Next Article in Journal
Comparison of 30 Cytokines in Human Breast Milk between 1989 and 2013 in Japan
Next Article in Special Issue
Redox Regulation in Age-Related Cataracts: Roles for Glutathione, Vitamin C, and the NRF2 Signaling Pathway
Previous Article in Journal
The Impact of Obesity on the Excretion of Steroid Metabolites in Boys and Girls: A Comparison with Normal-Weight Children
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Renal-Protective Roles of Lipoic Acid in Kidney Disease

1
Department of Pharmaceutical Sciences, College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
2
School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266071, China
3
Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(7), 1732; https://doi.org/10.3390/nu15071732
Submission received: 8 March 2023 / Revised: 27 March 2023 / Accepted: 30 March 2023 / Published: 1 April 2023
(This article belongs to the Special Issue Nutritional Regulation of Aging and Age-Related Diseases)

Abstract

:
The kidney is a crucial organ that eliminates metabolic waste and reabsorbs nutritious elements. It also participates in the regulation of blood pressure, maintenance of electrolyte balance and blood pH homeostasis, as well as erythropoiesis and vitamin D maturation. Due to such a heavy workload, the kidney is an energy-demanding organ and is constantly exposed to endogenous and exogenous insults, leading to the development of either acute kidney injury (AKI) or chronic kidney disease (CKD). Nevertheless, there are no therapeutic managements to treat AKI or CKD effectively. Therefore, novel therapeutic approaches for fighting kidney injury are urgently needed. This review article discusses the role of α-lipoic acid (ALA) in preventing and treating kidney diseases. We focus on various animal models of kidney injury by which the underlying renoprotective mechanisms of ALA have been unraveled. The animal models covered include diabetic nephropathy, sepsis-induced kidney injury, renal ischemic injury, unilateral ureteral obstruction, and kidney injuries induced by folic acid and metals such as cisplatin, cadmium, and iron. We highlight the common mechanisms of ALA’s renal protective actions that include decreasing oxidative damage, increasing antioxidant capacities, counteracting inflammation, mitigating renal fibrosis, and attenuating nephron cell death. It is by these mechanisms that ALA achieves its biological function of alleviating kidney injury and improving kidney function. Nevertheless, we also point out that more comprehensive, preclinical, and clinical studies will be needed to make ALA a better therapeutic agent for targeting kidney disorders.

1. Introduction

The kidney is a vital organ, participating in maintenance of electrolyte balance, blood pH stability, removal of metabolic waste, and reabsorption of nutrients and minerals [1,2]. The kidney is also involved in erythropoiesis, vitamin D maturation, and blood pressure regulation [3]. Under pathophysiological conditions such as fasting, long-term starvation, and insulin resistance, the kidney can also regenerate glucose via gluconeogenesis using precursor molecules such as glycerol, alanine, pyruvate, and lactate [4]. As such, the kidney always has a heavy workload and is thus exposed to numerous risk factors that can cause kidney disease or injuries. There are two types of kidney diseases: chronic kidney disease (CKD) [5] and acute kidney injury (AKI) [6]. CKD occurs when there’s a gradual decline in renal function for more than 3 months due to damage to glomerular filtration and tubular injuries [7]. It has been projected that by 2040, CKD will become the fifth leading cause of death worldwide [8]. On the other hand, AKI occurs when there’s a rapid decline in renal function for less than 3 months, and this is indicated by acidosis, fluid overload, and abnormalities with electrolytes and hematology changes [7]. It is also known that individuals with CKD can have an increased risk of AKI [8]. Currently, there are no pharmaceutical products to cure AKI or CKD. While renal dialysis is often used to prevent further damage to the kidneys and maintain their function, renal replacement may be the last resort for patient survival. Therefore, if uncontrolled, AKI and CKD can lead to kidney failure, which significantly increases morbidity and mortality [7,9]. Hence, there is an unmet need in fighting kidney disease.
To fight kidney diseases, numerous therapeutic approaches have been explored [10,11,12,13]. These include exogenous and endogenous compounds, dietary manipulations, modulation of metabolic pathways, stem cell approaches, and cell signaling processes [10,11,14,15,16,17,18,19,20,21]. In this review article, we will focus on the role of lipoic acid in preventing and ameliorating kidney injuries. In particular, we will focus on studies using animal models for exploring the protective effects of lipoic acid on kidney injuries. These animal models of kidney injury include diabetic kidney disease or diabetic nephropathy, renal ischemia-reperfusion injury, sepsis-induced kidney injury, unilateral ureteral obstruction (UUO)-induced kidney injury, cisplatin-induced kidney injury, cadmium-induced kidney injury, folic acid-induced kidney injury, and iron-induced acute kidney injury (Figure 1).

