Next Article in Journal
Predictors of Fasting Endogenous Erythritol and Erythronate Concentrations in Humans: Cross-Sectional and Post-Bariatric Surgery Analyses
Previous Article in Journal
Impact of Maternal Overweight and Obesity on Adipokines During Pregnancy and Lactation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Lipid Peroxidation in Diabetic Kidney Disease: Mechanism and Natural Solution

College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130000, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(19), 9764; https://doi.org/10.3390/ijms26199764
Submission received: 13 September 2025 / Revised: 5 October 2025 / Accepted: 6 October 2025 / Published: 7 October 2025
(This article belongs to the Section Molecular Biology)

Abstract

Diabetic kidney disease (DKD), as one of the most serious microvascular complications of diabetes, is the main cause of end-stage renal disease in the world. Lipid peroxidation plays a crucial role in the development and progression of DKD. Under conditions of high glucose and insulin resistance, renal lipid metabolism disorders result in abnormal accumulation of polyunsaturated fatty acids (PUFAs), which undergo lipid peroxidation via free radical chain reactions to generate reactive aldehydes. These substances not only directly damage the cell structure but can also be used as signaling molecules that activate pathways related to inflammation, fibrosis, and ferroptosis, eventually leading to glomerular sclerosis and tubulointerstitial fibrosis. Natural products have shown considerable application prospects in the treatment of DKD due to their multi-functional properties, including anti-inflammatory, antioxidant, and lipid-metabolism-regulating effects. To elucidate this, we conducted a systematic review of the literature available in electronic databases (including PubMed, Web of Science, and Scopus, and Google Scholar) from January 2000 to May 2025. This study further discusses the therapeutic effect and mechanism of natural products targeting lipid peroxidation in DKD. The results indicate that natural products are promising anti-lipid peroxidation drugs. Further clinical trials will be necessary to verify the safety and effectiveness of these natural compounds in clinical applications, thereby laying the foundation for developing novel treatment strategies for DKD.

1. Introduction

Diabetic kidney disease (DKD) is one of the most serious and common microvascular complications of diabetes. According to epidemiological statistics, approximately 537 million people globally suffered from diabetes in 2021, and more than 40% of them subsequently developed DKD, which has become the main cause of end-stage renal disease (ESRD) [1]. While current treatment methods, including strict glycemic and blood pressure control, sodium-dependent glucose transporters 2 inhibitors (SGLT2is), and renin–angiotensin–aldosterone system inhibitors (RAASis), can delay the progression of DKD to some extent, many patients still progress irreversibly to renal failure and require renal replacement therapy to sustain their lives [2,3]. Therefore, it remains a critical challenge to explore the underlying molecular mechanisms of DKD and identify effective therapeutic targets.
Studies have shown that lipid peroxidation is a key pathogenic process in DKD [4,5]. Under physiological conditions, intracellular free fatty acids, which serve as a key energy source, are efficiently metabolized in mitochondria mainly through the β-oxidation pathway. However, the hyperglycemic environment and insulin resistance in diabetes disrupt the balance between fatty acid synthesis and oxidation, leading to lipid metabolism disorders [6]. Lipid metabolism disorders contribute to abnormal deposition of polyunsaturated fatty acids (PUFAs) in renal cells, further exacerbating oxidative stress. The consequent massive release of reactive oxygen species (ROS) reacts with accumulated PUFAs in renal cells, initiating a free radical chain reaction of lipid peroxidation that ultimately generates multiple highly reactive aldehyde compounds [7]. These reactive aldehydes not only directly disrupt the cell membrane structure and compromise functional integrity but also act as signaling molecules that trigger inflammatory responses, promote fibrotic processes, and induce cell death such as ferroptosis, ultimately aggravating the pathological progression of glomerulosclerosis and tubulointerstitial fibrosis.
In this complex pathological network, natural products exhibit unique therapeutic potential owing to their multi-targeted and multi-pathway characteristics. Studies have shown that various natural active ingredients (such as flavonoids, terpenoids and polyphenols) can effectively interrupt the vicious circle of lipid peroxidation by modulating the balance of fatty acid synthesis and oxidation, enhancing the antioxidant defense system and inhibiting inflammatory signal transduction, thereby demonstrating broad application prospects in DKD treatment [8]. In this paper, we summarize the molecular mechanism of lipid peroxidation in DKD, with a focus on its driving factors, signaling pathways and downstream effects, and discuss the current status of natural components targeting lipid peroxidation in the treatment of DKD, aiming to provide new insights for DKD treatment strategies.

2. Mechanisms of Lipid Peroxidation in DKD

2.1. Lipid Metabolism Disorder

In the occurrence and development of DKD, lipid metabolism disorders play a critical role, with insulin resistance being recognized as the initiating factor [9]. Insulin exerts an anti-lipolysis function. However, in diabetic patients, impaired insulin sensitivity leads to enhanced lipolysis, resulting in the release of excessive free fatty acids (FFAs) into the circulatory system [10,11]. These surplus FFAs circulate to the kidney and are actively taken up into renal cells via upregulated scavenger receptors, such as cluster of differentiation 36 (CD36) [12]. Moreover, under the influence of persistent hyperglycemia and insulin resistance in DKD, fatty acid oxidation (FAO) is suppressed, while fatty acid synthesis is upregulated. Consequently, excess FFAs accumulate in the kidney in the form of triglycerides, which aggravates lipid metabolism disorders in DKD [7,13] (Figure 1).
Fatty acid oxidation (FAO) is a continuous biochemical process in which FFAs produce acetyl coenzyme A (Acetyl-CoA) and release ATP through a series of enzymatic reactions. Among them, fatty acid-β-oxidation represents the predominant form. FFAs are activated in the cytoplasm to form Acyl Coenzyme A (Acyl-CoA), which is then transported into mitochondria via carnitine palmitoyl transferase 1 (CPT1). Inside mitochondria, Acyl-CoA undergoes repeated β-oxidation to produce acetyl-CoA, which enters the tricarboxylic acid (TCA) cycle for complete oxidation, thereby generating energy through the mitochondrial electron transport chain (ETC) [14]. Mitochondria play an essential role throughout this process, particularly in highly energy-consuming renal tubular epithelial cells. FAO serves as the primary energy source sustaining the normal ion gradient, reabsorption function, and cellular viability of these cells [15]. Peroxisome proliferator-activated receptor alpha (PPARa) acts as the master regulator of FAO, governing the gene expression of a series of key enzymes in this pathway, while CPT1 functions as the principal carrier mediating fatty acid uptake into mitochondria [16]. However, renal biopsies from DKD patients have revealed downregulation of PPARa and its downstream target CPT1, accompanied by significant lipid deposition in renal tissues with an increase in intracellular lipid droplets [17]. Research on DKD animal models has confirmed that overexpression of CPT1 can down-regulate fibronectin expression, reduce inflammatory cell infiltration and mitochondrial damage, and improve renal function [18].
Fatty acid synthesis is a key metabolic pathway driving lipid deposition and is closely associated with hyperglycemia and insulin resistance in DKD. This process is primarily regulated by two key transcription factors: sterol regulatory element binding protein-1c (SREBP-1c) and carbohydrate response element binding protein (ChREBP) [19,20]. Under persistent high glucose conditions, both pathways are aberrantly activated and act synergistically to upregulate acetyl-CoA carboxylase (ACC) and fatty acid synthase (FAS), ultimately promoting fatty acid synthesis [21]. Studies have confirmed that the expression of lipogenic markers—SREBP-1c, ACC and FAS—is significantly up-regulated in DKD rats, and targeted therapy can mitigate the effect of hyperglycemia on renal lipogenesis [22,23].
Therefore, persistent hyperglycemia and insulin resistance in DKD can induce FFAs overload in the kidney, leading to subsequent esterification and triglyceride (TG) synthesis, and ultimately contributing to renal ectopic lipid deposition [24]. This ectopic lipid deposition impairs mitochondrial function and integrity, prompting excessive reactive oxygen species release, inducing oxidative stress and accelerating lipid peroxidation [25].

2.2. Oxidative Stress and Lipid Peroxidation

Under physiological conditions, fatty acid oxidation and fatty acid synthesis are maintained in a dynamic equilibrium. When energy supply is sufficient, fatty acids are synthesized within cells and subsequently esterified for storage. Conversely, when energy demand increases, fatty acids are oxidized to generate ATP [26]. However, in DKD, this lipid homeostasis is disrupted, leading to the accumulation of FFAs in the kidney. Hyperglycemia and renal lipid deposition further impair mitochondrial structure and function, promote oxidative stress, stimulate excessive ROS production, and trigger lipid peroxidation [25] (Figure 2).
Mitochondria serve as the primary site for cellular energy production. As an organ with high energy demand, the kidney is critically dependent on mitochondrial function [27]. However, studies have revealed that DKD is frequently associated with varying degrees of mitochondrial dysfunction and structural abnormalities, which may be closely linked to hyperglycemia and lipid metabolism disorders [28]. The persistent hyperglycemia in DKD upregulates glucose uptake via renal glucose transporter (GLUT), enhances glycolysis to generate pyruvate, and facilitates its entry into mitochondria through the mitochondrial pyruvate carrier (MPC) located on the inner mitochondrial membrane. Within the mitochondrial matrix, pyruvate is converted to acetyl coenzyme A (acetyl-CoA) by the pyruvate dehydrogenase complex [29]. Simultaneously, lipid metabolism disorders in DKD results in intracellular lipid accumulation, and a portion of FFAs enters mitochondria via CPT1 on the outer mitochondrial membrane to undergo β-oxidation, synergistically promoting the formation of acetyl-CoA. The acetyl-CoA derived from both glycolysis and fatty acid oxidation enters the TCA cycle, generating a substantial amount of energy carriers. This overload forces the ETC into a highly reduced, electron-saturated state, which favors the aberrant formation of ROS through the unintended transfer of electrons to molecular oxygen [30].
Advanced glycation end products (AGEs), resulting from hyperglycemia, also represent a major contributor to oxidative stress. AGEs are a class of heterogeneous molecules formed through the non-enzymatic glycation process, in which excess carbonyl groups of plasma glucose molecules react with amino groups of biomolecules such as lipids and proteins through nucleophilic addition [31]. In DKD, AGEs accumulate extensively in the kidney and bind to the receptor for AGE (RAGE). This binding triggers the recruitment and activation of NADPH oxidase (NOX), leading to the production of large amounts of ROS, which induces oxidative stress and exacerbates renal injury [32,33,34]. Furthermore, multiple studies have confirmed that inhibition of the AGE-RAGE signaling axis effectively suppresses NOX-mediated ROS production and alleviates oxidative renal damage, thereby further substantiating the pathogenic role of this pathway in DKD [35,36].
Beyond the classic oxidative stress pathways, alterations in cytochrome P450 (CYP) enzyme activity also serve as upstream drivers of oxidative stress in DKD. Under DKD conditions, the enhanced activity of renal CYP4A and CYP4F subfamilies shifts arachidonic acid (AA) metabolism toward the predominant production of 20-hydroxyeicosatetraenoic acid (20-HETE). As a key functional mediator, 20-HETE potently activates NOX, catalyzing ROS generation and aggravating lipid peroxidation damage [37,38].
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a central regulator of cellular redox homeostasis. Under basal conditions, Nrf2 combines with its endogenous inhibitor kelch-like ECH-associated protein 1 (KeaP1) to form a complex, while in the oxidative stress state, Nrf2 undergoes nuclear translocation and binds with antioxidant response element (ARE), thereby activating the transcription of a battery of antioxidant genes [39,40]. Nevertheless, study has indicated that the function of Nrf2 may be inhibited in DKD, leading to a compromised antioxidant defense system that exacerbates renal oxidative injury [41].
Lipid peroxidation proceeds via both enzymatic and non-enzymatic pathways, with the non-enzymatic, free radical-mediated mechanism playing a predominant role in DKD progression [42,43]. In renal cells of DKD, abundant free PUFAs are esterified by acyl-CoA synthetase long chain family member 4 (ACSL4) to form polyunsaturated fatty acid-phosphatidyl ethanolamine (PUFA-PE). The initiation of non-enzymatic lipid peroxidation depends on reactions between ROS and these PUFAs or PUFA-PE. ROS extract hydrogen atoms from the carbon chain of PUFAs or PUFA-PEs, generating unstable lipid free radicals (L·). This rapidly reacts with oxygen to form lipid peroxide free radicals (LOO·), which can abstract hydrogen atoms from adjacent lipids to yield lipid hydroperoxides (LOOH) and new L·, thereby forming a self-sustaining chain reaction [44,45]. Additionally, LOOH can be further degraded to reactive secondary products such as malondialdehyde (MDA) and 4-hydroxy-nonenal (4-HNE). These reactive aldehydes exhibit high reactivity and are capable of mediating diverse physiological and pathological processes [46,47,48].

