. Sandoval, V.; Sanz-Lamora, H.; Arias, G.; Marrero, P.F.; Haro, D.; Relat, J. Metabolic impact of flavonoids consumption in obesity: From central to peripheral. Nutrients 2020, 12, 2393. [Google Scholar] [CrossRef] [PubMed]
  103. Mirzaei, A.; Mirzaei, A.; Najjar Khalilabad, S.; Askari, V.R.; Baradaran Rahimi, V. Promising influences of hesperidin and hesperetin against diabetes and its complications: A systematic review of molecular, cellular, and metabolic effects. Excli J. 2023, 22, 1235–1263. [Google Scholar]
  104. Russo, B.; Picconi, F.; Malandrucco, I.; Frontoni, S. Flavonoids and insulin-resistance: From molecular evidences to clinical trials. Int. J. Mol. Sci. 2019, 20, 2061. [Google Scholar] [CrossRef] [PubMed]
  105. Yi, H.; Peng, H.; Wu, X.; Xu, X.; Kuang, T.; Zhang, J.; Du, L.; Fan, G. The therapeutic effects and mechanisms of quercetin on metabolic diseases: Pharmacological data and clinical evidence. Oxidative Med. Cell. Longev. 2021, 2021, 6678662. [Google Scholar] [CrossRef] [PubMed]
  106. Dong, J.; Zhang, X.; Zhang, L.; Bian, H.X.; Xu, N.; Bao, B.; Liu, J. Quercetin reduces obesity-associated ATM infiltration and inflammation in mice: A mechanism including AMPKα1/SIRT1. J. Lipid Res. 2014, 55, 363–374. [Google Scholar] [CrossRef] [PubMed]
  107. Tsai, C.F.; Chen, G.W.; Chen, Y.C.; Shen, C.K.; Lu, D.Y.; Yang, L.Y.; Chen, J.H.; Yeh, W.L. Regulatory Effects of Quercetin on M1/M2 Macrophage Polarization and Oxidative/Antioxidative Balance. Nutrients 2021, 14, 67. [Google Scholar] [CrossRef]
  108. Kohjima, M.; Higuchi, N.; Kato, M.; Kotoh, K.; Yoshimoto, T.; Fujino, T.; Yada, M.; Yada, R.; Harada, N.; Enjoji, M. SREBP-1c, regulated by the insulin and AMPK signaling pathways, plays a role in nonalcoholic fatty liver disease. Int. J. Mol. Med. 2008, 21, 507–511. [Google Scholar] [CrossRef]
  109. Lee, J.; Hong, S.-W.; Rhee, E.-J.; Lee, W.-Y. GLP-1 receptor agonist and non-alcoholic fatty liver disease. Diabetes Metab. J. 2012, 36, 262. [Google Scholar] [CrossRef]
  110. Li, S.; Zhang, Y.; Sun, Y.; Zhang, G.; Bai, J.; Guo, J.; Su, X.; Du, H.; Cao, X.; Yang, J. Naringenin improves insulin sensitivity in gestational diabetes mellitus mice through AMPK. Nutr. Diabetes 2019, 9, 28. [Google Scholar] [CrossRef]
  111. Nery, M.; Ferreira, P.S.; Gonçalves, D.R.; Spolidorio, L.C.; Manthey, J.A.; Cesar, T.B. Physiological effects of tangeretin and heptamethoxyflavone on obese C57BL/6J mice fed a high-fat diet and analyses of the metabolites originating from these two polymethoxylated flavones. Food Sci. Nutr. 2021, 9, 1997–2009. [Google Scholar] [CrossRef]
  112. La Spina, M.; Galletta, E.; Azzolini, M.; Gomez Zorita, S.; Parrasia, S.; Salvalaio, M.; Salmaso, A.; Biasutto, L. Browning effects of a chronic pterostilbene supplementation in mice fed a high-fat diet. Int. J. Mol. Sci. 2019, 20, 5377. [Google Scholar] [CrossRef]
  113. Chen, Q.; Wang, D.; Gu, Y.; Jiang, Z.; Zhou, Z. Tangeretin prevents obesity by modulating systemic inflammation, fat browning, and gut microbiota in high-fat diet-induced obese C57BL/6 mice. J. Nutr. Biochem. 2022, 101, 108943. [Google Scholar] [CrossRef]
  114. Gao, J.; Zhang, M.; Niu, R.; Gu, X.; Hao, E.; Hou, X.; Deng, J.; Bai, G. The combination of cinnamaldehyde and kaempferol ameliorates glucose and lipid metabolism disorders by enhancing lipid metabolism via AMPK activation. J. Funct. Foods 2021, 83, 104556. [Google Scholar] [CrossRef]
  115. Park, J.E.; Yoo, J.; Han, J.S. HM-Chromanone Alleviates Hyperglycemia by Activating AMPK and PI3K/AKT Pathways in Mice Fed a High-Fat Diet. Nutrients 2024, 16, 3972. [Google Scholar]
  116. Chang, W.-T.; Huang, S.-C.; Cheng, H.-L.; Chen, S.-C.; Hsu, C.-L. Rutin and gallic acid regulates mitochondrial functions via the SIRT1 pathway in C2C12 myotubes. Antioxidants 2021, 10, 286. [Google Scholar] [CrossRef] [PubMed]
  117. Alkhalidy, H.; Moore, W.; Wang, Y.; Luo, J.; McMillan, R.P.; Zhen, W.; Zhou, K.; Liu, D. The flavonoid kaempferol ameliorates streptozotocin-induced diabetes by suppressing hepatic glucose production. Molecules 2018, 23, 2338. [Google Scholar] [CrossRef]
  118. Hotamisligil, G.S.; Shargill, N.S.; Spiegelman, B.M. Adipose expression of tumor necrosis factor-alpha: Direct role in obesity-linked insulin resistance. Science 1993, 259, 87–91. [Google Scholar] [CrossRef]