2. Alpha-Lipoic Acid

ALA is a naturally occurring dithiol compound [22]. It is a cofactor for α-ketoglutarate dehydrogenase, branched-chain amino acid dehydrogenase, and pyruvate dehydrogenase [23,24,25] (Figure 2). Therefore, ALA is an energy modulator [26,27] (Figure 3). Moreover, due to its capability of exchanging thiol groups with other thiol-containing molecules, such as glutathione and protein’s cysteine residues, ALA is also known as a redox modulator [26,27,28] (Figure 3). ALA is often referred to as a universal antioxidant because it can act as an antioxidant in both lipophilic and hydrophilic settings to reduce byproducts of oxidative metabolism, such as reactive oxygen species (ROS) and reactive nitrogen species (RNS) (Figure 4) [29]. ALA can also chelate metals such as zinc, iron, and copper and regenerate endogenous antioxidants—such as glutathione—and exogenous vitamin antioxidants—such as vitamins C and E—with minimal side effects [30] (Figure 4). More importantly, ALA can also inhibit inflammation by targeting NF-KB and decreasing the release of inflammatory cytokines (Figure 4). Therefore, it has been demonstrated that by scavenging oxygen free radicals, ALA could increase glomerular function and decrease renal inflammation [31]. Studies have also shown that treatment with ALA can decrease acute kidney injury by lowering serum blood urea nitrogen, creatinine levels, tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1 beta (IL-1β), thereby decreasing endothelin-1 vasoconstriction, neutrophil diffusion, and inflammation in the kidneys.

3. Protective Roles of α-Lipoic Acid (ALA) in Kidney Injury

Kidney disorders can be induced by a number of insults, such as diabetes, ischemic reperfusion, drug toxicity, contrast media, and medications [7]. If not controlled, AKI can lead to renal failure with a 20% mortality rate [25]. AKI is characterized by an increase in serum creatinine, oliguria, and the presence of kidney damage markers such as albuminuria, electrolyte abnormalities due to tubular disorders, or structural damage seen in imaging or histology [9]. ALA prophylaxis has been shown to decrease renal tubular injury scores, urinary damage markers, serum creatinine structural damage, and increase glomerular filtration [35]. In the following sections, we will discuss the nephroprotective effects and the underlying protective mechanisms of ALA in a variety of animal models of kidney injury, as indicated in Figure 1.

3.1. Diabetic Nephropathy

Diabetic nephropathy (DN), also known as diabetic kidney disease (DKD) [36,37,38], is a major cause of CKD and end-stage kidney failure in diabetic patients [39,40,41]. It has been well-established that mitochondrial dysfunction contributes to DKD, and the mitochondrion is a target for fighting DKD [42,43,44]. DN can be considered a microvascular complication from type 1 or type 2 diabetes mellitus [7,41]. This diabetic kidney disease is characterized by a decrease in glomerular filtration, proteinuria, and renal fibrosis [7]. Hyperglycemia increases oxidative stress, leading to the early overproduction of reactive oxygen species (ROS) [45] and dysregulation of glutathione metabolic pathways [7]. Malondialdehyde (MDA) is the end product of lipid peroxidation and has been commonly used as a good marker of free radicals and oxidative stress [46,47,48,49]. Studies have shown that pretreatment with ALA decreased MDA content and ameliorated renal oxidative stress [32,50]. Lipoamide, a derivative of ALA, has been demonstrated to inhibit kidney fibrosis in diabetes by enhancing mitochondrial function and regulating the expression of activation of transcriptional factor retinal X receptor alpha [51]. In a rat model of diabetes induced by nicotinamide together with streptozotocin, a less time-consuming approach in creating rodent models of diabetes [52], Dugbartey et al. have also demonstrated that ALA’s renal protective mechanism involves activation of the renal cystathionine γ-lyase/hydrogen sulfide system [53]. It has also been demonstrated that ALA has a synergistic effect on attenuating serum levels of inflammatory cytokines and improving kidney function in diabetic animals when combined with the angiotensin II receptor inhibitor valsartan [54]. Figure 5 shows compelling evidence of histological staining that ALA exhibits strong protective effects on diabetic kidneys in a type 2 diabetes animal model [55]. Another potential mechanism of ALA’s nephroprotection in diabetes is its ability to activate the Nrf2 signaling pathway, leading to upregulation of the second-phase cytoprotective proteins such as heme oxygenase-1 (HO-1) and NAD(P)H quinone dehydrogenase 1 (NQO1) [56,57,58]. It should be pointed out that while ALA is nephroprotective in diabetic kidney disease, it may impose pro-oxidant or toxic effects and could fail to serve as an Nrf2 inducer under certain pathophysiological conditions [59,60,61]. Additionally, while ALA has been thought to activate insulin signaling pathways to combat diabetes [28] and has been shown to prevent high fructose-induced cardiometabolic disorders and renal dysfunction [62], it has also been reported that ALA could only attenuate proteinuria and oxidative stress without slowing progression of diabetic renal failure [63].

3.2. Sepsis-Induced Kidney Injury

Sepsis is potentially a life-threatening pathological condition due to an overdriving inflammatory response to bacterial infection [64,65,66]. Severe sepsis can cause multi-organ failures, with the kidney being the most-affected organ [67,68]. It has been estimated that nearly 50% of septic patients would develop acute kidney injury, and there are no effective treatments for septic AKI [69]. In this regard, numerous investigators have comprehensively evaluated ALA’s protective and therapeutic values in sepsis-induced AKI modeled by injection of lipopolysaccharides, though cacal puncture and ligation (CPL)-induced septic AKI have been occasionally used [22,70]. It has been demonstrated that ALA can protect against septic kidney injury by enhancing autophagy [71]. Moreover, ALA may also ameliorate sepsis-induced AKI by counteracting inflammation via inhibition of the NF-KB signaling pathway [72], attenuating mitochondrial oxidative stress, and preserving the type 3 Na+/H+ exchanger and aquaporin 2 expression in the kidney [73]. ALA can also inhibit the release of tumor necrosis factor-α, interleukin (IL)-6, and IL-1β into the serum and suppress the expression of inducible nitric oxide synthase [22] in septic AKI. Therefore, ALA could be a promising natural product for the treatment of septic AKI.