2.3. Lipid Peroxide-Mediated Renal Injury

Lipid peroxides (LPOs) generated through lipid peroxidation serve as both direct effector substances and signaling molecules of cellular damage. The cross-linking of LPO with phosphatidylcholine compromises membrane fluidity and selective permeability, thereby directly impairing normal cellular physiological functions [46,49]. In addition, reactive aldehydes derived from LPO, such as MDA and 4-HNE, exhibit strong electrophilic properties, enabling them to react readily with sulfhydryl or amino groups in proteins. This interaction leads to the formation of covalent adducts and subsequent protein dysfunction [50]. Therefore, LPOs also function as bioactive signaling molecules that modulate multiple pathways implicated in inflammation, oxidative stress and cell death in kidney diseases [51,52] (Figure 3).
Nuclear factor kappa-B (NF-κB) is an inducible transcription factor that directly regulates the expression of inflammatory mediators and plays a key role in inflammatory and immune responses in kidney diseases [53,54]. The activation of this signaling pathway primarily relies on phosphorylation of the inhibitor of NF-κB (IκB) mediated by the inhibitor of kappa B kinase (IKK) complex. Notably, LPO produced by lipid peroxidation can covalently modify IKK, forming adducts that promote IκB phosphorylation and subsequent nuclear translocation of NF-κB, thereby initiating transcription of proinflammatory cytokines and chemokines, and amplifying inflammatory injury [55,56]. Simultaneously, protein adducts derived from lipid peroxidation can act as damage associated molecular patterns (DAMPS) that bine to pattern recognition receptors (PRR) and activate NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome [57]. Upon abnormal activation of the NLRP3 inflammasome, pro-caspase-1 is cleaved into caspase-1, which in turn promotes the release of the potent pro-inflammatory cytokines IL-1β and IL-18 [58,59,60]. Additionally, caspase-1 cleaves gasdermin D (GSDMD), generating N-terminal fragment of GSDMD (N-GSDMD) that forms membrane pores, and induces pyroptosis [61]. Consistent with these mechanisms, studies have confirmed that high glucose activates the NF-κB signaling pathway, upregulates the expression of IL-6, and contributes to glomerulosclerosis in DKD [62]. In high glucose-cultured glomerular mesangial cells, the phosphorylation level of IκBα is enhanced, further activating NF-κB signaling pathway, exacerbating inflammation, and promoting mesangial cell proliferation [63].
The abundant free radicals generated during the lipid peroxidation chain reaction can directly damage proteins, DNA and lipids, disrupt cellular redox homeostasis, and aggravate oxidative stress. Meanwhile, reactive aldehydes derived from unstable lipid peroxides covalently modify mitochondrial respiratory chain complexes, leading to ETC dysfunction and promoting mitochondrial ROS production [64]. Under physiological conditions, glutathione peroxidase (GPx) serves as a critical endogenous antioxidant enzyme that eliminates lipid hydroperoxides and terminates lipid peroxidation cascades [65]. However, studies have revealed that reactive aldehydes can also modify the glutathione (GSH) binding site, thereby inhibiting GPx activity, compromising the cellular antioxidant defense system, and contributing to the progression of oxidative stress [66,67].
Interestingly, the inhibition of GPx by reactive aldehydes may synergistically enhance ferroptosis in DKD. In the cellular iron cycle, Fe3+ bound to transferrin enters renal cells via transferrin receptor (TFR), is then reduced to Fe2+ by ferric reductases and stored within ferritin (FTN) to maintain cellular iron homeostasis [68]. However, in DKD, this homeostasis is disrupted. Excess labile iron promotes the Fenton reaction, generating highly reactive free radicals that initiate lipid peroxidation and trigger a unique form of regulated cell death known as ferroptosis, which is characterized by iron-dependent lipid peroxidation [69,70]. Studies in DKD animal models have demonstrated suppressed expression of GPX4 and GSH, accompanied by upregulation of TFR1, elevated levels of iron and MDA, collectively substantiating the involvement of ferroptosis in renal injury [71].

3. Natural Intervention Strategies for Lipid Peroxidation in Diabetic Kidney Disease

In the therapeutic strategy for DKD, natural products demonstrate significant potential. Studies have indicated that natural products can effectively counter lipid metabolism disorders in DKD, alleviate renal ectopic lipid deposition, mitigate oxidative stress status, and enhance antioxidant defenses, thereby suppressing lipid peroxidation. Furthermore, aberrant activation of lipid peroxidation exacerbates the progression of DKD by promoting inflammation, oxidative stress, ferroptosis, and other processes, while natural products represent a promising strategy to ameliorate this pathogenic cycle (Table 1).

3.1. Cutting Off the Substrate Supply: Restoring Lipid Metabolic Homeostasis

Resveratrol, a natural phytoalexin obtained from Reynoutria japonica Houtt (Polygonaceae), exhibits potent bioactivity. As a well-established Sirtuin 1 (SIRT1) agonist, it confers broad protection against lipid metabolism disorders [72,73]. Mechanistically, resveratrol activates SIRT1, suppressing lipogenic transcription factors SREBP-1 and ChREBP and significantly ameliorating renal ectopic lipid deposition [74]. In diabetic animal models, it also activates the SIRT1/PGC1 axis, upregulating PPARa and CPT1 to promote fatty acid-β-oxidation and ameliorate renal injury [75]. Notably, resveratrol suppresses the AGE/RAGE axis under high glucose conditions, thereby alleviating renal oxidative stress, inhibiting lipid peroxidation, significantly reducing 4-HNE levels, and attenuating DKD progression [76].
Curcumin, a natural polyphenol extracted from the rhizome of Curcuma longa L. (Zingiberaceae), exhibits hypolipidemic, anti-inflammatory, and antioxidant properties with therapeutic potential for endocrine and immune-related diseases [77]. It enhances adenosine 5′-monophosphate (AMP)-activated protein kinase (AMPK) phosphorylation, leading to downregulation of SREBP-1 and phosphorylation-mediated inactivation of ACC, thereby inhibiting fatty acid synthesis, reducing renal lipid deposition, and ameliorating glomerular basement membrane thickening in DKD [78].
Apigenin, a naturally abundant flavonoid in Apium graveolens L. (Apiaceae), improves insulin resistance and regulates glucose and lipid metabolism [79]. It suppresses SREBP-1c and SREBP-2 expression, markedly down-regulating FAS activity and attenuating ectopic lipid deposition [80]. Furthermore, studies have revealed that apigenin downregulates GLUT1 expression, curbs transmembrane glucose uptake, lowers serum TG and total cholesterol, collectively improving both insulin sensitivity and lipid homeostasis [81].
Soy isoflavones, polyphenolic compounds extracted from the seeds of Glycine max (L.) Merr (Fabaceae), demonstrate promise in preventing and managing of metabolic diseases. Studies have revealed that soy isoflavones activate PPARa via enhanced ligand binding, promoting transcription of target genes and enhancing fatty acid oxidation, thereby countering insulin resistance-induced lipid metabolism disorders [82]. In diabetic rats, soy isoflavones suppress renal SREBP-1c expression, inhibit fatty acid synthesis, and ameliorate glomerulosclerosis [83].
Anthocyanin, a potent natural antioxidant abundant in berries such as Vaccinium uliginosum L. (Ericaceae), exhibits strong free radical scavenging capacity and antioxidant activity. Additionally, anthocyanin can modulate lipid-metabolizing enzymes, thereby ameliorating lipid metabolism disorders and inhibiting lipid peroxidation [84]. Studies have found that anthocyanin regulates glucose and lipid metabolism primarily through AMPK activation, which suppresses the expression of key fatty acid synthesis enzymes SREBP-1c and ACC, inhibits gluconeogenesis and improves insulin sensitivity, ultimately ameliorating glucose and lipid metabolism disorders [85].
Myricetin, a major active component extracted from Morella rubra Lour (Myricaceae), also demonstrates lipid metabolism regulatory effects. It downregulates SREBP-1c and SREBP-2 while upregulating PPARa, exerting significant protective effects against diabetes-induced lipid metabolism disorders. Concurrently, myricetin suppresses the overexpression of transforming growth factor-β (TGF-β), thereby ameliorating glomerulosclerosis and interstitial fibrosis in DKD [86].