  119. Hotamisligil, G.S.; Arner, P.; Caro, J.F.; Atkinson, R.L.; Spiegelman, B.M. Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. J. Clin. Invest. 1995, 95, 2409–2415. [Google Scholar] [CrossRef]
  120. Shimu, S.J.; Islam, S. Gender Differences in Drug Addiction: Neurobiological, Social, and Psychological Perspectives in Women–A Systematic Review. J. Primeasia 2025, 6, 1–13. [Google Scholar]
  121. Gao, Z.; Hwang, D.; Bataille, F.; Lefevre, M.; York, D.; Quon, M.J.; Ye, J. Serine phosphorylation of insulin receptor substrate 1 by inhibitor kappa B kinase complex. J. Biol. Chem. 2002, 277, 48115–48121. [Google Scholar] [CrossRef] [PubMed]
  122. Kanety, H.; Feinstein, R.; Papa, M.Z.; Hemi, R.; Karasik, A. Tumor necrosis factor alpha-induced phosphorylation of insulin receptor substrate-1 (IRS-1). Possible mechanism for suppression of insulin-stimulated tyrosine phosphorylation of IRS-1. J. Biol. Chem. 1995, 270, 23780–23784. [Google Scholar] [CrossRef]
  123. Martínez Báez, A.; Ayala, G.; Pedroza-Saavedra, A.; González-Sánchez, H.M.; Chihu Amparan, L. Phosphorylation codes in IRS-1 and IRS-2 are associated with the activation/inhibition of insulin canonical signaling pathways. Curr. Issues Mol. Biol. 2024, 46, 634–649. [Google Scholar] [CrossRef]
  124. Baker, R.G.; Hayden, M.S.; Ghosh, S. NF-κB, inflammation, and metabolic disease. Cell Metab. 2011, 13, 11–22. [Google Scholar] [CrossRef]
  125. Kanda, H.; Tateya, S.; Tamori, Y.; Kotani, K.; Hiasa, K.-I.; Kitazawa, R.; Kitazawa, S.; Miyachi, H.; Maeda, S.; Egashira, K. MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity. J. Clin. Investig. 2006, 116, 1494–1505. [Google Scholar] [CrossRef] [PubMed]
  126. Apaijit, K.; Pakdeechote, P.; Maneesai, P.; Meephat, S.; Prasatthong, P.; Bunbupha, S. Hesperidin Alleviates Vascular Dysfunction and Remodelling in High-Fat/High-Fructose Diet-Fed Rats by Modulating Oxidative Stress, Inflammation, AdipoR1, and eNOS Expression. Tissue Cell 2022, 78, 101901. [Google Scholar] [CrossRef] [PubMed]
  127. Alam, M.A.; Subhan, N.; Rahman, M.M.; Uddin, S.J.; Reza, H.M.; Sarker, S.D. Effect of citrus flavonoids, naringin and naringenin, on metabolic syndrome and their mechanisms of action. Adv. Nutr. 2014, 5, 404–417. [Google Scholar] [CrossRef] [PubMed]
  128. Shen, C.-Y.; Lu, C.-H.; Wu, C.-H.; Li, K.-J.; Kuo, Y.-M.; Hsieh, S.-C.; Yu, C.-L. The development of maillard reaction, and advanced glycation end product (AGE)-receptor for AGE (RAGE) signaling inhibitors as novel therapeutic strategies for patients with AGE-related diseases. Molecules 2020, 25, 5591. [Google Scholar] [CrossRef]
  129. Syed, A.A.; Reza, M.I.; Shafiq, M.; Kumariya, S.; Singh, P.; Husain, A.; Hanif, K.; Gayen, J.R. Naringin ameliorates type 2 diabetes mellitus-induced steatohepatitis by inhibiting RAGE/NF-κB mediated mitochondrial apoptosis. Life Sci. 2020, 257, 118118. [Google Scholar] [CrossRef]
  130. Cepas, V.; Collino, M.; Mayo, J.C.; Sainz, R.M. Redox Signaling and Advanced Glycation Endproducts (AGEs) in Diet-Related Diseases. Antioxidants 2020, 9, 142. [Google Scholar] [CrossRef]
  131. Al-Aubaidy, H.A.; Dayan, A.; Deseo, M.A.; Itsiopoulos, C.; Jamil, D.; Hadi, N.R.; Thomas, C.J. Twelve-week mediterranean diet intervention increases citrus bioflavonoid levels and reduces inflammation in people with type 2 diabetes mellitus. Nutrients 2021, 13, 1133. [Google Scholar] [CrossRef]
  132. Passaro, A.; Sanz, J.M.; Naumovski, N.; Sergi, D. The complex interplay between oxinflammation, mitochondrial dysfunction and lipotoxicity: Focus on their role in the pathogenesis of skeletal muscle insulin resistance and modulation by dietary fatty acids. Adv. Redox Res. 2024, 11, 100100. [Google Scholar] [CrossRef]
  133. Cavaliere, G.; Cimmino, F.; Trinchese, G.; Catapano, A.; Petrella, L.; D’Angelo, M.; Lucchin, L.; Mollica, M.P. From obesity-induced low-grade inflammation to lipotoxicity and mitochondrial dysfunction: Altered multi-crosstalk between adipose tissue and metabolically active organs. Antioxidants 2023, 12, 1172. [Google Scholar] [PubMed]
  134. Kicinska, A.; Jarmuszkiewicz, W. Flavonoids and mitochondria: Activation of cytoprotective pathways? Molecules 2020, 25, 3060. [Google Scholar] [CrossRef] [PubMed]
  135. Armani, A.; Feraco, A.; Camajani, E.; Gorini, S.; Lombardo, M.; Caprio, M. Nutraceuticals in Brown Adipose Tissue Activation. Cells 2022, 11, 3996. [Google Scholar] [CrossRef]