3.3. Renal Ischemic Reperfusion

Ischemia occurs when there is a decrease in blood perfusion and the blood flow to organs is reduced [74]; this is due to many causes, such as thrombi, trauma, and atherosclerosis [75]. To prevent tissue damage and necrosis, ischemia is resolved by reperfusion [75]. Although ischemic reperfusion is essential to prevent tissue necrosis, it can also cause inflammation and an increase in reactive oxygen species and reactive nitrogen species [75,76,77]. Studies have shown that pretreatment of ALA can ameliorate damage to the kidneys, retina, nervous system, liver, intestines, and more [75]. The underlying protective mechanisms of ALA in renal ischemia reperfusion injury may be multifactorial, including counteracting oxidative damage [32,75] and downregulation of channels, enzymes, and transporters, such as aquaporins and sodium transporters, as well as sodium-potassium ATPase and nitric oxide synthase isoforms [78]. ALA may also protect renal ischemia reperfusion injury by mitigating neutrophil infiltration and inhibiting the release of inflammatory mediators [79]. Studies have also demonstrated that renal cortical structure damage induced by limb ischemia-reperfusion injury can be ameliorated by ALA [80] and ALA, and when combined with the xanthine oxidase inhibitor febuxostat, shows superior protective effects on renal ischemia reperfusion injury [81]. ALA may also prevent renal dysfunction and kidney injury by suppressing the overexpression of endothelin-1 in renal ischemia reperfusion injury [82].

3.4. Unilateral Ureteral Obstruction (UUO)-Induced Kidney Injury

The unilateral ureteral obstruction (UUO) animal model of kidney injury has been widely used to investigate the mechanisms of kidney injury and the therapeutic values of numerous agents [83,84,85,86,87]. This model has certain advantages in that it is a nonuremic normotensive disorder in the absence of any apparent inflammatory or toxic insult to the kidneys [88]. Moreover, the UUO model of kidney injury is also a good model for studying the pathophysiology of renal fibrosis [89,90,91,92,93]. Therefore, the UUO model of kidney injury may closely mimic the underlying pathophysiology of human obstructive kidney injury [94,95]. ALA has been shown to be renoprotective against UUO-induced kidney injury [88]. Wongmekiat et al. have found that when ALA (60/mg/kg body weight) was given to rats via i.p. injection two days before UUO induction and continued for a week after UUO, UUO-induced renal dysfunction, oxidative stress, and production of nitric oxide and transforming factor-1 were greatly attenuated by ALA treatment [88]. Additionally, ALA has also been demonstrated to ameliorate the epithelial-mesenchymal transition in a mouse model of UUO renal injury [96]. These studies thus demonstrate that ALA is nephroprotective against UUO-induced kidney injury.

3.5. Cisplatin-Induced Nephrotoxicity

Cisplatin is a chemotherapy drug that treats cancer by entering the tumor cells, releasing chloride ions, and becoming hydrated to crosslink with DNA and form DNA adducts to inhibit tumor cell replication [97,98]. However, cisplatin medication has many side effects, such as ototoxicity, neurotoxicity, nephrotoxicity, nausea, and vomiting [35,97,98,99]. Nephrotoxicity occurs because cisplatin is taken up by the proximal tubular cells and can concentrate up to five times more than the cisplatin in the serum [35,97]. Cisplatin nephrotoxicity leads to decreased creatinine clearance, increased serum creatinine levels, increased urea levels, increased urine output, and decreased glomerular filtration rate [100,101]. Cisplatin also decreases the antioxidants glutathione S-transferase, glutathione peroxidase, and superoxide dismutase, causing an elevation in ROS and oxidative markers such as MDA derived from lipid peroxidation [35,100,102].
Proximal tubules in the kidneys contain large amounts of mitochondria [35]. Hydrolyzed cisplatin creates a metabolite with a positive charge, and this accumulates within mitochondria due to mitochondria’s negatively charged molecules, creating high levels of oxidative stress [35]. To manage this, mitochondria use endogenous antioxidants such as lipoic acid to reduce ROS [35]. Studies have shown that ALA protects renal cells against cisplatin’s toxicity [98], decreases structural proximal tubular damage, and increases glomerular filtration in kidneys [99,103]. ALA has also been shown to lower plasma creatinine levels and urine output, increase creatinine clearance and urine osmolality, and normalize sodium excretion in cisplatin kidney injury [101].

3.6. Folic Acid-Induced Nephrotoxicity

High levels of folic acid can cause tubular damage by the detachment and dilation of tubular cells [104], leading to cell death [105]. Ferroptosis is a type of cell death caused by iron and lipid peroxidation that occurs during folic acid-induced AKI [106]. When large amounts of iron are present in the body, ROS are generated, causing lipid peroxidation, damaging the lipid membranes, and causing cell death [105]. It has been established that ALA’s antioxidant effects resulted in renoprotection against folic acid-induced kidney damage [105]. In the study by Li et al., there was no significant difference between low doses and high doses of ALA, indicating that the benefits of ALA are not dose-dependent [105]. Furthermore, ALA may work by blocking p53 from causing ferroptosis and by upregulating ferritin and ferroportin iron exporters, thus preventing folic acid-induced AKI [105]. Figure 6 shows histologically the visualization of ALA protection against renal injury induced by folic acid [105].