3.2. Reducing Triggers: Reinforcing the Antioxidant Barrier

Hesperidin, a flavonoid abundant in the peel of Citrus reticulata Blanco (Rutaceae), possesses potent free radical-scavenging capacity and enhances the expression of Nrf2 [87]. Research has shown that hesperidin augments Nrf2/ARE signaling transduction, promotes glyoxalase 1 overexpression, and subsequently suppresses AGE/RAGE activation, thereby ameliorating oxidative stress-induced renal pathology in DKD [88]. Furthermore, hesperidin significantly downregulates renal NF-κB and NLRP3 expression, reduces pro-inflammatory cytokines IL-6 and IL-1β, and mitigates renal injury driven by diabetic hyperglycemia and hyperlipidemia [89].
Quercetin, a flavonol widely present in various herbal plants, protects against tissue damage through its potent antioxidant capacity. In diabetic kidney disease, ROS derived primarily from mitochondrial and NOX activities contribute to oxidative stress, while quercetin acts as an effective ROS scavenger to maintain redox homeostasis [90]. It also activates Nrf2 signaling and alleviates high glucose-induced overexpression of TFR1 in renal tubular epithelial cells and upregulates GPx4 to ameliorate intracellular iron overload and lipid peroxidation [91]. Moreover, studies have demonstrated that quercetin significantly restores the activities of antioxidant enzymes—including catalase (CAT), superoxide dismutase (SOD), and glutathione S-transferase (GST)—under diabetic conditions. By enhancing antioxidant capacity, it inhibits lipid peroxidation, reduces levels of MDA, and ameliorates renal fibrosis, thus exerting renal protection [92].
Astragaloside IV, a triterpenoid extracted from the dried roots of Astragalus membranaceus (Fisch.) Bunge (Fabaceae), binds to Keap1 and promotes Nrf2 release and nuclear translocation. Within the nucleus, it binds to the ARE, synergistically upregulating multiple antioxidant enzymes. Additionally, it helps maintain mitochondrial homeostasis, restores the activity of mitochondrial respiratory chain complexes, and reduces the release of ROS [93]. Studies have found that astragaloside IV effectively ameliorates palmitate-induced oxidative stress damage in glomerular mesangial cells by downregulating CD36 expression, reducing active uptake of free fatty acids, suppressing the NOX subunit p22phox, and inducing NOX degradation, thereby alleviating lipid deposition and curbing ROS production [94,95]. Moreover, astragaloside IV significantly inhibits NLRP3 inflammasome activation, reduces IL-1β, TNF-α, and MCP-1 levels, and improves renal function and podocyte injury in DKD [96].
Sinapic acid, a phenolic compound found in various plants such as fruits, vegetables, and herbs, functions as a natural free radical scavenger due to its unique molecular structure. Its phenolic hydroxyl groups donate hydrogen atoms to neutralize free radicals, forming stable intermediates that terminate radical chain reactions and inhibit lipid peroxidation [97]. In DKD rats, the administration of sinapic acid activates Nrf2, upregulates SOD and GPx4, thereby significantly suppressing lipid peroxidation. Concurrently, it downregulates NF-κB signaling, inhibiting the release of IL-6 and TNF-α [98]. Furthermore, sinapic acid activates PPAR and CPT1, suppresses SREBP-1, ACC, and FAS expression, ameliorates lipid metabolism disorders, and enhances total antioxidant capacity, thereby mitigating oxidative stress [99].
Chlorogenic acid, a major phenolic compound found in Lonicera japonica Thunb. (Caprifoliaceae), also exhibits remarkable antioxidant capacity. Studies have reported that the protective effect of chlorogenic acid against DKD involves the interaction between the Nrf2/Heme Oxygenase-1 (HO-1) and NF-κB pathways. Upregulated Nrf2 activates HO-1, enhancing antioxidant enzyme expression, while HO-1 downregulates NF-κB and inhibits the release of pro-inflammatory cytokines, significantly ameliorating diabetic kidney injury [100]. Meanwhile, chlorogenic acid can also modulate the NLRP3 inflammasome via Nrf2, exerting anti-inflammatory effects [101].
Proanthocyanidin, obtained from the seeds of Vitis vinifera L. (Vitaceae), serves as an effective free radical scavenger with potent antioxidant activity. Studies have reported that proanthocyanidin activates the Keap1/Nrf2 signaling pathway, upregulates GSH and SOD, and mitigates lipid peroxidation-induced toxicity in DKD [102]. Furthermore, proanthocyanidin inhibits AGE formation. The phenolic hydroxyl groups of proanthocyanidin can trap the carbonyl groups of dicarbonyl compounds, which are precursors to AGEs, forming stable adducts and thereby blocking AGE generation. This action suppresses NOX activation via the AGE/RAGE axis, contributing to oxidative stress amelioration [103].
Geniposide, a natural bioactive compound isolated from Gardenia jasminoides J. Ellis (Rubiaceae), exhibits multiple pharmacological effects including anti-inflammatory, antioxidant, antidiabetic, and neuroprotective properties. Studies have demonstrated that geniposide activates protein kinase A (PKA) and enhances AMPK phosphorylation, thereby promoting autophagy and subsequently inhibiting oxidative stress [104]. Additionally, phosphorylated AMPK further activates SIRT1 and suppresses NF-κB, reducing inflammatory cell infiltration and glomerular basement membrane thickening [105].
Lycopene, a potent antioxidant obtained from Solanum lycopersicum L. (Solanaceae), effectively inhibits lipid peroxidation. It upregulates Nrf2, enhances renal antioxidant capacity, suppresses NF-κB signaling, lowers IL-1β and TNF-α levels, and improves renal inflammation and oxidative stress damage [106]. Concurrently, lycopene also reduces renal AGEs, upregulates SOD and CAT, promotes GPx expression, effectively scavenges free radicals, and alleviates oxidative stress, thereby inhibiting lipid peroxidation in DKD [107].
Coumarin derivatives extracted from the seeds of Dipteryx odorata (Aubl.) Willd. (Fabaceae) exert multi-target therapeutic effects by alleviating oxidative stress and fibrosis. They activate the Nrf2 signaling pathway to enhance antioxidant capacity and effectively scavenge ROS, and they also function as natural SIRT3 activators, promoting the deacetylation of forkhead box O3A (FOXO3a) to enhance manganese superoxide dismutase (MnSOD) activity, thereby establishing a dual antioxidant defense. Concurrently, coumarin derivatives inhibit Smad2/3 phosphorylation, reducing the secretion of fibronectin and delaying renal fibrosis. Furthermore, they downregulate renal SREBP expression, suppress fatty acid synthesis, and ameliorate renal ectopic lipid deposition [108,109].

3.3. Ameliorating Renal Damage: Delaying the Progression of Kidney Pathology

Silymarin, an extract obtained from the dried fruits and seeds of Silybum marianum (L.) Gaertn (Asteraceae), exhibits anti-inflammatory, antioxidant, and anti-fibrotic properties. Studies have found that silymarin ameliorates renal fibrosis in DKD, potentially through inhibition of the TGF-β/Smad signaling pathway [110]. TGF-β, a major driver of fibrosis, is significantly upregulated in DKD, leading to Smad protein activation, nuclear translocation, and subsequent expression of fibrotic genes [111]. Additionally, silymarin upregulates antioxidant enzymes including CAT and GPx4, thereby alleviating lipid peroxidation in DKD [112].
α-Actinin-4 (ACTN4), an actin-binding protein, is critical for maintaining the structural and functional integrity of glomerular podocytes [113]. In DKD, however, hypermethylation of ACTN4 occurs, compromising the glomerular filtration barrier and promoting scar formation [114]. Epigallocatechin gallate, a compound isolated from Camellia sinensis (L.) Kuntze (Theaceae), exhibits natural demethylating activity and demonstrates therapeutic potential in DKD. Studies have confirmed that Epigallocatechin gallate inhibits DNA methyltransferases, reverses ACTN4 hypermethylation, and suppresses the expression of NF-κB and IκBα. This leads to reduced levels of the fibrosis marker α-smooth muscle actin (α-SMA) and inflammatory cytokines, thereby ameliorating renal damage [115]. Additionally, Epigallocatechin gallate inhibits the overexpression of NLRP3 and blocks the release of IL-1β, thereby attenuating DKD-associated inflammatory injury [116].
Isoliquiritigenin, a bioactive compound extracted from the roots of Glycyrrhiza uralensis Fisch (Fabaceae), demonstrates potent free radical scavenging capacity, ameliorates oxidative stress, and inhibits lipid peroxidation. Additionally, it suppresses NLRP3 and NF-κB signaling pathways, exerting anti-inflammatory and antioxidant effects that alleviate renal damage in DKD [89]. Furthermore, 4-HNE, an active mediator linking lipid peroxidation and inflammatory responses, has been demonstrated in other disease models to modify the granulocyte-macrophage colony-stimulating factor (GM-CSF) receptor, leading to sustained activation of the downstream janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathway and amplified inflammatory responses [117,118]. The role of JAK/STAT signaling in DKD-related inflammatory damage is well-established [119]. Studies have found that isoliquiritigenin ameliorates renal inflammation and fibrosis partly through inhibition of the JAK/STAT signaling pathway. In DKD, upregulated JAK promotes the phosphorylation and nuclear translocation of STAT, regulating pro-inflammatory cytokines and fibrotic markers. Isoliquiritigenin treatment significantly inhibits the JAK2/STAT3 signaling pathway, downregulates TGF-β, fibronectin, and collagen expression, and improves renal pathological damage in DKD [120].
Andrographolide, the primary active constituent obtained from Andrographis paniculata (Burm. f.) Nees (Acanthaceae), is known as a “natural antibiotic” for its notable anti-inflammatory and antiviral properties. Studies have revealed that andrographolide ameliorates renal pathology in DKD by suppressing the STAT3-mediated phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling pathway. Aberrant activation of PI3K promotes DKD progression by recruiting and phosphorylating Akt, thereby promoting inflammation and inducing epithelial–mesenchymal transition (EMT) in renal tubular epithelial cells. Andrographolide effectively reverses this process [121]. Moreover, andrographolide induced Akt inhibition reduces NF-κB transcriptional activity, blocks pro-inflammatory cytokines release, and mitigates inflammatory injury in DKD [122].
Berberine, an alkaloid isolated from the roots of Coptis chinensis Franch (Ranunculaceae), exhibits hypoglycemic and lipid-lowering, and insulin-sensitizing effects, demonstrating considerable potential for treating diabetes and its complications [123]. Furthermore, studies have shown that berberine also inhibits ferroptosis by upregulating GPx4 and Nrf2, thereby effectively inhibiting ferroptosis [124].
Ginkgolide B, primarily extracted from the dried leaves of Ginkgo biloba L. (Ginkgoaceae), is well-known for its specific platelet-activating factor receptor antagonist with recently demonstrated renal protective effects. In vitro, ginkgolide B inhibits TFR1, enhances FTH1 expression, reduces overloaded intracellular free iron levels, and restores renal iron homeostasis. It also suppresses ubiquitination-mediated degradation of GPx4, thereby improving antioxidant capacity and inhibiting oxidative stress and ferroptosis in DKD [125]. Furthermore, ginkgolide B activates PPARa to promote fatty acid oxidation and upregulates the Nrf2-mediated antioxidant system, thus ameliorating lipid metabolism disorders [126].
Tanshinone IIA, a diterpenoid quinone found in the roots of Salvia miltiorrhiza Bunge (Lamiaceae), is used in treating cardiovascular and cerebrovascular diseases. Studies have shown that tanshinone IIA exhibits renal protective effects, including anti-inflammatory, antioxidant, and anti-fibrotic activities. In DKD models, tanshinone IIA inhibits the activation of NLRP3, blocking caspase-1-mediated release of pro-inflammatory cytokines and cleavage of GSDMD, thereby suppressing pyroptosis [127]. Furthermore, it also downregulates ACSL4 expression, attenuates high glucose-induced ferroptosis, upregulates GSH to scavenge free radicals, and collectively ameliorates renal pathological damage in DKD [128].
Table 1. Natural products regulating lipid peroxidation in DKD.
Table 1. Natural products regulating lipid peroxidation in DKD.
Natural ProductSourceApplication ModelMechanismRefs.
ResveratrolReynoutria japonica HouttHFD induced C57BL/6J mice
C57BL/KsJ db/db mice
HG induced NMS2 cells
STZ injected male Wistar rats
Activate SIRT1/PGC1, upregulate PPARa/CPT1, suppress SREBP-1 and ChREBP, suppress AGE/RAGE, reduce 4-HNE[74,75,76]
CurcuminCurcuma longa L.OLETF ratsEnhance AMPK phosphorylation, downregulate SREBP-1 and promote ACC phosphorylation[78]
ApigeninApium graveolens L.HFD induced male C57BL/6J mice
HF induced SD rats
Suppress SREBP-1c and SREBP-2, downregulate FAS and GLUT1[80,81]
Soy isoflavonesGlycine max (L.) Merrobese Zucker rats
HFD + STZ injected male SD rats
Active PPARa and suppress SREBP-1c[82,83]
AnthocyaninVaccinium uliginosum L.HFSD + STZ injected male C57BL/6J miceScavenge free radicals, activate AMPK, suppress SREBP-1c and ACC[85]
MyricetinMorella rubra LourSTZ-Cd induced male albino Wistar ratsDownregulate SREBP-1c and SREBP-2, upregulate PPARa, suppress TGF-β[86]
HesperidinCitrus reticulata BlancoSTZ injected male SD rats
HFD + STZ injected Wistar rats
Scavenge free radicals, activate Nrf2/ARE signaling pathway, promote Glo-1 and suppress AGE/RAGE, downregulate NF-κB and NLRP3, reduces IL-6 and IL-1β[88,89]
QuercetinVarious herbal plantsHG induced HK-2 cells
Male C57BL/KsJ db/db mice
Scavenge ROS, activate Nrf2, downregulate TFR1 and upregulate GPx4, enhance CAT, SOD and GST[91]
Astragaloside IVAstragalus membranaceus (Fisch.) BungeSTZ injected male C57BL/6J rats
HG induced MPC cells
HFD + STZ injected Sprague Dawley rats
PA induced HMC cells
BKS-db/db mice
HG induced MPC cells
Promote Nrf2, downregulate CD36, suppress NOX, inhibit NLRP3 inflammasome, reduce IL-1β, TNF-α, and MCP-1[93,94,96]
Sinapic acidVarious herbal plantsSTZ injected male Wistar rats
HFD induced male Syrian hamsters
Scavenge ROS, activate Nrf2, upregulate SOD and GPx4, downregulate NF-κB signaling pathway, promote PPAR and CPT1, suppress SREBP-1, ACC, and FAS[98,99]
Chlorogenic acidLonicera japonica Thunb.STZ injected HBZY-1 cells
HFD + STZ injected Wistar rats
HG induced HK-2 cells
Upregulate Nrf2/HO-1, downregulate NF-κB and NLRP3 inflammasome signaling pathway[100,101]
ProanthocyanidinVitis vinifera L.Cd + HFSD induced male KM miceScavenge ROS, activate Keap1/Nrf2 signaling pathway, upregulate GSH and SOD, suppress NOX via the AGE/RAGE axis[102]
GeniposideGardenia jasminoides J. EllisHFD + UNx + STZ injected C57BL/6 male mice
HFD + STZ injected C57BL/6 mice
Activate PKA, enhance AMPK and autophagy, activate SIRT1 and suppress NF-κB[104,105]
LycopeneSolanum lycopersicum L.HFD induced male Wistar rats
STZ injected male Wistar rats
upregulate Nrf2, suppress NF-κB, reduce IL-1β and TNF-α, suppress AGEs, upregulate SOD and CAT, promotes GPx[106,107]
Coumarin derivativesDipteryx odorata (Aubl.) Willd.STZ injected male Wistar rats
HG induced HBZY-1 cells
HFD + STZ injected male C57BL/6 mice
AGEs induced HK2 cells
Activate Nrf2, scavenge ROS, activate SIRT3, promote FOXO3a, enhance MnSOD, inhibit Smad2/3, reduce fibronectin, downregulate SREBP[108,109]
SilymarinSilybum marianum (L.) GaertnHG induced MPC5 cells
STZ injected male SD rats
Inhibit TGF-β/Smad signaling pathway, upregulate CAT and GPx4[110,112]
Epigallocatechin gallateCamellia sinensis (L.) KuntzeHG induced HPC cells
Male C57BL/KsJ db/db mice
Inhibit DNA methyltransferase, reverse ACTN4 hypermethylation, suppress NF-κB and IκBα, reduce α-SMA and inhibit NLRP3[115,116]
IsoliquiritigeninGlycyrrhiza uralensis FischHFD + STZ injected Wistar rats
HG induced HK-2 cells
HFD + STZ injected male SD rats
Scavenge free radicals, suppress NLRP3, NF-κB and JAK/STAT signaling pathways, downregulate TGF-β, fibronectin, and collagen[89,120]
AndrographolideAndrographis paniculata (Burm. f.) NeesHFD + STZ induced SD rats
AGEs induced MPC5 cells
HFD + STZ injected C57BL/6 mice
Suppress STAT3/PI3K/Akt signaling pathway, inhibit NF-κB[121,122]
BerberineCoptis chinensis FranchHFD induced C57BL/6J mice and
AopE−/− mice
Upregulate GPx4 and Nrf2[124]
Ginkgolide BGinkgo biloba L.Male C57BL/KsJ db/db mice
PA-G induced MPC5 cells
HFD induced SD rats
Inhibit TFR1, enhance FTH1, reduce intracellular free iron, suppress the ubiquitination of GPx4, activate PPARa and upregulate Nrf2[125,126]
Tanshinone IIASalvia miltiorrhiza Bungedb/db mice
HG induced HRGEC cells
HG induced MPC5 cells
Inhibit NLRP3/caspase-1/GSDMD mediated pyroptosis, downregulate ACSL4, upregulate GSH and scavenge free radicals[127,128]