  136. Zong, Y.; Li, H.; Liao, P.; Chen, L.; Pan, Y.; Zheng, Y.; Zhang, C.; Liu, D.; Zheng, M.; Gao, J. Mitochondrial dysfunction: Mechanisms and advances in therapy. Signal Transduct. Target. Ther. 2024, 9, 124. [Google Scholar] [CrossRef]
  137. Muoio, D.M.; Neufer, P.D. Lipid-induced mitochondrial stress and insulin action in muscle. Cell Metab. 2012, 15, 595–605. [Google Scholar] [CrossRef]
  138. Narendra, D.P.; Jin, S.M.; Tanaka, A.; Suen, D.-F.; Gautier, C.A.; Shen, J.; Cookson, M.R.; Youle, R.J. PINK1 is selectively stabilized on impaired mitochondria to activate Parkin. PLoS Biol. 2010, 8, e1000298. [Google Scholar] [CrossRef]
  139. Lemos, G.d.O.; Torrinhas, R.S.; Waitzberg, D.L. Nutrients, physical activity, and mitochondrial dysfunction in the setting of metabolic syndrome. Nutrients 2023, 15, 1217. [Google Scholar] [CrossRef]
  140. Ouyang, Y.B.; Xu, L.J.; Emery, J.F.; Lee, A.S.; Giffard, R.G. Overexpressing GRP78 influences Ca2+ handling and function of mitochondria in astrocytes after ischemia-like stress. Mitochondrion 2011, 11, 279–286. [Google Scholar] [CrossRef]
  141. Chen, X.; Shi, C.; He, M.; Xiong, S.; Xia, X. Endoplasmic reticulum stress: Molecular mechanism and therapeutic targets. Signal Transduct. Target. Ther. 2023, 8, 352. [Google Scholar] [CrossRef]
  142. Alotaibi, G.; Alkhammash, A. Pharmacological landscape of endoplasmic reticulum stress: Uncovering therapeutic avenues for metabolic diseases. Eur. J. Pharmacol. 2025, 998, 177509. [Google Scholar] [CrossRef] [PubMed]
  143. Lenghel, A.; Gheorghita, A.M.; Vacaru, A.M.; Vacaru, A.-M. What is the sweetest UPR flavor for the β-cell? That is the question. Front. Endocrinol. 2021, 11, 614123. [Google Scholar] [CrossRef]
  144. Lee, J.-H.; Lee, J. Endoplasmic reticulum (ER) stress and its role in pancreatic β-cell dysfunction and senescence in type 2 diabetes. Int. J. Mol. Sci. 2022, 23, 4843. [Google Scholar] [CrossRef] [PubMed]
  145. Zhao, T.; Du, J.; Zeng, H. Interplay between endoplasmic reticulum stress and non-coding RNAs in cancer. J. Hematol. Oncol. 2020, 13, 163. [Google Scholar] [CrossRef] [PubMed]
  146. Amodio, G.; Pagliara, V.; Moltedo, O.; Remondelli, P. Structural and functional significance of the endoplasmic reticulum unfolded protein response transducers and chaperones at the mitochondria–ER contacts: A cancer perspective. Front. Cell Dev. Biol. 2021, 9, 641194. [Google Scholar] [CrossRef]
  147. Park, S.; Lim, W.; Bazer, F.W.; Song, G. Naringenin induces mitochondria-mediated apoptosis and endoplasmic reticulum stress by regulating MAPK and AKT signal transduction pathways in endometriosis cells. MHR: Basic. Sci. Reprod. Med. 2017, 23, 842–854. [Google Scholar]
  148. Li, X.; Lin, Q.; Gou, F.; Zhu, J.; Yu, M.; Hong, Q.; Hu, C. Effects of hesperidin on mitochondrial function, mitochondria-associated endoplasmic reticulum membranes and IP3R–MCU calcium axis in the intestine of piglets exposed to deoxynivalenol. Food Funct. 2024, 15, 6459–6474. [Google Scholar] [CrossRef] [PubMed]
  149. Gou, F.; Lin, Q.; Tu, X.; Zhu, J.; Li, X.; Chen, S.; Hu, C. Hesperidin alleviated intestinal barrier injury, mitochondrial dysfunction, and disorder of endoplasmic reticulum mitochondria contact sites under oxidative stress. J. Agric. Food Chem. 2024, 72, 16276–16286. [Google Scholar] [CrossRef]
  150. Khoi, C.-S.; Lin, T.-Y.; Chiang, C.-K. Targeting Insulin Resistance, Reactive Oxygen Species, Inflammation, Programmed Cell Death, ER Stress, and Mitochondrial Dysfunction for the Therapeutic Prevention of Free Fatty Acid-Induced Vascular Endothelial Lipotoxicity. Antioxidants 2024, 13, 1486. [Google Scholar] [CrossRef]
  151. Karunakaran, U.; Park, K.G. A systematic review of oxidative stress and safety of antioxidants in diabetes: Focus on islets and their defense. Diabetes Metab. J. 2013, 37, 106–112. [Google Scholar] [CrossRef]
  152. Sergi, D.; Naumovski, N.; Heilbronn, L.K.; Abeywardena, M.; O’Callaghan, N.; Lionetti, L.; Luscombe-Marsh, N. Mitochondrial (dys) function and insulin resistance: From pathophysiological molecular mechanisms to the impact of diet. Front. Physiol. 2019, 10, 449821. [Google Scholar] [CrossRef]
  153. González, P.; Lozano, P.; Ros, G.; Solano, F. Hyperglycemia and oxidative stress: An integral, updated and critical overview of their metabolic interconnections. Int. J. Mol. Sci. 2023, 24, 9352. [Google Scholar] [CrossRef] [PubMed]