3.7. Cadmium-Induced Nephrotoxicity

Cadmium is a naturally occurring toxic heavy metal that causes nephrotoxicity [104,107,108,109]. Cadmium-induced nephrotoxicity increases MDA levels, causing damage to renal mitochondria and the renal cortex [110]. Cadmium has also been shown to decrease glutathione antioxidant (GSH), glutathione peroxidase, catalase, and superoxide dismutase (SOD) [110]. Treatment with ALA functioned as an antioxidant to decrease MDA and apoptosis and chelate cadmium to lower cadmium damage to the kidneys [111]. ALA also lowered oxidative stress, promoted glutathione-related endogenous enzymes, and prevented mitochondrial apoptosis in cadmium-induced kidney injury [110,111,112].

3.8. Iron-Induced Acute Kidney Injury

Iron has been thought to contribute to both AKI and CKD [113,114,115,116,117,118]. Indeed, it has been observed that renal tubules are exposed to elevated levels of iron in patients with kidney disease, which is likely due to increased filtration of iron and iron-containing proteins through the glomerular apparatus [114,117,119]. Iron can also aggravate diabetic kidney disease by increasing oxidative stress [120,121]. Therefore, iron-induced animal models of kidney injury have been a valuable tool in studying the mechanisms of iron-induced kidney injury and testing the therapeutic effects of natural products or pharmaceutical drugs [122,123,124,125]. In this regard, ALA has been shown to be nephroprotective in ion-induced kidney injury [126,127]. In an iron-overloading rat model of kidney injury, ALA was found to exhibit antioxidant effects by attenuating oxidative damage [128]. ALA was also found to inhibit p38 MAPK signaling and NADPH oxidase 4 expression in iron-induced kidney injury [127]. It should be noted that all of these protective effects of ALA in iron-induced kidney injury may be partly due to its iron-chelating property [128,129,130,131], which lowers the availability of free iron.

4. Miscellaneous

1. Aging is linked to functional decline in the kidneys [132,133]. It has been established that there are numerous changes in the molecular, structural, and morphological levels in the kidney [134,135]. In aging kidneys, there are increased levels of oxidative stress, as reflected by increased lipid peroxidation, mitochondrial dysfunction, and decreased levels of antioxidants, including superoxide dismutase, catalase, and glutathione peroxidase [136,137,138,139]. Not surprisingly, ALA could alleviate all of these deleterious changes in aged kidneys [136,137,138]. Additionally, ALA given as a dietary supplement has been shown to be able to reverse age-related decline in kidney function and serum total proteins [140]. These studies demonstrate the protective and preventative effects of ALA on kidney aging.
2. Sleep apnea is a disorder that causes intermittent hypoxia, which can further cause hypoxia-associated renal injury [141,142,143,144]. ALA has been found to be protective against renal injury induced by sleep apnea hypoxia [145]. In a study using a mouse model of sleep apnea, Abuyassin et al. have demonstrated that in the animals that underwent intermittent hypoxia and were treated with an ALA-enhanced diet, renal oxidative stress and inflammation were lower than those exposed to intermittent hypoxia only. Moreover, renal cell death and tubular injury were also deceased in the intermittent hypoxia + ALA group, and treatment with ALA mitigated intermittent hypoxia-induced glomerular hypertrophy and decreased albuminuria [145]. Therefore, ALA is nephroprotective in hypoxia-related kidney injury induced by sleep apnea.
3. Functional impairment of the kidneys is also a frequent disorder in the presence of high blood pressure, also known as hypertension. It has been thought that renal damage linked to hypertension is caused by oxidative stress [146,147,148]. In this regard, ALA has also been tested for its antioxidant role in hypertension-induced kidney injury. Martinelli et al. [149] have found that when spontaneous hypertensive rats with high blood pressure were treated by a racemic mixture of ALA, renal oxidative damage was attenuated, with a significant improvement in kidney function accompanied by ameliorated glomerular and tubular injury. This study also indicates that when exogenous ALA is administered, a racemic mixture of ALA is often used [149].
4. Chronic kidney disease (CKD) can be created by feeding animals with a high concentration of adenine [150,151,152]. In fact, adenine-induced CKD is a popular model for studying the pathophysiology of chronic kidney injury and the therapeutic effects of a variety of natural products or drugs [153,154,155]. Nonetheless, the nephroprotective role of ALA has not been comprehensively evaluated in this animal model, which should be investigated in the future. Additionally, the protective effects of ALA on each of the five stages of chronic kidney disease [156,157,158] should also be studied in this animal model of CKD.
5. IgA nephropathy (IgAN) is known to cause glomerulonephritis due to deposition of IgA 1 [159,160,161] and is a prevalent chronic kidney disease [162,163,164]. Its major feature is mesangial cells and mononuclear leukocyte infiltration in renal interstitial tissues and the glomerulus. It has been demonstrated that oxidative stress-induced protein oxidation and lipid peroxidation is one of the underlying pathogenic mechanisms [165,166,167,168,169]. While animal models of IgAN are available for studying the pathogenesis of IgAN and exploring therapeutic approaches [170,171], the potential effects of ALA in this kidney disorder have not been evaluated. Nonetheless, it is conceivable that ALA would exhibit nephroprotective effects in IgAN, given its powerful antioxidant capacity.
6. It is also worth noting that studies comparing the renoprotective effect of ALA with other agents have also been conducted in recent years. For example, ALA was compared with a traditional Chinese medicine Huangkui capsule in rats with diabetic nephropathy [172]. The authors found that ALA is equivalent to the Huangkui capsule in renoprotection against diabetic kidney injury, and both agents improve kidney function by attenuating oxidative stress and downregulating the activation of the p38MAPK and Akt pathways. ALA has also been compared with N-acetylcysteine (NAC) in one study, whereby the authors found that NAC is better than ALA in protecting oxidative kidney injury induced by the chemotherapeutic drug ifosfamide, which is highly toxic to the kidney [173,174,175,176]. However, the authors used an NAC concentration (200 mg/kg) that was twice that of ALA (100 mg/kg) [176]. Hence, the conclusion that NAC is more renoprotective than ALA in this animal model of kidney injury may not be definitive. It should also be noted that a recent report indicates that ALA can also minimize renal toxicity induced by gold nanoparticles, which are often used as drug carriers [177]. In fact, poly(lipoic acid) nanoparticles themselves can also be used as a therapeutic tool for delivering active compounds [178].