4. Conclusions

Diabetic kidney disease (DKD) is a complex pathological process characterized by the interplay of chronic hyperglycemia and lipid metabolism disorders, involving multiple interconnected mechanisms. In recent years, lipid metabolism disorders, particularly ectopic lipid deposition in the renal tissue and the resulting lipid peroxidation, have emerged as a core factor promoting the progression of DKD. This process accelerates glomerulosclerosis and tubulointerstitial fibrosis by disrupting cellular structures, amplifying inflammatory responses, exacerbating oxidative stress, and inducing programmed cell death, ultimately leading to irreversible renal injury. Therefore, targeting lipid peroxidation and its associated injury network represents a highly promising therapeutic strategy for DKD.
Although current studies have made significant progress, the pathogenic network of DKD requires further systematic elucidation. For instance, 4-HNE is not merely a terminal product of lipid peroxidation but also functions as a highly reactive signaling molecule that actively contributes to disease progression. However, its specific mechanisms of action in DKD still need further exploration. Future studies should prioritize elucidating the molecular regulatory network of lipid peroxidation in DKD, systematically identifying the specific targets, signaling pathways, and interactions of 4-HNE with other pathological processes. A deeper understanding of these mechanisms will not only refine the pathological framework of DKD but also provide novel insights and targets for developing natural medicines that intervene in lipid peroxidation and its downstream effects.
Traditional Chinese herbal medicine possesses a rich historical foundation in disease treatment. However, the poorly defined active constituents have often limited its application and promotion in modern medicine. With advances in extraction and isolation technologies, our understanding of bioactive compounds obtained from natural plants has considerably improved. Natural compounds extracted from medicinal herbs have demonstrated remarkable efficacy in mitigating lipid peroxidation in DKD. These components ameliorate renal pathology through core signaling pathways such as FAO, FAS, Nrf2, GPx4, and NF-κB, which represent key regulators of lipid peroxidation in DKD. Preclinical studies have preliminarily confirmed the potential of natural compounds in alleviating lipid peroxidation in DKD. These compounds exhibit diverse mechanisms of action and represent promising prospects for novel therapeutic strategies, given their capacity to modulate multiple targets and pathways in a synergistic manner.
It is noteworthy that the multi-target and multi-pathway characteristics of natural products, while offering comprehensive therapeutic advantages, also entail potential off-target risks, complicating their clinical application. Moreover, most current evidence comes from animal or cell-based studies, and clinical translation still faces multiple challenges. The complexity of human physiology necessitates further validation of critical issues such as dose–response relationships, long-term safety, and potential drug interactions. Additionally, standardized extraction processes and rigorous quality control are essential prerequisites for clinical application. Nevertheless, the considerable potential of natural products in mitigating lipid peroxidation in DKD should not be overlooked.
Future studies should focus on further elucidating the molecular mechanisms underlying lipid peroxidation in DKD to identify novel therapeutic targets. Moreover, natural products often suffer from limitations such as low oral bioavailability and rapid systemic clearance, which hinder their therapeutic efficacy. Therefore, research efforts should be directed toward developing advanced drug delivery systems—such as nanoparticles, liposomes, or kidney-targeting peptide carriers—to improve bioavailability and renal targeting. At the same time, it is crucial to conduct large-scale, multicenter, randomized controlled clinical trials to systematically evaluate the efficacy, long-term safety, and potential side effects of natural products in DKD patients, thereby providing a robust evidence base for their clinical translation.

Author Contributions

Y.D. drafted the manuscript. Y.T. discussed and critically revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
4-HNE4-Hydroxy-nonenal
ACCAcetyl-CoA carboxylase
ACSL4Acyl-CoA synthetase long chain family member 4
AMPKAdenosine 5′-monophosphate (AMP)-activated protein kinase
AGEAdvanced glycation end product
AREAntioxidant response element
ChREBPCarbohydrate response element binding protein
CPT1Carnitine palmitoyl transferase 1
CATCatalase
CD36Cluster of differentiation 36
DKDDiabetic kidney disease
ETCElectron transport chain
ESRDEnd-stage renal disease
EMTEpithelial–mesenchymal transition
FAOFatty acid oxidation
FASFatty acid synthase
FTNFerritin
GSDMDGasdermin D
GLUTGlucose transporter
GSHGlutathione
GPxGlutathione peroxidase
GSTGlutathione S-transferase
HO-1Heme oxygenase-1
IKKInhibitor of kappa B kinase
IκBInhibitor of NF-κB
JAKJanus kinase
KeaP1Kelch-like ECH-associated protein 1
LOOHLipid hydroperoxides
MDAMalondialdehyde
MPCMitochondrial pyruvate carrier
NOXNADPH oxidase
NLRP3NOD-like receptor thermal protein domain associated protein 3
Nrf2Nuclear factor erythroid 2-related factor 2
NF-κBNuclear factor kappa-B
PRRPattern recognition receptors
PPARaPerixisome proliferator-activated receptor alpha
PI3KPhosphoinositide 3-kinase
PUFA-PEPolyunsaturated fatty acid-phosphatidyl ethanolamine
PUFAPolyunsaturated fatty acid
PKAProtein kinase A
AktProtein kinase B
ROSReactive oxygen species
RAGEReceptor for AGE
RAASiRenin–angiotensin–aldosterone system inhibitor
STATSignal transducer and activator of transcription
SIRT1Sirtuin 1
SGLT2iSodium-dependent glucose transporters 2 inhibitor
SREBP-1cSterol regulatory element binding protein-1c
SODSuperoxide dismutase
TFRTransferrin receptor
TGTriglycerides
ACTN4α-actinin-4
α-SMAα-smooth muscle actin