  154. Ali, A.M.; Gabbar, M.A.; Abdel-Twab, S.M.; Fahmy, E.M.; Ebaid, H.; Alhazza, I.M.; Ahmed, O.M. Antidiabetic Potency, Antioxidant Effects, and Mode of Actions of Citrus reticulata Fruit Peel Hydroethanolic Extract, Hesperidin, and Quercetin in Nicotinamide/Streptozotocin-Induced Wistar Diabetic Rats. Oxid. Med. Cell Longev. 2020, 2020, 1730492. [Google Scholar] [PubMed]
  155. Korac, B.; Kalezic, A.; Pekovic-Vaughan, V.; Korac, A.; Jankovic, A. Redox changes in obesity, metabolic syndrome, and diabetes. Redox Biol. 2021, 42, 101887. [Google Scholar] [CrossRef]
  156. Park, H.J.; Jung, U.J.; Cho, S.J.; Jung, H.K.; Shim, S.; Choi, M.S. Citrus unshiu peel extract ameliorates hyperglycemia and hepatic steatosis by altering inflammation and hepatic glucose- and lipid-regulating enzymes in db/db mice. J. Nutr. Biochem. 2013, 24, 419–427. [Google Scholar]
  157. Kim, B.M.; Cho, B.O.; Jang, S.I. Anti-obesity effects of Diospyros lotus leaf extract in mice with high-fat diet-induced obesity. Int. J. Mol. Med. 2019, 43, 603–613. [Google Scholar]
  158. L Suraweera, T.; Rupasinghe, H.P.V.; Dellaire, G.; Xu, Z. Regulation of Nrf2/ARE Pathway by Dietary Flavonoids: A Friend or Foe for Cancer Management? Antioxidants 2020, 9, 973. [Google Scholar] [CrossRef]
  159. Tsai, H.L.; Chang, S.K.; Chang, S.J. Antioxidant content and free radical scavenging ability of fresh red pummelo [Citrus grandis (L.) Osbeck] juice and freeze-dried products. J. Agric. Food Chem. 2007, 55, 2867–2872. [Google Scholar] [CrossRef]
  160. Namkhah, Z.; Naeini, F.; Mahdi Rezayat, S.; Mehdi, Y.; Mansouri, S.; Javad Hosseinzadeh-Attar, M. Does naringenin supplementation improve lipid profile, severity of hepatic steatosis and probability of liver fibrosis in overweight/obese patients with NAFLD? A randomised, double-blind, placebo-controlled, clinical trial. Int. J. Clin. Pract. 2021, 75, e14852. [Google Scholar]
  161. Naeini, F.; Namkhah, Z.; Tutunchi, H.; Rezayat, S.M.; Mansouri, S.; Jazayeri-Tehrani, S.A.; Yaseri, M.; Hosseinzadeh-Attar, M.J. Effects of naringenin supplementation in overweight/obese patients with non-alcoholic fatty liver disease: Study protocol for a randomized double-blind clinical trial. Trials 2021, 22, 801. [Google Scholar] [CrossRef]
  162. Goldwasser, J.; Cohen, P.Y.; Yang, E.; Balaguer, P.; Yarmush, M.L.; Nahmias, Y. Transcriptional regulation of human and rat hepatic lipid metabolism by the grapefruit flavonoid naringenin: Role of PPARalpha, PPARgamma and LXRalpha. PLoS ONE 2010, 5, e12399. [Google Scholar]
  163. Sleiman, D.; Al-Badri, M.R.; Azar, S.T. Effect of mediterranean diet in diabetes control and cardiovascular risk modification: A systematic review. Front. Public. Health 2015, 3, 69. [Google Scholar]
  164. Carpenito, M.; Coletti, F.; Muscoli, S.; Guarino, L.; Di Cristo, A.; Cammalleri, V.; Mega, S.; Emerenziani, S.; Cicala, M.; Fanali, C.; et al. Unveiling the Power of Bergamot: Beyond Lipid-Lowering Effects. Nutrients 2025, 17, 1871. [Google Scholar] [CrossRef]
  165. Fogacci, F.; Giovannini, M.; Imbalzano, E.; Grandi, E.; Rizzoli, E.; D’Addato, S.; Cicero, A.F.G. Metabolic and vascular effect of a new standardized bergamot phytocomplex: A three-arm, placebocontrolled, double-blind clinical trial. Arch. Med. Sci. 2023, 19, 1228–1235. [Google Scholar] [CrossRef] [PubMed]
  166. Ferro, Y.; Maurotti, S.; Mazza, E.; Pujia, R.; Sciacqua, A.; Musolino, V.; Mollace, V.; Pujia, A.; Montalcini, T. Citrus Bergamia and Cynara Cardunculus Reduce Serum Uric Acid in Individuals with Non-Alcoholic Fatty Liver Disease. Medicina 2022, 58, 1728. [Google Scholar] [CrossRef]
  167. Jalili, F.; Moradi, S.; Talebi, S.; Mehrabani, S.; Ghoreishy, S.M.; Wong, A.; Jalalvand, A.R.; Kermani, M.A.H.; Jalili, C. The effects of citrus flavonoids supplementation on endothelial function: A systematic review and dose-response meta-analysis of randomized clinical trials. Phytother. Res. 2024, 38, 2847–2859. [Google Scholar] [CrossRef]
  168. Sost, M.M.; Ahles, S.; Verhoeven, J.; Verbruggen, S.; Stevens, Y.; Venema, K. A Citrus Fruit Extract High in Polyphenols Beneficially Modulates the Gut Microbiota of Healthy Human Volunteers in a Validated In Vitro Model of the Colon. Nutrients 2021, 13, 3915. [Google Scholar] [CrossRef] [PubMed]