5. Summary

In this article, we have reviewed the protective mechanisms of ALA in various animal models of kidney injury. These models cover both AKI and CKD, which include DKD, ischemia-reperfusion-induced kidney injury, sepsis-induced kidney injury, and kidney disorders induced by UUO, cisplatin, cadmium, folic acid, and iron. Common underlying mechanisms of ALA’s renoprotection are summarized in Figure 7.
As further summarized in Table 1, these mechanisms include decreasing oxidative stress, increasing endogenous antioxidant defense capacities, counteracting inflammation by inhibiting NF-kB and release of inflammatory cytokines, mitigating renal fibrosis, and decreasing cell death, such as apoptosis, ferroptosis, and necrosis. The eventual outcome of ALA treatment, regardless of the kidney injury models and the protective mechanisms unraveled, is the improvement of kidney function. Therefore, ALA is a promising agent targeting kidney disorders.

6. Future Perspectives

Most studies discussed in this review article utilized systemic administration of ALA in their studies of the protective effects of ALA on the respective animal models of kidney injury. Systemic administration of ALA will certainly result in distribution of ALA to the organs or tissues that may not need ALA, whereby excess ALA may pose deleterious effects [59]. Therefore, future studies will need to focus on developing approaches by which ALA will only be delivered to the kidneys. Such studies of target delivery of ALA to the kidney will certainly provide more insights into the protective mechanisms of ALA in different kidney injury models, elucidating both redox- and energy-modulatory properties of ALA. In this regard, nanoparticle delivery or nanomedicines of ALA targeting the kidney could be a promising approach [179,180,181,182,183,184]. Additionally, the combination of ALA with other natural products in treating kidney injuries and preventing the AKI to CKD transition will be interesting to investigate. Moreover, comprehensive preclinical and human studies are needed to evaluate the efficacy of ALA in the settings of AKI and CKD, as well as the AKI to CKD transition.

Author Contributions

Conceptualization, S.F.K. and L.-J.Y.; original draft preparation, S.F.K.; review and editing, J.L. and L.-J.Y. All authors have read and agreed to the published version of the manuscript.