References

  1. Kidney Disease: Improving Global Outcomes (KDIGO) Diabetes Work Group. KDIGO 2022 clinical practice guideline for diabetes management in chronic kidney disease. Kidney Int. 2022, 102, S1–S127. [Google Scholar] [CrossRef]
  2. van Raalte, D.H.; Bjornstad, P.; Cherney, D.Z.I.; de Boer, I.H.; Fioretto, P.; Gordin, D.; Persson, F.; Rosas, S.E.; Rossing, P.; Schaub, J.A.; et al. Combination therapy for kidney disease in people with diabetes mellitus. Nat. Rev. Nephrol. 2024, 20, 433–446. [Google Scholar] [CrossRef]
  3. Danta, C.C.; Boa, A.N.; Bhandari, S.; Sathyapalan, T.; Xu, S.Z. Recent advances in drug discovery for diabetic kidney disease. Expert Opin. Drug Discov. 2021, 16, 447–461. [Google Scholar] [CrossRef]
  4. Njeim, R.; Alkhansa, S.; Fornoni, A. Unraveling the crosstalk between lipids and NADPH oxidases in diabetic kidney disease. Pharmaceutics 2023, 15, 1360. [Google Scholar] [CrossRef]
  5. Hou, Y.; Tan, E.; Shi, H.; Ren, X.; Wan, X.; Wu, W.; Chen, Y.; Niu, H.; Zhu, G.; Li, J.; et al. Mitochondrial oxidative damage reprograms lipid metabolism of renal tubular epithelial cells in the diabetic kidney. Cell. Mol. Life Sci. 2024, 81, 23. [Google Scholar] [CrossRef] [PubMed]
  6. Raz, I.; Eldor, R.; Cernea, S.; Shafrir, E. Diabetes: Insulin resistance and derangements in lipid metabolism. Cure through intervention in fat transport and storage. Diabetes Metab. Res. Rev. 2005, 21, 3–14. [Google Scholar] [CrossRef] [PubMed]
  7. Gai, Z.; Wang, T.; Visentin, M.; Kullak-Ublick, G.A.; Fu, X.; Wang, Z. Lipid accumulation and chronic kidney disease. Nutrients 2019, 11, 722. [Google Scholar] [CrossRef] [PubMed]
  8. Zhou, K.; Zi, X.; Song, J.; Zhao, Q.; Liu, J.; Bao, H.; Li, L. Molecular mechanistic pathways targeted by natural compounds in the prevention and treatment of diabetic kidney disease. Molecules 2022, 27, 6221. [Google Scholar] [CrossRef]
  9. Zhao, X.; An, X.; Yang, C.; Sun, W.; Ji, H.; Lian, F. The crucial role and mechanism of insulin resistance in metabolic disease. Front. Endocrinol. 2023, 14, 1149239. [Google Scholar] [CrossRef]
  10. Petersen, M.C.; Shulman, G.I. Mechanisms of insulin action and insulin resistance. Physiol. Rev. 2018, 98, 2133–2223. [Google Scholar] [CrossRef]
  11. Lu, X.; Xie, Q.; Pan, X.; Zhang, R.; Zhang, X.; Peng, G.; Zhang, Y.; Shen, S.; Tong, N. Type 2 diabetes mellitus in adults: Pathogenesis, prevention and therapy. Signal Transduct. Target. Ther. 2024, 9, 262. [Google Scholar] [CrossRef] [PubMed]
  12. Puchałowicz, K.; Rać, M.E. The multifunctionality of CD36 in diabetes mellitus and its complications-update in pathogenesis, treatment and monitoring. Cells 2020, 9, 1877. [Google Scholar] [CrossRef] [PubMed]
  13. Lopaschuk, G.D. Fatty acid oxidation and its relation with insulin resistance and associated disorders. Ann. Nutr. Metab. 2016, 68 (Suppl. 3), 15–20. [Google Scholar] [CrossRef] [PubMed]
  14. Schlaepfer, I.R.; Joshi, M. CPT1A-mediated fat oxidation, mechanisms, and therapeutic potential. Endocrinology 2020, 161, bqz046. [Google Scholar] [CrossRef]
  15. Jang, H.S.; Noh, M.R.; Kim, J.; Padanilam, B.J. Defective mitochondrial fatty acid oxidation and lipotoxicity in kidney diseases. Front. Med. 2020, 7, 65. [Google Scholar] [CrossRef]
  16. Bjørndal, B.; Alterås, E.K.; Lindquist, C.; Svardal, A.; Skorve, J.; Berge, R.K. Associations between fatty acid oxidation, hepatic mitochondrial function, and plasma acylcarnitine levels in mice. Nutr. Metab. 2018, 15, 10. [Google Scholar] [CrossRef]
  17. Herman-Edelstein, M.; Scherzer, P.; Tobar, A.; Levi, M.; Gafter, U. Altered renal lipid metabolism and renal lipid accumulation in human diabetic nephropathy. J. Lipid Res. 2014, 55, 561–572. [Google Scholar] [CrossRef]
  18. Miguel, V.; Tituaña, J.; Herrero, J.I.; Herrero, L.; Serra, D.; Cuevas, P.; Barbas, C.; Puyol, D.R.; Márquez-Expósito, L.; Ruiz-Ortega, M.; et al. Renal tubule CPT1A overexpression protects from kidney fibrosis by restoring mitochondrial homeostasis. J. Clin. Invest. 2021, 131, e140695. [Google Scholar] [CrossRef]
  19. Nakamura, M.T.; Nara, T.Y. Essential fatty acid synthesis and its regulation in mammals. Prostaglandins Leukot. Essent. Fat. Acids 2003, 68, 145–150. [Google Scholar] [CrossRef]
  20. Li, L.; Long, J.; Mise, K.; Poungavrin, N.; Lorenzi, P.L.; Mahmud, I.; Tan, L.; Saha, P.K.; Kanwar, Y.S.; Chang, B.H.; et al. The transcription factor ChREBP links mitochondrial lipidomes to mitochondrial morphology and progression of diabetic kidney disease. J. Biol. Chem. 2023, 299, 105185. [Google Scholar] [CrossRef]
  21. Wedan, R.J.; Longenecker, J.Z.; Nowinski, S.M. Mitochondrial fatty acid synthesis is an emergent central regulator of mammalian oxidative metabolism. Cell Metab. 2024, 36, 36–47. [Google Scholar] [CrossRef]
  22. Wang, H.; Zhu, L.; Hao, J.; Duan, H.; Liu, S.; Zhao, S.; Liu, Q.; Liu, W. Co-regulation of SREBP-1 and mTOR ameliorates lipid accumulation in kidney of diabetic mice. Exp. Cell Res. 2015, 336, 76–84. [Google Scholar] [CrossRef]
  23. Tao, J.; Zhang, X.Y.; Tan, H.B.; Huang, M.Y.; Yang, Y.Q.; Guo, J. FTZ alleviates lipid deposition in diabetic kidney disease by AMPK/ACC/SREBP signaling pathway. Acta Diabetol. 2025, 62, 1–10. [Google Scholar] [CrossRef] [PubMed]
  24. Mitrofanova, A.; Merscher, S.; Fornoni, A. Kidney lipid dysmetabolism and lipid droplet accumulation in chronic kidney disease. Nat. Rev. Nephrol. 2023, 19, 629–645. [Google Scholar] [CrossRef] [PubMed]
  25. Ducasa, G.M.; Mitrofanova, A.; Fornoni, A. Crosstalk between lipids and mitochondria in diabetic kidney disease. Curr. Diab. Rep. 2019, 19, 144. [Google Scholar] [CrossRef] [PubMed]
  26. Nakamura, M.T.; Yudell, B.E.; Loor, J.J. Regulation of energy metabolism by long-chain fatty acids. Prog. Lipid Res. 2014, 53, 124–144. [Google Scholar] [CrossRef]
  27. Bhargava, P.; Schnellmann, R.G. Mitochondrial energetics in the kidney. Nat. Rev. Nephrol. 2017, 13, 629–646. [Google Scholar] [CrossRef]
  28. Yao, L.; Liang, X.; Qiao, Y.; Chen, B.; Wang, P.; Liu, Z. Mitochondrial dysfunction in diabetic tubulopathy. Metabolism 2022, 131, 155195. [Google Scholar] [CrossRef]
  29. Ito, M.; Gurumani, M.Z.; Merscher, S.; Fornoni, A. Glucose- and non-glucose-induced mitochondrial dysfunction in diabetic kidney disease. Biomolecules 2022, 12, 351. [Google Scholar] [CrossRef]
  30. Braga, P.C.; Alves, M.G.; Rodrigues, A.S.; Oliveira, P.F. Mitochondrial pathophysiology on chronic kidney disease. Int. J. Mol. Sci. 2022, 23, 1776. [Google Scholar] [CrossRef]
  31. Efiong, E.E.; Bazireh, H.; Fuchs, M.; Amadi, P.U.; Effa, E.; Sharma, S.; Schmaderer, C. Crosstalk of hyperglycaemia and cellular mechanisms in the pathogenesis of diabetic kidney disease. Int. J. Mol. Sci. 2024, 25, 10882. [Google Scholar] [CrossRef]
  32. Sanajou, D.; Ghorbani Haghjo, A.; Argani, H.; Aslani, S. AGE-RAGE axis blockade in diabetic nephropathy: Current status and future directions. Eur. J. Pharmacol. 2018, 833, 158–164. [Google Scholar] [CrossRef] [PubMed]
  33. Pathomthongtaweechai, N.; Chutipongtanate, S. AGE/RAGE signaling-mediated endoplasmic reticulum stress and future prospects in non-coding RNA therapeutics for diabetic nephropathy. Biomed. Pharmacother. 2020, 131, 110655. [Google Scholar] [CrossRef] [PubMed]
  34. Guo, J.; Ananthakrishnan, R.; Qu, W.; Lu, Y.; Reiniger, N.; Zeng, S.; Ma, W.; Rosario, R.; Yan, S.F.; Ramasamy, R.; et al. RAGE mediates podocyte injury in adriamycin-induced glomerulosclerosis. J. Am. Soc. Nephrol. 2008, 19, 961–972. [Google Scholar] [CrossRef] [PubMed]
  35. Yiu, W.H.; Wong, D.W.; Wu, H.J.; Li, R.X.; Yam, I.; Chan, L.Y.; Leung, J.C.; Lan, H.Y.; Lai, K.N.; Tang, S.C. Kallistatin protects against diabetic nephropathy in db/db mice by suppressing AGE-RAGE-induced oxidative stress. Kidney Int. 2016, 89, 386–398. [Google Scholar] [CrossRef]
  36. Kaseda, K.; Kai, Y.; Tajima, M.; Suematsu, M.; Iwata, S.; Miyata, M.; Mifude, C.K.; Yamashita, N.; Seiryu, W.A.; Fukada, M.; et al. Oral administration of spa-derived green alga improves insulin resistance in overweight subjects: Mechanistic insights from fructose-fed rats. Pharmacol. Res. 2020, 152, 104633. [Google Scholar] [CrossRef]
  37. Eid, A.A.; Gorin, Y.; Fagg, B.M.; Maalouf, R.; Barnes, J.L.; Block, K.; Abboud, H.E. Mechanisms of podocyte injury in diabetes: Role of cytochrome P450 and NADPH oxidases. Diabetes 2009, 58, 1201–1211. [Google Scholar] [CrossRef]
  38. Kawamura, M.; Kobashi, Y.; Tanaka, H.; Bohno-Mikami, A.; Hamada, M.; Ito, Y.; Hirata, T.; Ohara, H.; Kojima, N.; Koretsune, H.; et al. Discovery of novel pyrazolylpyridine derivatives for 20-hydroxyeicosatetraenoic acid synthase inhibitors with selective CYP4A11/4F2 inhibition. J. Med. Chem. 2022, 65, 14599–14613. [Google Scholar] [CrossRef]
  39. Bellezza, I.; Giambanco, I.; Minelli, A.; Donato, R. Nrf2-Keap1 signaling in oxidative and reductive stress. Biochim. Biophys. Acta Mol. Cell Res. 2018, 1865, 721–733. [Google Scholar] [CrossRef]
  40. Liu, Y.; Uruno, A.; Saito, R.; Matsukawa, N.; Hishinuma, E.; Saigusa, D.; Liu, H.; Yamamoto, M. Nrf2 deficiency deteriorates diabetic kidney disease in Akita model mice. Redox Biol. 2022, 58, 102525. [Google Scholar] [CrossRef]
  41. Hashemi, M.; Zandieh, M.A.; Ziaolhagh, S.; Mojtabavi, S.; Sadi, F.H.; Koohpar, Z.K.; Ghanbarirad, M.; Haghighatfard, A.; Behroozaghdam, M.; Khorrami, R.; et al. Nrf2 signaling in diabetic nephropathy, cardiomyopathy and neuropathy: Therapeutic targeting, challenges and future prospective. Biochim. Biophys. Acta Mol. Basis Dis. 2023, 1869, 166714. [Google Scholar] [CrossRef] [PubMed]