  169. Sánchez Macarro, M.; Rodríguez, J.P.M.; Morell, E.B.; Pérez-Piñero, S.; Victoria-Montesinos, D.; García-Muñoz, A.M.; García, F.C.; Sánchez, J.C.; López-Román, F.J. Effect of a Combination of Citrus Flavones and Flavanones and Olive Polyphenols for the Reduction of Cardiovascular Disease Risk: An Exploratory Randomized, Double-Blind, Placebo-Controlled Study in Healthy Subjects. Nutrients 2020, 12, 1475. [Google Scholar] [CrossRef]
  170. Victoria-Montesinos, D.; Sánchez-Macarro, M.; Gabaldón-Hernández, J.A.; Abellán-Ruiz, M.S.; Querol-Calderón, M.; Luque-Rubia, A.J.; Bernal-Morell, E.; Ávila-Gandía, V.; López-Román, F.J. Effect of Dietary Supplementation with a Natural Extract of Sclerocarya birrea on Glycemic Metabolism in Subjects with Prediabetes: A Randomized Double-Blind Placebo-Controlled Study. Nutrients 2021, 13, 1948. [Google Scholar] [CrossRef]
  171. Morand, C.; DuBray, C.; Milenkovic, D.; Lioger, D.; Martin, J.F.; Scalbert, A.; Mazur, A. Hesperidin contributes to the vascular protective effects of orange juice: A randomized crossover study in healthy volunteers. Am. J. Clin. Nutr. 2011, 93, 73–80. [Google Scholar] [CrossRef]
  172. Yari, Z.; Movahedian, M.; Imani, H.; Alavian, S.M.; Hedayati, M.; Hekmatdoost, A. The effect of hesperidin supplementation on metabolic profiles in patients with metabolic syndrome: A randomized, double-blind, placebo-controlled clinical trial. Eur. J. Nutr. 2020, 59, 2569–2577. [Google Scholar] [CrossRef]
  173. Khorasanian, A.S.; Fateh, S.T.; Gholami, F.; Rasaei, N.; Gerami, H.; Khayyatzadeh, S.S.; Shiraseb, F.; Asbaghi, O. The effects of hesperidin supplementation on cardiovascular risk factors in adults: A systematic review and dose-response meta-analysis. Front. Nutr. 2023, 10, 1177708. [Google Scholar] [CrossRef]
  174. Notarnicola, M.; Tutino, V.; De Nunzio, V.; Cisternino, A.M.; Cofano, M.; Donghia, R.; Giannuzzi, V.; Zappimbulso, M.; Milella, R.A.; Giannelli, G.; et al. Daily Orange Consumption Reduces Hepatic Steatosis Prevalence in Patients with Metabolic Dysfunction-Associated Steatotic Liver Disease: Exploratory Outcomes of a Randomized Clinical Trial. Nutrients 2024, 16, 3191. [Google Scholar] [CrossRef]
  175. Navajas-Porras, B.; Bosch-Sierra, N.; Valle, C.G.-D.; Salazar, J.D.; Marqués-Cardete, R.; Sáez, G.; Morillas, C.; Bañuls, C. Effects of a flavonoid-enriched orange juice on antioxidant capacity, lipid profile, and inflammation in obese patients: A randomized placebo-controlled trial. Food Res. Int. 2025, 217, 116759. [Google Scholar]
  176. Cesar, T.B.; Ramos, F.M.M.; Ribeiro, C.B. Nutraceutical Eriocitrin (Eriomin) Reduces Hyperglycemia by Increasing Glucagon-Like Peptide 1 and Downregulates Systemic Inflammation: A Crossover-Randomized Clinical Trial. J. Med. Food 2022, 25, 1050–1058. [Google Scholar] [CrossRef] [PubMed]
  177. Wang, X.C.; Song, L.; Wang, X.H. Efficacy of dietary polyphenol supplement in patients with non-alcoholic fatty liver disease: A network meta-analysis. Front. Nutr. 2025, 12, 1582861. [Google Scholar] [CrossRef] [PubMed]
  178. Hirano, T. Pathophysiology of Diabetic Dyslipidemia. J. Atheroscler. Thromb. 2018, 25, 771–782. [Google Scholar] [CrossRef] [PubMed]
  179. Chen, J.; Fang, Z.; Luo, Q.; Wang, X.; Warda, M.; Das, A.; Oldoni, F.; Luo, F. Unlocking the Mysteries of VLDL: Exploring Its Production, Intracellular Trafficking, and Metabolism as Therapeutic Targets. Lipids Health Dis. 2024, 23, 14. [Google Scholar] [CrossRef]
  180. Gluba-Sagr, A.; Olszewski, R.; Franczyk, B.; Młynarska, E.; Rysz-Górzyńska, M.; Rysz, J.; Surma, S.; Sohum, S.; Banach, M.; Toth, P.P. High-Density Lipoproteins. Part 2. Impact of Disease States on Functionality. Am. J. Prev. Cardiol. 2025, 23, 101073. [Google Scholar] [CrossRef]
  181. Nauman, M.C.; Johnson, J.J. Clinical Application of Bergamot (Citrus Bergamia) for Reducing High Cholesterol and Cardiovascular Disease Markers. Integr. Food Nutr. Metab. 2019, 6, 10.15761/IFNM.1000249. [Google Scholar] [CrossRef]
  182. Mulvihill, E.E.; Assini, J.M.; Lee, J.K.; Allister, E.M.; Sutherland, B.G.; Koppes, J.B.; Sawyez, C.G.; Edwards, J.Y.; Telford, D.E.; Charbonneau, A.; et al. Nobiletin Attenuates VLDL Overproduction, Dyslipidemia, and Atherosclerosis in Mice With Diet-Induced Insulin Resistance. Diabetes 2011, 60, 1446–1457. [Google Scholar] [CrossRef]