Funding

Jiankang Liu was supported by the National Natural Science Foundation of China Integrated Project of the Major Research Plan 92249303 and General Projects 32171102 and 31770917.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rennke, H.G.; Denker, B.M. Renal Pathology: The Essentials, 5th ed.; Wolters Kluwer: New York, NY, USA, 2020. [Google Scholar]
  2. Koeppen, B.M.; Stanton, B.A. Renal Physiology, 5th ed.; Elsevier: Philadelphia, PA, USA, 2013. [Google Scholar]
  3. Dipiro, J.T.; Talbet, R.L.; Yee, G.C.; Matzke, G.R.; Wells, B.G.; Posey, L.M. Pharmacotherapy: A Pathophysiological Approach, 9th ed.; McGraw-Hill Education: New York, NY, USA, 2014. [Google Scholar]
  4. Lieberman, M.; Marks, A.D. Marks’ Basic Medical Biochemistry: A Clinical Approach, 4th ed.; Wolters Kluwer/Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2013. [Google Scholar]
  5. Ammirati, A.L. Chronic kidney disease. Rev. Assoc. Med. Bras. (1992) 2020, 66 (Suppl. S1), s03–s09. [Google Scholar] [CrossRef] [PubMed]
  6. Turgut, F.; Awad, A.S.; Abdel-Rahman, E.M. Acute kidney injury: Medical causes and pathogenesis. J. Clin. Med. 2023, 12, 375. [Google Scholar] [CrossRef] [PubMed]
  7. Duann, P.; Lin, P.H. Mitochondria damage and kidney disease. Adv. Exp. Med. Biol. 2017, 982, 529–551. [Google Scholar] [PubMed]
  8. Sanz, A.B.; Sanchez-Nino, M.D.; Ramos, A.M.; Ortiz, A. Regulated cell death pathways in kidney disease. Nat. Rev. Nephrol. 2023, 1–19. [Google Scholar] [CrossRef]
  9. Levey, A.S.; Becker, C.; Inker, L.A. Glomerular filtration rate and albuminuria for detection and staging of acute and chronic kidney disease in adults: A systematic review. JAMA 2015, 313, 837–846. [Google Scholar] [CrossRef] [Green Version]
  10. Mima, A. A narrative review of diabetic kidney disease: Previous and current evidence-based therapeutic approaches. Adv. Ther. 2022, 39, 3488–3500. [Google Scholar] [CrossRef]
  11. Mima, A. Mitochondria-targeted drugs for diabetic kidney disease. Heliyon 2022, 8, e08878. [Google Scholar] [CrossRef]
  12. Bhatia, D.; Capili, A.; Choi, M.E. Mitochondrial dysfunction in kidney injury, inflammation, and disease: Potential therapeutic approaches. Kidney Res. Clin. Pract. 2020, 39, 244–258. [Google Scholar] [CrossRef]
  13. Bhatia, D.; Choi, M.E. Autophagy in kidney disease: Advances and therapeutic potential. Prog. Mol. Biol. Transl. Sci. 2020, 172, 107–133. [Google Scholar]
  14. Chen, J.H.; Wu, C.H.; Chiang, C.K. Therapeutic approaches targeting proteostasis in kidney disease and fibrosis. Int. J. Mol. Sci. 2021, 22, 8674. [Google Scholar] [CrossRef]
  15. Tanase, D.M.; Gosav, E.M.; Anton, M.I.; Floria, M.; Seritean Isac, P.N.; Hurjui, L.L.; Tarniceriu, C.C.; Costea, C.F.; Ciocoiu, M.; Rezus, C. Oxidative stress and nrf2/keap1/are pathway in diabetic kidney disease (dkd): New perspectives. Biomolecules 2022, 12, 1227. [Google Scholar] [CrossRef] [PubMed]
  16. Swaminathan, S. Iron homeostasis pathways as therapeutic targets in acute kidney injury. Nephron 2018, 140, 156–159. [Google Scholar] [CrossRef]
  17. Pieretti, J.C.; Junho, C.V.C.; Carneiro-Ramos, M.S.; Seabra, A.B. H2S- and NO-releasing gasotransmitter platform: A crosstalk signaling pathway in the treatment of acute kidney injury. Pharmacol. Res. 2020, 161, 105121. [Google Scholar] [CrossRef] [PubMed]
  18. Ali, A.; Lima Sampaio, T.; Khan, H.; Jeandet, P.; Kupeli Akkol, E.; Bahadar, H.; Costa Martins, A.M. Plants with therapeutic potential for ischemic acute kidney injury: A systematic review. Evid. Based Complement. Altern. Med. 2022, 2022, 6807700. [Google Scholar] [CrossRef] [PubMed]
  19. Zhao, S.; Wu, W.; Liao, J.; Zhang, X.; Shen, M.; Li, X.; Lin, Q.; Cao, C. Molecular mechanisms underlying the renal protective effects of coenzyme q10 in acute kidney injury. Cell. Mol. Biol. Lett. 2022, 27, 57. [Google Scholar] [CrossRef] [PubMed]
  20. Liu, Y.; Han, J.; Fang, J.; Li, R. The beneficial effects of mesenchymal stem cells in acute kidney injury: A narrative review. Curr. Stem Cell Res. Ther. 2023. [Google Scholar] [CrossRef] [PubMed]
  21. Singh, G.; Krishan, P. Dietary restriction regimens for fighting kidney disease: Insights from rodent studies. Exp. Gerontol. 2019, 128, 110738. [Google Scholar] [CrossRef]
  22. Li, G.; Gao, L.; Jia, J.; Gong, X.; Zang, B.; Chen, W. α-Lipoic acid prolongs survival and attenuates acute kidney injury in a rat model of sepsis. Clin. Exp. Pharmacol. Physiol. 2014, 41, 459–468. [Google Scholar] [CrossRef]