  42. Asmat, U.; Abad, K.; Ismail, K. Diabetes mellitus and oxidative stress-A concise review. Saudi Pharm. J. 2016, 24, 547–553. [Google Scholar] [CrossRef] [PubMed]
  43. Niedowicz, D.M.; Daleke, D.L. The role of oxidative stress in diabetic complications. Cell Biochem. Biophys. 2005, 43, 289–330. [Google Scholar] [CrossRef]
  44. Yin, H.; Xu, L.; Porter, N.A. Free radical lipid peroxidation: Mechanisms and analysis. Chem. Rev. 2011, 111, 5944–5972. [Google Scholar] [CrossRef]
  45. Su, L.J.; Zhang, J.H.; Gomez, H.; Murugan, R.; Hong, X.; Xu, D.; Jiang, F.; Peng, Z.Y. Reactive oxygen species-induced lipid peroxidation in apoptosis, autophagy, and ferroptosis. Oxid. Med. Cell. Longev. 2019, 2019, 5080843. [Google Scholar] [CrossRef]
  46. Ayala, A.; Muñoz, M.F.; Argüelles, S. Lipid peroxidation: Production, metabolism, and signaling mechanisms of malondialdehyde and 4-hydroxy-2-nonenal. Oxid. Med. Cell. Longev. 2014, 2014, 360438. [Google Scholar] [CrossRef]
  47. Medina-Navarro, R.; Nieto-Aguilar, R.; Alvares-Aguilar, C. Protein conjugated with aldehydes derived from lipid peroxidation as an independent parameter of the carbonyl stress in the kidney damage. Lipids Health Dis. 2011, 10, 201. [Google Scholar] [CrossRef]
  48. Vazdar, K.; Škulj, S.; Bakarić, D.; Margetić, D.; Vazdar, M. Chemistry and reactivity of 4-hydroxy-2-nonenal (HNE) in model biological systems. Mini Rev. Med. Chem. 2021, 21, 1394–1405. [Google Scholar] [CrossRef]
  49. Epizzimenti, S.; Ciamporcero, E.S.; Edaga, M.; Epettazzoni, P.; Earcaro, A.; Ecetrangolo, G.; Eminelli, R.; Edianzani, C.; Elepore, A.; Egentile, F.; et al. Interaction of aldehydes derived from lipid peroxidation and membrane proteins. Front. Physiol. 2013, 4, 242. [Google Scholar] [CrossRef]
  50. Schaur, R.J.; Siems, W.; Bresgen, N.; Eckl, P.M. 4-Hydroxy-nonenal-a bioactive lipid peroxidation product. Biomolecules 2015, 5, 2247–2337. [Google Scholar] [CrossRef]
  51. Soulage, C.O.; Pelletier, C.C.; Florens, N.; Lemoine, S.; Dubourg, L.; Juillard, L.; Guebre-Egziabher, F. Two toxic lipid aldehydes, 4-hydroxy-2-hexenal (4-HHE) and 4-hydroxy-2-nonenal (4-HNE), accumulate in patients with chronic kidney disease. Toxins 2020, 12, 567. [Google Scholar] [CrossRef]
  52. Mu, F.; Luo, P.; Zhu, Y.; Nie, P.; Li, B.; Bai, X. Iron metabolism and ferroptosis in diabetic kidney disease. Cell Biochem. Funct. 2025, 43, e70067. [Google Scholar] [CrossRef] [PubMed]
  53. Barnabei, L.; Laplantine, E.; Mbongo, W.; Rieux-Laucat, F.; Weil, R. NF-κB: At the borders of autoimmunity and inflammation. Front. Immunol. 2021, 12, 716469. [Google Scholar] [CrossRef] [PubMed]
  54. White, S.; Lin, L.; Hu, K. NF-κB and tPA signaling in kidney and other diseases. Cells 2020, 9, 1348. [Google Scholar] [CrossRef] [PubMed]
  55. Nakashima, I.; Liu, W.; Akhand, A.A.; Takeda, K.; Kawamoto, Y.; Kato, M.; Suzuki, H. 4-hydroxynonenal triggers multistep signal transduction cascades for suppression of cellular functions. Mol. Asp. Med. 2003, 24, 231–238. [Google Scholar] [CrossRef]
  56. Ioannidis, M.; Tjepkema, J.; Uitbeijerse, M.R.P.; van den Bogaart, G. Immunomodulatory effects of 4-hydroxynonenal. Redox Biol. 2025, 85, 103719. [Google Scholar] [CrossRef]
  57. Uchida, K. Redox-derived damage-associated molecular patterns: Ligand function of lipid peroxidation adducts. Redox Biol. 2013, 1, 94–96. [Google Scholar] [CrossRef]
  58. Tang, S.C.W.; Yiu, W.H. Innate immunity in diabetic kidney disease. Nat. Rev. Nephrol. 2020, 16, 206–222. [Google Scholar] [CrossRef]
  59. Fu, J.; Wu, H. Structural mechanisms of NLRP3 inflammasome assembly and activation. Annu. Rev. Immunol. 2023, 41, 301–316. [Google Scholar] [CrossRef]
  60. Henedak, N.T.; El-Abhar, H.S.; Soubh, A.A.; Abdallah, D.M. NLRP3 inflammasome: A central player in renal pathologies and nephropathy. Life Sci. 2024, 351, 122813. [Google Scholar] [CrossRef]
  61. Liu, P.; Zhang, Z.; Li, Y. Relevance of the pyroptosis-related inflammasome pathway in the pathogenesis of diabetic kidney disease. Front. Immunol. 2021, 12, 603416. [Google Scholar] [CrossRef]
  62. Foresto-Neto, O.; Albino, A.H.; Arias, S.C.A.; Faustino, V.D.; Zambom, F.F.F.; Cenedeze, M.A.; Elias, R.M.; Malheiros, D.M.A.C.; Camara, N.O.S.; Fujihara, C.K.; et al. NF-κB system is chronically activated and promotes glomerular injury in experimental type 1 diabetic kidney disease. Front. Physiol. 2020, 11, 84. [Google Scholar] [CrossRef]
  63. Wu, K.; Zha, H.; Wu, T.; Liu, H.; Peng, R.; Lin, Z.; Lv, D.; Liao, X.; Sun, Y.; Zhang, Z. Cytosolic Hmgb1 accumulation in mesangial cells aggravates diabetic kidney disease progression via NFκB signaling pathway. Cell. Mol. Life Sci. 2024, 81, 408. [Google Scholar] [CrossRef]
  64. Sharma, S.; Sharma, P.; Bailey, T.; Bhattarai, S.; Subedi, U.; Miller, C.; Ara, H.; Kidambi, S.; Sun, H.; Panchatcharam, M.; et al. Electrophilic aldehyde 4-hydroxy-2-nonenal mediated signaling and mitochondrial dysfunction. Biomolecules 2022, 12, 1555. [Google Scholar] [CrossRef] [PubMed]
  65. Maiorino, M.; Conrad, M.; Ursini, F. GPx4, lipid oeroxidation, and cell death: Discoveries, rediscoveries, and open issues. Antioxid. Redox Signal 2018, 29, 61–74. [Google Scholar] [CrossRef] [PubMed]
  66. Lee, S.H.; Takahashi, K.; Hatakawa, Y.; Oe, T. Lipid peroxidation-derived modification and its effect on the activity of glutathione peroxidase 1. Free Radic. Biol. Med. 2023, 208, 252–259. [Google Scholar] [CrossRef] [PubMed]
  67. Bosch-Morell, F.; Flohé, L.; Marín, N.; Romero, F.J. 4-Hydroxynonenal inhibits glutathione peroxidase: Protection by glutathione. Free Radic. Biol. Med. 1999, 26, 1383–1387. [Google Scholar] [CrossRef]
  68. Gao, G.; Li, J.; Zhang, Y.; Chang, Y.Z. Cellular iron metabolism and regulation. Adv. Exp. Med. Biol. 2019, 1173, 21–32. [Google Scholar] [CrossRef]
  69. Zhong, S.; Wang, N.; Zhang, C. Podocyte death in diabetic kidney disease: Potential molecular mechanisms and therapeutic targets. Int. J. Mol. Sci. 2024, 25, 9035. [Google Scholar] [CrossRef]
  70. Wang, H.; Liu, D.; Zheng, B.; Yang, Y.; Qiao, Y.; Li, S.; Pan, S.; Liu, Y.; Feng, Q.; Liu, Z. Emerging role of ferroptosis in diabetic kidney disease: Molecular mechanisms and therapeutic opportunities. Int. J. Biol. Sci. 2023, 19, 2678–2694. [Google Scholar] [CrossRef]
  71. Wu, K.; Zhu, E.; Chen, J.; Kuang, Q.; Lin, J.; Zhao, S.; Xu, X.; Li, S.; Sui, Y.; Huang, M.; et al. Overexpression of GPX4 in diabetic rat kidney alleviates renal injury induced by ferroptosis. Biometals 2025, 38, 1281–1297. [Google Scholar] [CrossRef]
  72. Khamis, M.M.; Moselhy, S.S.; Rihan, S. Role of trans-resveratrol in ameliorating biochemical and molecular alterations in obese rats induced by a high fructose/fat diet. Sci. Rep. 2025, 15, 7879. [Google Scholar] [CrossRef]
  73. Sikur, N.; Böröczky, C.; Paszternák, A.; Gyöngyössy, R.; Szökő, É.; Varga, K.; Tábi, T. Resveratrol and its derivatives diminish lipid accumulation in adipocytes in vitro-mechanism of action and structure-activity relationship. Nutrients 2024, 16. [Google Scholar] [CrossRef]
  74. Gu, W.; Wang, X.; Zhao, H.; Geng, J.; Li, X.; Zheng, K.; Guan, Y.; Hou, X.; Wang, C.; Song, G. Resveratrol ameliorates diabetic kidney injury by reducing lipotoxicity and modulates expression of components of the junctional adhesion molecule-like/sirtuin 1 lipid metabolism pathway. Eur. J. Pharmacol. 2022, 918, 174776. [Google Scholar] [CrossRef] [PubMed]
  75. Kim, M.Y.; Lim, J.H.; Youn, H.H.; Hong, Y.A.; Yang, K.S.; Park, H.S.; Chung, S.; Ko, S.H.; Shin, S.J.; Choi, B.S.; et al. Resveratrol prevents renal lipotoxicity and inhibits mesangial cell glucotoxicity in a manner dependent on the AMPK-SIRT1-PGC1α axis in db/db mice. Diabetologia 2013, 56, 204–217. [Google Scholar] [CrossRef] [PubMed]
  76. Al-Hussaini, H.; Kilarkaje, N. Trans-resveratrol mitigates type 1 diabetes-induced oxidative DNA damage and accumulation of advanced glycation end products in glomeruli and tubules of rat kidneys. Toxicol. Appl. Pharmacol. 2018, 339, 97–109. [Google Scholar] [CrossRef] [PubMed]
  77. Witkin, J.M.; Li, X. Curcumin, an active constiuent of the ancient medicinal herb Curcuma longa L.: Some uses and the establishment and biological basis of medical efficacy. CNS Neurol. Disord. Drug Targets 2013, 12, 487–497. [Google Scholar] [CrossRef]
  78. Kim, B.H.; Lee, E.S.; Choi, R.; Nawaboot, J.; Lee, M.Y.; Lee, E.Y.; Kim, H.S.; Chung, C.H. Protective effects of curcumin on renal oxidative stress and lipid metabolism in a rat model of type 2 diabetic nephropathy. Yonsei Med. J. 2016, 57, 664–673. [Google Scholar] [CrossRef]
  79. Allemailem, K.S.; Almatroudi, A.; Alharbi, H.O.A.; AlSuhaymi, N.; Alsugoor, M.H.; Aldakheel, F.M.; Khan, A.A.; Rahmani, A.H. Apigenin: A bioflavonoid with a promising role in disease prevention and treatment. Biomedicines 2024, 12, 1353. [Google Scholar] [CrossRef]
  80. Wu, L.; Guo, T.; Deng, R.; Liu, L.; Yu, Y. Apigenin ameliorates insulin resistance and lipid accumulation by endoplasmic reticulum stress and SREBP-1c/SREBP-2 pathway in palmitate-induced HepG2 cells and high-fat diet-fed mice. J. Pharmacol. Exp. Ther. 2021, 377, 146–156. [Google Scholar] [CrossRef]
  81. Chen, X.; Tan, J.; Zhang, L.; Liu, Y.; Cheng, Y.; Zhang, Q.; Ding, H. Apigenin ameliorates vascular injury in rats with high fructose-induced metabolic disturbance by inhibiting PI3K/AKT/GLUT1. RSC Adv. 2018, 8, 24470–24476. [Google Scholar] [CrossRef] [PubMed]