  183. Zhang, S.; Xu, M.; Zhang, W.; Liu, C.; Chen, S. Natural Polyphenols in Metabolic Syndrome: Protective Mechanisms and Clinical Applications. Int. J. Mol. Sci. 2021, 22, 6110. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of the mechanisms linking obesity to β-cell failure and T2DM. Chronic caloric excess induces adipocyte hypertrophy and local hypoxia, activating HIF-1α and promoting the release of FFAs, TNF-α, IL-6, and MCP-1, while reducing adiponectin. This triggers M1 macrophage polarization and inflammation in adipose tissue. Circulating FFAs and lipid intermediates reach the liver, activating PKC/JNK pathways and impairing IRS1–PI3K–Akt signaling, resulting in increased gluconeogenesis. Concurrently, impaired β-oxidation and mitochondrial dysfunction lead to ROS accumulation, ER stress, and apoptosis. In the pancreas, inflammatory and ER stress signals impair insulin synthesis and activate mitochondrial apoptotic pathways, reducing β-cell mass and insulin secretion—contributing to hyperglycemia and T2DM. (Image created with biorender.com).
Figure 1. Schematic representation of the mechanisms linking obesity to β-cell failure and T2DM. Chronic caloric excess induces adipocyte hypertrophy and local hypoxia, activating HIF-1α and promoting the release of FFAs, TNF-α, IL-6, and MCP-1, while reducing adiponectin. This triggers M1 macrophage polarization and inflammation in adipose tissue. Circulating FFAs and lipid intermediates reach the liver, activating PKC/JNK pathways and impairing IRS1–PI3K–Akt signaling, resulting in increased gluconeogenesis. Concurrently, impaired β-oxidation and mitochondrial dysfunction lead to ROS accumulation, ER stress, and apoptosis. In the pancreas, inflammatory and ER stress signals impair insulin synthesis and activate mitochondrial apoptotic pathways, reducing β-cell mass and insulin secretion—contributing to hyperglycemia and T2DM. (Image created with biorender.com).
Medsci 13 00180 g001
Figure 2. Multi-organ effects of citrus fruit polyphenols in obesity-induced diabetes. In the liver, polyphenols activate AMPK and PI3K/AKT signaling, suppress SREBP1c and HMGCR, reduce lipogenesis, and inhibit gluconeogenic enzymes (G6Pase, FOXO1), thereby lowering hepatic glucose output. In adipocytes, they enhance PI3K–AKT–GLUT4 translocation to improve glucose uptake and downregulate PPARγ, FAS, and SREBP1c to reduce adipogenesis and lipid accumulation. In pancreatic β-cells, polyphenols upregulate antioxidant enzymes (SOD, GSH-Px), reduce ROS, inhibit caspase-3, and promote Ca2+ influx, enhancing insulin secretion and survival. In the intestine, they inhibit α-amylase and α-glucosidase, slowing carbohydrate digestion and reducing postprandial glycemia via SGLT1/GLUT2 downregulation. (Image created with biorender.com).
Figure 2. Multi-organ effects of citrus fruit polyphenols in obesity-induced diabetes. In the liver, polyphenols activate AMPK and PI3K/AKT signaling, suppress SREBP1c and HMGCR, reduce lipogenesis, and inhibit gluconeogenic enzymes (G6Pase, FOXO1), thereby lowering hepatic glucose output. In adipocytes, they enhance PI3K–AKT–GLUT4 translocation to improve glucose uptake and downregulate PPARγ, FAS, and SREBP1c to reduce adipogenesis and lipid accumulation. In pancreatic β-cells, polyphenols upregulate antioxidant enzymes (SOD, GSH-Px), reduce ROS, inhibit caspase-3, and promote Ca2+ influx, enhancing insulin secretion and survival. In the intestine, they inhibit α-amylase and α-glucosidase, slowing carbohydrate digestion and reducing postprandial glycemia via SGLT1/GLUT2 downregulation. (Image created with biorender.com).
Medsci 13 00180 g002
Figure 3. Citrus polyphenols regulate mitochondrial and endoplasmic reticulum (er) homeostasis under metabolic stress. This diagram illustrates the coordinated protective effects of citrus fruit polyphenols on mitochondrial and ER function. In mitochondria, polyphenols activate the AMPK–SIRT1–PGC-1α pathway, enhancing mitochondrial biogenesis via NRF1/2 and TFAM, and reducing ROS through upregulation of MnSOD and GPx. They stabilize the mitochondrial membrane potential, support ATP production, and improve complex I/IV activity while reducing proton leak and oxidative stress. In the ER, citrus polyphenols alleviate ER stress by modulating UPR path ways (PERK, IRE1α, ATF6), reducing expression of stress markers (CHOP, p-PERK), enhancing SERCA-mediated Ca2+ influx, and promoting ER–mitochondria Ca2+ transfer. They also inhibit apoptosis by increasing Bcl-2 and suppressing Bax and caspase activation. Together, these actions maintain cellular homeostasis and prevent metabolic dysfunction. (Image created with biorender.com).