  23. Yan, L.J.; Thangthaeng, N.; Sumien, N.; Forster, M.J. Serum dihydrolipoamide dehydrogenase is a labile enzyme. J. Biochem. Pharmacol. Res. 2013, 1, 30–42. [Google Scholar]
  24. Yang, X.; Song, J.; Yan, L.J. Chronic inhibition of mitochondrial dihydrolipoamide dehydrogenase (dldh) as an approach to managing diabetic oxidative stress. Antioxidants 2019, 8, 32. [Google Scholar] [CrossRef] [Green Version]
  25. Zaazaa, A.M.; Motelp, B.; Aniss, N.N. Potential protective role of rutin and alpha-lipoic acid against cisplatin-induced nephrotoxicity in rats. Pak. J. Biol. Sci. 2019, 22, 361–371. [Google Scholar] [CrossRef] [PubMed]
  26. Packer, L.; Cadenas, E. Lipoic acid: Energy metabolism and redox regulation of transcription and cell signaling. J. Clin. Biochem. Nutr. 2011, 48, 26–32. [Google Scholar] [CrossRef] [Green Version]
  27. Solmonson, A.; DeBerardinis, R.J. Lipoic acid metabolism and mitochondrial redox regulation. J. Biol. Chem. 2018, 293, 7522–7530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Rochette, L.; Ghibu, S.; Muresan, A.; Vergely, C. Alpha-lipoic acid: Molecular mechanisms and therapeutic potential in diabetes. Can. J. Physiol. Pharmacol. 2015, 93, 1021–1027. [Google Scholar] [CrossRef] [PubMed]
  29. Khalifa, E.A.; Nabil Ahmed, A.; Hashem, K.S.; Allah, A.G. Therapeutic effects of the combination of alpha-lipoic acid (ala) and coenzyme q10 (coq10) on cisplatin-induced nephrotoxicity. Int. J. Inflam. 2020, 2020, 5369797. [Google Scholar] [CrossRef]
  30. Tong, Z.J.; Kuo, C.W.; Yen, P.C.; Lin, C.C.; Tsai, M.T.; Lu, S.H.; Chang, Y.P.; Liu, W.S.; Tsou, H.H.; Cheng, H.W.; et al. Acrolein plays a culprit role in the pathogenesis of diabetic nephropathy in vitro and in vivo. Eur. J. Endocrinol. 2022, 187, 579–592. [Google Scholar] [CrossRef]
  31. Gao, W.W.; Chun, S.Y.; Kim, B.S.; Ha, Y.S.; Lee, J.N.; Lee, E.H.; Kim, I.Y.; You, S.; Kwon, T.G. Locally transplanted human urine-induced nephron progenitor cells contribute to renal repair in mice kidney with diabetic nephropathy. Biochem. Biophys. Res. Commun. 2022, 629, 128–134. [Google Scholar] [CrossRef]
  32. Wu, J.; Li, R.; Li, W.; Ren, M.; Thangthaeng, N.; Sumien, N.; Liu, R.; Yang, S.; Simpkins, J.W.; Forster, M.J.; et al. Administration of 5-methoxyindole-2-carboxylic acid that potentially targets mitochondrial dihydrolipoamide dehydrogenase confers cerebral preconditioning against ischemic stroke injury. Free Radic. Biol. Med. 2017, 113, 244–254. [Google Scholar] [CrossRef] [Green Version]
  33. Yan, L.J.; Thangthaeng, N.; Forster, M.J. Changes in dihydrolipoamide dehydrogenase expression and activity during postnatal development and aging in the rat brain. Mech. Ageing Dev. 2008, 129, 282–290. [Google Scholar] [CrossRef] [Green Version]
  34. Yan, L.J.; Yang, S.H.; Shu, H.; Prokai, L.; Forster, M.J. Histochemical staining and quantification of dihydrolipoamide dehydrogenase diaphorase activity using blue native page. Electrophoresis 2007, 28, 1036–1045. [Google Scholar] [CrossRef]
  35. Zhang, J.; McCullough, P.A. Lipoic acid in the prevention of acute kidney injury. Nephron 2016, 134, 133–140. [Google Scholar] [CrossRef] [PubMed]
  36. Oktan, M.A.; Heybeli, C.; Ural, C.; Kocak, A.; Bilici, G.; Cavdar, Z.; Ozbal, S.; Arslan, S.; Yilmaz, O.; Cavdar, C. Alpha-lipoic acid alleviates colistin nephrotoxicity in rats. Hum. Exp. Toxicol. 2021, 40, 761–771. [Google Scholar] [CrossRef] [PubMed]
  37. Chen, H.; Busse, L.W. Novel therapies for acute kidney injury. Kidney Int. Rep. 2017, 2, 785–799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Putra, I.; Fakhrudin, N.; Nurrochmad, A.; Wahyuono, S. A review of medicinal plants with renoprotective activity in diabetic nephropathy animal models. Life 2023, 13, 560. [Google Scholar] [CrossRef] [PubMed]
  39. Xu, J.; Kitada, M.; Koya, D. NAD+ homeostasis in diabetic kidney disease. Front. Med. 2021, 8, 703076. [Google Scholar] [CrossRef] [PubMed]
  40. Ahmad, A.A.; Draves, S.O.; Rosca, M. Mitochondria in diabetic kidney disease. Cells 2021, 10, 2945. [Google Scholar] [CrossRef]
  41. Yan, L.J. Nadh/NAD+ redox imbalance and diabetic kidney disease. Biomolecules 2021, 11, 730. [Google Scholar] [CrossRef]
  42. Song, Y.; Yu, H.; Sun, Q.; Pei, F.; Xia, Q.; Gao, Z.; Li, X. Grape seed proanthocyanidin extract targets p66shc to regulate mitochondrial biogenesis and dynamics in diabetic kidney disease. Front. Pharmacol. 2022, 13, 1035755. [Google Scholar] [CrossRef]