  82. Mezei, O.; Banz, W.J.; Steger, R.W.; Peluso, M.R.; Winters, T.A.; Shay, N. Soy isoflavones exert antidiabetic and hypolipidemic effects through the PPAR pathways in obese Zucker rats and murine RAW 264.7 cells. J. Nutr. 2003, 133, 1238–1243. [Google Scholar] [CrossRef] [PubMed]
  83. Misiakiewicz-Has, K.; Maciejewska-Markiewicz, D.; Szypulska-Koziarska, D.; Kolasa, A.; Wiszniewska, B. The influence of soy isoflavones and soy isoflavones with inulin on kidney morphology, fatty acids, and associated parameters in rats with and without induced diabetes type 2. Int. J. Mol. Sci. 2024, 25, 5418. [Google Scholar] [CrossRef] [PubMed]
  84. Danielewski, M.; Matuszewska, A.; Szeląg, A.; Sozański, T. The impact of anthocyanins and iridoids on transcription factors crucial for lipid and cholesterol homeostasis. Int. J. Mol. Sci. 2021, 22, 6074. [Google Scholar] [CrossRef]
  85. Herrera-Balandrano, D.D.; Chai, Z.; Hutabarat, R.P.; Beta, T.; Feng, J.; Ma, K.; Li, D.; Huang, W. Hypoglycemic and hypolipidemic effects of blueberry anthocyanins by AMPK activation: In vitro and in vivo studies. Redox Biol. 2021, 46, 102100. [Google Scholar] [CrossRef]
  86. Kandasamy, N.; Ashokkumar, N. Renoprotective effect of myricetin restrains dyslipidemia and renal mesangial cell proliferation by the suppression of sterol regulatory element binding proteins in an experimental model of diabetic nephropathy. Eur. J. Pharmacol. 2014, 743, 53–62. [Google Scholar] [CrossRef]
  87. Parhiz, H.; Roohbakhsh, A.; Soltani, F.; Rezaee, R.; Iranshahi, M. Antioxidant and anti-inflammatory properties of the citrus flavonoids hesperidin and hesperetin: An updated review of their molecular mechanisms and experimental models. Phytother. Res. 2015, 29, 323–331. [Google Scholar] [CrossRef]
  88. Chen, Y.J.; Kong, L.; Tang, Z.Z.; Zhang, Y.M.; Liu, Y.; Wang, T.Y.; Liu, Y.W. Hesperetin ameliorates diabetic nephropathy in rats by activating Nrf2/ARE/glyoxalase 1 pathway. Biomed. Pharmacother. 2019, 111, 1166–1175. [Google Scholar] [CrossRef]
  89. Meenu; Bano, U.; Mujeeb, M.; Akhtar, M.; Aqil, M.; Haque, S.E.U.; Najmi, A.K. Hesperetin and isoliquiritigenin attenuate the progression of diabetic nephropathy via inhibition of NF-κB/NLRP3 pathways in type 2 diabetic rats. ACS Omega 2025, 10, 34342–34351. [Google Scholar] [CrossRef]
  90. Muñoz-Reyes, D.; Morales, A.I.; Prieto, M. Transit and metabolic pathways of quercetin in tubular cells: Involvement of its antioxidant properties in the kidney. Antioxidants 2021, 10, 909. [Google Scholar] [CrossRef]
  91. Feng, Q.; Yang, Y.; Qiao, Y.; Zheng, Y.; Yu, X.; Liu, F.; Wang, H.; Zheng, B.; Pan, S.; Ren, K.; et al. Quercetin ameliorates diabetic kidney injury by inhibiting ferroptosis via activating Nrf2/HO-1 signaling pathway. Am. J. Chin. Med. 2023, 51, 997–1018. [Google Scholar] [CrossRef] [PubMed]
  92. Rahmani, A.H.; Alsahli, M.A.; Khan, A.A.; Almatroodi, S.A. Quercetin, a plant flavonol attenuates diabetic complications, renal tissue damage, renal oxidative stress and inflammation in streptozotocin-induced diabetic rats. Metabolites 2023, 13, 130. [Google Scholar] [CrossRef] [PubMed]
  93. Shen, Q.; Fang, J.; Guo, H.; Su, X.; Zhu, B.; Yao, X.; Wang, Y.; Cao, A.; Wang, H.; Wang, L. Astragaloside IV attenuates podocyte apoptosis through ameliorating mitochondrial dysfunction by up-regulated Nrf2-ARE/TFAM signaling in diabetic kidney disease. Free Radic. Biol. Med. 2023, 203, 45–57. [Google Scholar] [CrossRef] [PubMed]
  94. Su, Y.; Chen, Q.; Ma, K.; Ju, Y.; Ji, T.; Wang, Z.; Li, W.; Li, W. Astragaloside IV inhibits palmitate-mediated oxidative stress and fibrosis in human glomerular mesangial cells via downregulation of CD36 expression. Pharmacol. Rep. 2019, 71, 319–329. [Google Scholar] [CrossRef]
  95. Zang, J.; Peters, F.; Cambet, Y.; Cifuentes-Pagano, E.; Hissabu, M.M.S.; Dustin, C.M.; Svensson, L.H.; Olesen, M.M.; Poulsen, M.F.L.; Jacobsen, S.; et al. Targeting NOX2 with bivalent small-molecule p47phox-p22phox inhibitors. J. Med. Chem. 2023, 66, 14963–15005. [Google Scholar] [CrossRef]
  96. Feng, H.; Zhu, X.; Tang, Y.; Fu, S.; Kong, B.; Liu, X. Astragaloside IV ameliorates diabetic nephropathy in db/db mice by inhibiting NLRP3 inflammasome-mediated inflammation. Int. J. Mol. Med. 2021, 48, 164. [Google Scholar] [CrossRef]
  97. Chen, C. Sinapic acid and its derivatives as medicine in oxidative stress-induced diseases and aging. Oxid. Med. Cell Longev. 2016, 2016, 3571614. [Google Scholar] [CrossRef]
  98. Alaofi, A.L. Sinapic acid ameliorates the progression of streptozotocin (STZ)-induced diabetic nephropathy in rats via NRF2/HO-1 mediated pathways. Front. Pharmacol. 2020, 11, 1119. [Google Scholar] [CrossRef]
  99. Wang, K.; Liang, C.; Cao, W.; Luo, G.; Zhong, S.; Zeng, Z.; Dai, L.; Song, J.L. Dietary sinapic acid attenuated high-fat diet-induced lipid metabolism and oxidative stress in male Syrian hamsters. J. Food Biochem. 2022, 46, e14203. [Google Scholar] [CrossRef]
  100. Bao, L.; Li, J.; Zha, D.; Zhang, L.; Gao, P.; Yao, T.; Wu, X. Chlorogenic acid prevents diabetic nephropathy by inhibiting oxidative stress and inflammation through modulation of the Nrf2/HO-1 and NF-ĸB pathways. Int. Immunopharmacol. 2018, 54, 245–253. [Google Scholar] [CrossRef]
  101. Bao, L.; Gong, Y.; Xu, W.; Dao, J.; Rao, J.; Yang, H. Chlorogenic acid inhibits NLRP3 inflammasome activation through Nrf2 activation in diabetic nephropathy. PLoS ONE 2025, 20, e0316615. [Google Scholar] [CrossRef] [PubMed]
  102. Gong, P.; Wang, P.; Pi, S.; Guo, Y.; Pei, S.; Yang, W.; Chang, X.; Wang, L.; Chen, F. Proanthocyanidins protect against cadmium-induced diabetic nephropathy through p38 MAPK and Keap1/Nrf2 signaling pathways. Front. Pharmacol. 2021, 12, 801048. [Google Scholar] [CrossRef] [PubMed]
  103. Peng, X.; Cheng, K.W.; Ma, J.; Chen, B.; Ho, C.T.; Lo, C.; Chen, F.; Wang, M. Cinnamon bark proanthocyanidins as reactive carbonyl scavengers to prevent the formation of advanced glycation endproducts. J. Agric. Food Chem. 2008, 56, 1907–1911. [Google Scholar] [CrossRef] [PubMed]
  104. Dusabimana, T.; Park, E.J.; Je, J.; Jeong, K.; Yun, S.P.; Kim, H.J.; Kim, H.; Park, S.W. Geniposide improves diabetic nephropathy by enhancing ULK1-mediated autophagy and reducing oxidative stress through AMPK activation. Int. J. Mol. Sci. 2021, 22, 1651. [Google Scholar] [CrossRef]
  105. Li, F.; Chen, Y.; Li, Y.; Huang, M.; Zhao, W. Geniposide alleviates diabetic nephropathy of mice through AMPK/SIRT1/NF-κB pathway. Eur. J. Pharmacol. 2020, 886, 173449. [Google Scholar] [CrossRef]
  106. Albrahim, T.; Robert, A.A. Lycopene effects on metabolic syndrome and kidney injury in rats fed a high-fat diet: An experimental study. ACS Omega 2022, 7, 30930–30938. [Google Scholar] [CrossRef]
  107. Figueiredo, I.D.; Lima, T.F.O.; Inácio, M.D.; Costa, M.C.; Assis, R.P.; Brunetti, I.L.; Baviera, A.M. Lycopene improves the metformin effects on glycemic control and decreases biomarkers of glycoxidative stress in diabetic rats. Diabetes Metab. Syndr. Obes. 2020, 13, 3117–3135. [Google Scholar] [CrossRef]
  108. Sen, Z.; Weida, W.; Jie, M.; Li, S.; Dongming, Z.; Xiaoguang, C. Coumarin glycosides from Hydrangea paniculata slow down the progression of diabetic nephropathy by targeting Nrf2 anti-oxidation and smad2/3-mediated profibrosis. Phytomedicine 2019, 57, 385–395. [Google Scholar] [CrossRef]
  109. Liu, M.; Xuan, A.; Zheng, L.; Li, D.; Chen, C.; Liu, H.; Lu, G.; Cheng, Z.; Zou, Y.; Zhi, S.; et al. Novel coumarin derivative SZC-6 as an allosteric activator of SIRT3 alleviates diabetic kidney disease via the SIRT3-Foxo3a signaling axis. Free Radic. Biol. Med. 2025, 240, 29–45. [Google Scholar] [CrossRef]
  110. Chen, Y.; Chen, L.; Yang, T. Silymarin nanoliposomes attenuate renal injury on diabetic nephropathy rats via co-suppressing TGF-β/Smad and JAK2/STAT3/SOCS1 pathway. Life Sci. 2021, 271, 119197. [Google Scholar] [CrossRef]
  111. Zhang, Y.; Jin, D.; Kang, X.; Zhou, R.; Sun, Y.; Lian, F.; Tong, X. Signaling pathways involved in diabetic renal fibrosis. Front. Cell Dev. Biol. 2021, 9, 696542. [Google Scholar] [CrossRef] [PubMed]
  112. Vessal, G.; Akmali, M.; Najafi, P.; Moein, M.R.; Sagheb, M.M. Silymarin and milk thistle extract may prevent the progression of diabetic nephropathy in streptozotocin-induced diabetic rats. Ren. Fail. 2010, 32, 733–739. [Google Scholar] [CrossRef] [PubMed]
  113. Feng, D.; Kumar, M.; Muntel, J.; Gurley, S.B.; Birrane, G.; Stillman, I.E.; Ding, L.; Wang, M.; Ahmed, S.; Schlondorff, J.; et al. Phosphorylation of ACTN4 leads to podocyte vulnerability and proteinuric glomerulosclerosis. J. Am. Soc. Nephrol. 2020, 31, 1479–1495. [Google Scholar] [CrossRef] [PubMed]
  114. Kato, M.; Natarajan, R. Epigenetics and epigenomics in diabetic kidney disease and metabolic memory. Nat. Rev. Nephrol. 2019, 15, 327–345. [Google Scholar] [CrossRef]
  115. He, C.; Wang, D.; Wang, R.; Huang, Y.; Huang, X.; Shen, S.; Lv, J.; Wu, M. Epigallocatechin gallate induces the demethylation of actinin alpha 4 to inhibit diabetic nephropathy renal fibrosis via the NF-KB signaling pathway in vitro. Dose Response 2022, 20, 15593258221105704. [Google Scholar] [CrossRef]
  116. Wang, Y.; Wang, Q.; Wang, M.; Wang, X.; Liu, Q.; Lv, S.; Nie, H.; Liu, G. Epigallocatechin-3-gallate ameliorates diabetic kidney disease by inhibiting the TXNIP/NLRP3/IL-1β signaling pathway. Food Sci. Nutr. 2024, 12, 10800–10815. [Google Scholar] [CrossRef]