Figure 3. Citrus polyphenols regulate mitochondrial and endoplasmic reticulum (er) homeostasis under metabolic stress. This diagram illustrates the coordinated protective effects of citrus fruit polyphenols on mitochondrial and ER function. In mitochondria, polyphenols activate the AMPK–SIRT1–PGC-1α pathway, enhancing mitochondrial biogenesis via NRF1/2 and TFAM, and reducing ROS through upregulation of MnSOD and GPx. They stabilize the mitochondrial membrane potential, support ATP production, and improve complex I/IV activity while reducing proton leak and oxidative stress. In the ER, citrus polyphenols alleviate ER stress by modulating UPR path ways (PERK, IRE1α, ATF6), reducing expression of stress markers (CHOP, p-PERK), enhancing SERCA-mediated Ca2+ influx, and promoting ER–mitochondria Ca2+ transfer. They also inhibit apoptosis by increasing Bcl-2 and suppressing Bax and caspase activation. Together, these actions maintain cellular homeostasis and prevent metabolic dysfunction. (Image created with biorender.com).
Medsci 13 00180 g003
Figure 4. Citrus fruit polyphenols mitigate oxidative stress in the liver. This schematic depicts how citrus polyphenols exert antioxidant and anti-inflammatory effects to protect hepatic tissue from oxidative damage. On the left, citrus polyphenols directly scavenge reactive oxygen species (ROS), DPPH+ (2,2-diphenyl-1-picrylhydrazyl), and NO radicals via hydrogen atom donation, preventing lipid peroxidation and radical propagation. Simultaneously, they activate the Keap1–Nrf2 pathway, promoting Nrf2 release and nuclear translocation. Nrf2 then binds to antioxidant response elements (ARE), upregulating endogenous antioxidants such as SOD, CAT, GPx, and HO-1. This enhances hepatic ROS detoxification and reduces oxidative stress markers like malondialdehyde (MDA) and 8-hydroxy-2′-deoxyguanosine (8-OHdG). On the right, citrus polyphenols inhibit xanthine oxidase as well as COX-1/COX-2 and LOX enzymes, decreasing prostaglandin and leukotriene synthesis and limiting inflammatory cell infiltration. Together, these mechanisms mitigate oxidative DNA damage and inflammation, preventing chronic liver injury. (Image created with biorender.com).
Figure 4. Citrus fruit polyphenols mitigate oxidative stress in the liver. This schematic depicts how citrus polyphenols exert antioxidant and anti-inflammatory effects to protect hepatic tissue from oxidative damage. On the left, citrus polyphenols directly scavenge reactive oxygen species (ROS), DPPH+ (2,2-diphenyl-1-picrylhydrazyl), and NO radicals via hydrogen atom donation, preventing lipid peroxidation and radical propagation. Simultaneously, they activate the Keap1–Nrf2 pathway, promoting Nrf2 release and nuclear translocation. Nrf2 then binds to antioxidant response elements (ARE), upregulating endogenous antioxidants such as SOD, CAT, GPx, and HO-1. This enhances hepatic ROS detoxification and reduces oxidative stress markers like malondialdehyde (MDA) and 8-hydroxy-2′-deoxyguanosine (8-OHdG). On the right, citrus polyphenols inhibit xanthine oxidase as well as COX-1/COX-2 and LOX enzymes, decreasing prostaglandin and leukotriene synthesis and limiting inflammatory cell infiltration. Together, these mechanisms mitigate oxidative DNA damage and inflammation, preventing chronic liver injury. (Image created with biorender.com).
Medsci 13 00180 g004
Table 1. Citrus polyphenols and metabolic reprogramming in animal and cell studies. The table lists the compound studied, experimental model, intervention characteristics, and major outcomes on insulin signaling, oxidative stress, lipid metabolism, or mitochondrial/ER function.
Table 1. Citrus polyphenols and metabolic reprogramming in animal and cell studies. The table lists the compound studied, experimental model, intervention characteristics, and major outcomes on insulin signaling, oxidative stress, lipid metabolism, or mitochondrial/ER function.
CompoundModel/SystemIntervention Details (Dose and Duration)Primary Outcomes/MechanismsSource
NeohesperidinHFD-fed mice50–100 mg/kg/day, 8–12 weeks↑ AMPK–PGC-1α → ↑ mitochondrial biogenesis, ↓ steatosis[46]
NobiletinHFD-fed mice100 mg/kg/day, 8 weeks↑ FA oxidation, ↑ energy expenditure; AMPK-independent[47]
NobiletinHepatocytes10–50 μM, 24–48 hRestores Bmal1 → ↑ lipid/OXPHOS metabolism[48]
NobiletinInsulin-resistant mice50 mg/kg/day, 6 weeks↓ VLDL secretion; improved lipid/glucose metabolism[47]
NobiletinHepG2 cells25 μM, 24 h↑ PGC1α, CPT1, UCP2 → ↑ β-oxidation[47]
Nobiletinob/ob mice100 mg/kg/day, 6 weeks↑ GLUT4, ↑ Akt phosphorylation → improved insulin sensitivity[49]
NaringeninMCD or HFD mice50–100 mg/kg/day, 8–12 weeks↑ AMPK → ↑ autophagy, ↑ mitochondrial biogenesis[50]
NaringeninHepatocytes/mice10–50 μM in vitro; 100 mg/kg/day in vivo↑ AMPK, ↑ ATF3 → ↓ metabolic inflammation[51]
NaringinHFD-fed mice100 mg/kg/day, 10 weeks↑ AMPK → ↓ SREBP-1c/FAS, ↑ redox balance[52]
NaringinFructose-fed rats40 mg/kg/day, 8 weeks↑ Nrf2/HO-1 → antioxidant response; ↓ ChREBP/SREBP-1c[53]
NaringinHFD mice100 mg/kg/day, 12 weeks↑ TFEB → lipophagy → ↓ hepatic lipid droplets[54]
HesperidinLO2 hepatocytes (HG)25–100 μM, 24–48 h↑ ATP, restores ΔΨm via AKT/GSK3β[55]
HesperidinHyperlipidemic rats100 mg/kg/day, 6 weeks↑ SOD, ↑ catalase; preserved mitochondrial enzymes[56]
HesperidinNeurons (hyperglycemia)25 μM, 24–48 hImproves ATP/redox; ↓ mitochondrial dysfunction[57]
HesperetinAging mice50 mg/kg/day, 8 weeks↑ Cisd2 expression → maintenance of metabolic health[58]
LimoneneMice model100 mg/kg/day, 6 weeks↑ mitochondrial respiration, ↓ ROS[59,60]
EriocitrinHFD rats25–50 mg/kg/day, 8 weeks↑ mitochondrial biogenesis, ↓ steatosis[61]
SudachitinC57BL/6J, db/db mice50 mg/kg/day, 8 weeks↑ β-oxidation, ↑ mitochondrial biogenesis[62]
TangeretinDiabetic rats100 mg/kg/day, 6 weeks↑ GLUT4, ↑ antioxidant enzymes[63]
NaringeninNAFLD mice100 mg/kg/day, 10 weeks↓ NLRP3/NF-κB, ↓ IL-1β → metabolic reprogramming[64]
NaringeninNAFLD mice (metabolomics)100 mg/kg/day, 12 weeksModulates gut microbiota → improved host metabolism[65]
NaringeninMuscle cells25–50 μM, 24 h↑ p-AMPK → ↑ glucose uptake, ↑ mitochondrial content[66]
NaringinHepatocytes, HFD mice25 μM in vitro; 100 mg/kg/day, 8 weeksAMPK–IRS1–MAPK pathway → improved insulin signaling[67]
NaringeninMASLD mice100 mg/kg/day, 12 weeks↑ PPAR, ↑ lipid oxidation, gut microbiota shift[68]
NaringeninMice (aerobic fitness)100 mg/kg/day, 4 weeks↑ oxidative fibers, ↑ aerobic metabolism[69]
NaringinKK-A(y) mice100 mg/kg/day, 8 weeks↑ AMPK → ↓ glucose/lipids, ↑ insulin sensitivity[70]
NeohesperidinDIO mice, HepG2 cells50–100 mg/kg/day, 12 weeks; 25 μM in vitro↑ FGF21, ↑ AMPK → improved lipid regulation[71]
HesperidinMASLD mice100 mg/kg/day, 8 weeks↓ insulin resistance, ↓ oxidative stress[72]
NobiletinHepG2 cells25 μM, 24 h↑ AMPK, ↓ lipogenesis[73]
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

Shimu, S.J.; Mahir, J.U.K.; Shakib, F.A.F.; Ridoy, A.A.; Samir, R.A.; Jahan, N.; Hasan, M.F.; Sazzad, S.; Akter, S.; Mohiuddin, M.S.; et al. Metabolic Reprogramming Through Polyphenol Networks: A Systems Approach to Metabolic Inflammation and Insulin Resistance. Med. Sci. 2025, 13, 180. https://doi.org/10.3390/medsci13030180

AMA Style

Shimu SJ, Mahir JUK, Shakib FAF, Ridoy AA, Samir RA, Jahan N, Hasan MF, Sazzad S, Akter S, Mohiuddin MS, et al. Metabolic Reprogramming Through Polyphenol Networks: A Systems Approach to Metabolic Inflammation and Insulin Resistance. Medical Sciences. 2025; 13(3):180. https://doi.org/10.3390/medsci13030180

Chicago/Turabian Style

Shimu, Shakila Jahan, Jawad Ul Karim Mahir, Fardin Al Fahad Shakib, Arafath Amin Ridoy, Ratin Al Samir, Nadia Jahan, Md Fahim Hasan, Sadman Sazzad, Shamima Akter, Mohammad Sarif Mohiuddin, and et al. 2025. "Metabolic Reprogramming Through Polyphenol Networks: A Systems Approach to Metabolic Inflammation and Insulin Resistance" Medical Sciences 13, no. 3: 180. https://doi.org/10.3390/medsci13030180

APA Style

Shimu, S. J., Mahir, J. U. K., Shakib, F. A. F., Ridoy, A. A., Samir, R. A., Jahan, N., Hasan, M. F., Sazzad, S., Akter, S., Mohiuddin, M. S., Shawon, M. J. A., Shariare, M. H., Mohib, M. M., & Uddin, M. B. (2025). Metabolic Reprogramming Through Polyphenol Networks: A Systems Approach to Metabolic Inflammation and Insulin Resistance. Medical Sciences, 13(3), 180. https://doi.org/10.3390/medsci13030180

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