  117. Skorokhod, O.; Barrera, V.; Mandili, G.; Costanza, F.; Valente, E.; Ulliers, D.; Schwarzer, E. Malaria pigment hemozoin impairs GM-CSF receptor expression and function by 4-hydroxynonenal. Antioxidants 2021, 10, 1259. [Google Scholar] [CrossRef]
  118. Achuthan, A.A.; Lee, K.M.C.; Hamilton, J.A. Targeting GM-CSF in inflammatory and autoimmune disorders. Semin. Immunol. 2021, 54, 101523. [Google Scholar] [CrossRef]
  119. Liu, Y.; Wang, W.; Zhang, J.; Gao, S.; Xu, T.; Yin, Y. JAK/STAT signaling in diabetic kidney disease. Front. Cell Dev. Biol. 2023, 11, 1233259. [Google Scholar] [CrossRef]
  120. Sun, L.; Yang, Z.; Zhang, J.; Wang, J. Isoliquiritigenin attenuates acute renal injury through suppressing oxidative stress, fibrosis and JAK2/STAT3 pathway in streptozotocin-induced diabetic rats. Bioengineered 2021, 12, 11188–11200. [Google Scholar] [CrossRef]
  121. Yin, Y.; He, J.; Fang, Y.; Wei, M.; Zhang, W. Andrographolide as a multi-target therapeutic agent in diabetic nephropathy: Insights into STAT3/PI3K/Akt pathway modulation. Biomol. Ther. 2025, 33, 529–543. [Google Scholar] [CrossRef] [PubMed]
  122. Ji, X.; Li, C.; Ou, Y.; Li, N.; Yuan, K.; Yang, G.; Chen, X.; Yang, Z.; Liu, B.; Cheung, W.W.; et al. Andrographolide ameliorates diabetic nephropathy by attenuating hyperglycemia-mediated renal oxidative stress and inflammation via Akt/NF-κB pathway. Mol. Cell. Endocrinol. 2016, 437, 268–279. [Google Scholar] [CrossRef]
  123. Ma, X.; Chen, Z.; Wang, L.; Wang, G.; Wang, Z.; Dong, X.; Wen, B.; Zhang, Z. The pathogenesis of diabetes mellitus by oxidative stress and inflammation: Its inhibition by berberine. Front. Pharmacol. 2018, 9, 782. [Google Scholar] [CrossRef]
  124. Wang, T.T.; Yu, L.L.; Zheng, J.M.; Han, X.Y.; Jin, B.Y.; Hua, C.J.; Chen, Y.S.; Shang, S.S.; Liang, Y.Z.; Wang, J.R. Berberine inhibits ferroptosis and stabilizes atherosclerotic plaque through NRF2/SLC7A11/GPX4 pathway. Chin. J. Integr. Med. 2024, 30, 906–916. [Google Scholar] [CrossRef]
  125. Chen, J.; Ou, Z.; Gao, T.; Yang, Y.; Shu, A.; Xu, H.; Chen, Y.; Lv, Z. Ginkgolide B alleviates oxidative stress and ferroptosis by inhibiting GPX4 ubiquitination to improve diabetic nephropathy. Biomed. Pharmacother. 2022, 156, 113953. [Google Scholar] [CrossRef]
  126. Liu, Y.; Wang, F.; Xu, H.; Wang, H.; Lu, M.; Cheng, L. Ginkgolide B attenuates hyperlipidemia by restoring sphingolipid homeostasis and activating PPARα and Nrf2 pathways. Sci. Rep. 2025, 15, 28774. [Google Scholar] [CrossRef] [PubMed]
  127. Wu, Q.; Guan, Y.B.; Zhang, K.J.; Li, L.; Zhou, Y. Tanshinone IIA mediates protection from diabetes kidney disease by inhibiting oxidative stress induced pyroptosis. J. Ethnopharmacol. 2023, 316, 116667. [Google Scholar] [CrossRef] [PubMed]
  128. Zhu, S.; Kang, Z.; Zhang, F. Tanshinone IIA suppresses ferroptosis to attenuate renal podocyte injury in diabetic nephropathy through the embryonic lethal abnormal visual-like protein 1 and acyl-coenzyme A synthetase long-chain family member 4 signaling pathway. J. Diabetes Investig. 2024, 15, 1003–1016. [Google Scholar] [CrossRef]
Figure 1. Lipid metabolism disorder in DKD. Hyperglycemia and insulin resistance in diabetes promote fatty acid synthesis and inhibit fatty acid oxidation, further esterifying to form triglycerides, leading to renal ectopic lipid deposition. Abbreviations: SREBP-1c, sterol regulatory element binding protein-1c; ChREBP, carbohydrate response element binding protein; ACC, acetyl-CoA carboxylase; FAS, fatty acid synthase; FFAs, free fatty acids; PPARa, peroxisome proliferator-activated receptor alpha; CPT1, carnitine palmitoyl transferase 1; TCA, tricarboxylic acid; ETC, electronic transfer chain; TG, triglycerides.
Figure 1. Lipid metabolism disorder in DKD. Hyperglycemia and insulin resistance in diabetes promote fatty acid synthesis and inhibit fatty acid oxidation, further esterifying to form triglycerides, leading to renal ectopic lipid deposition. Abbreviations: SREBP-1c, sterol regulatory element binding protein-1c; ChREBP, carbohydrate response element binding protein; ACC, acetyl-CoA carboxylase; FAS, fatty acid synthase; FFAs, free fatty acids; PPARa, peroxisome proliferator-activated receptor alpha; CPT1, carnitine palmitoyl transferase 1; TCA, tricarboxylic acid; ETC, electronic transfer chain; TG, triglycerides.
Ijms 26 09764 g001
Figure 2. Oxidative stress and lipid peroxidation in DKD. Oxidative stress serves as the key driver of lipid peroxidation. Hyperglycemia and renal ectopic lipid deposition impair mitochondrial function. Furthermore, advanced glycation end products (AGEs) produced by plasma glucose bind to the receptor for AGE (RAGE), along with alterations in cytochrome P450 (CYP) enzyme activity, exacerbating oxidative stress and generating excessive reactive oxygen species (ROS). Concurrently, impaired nuclear factor erythroid 2-related factor 2 (Nrf2) function in DKD disrupts the antioxidant defense system. The massive ROS triggers a chain reaction of non-enzymatic lipid peroxidation, promoting the production of lipid peroxides. Abbreviations: GLUT, glucose transporter; MPC, mitochondrial pyruvate carrier; NOX, NADPH oxidase; Keap1, kelch-like ECH-associated protein 1; ARE, antioxidant response element; PUFA, polyunsaturated fatty acid; PUFA-PE, polyunsaturated fatty acid-phosphatidyl ethanolamine; ACSL4, Acyl-CoA synthetase long chain family member 4; AA, arachidonic acid; 20-HETE, 20-hydroxyeicosatetraenoic acid.
Figure 2. Oxidative stress and lipid peroxidation in DKD. Oxidative stress serves as the key driver of lipid peroxidation. Hyperglycemia and renal ectopic lipid deposition impair mitochondrial function. Furthermore, advanced glycation end products (AGEs) produced by plasma glucose bind to the receptor for AGE (RAGE), along with alterations in cytochrome P450 (CYP) enzyme activity, exacerbating oxidative stress and generating excessive reactive oxygen species (ROS). Concurrently, impaired nuclear factor erythroid 2-related factor 2 (Nrf2) function in DKD disrupts the antioxidant defense system. The massive ROS triggers a chain reaction of non-enzymatic lipid peroxidation, promoting the production of lipid peroxides. Abbreviations: GLUT, glucose transporter; MPC, mitochondrial pyruvate carrier; NOX, NADPH oxidase; Keap1, kelch-like ECH-associated protein 1; ARE, antioxidant response element; PUFA, polyunsaturated fatty acid; PUFA-PE, polyunsaturated fatty acid-phosphatidyl ethanolamine; ACSL4, Acyl-CoA synthetase long chain family member 4; AA, arachidonic acid; 20-HETE, 20-hydroxyeicosatetraenoic acid.
Ijms 26 09764 g002
Figure 3. Lipid peroxide-mediated renal injury through (A) Pyroptosis, (B) Inflammation, and (C) Ferroptosis. Lipid peroxide (LPO) is a key mediator leading to renal damage. It exacerbates kidney injury by disrupting cell membrane structure, modifying protein function, and acting as a signaling molecule. LPO can activate the nuclear factor kappa-B (NF-κB) and NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome pathways, triggering caspase-1-mediated cleavage of GSDMD and promoting inflammatory responses and pyroptosis. Meanwhile, LPO inhibits the activity of glutathione peroxidase 4 (GPx4), weakening the antioxidant defense system. It also synergizes with intracellular iron overload to drive ferroptosis, a specific form of programmed cell death, thereby playing a central role in the progression of DKD. Abbreviations: PRR, pattern recognition receptors; GSDMD, gasdermin D; IKK, inhibitor of kappa B kinase; IκB, inhibitor of NF-κB; TFR, transferrin receptor; FTN, ferritin; GSH, glutathione.
Figure 3. Lipid peroxide-mediated renal injury through (A) Pyroptosis, (B) Inflammation, and (C) Ferroptosis. Lipid peroxide (LPO) is a key mediator leading to renal damage. It exacerbates kidney injury by disrupting cell membrane structure, modifying protein function, and acting as a signaling molecule. LPO can activate the nuclear factor kappa-B (NF-κB) and NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome pathways, triggering caspase-1-mediated cleavage of GSDMD and promoting inflammatory responses and pyroptosis. Meanwhile, LPO inhibits the activity of glutathione peroxidase 4 (GPx4), weakening the antioxidant defense system. It also synergizes with intracellular iron overload to drive ferroptosis, a specific form of programmed cell death, thereby playing a central role in the progression of DKD. Abbreviations: PRR, pattern recognition receptors; GSDMD, gasdermin D; IKK, inhibitor of kappa B kinase; IκB, inhibitor of NF-κB; TFR, transferrin receptor; FTN, ferritin; GSH, glutathione.
Ijms 26 09764 g003
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

Dong, Y.; Tong, Y. Lipid Peroxidation in Diabetic Kidney Disease: Mechanism and Natural Solution. Int. J. Mol. Sci. 2025, 26, 9764. https://doi.org/10.3390/ijms26199764

AMA Style

Dong Y, Tong Y. Lipid Peroxidation in Diabetic Kidney Disease: Mechanism and Natural Solution. International Journal of Molecular Sciences. 2025; 26(19):9764. https://doi.org/10.3390/ijms26199764

Chicago/Turabian Style

Dong, Yuxin, and Yanqing Tong. 2025. "Lipid Peroxidation in Diabetic Kidney Disease: Mechanism and Natural Solution" International Journal of Molecular Sciences 26, no. 19: 9764. https://doi.org/10.3390/ijms26199764

APA Style

Dong, Y., & Tong, Y. (2025). Lipid Peroxidation in Diabetic Kidney Disease: Mechanism and Natural Solution. International Journal of Molecular Sciences, 26(19), 9764. https://doi.org/10.3390/ijms26199764

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