  43. Cleveland, K.H.; Schnellmann, R.G. Pharmacological targeting of mitochondria in diabetic kidney disease. Pharmacol. Rev. 2023, 75, 250–262. [Google Scholar] [CrossRef]
  44. Mohandes, S.; Doke, T.; Hu, H.; Mukhi, D.; Dhillon, P.; Susztak, K. Molecular pathways that drive diabetic kidney disease. J. Clin. Invest. 2023, 133, e165654. [Google Scholar] [CrossRef]
  45. Obrosova, I.G.; Fathallah, L.; Liu, E.; Nourooz-Zadeh, J. Early oxidative stress in the diabetic kidney: Effect of dl-alpha-lipoic acid. Free Radic. Biol. Med. 2003, 34, 186–195. [Google Scholar] [CrossRef] [PubMed]
  46. Pop, C.; Stefan, M.G.; Muntean, D.M.; Stoicescu, L.; Gal, A.F.; Kiss, B.; Morgovan, C.; Loghin, F.; Rochette, L.; Lauzier, B.; et al. Protective effects of a discontinuous treatment with alpha-lipoic acid in obesity-related heart failure with preserved ejection fraction, in rats. Antioxidants 2020, 9, 1073. [Google Scholar] [CrossRef]
  47. Yan, L.J.; Lodge, J.K.; Traber, M.G.; Matsugo, S.; Packer, L. Comparison between copper-mediated and hypochlorite-mediated modifications of human low density lipoproteins evaluated by protein carbonyl formation. J. Lipid Res. 1997, 38, 992–1001. [Google Scholar] [CrossRef] [PubMed]
  48. Yan, L.J.; Lodge, J.K.; Traber, M.G.; Packer, L. Apolipoprotein b carbonyl formation is enhanced by lipid peroxidation during copper-mediated oxidation of human low-density lipoproteins. Arch. Biochem. Biophys. 1997, 339, 165–171. [Google Scholar] [CrossRef] [PubMed]
  49. Wang, L.; Wu, C.G.; Fang, C.Q.; Gao, J.; Liu, Y.Z.; Chen, Y.; Chen, Y.N.; Xu, Z.G. The protective effect of alpha-lipoic acid on mitochondria in the kidney of diabetic rats. Int. J. Clin. Exp. Med. 2013, 6, 90–97. [Google Scholar] [PubMed]
  50. Golbidi, S.; Badran, M.; Laher, I. Diabetes and alpha lipoic acid. Front. Pharmacol. 2011, 2, 69. [Google Scholar] [CrossRef] [Green Version]
  51. Zhang, H.F.; Liu, H.M.; Xiang, J.Y.; Zhou, X.C.; Wang, D.; Chen, R.Y.; Tan, W.L.; Liang, L.Q.; Liu, L.L.; Shi, M.J.; et al. Alpha lipoamide inhibits diabetic kidney fibrosis via improving mitochondrial function and regulating rxralpha expression and activation. Acta Pharmacol. Sin. 2022, 1–15. [Google Scholar] [CrossRef]
  52. Yan, L.J. The nicotinamide/streptozotocin rodent model of type 2 diabetes: Renal pathophysiology and redox imbalance features. Biomolecules 2022, 12, 1225. [Google Scholar] [CrossRef]
  53. Dugbartey, G.J.; Alornyo, K.K.; Diaba, D.E.; Adams, I. Activation of renal cse/H2S pathway by alpha-lipoic acid protects against histological and functional changes in the diabetic kidney. Biomed. Pharm. 2022, 153, 113386. [Google Scholar] [CrossRef]
  54. Jiang, Z.; Tan, Z.; Meng, F.; Li, X. Curative effects of valsartan alone or combined with alpha-lipoic acid on inflammatory cytokines and renal function in early-stage diabetic kidney disease. J. Coll. Physicians Surg. Pak. 2019, 29, 1009–1011. [Google Scholar] [CrossRef]
  55. Feng, B.; Yan, X.F.; Xue, J.L.; Xu, L.; Wang, H. The protective effects of alpha-lipoic acid on kidneys in type 2 diabetic goto-kakisaki rats via reducing oxidative stress. Int. J. Mol. Sci. 2013, 14, 6746–6756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Louhelainen, M.; Merasto, S.; Finckenberg, P.; Lapatto, R.; Cheng, Z.J.; Mervaala, E.M. Lipoic acid supplementation prevents cyclosporine-induced hypertension and nephrotoxicity in spontaneously hypertensive rats. J. Hypertens 2006, 24, 947–956. [Google Scholar] [CrossRef] [PubMed]
  57. Liu, J. The effects and mechanisms of mitochondrial nutrient alpha-lipoic acid on improving age-associated mitochondrial and cognitive dysfunction: An overview. Neurochem. Res. 2008, 33, 194–203. [Google Scholar] [CrossRef] [PubMed]
  58. Granata, S.; Dalla Gassa, A.; Tomei, P.; Lupo, A.; Zaza, G. Mitochondria: A new therapeutic target in chronic kidney disease. Nutr. Metab. 2015, 12, 49. [Google Scholar] [CrossRef] [Green Version]
  59. Bhatti, F.; Mankhey, R.W.; Asico, L.; Quinn, M.T.; Welch, W.J.; Maric, C. Mechanisms of antioxidant and pro-oxidant effects of alpha-lipoic acid in the diabetic and nondiabetic kidney. Kidney Int. 2005, 67, 1371–1380. [Google Scholar] [CrossRef] [Green Version]
  60. Halabi, Z.; El Helou, C.; Al Balushi, H.; Gittinger, M.; Steck, A.R.; Kaakour, A.; Abu-Alfa, A.; El Zahran, T. Alpha lipoic acid toxicity: The first reported mortality in an adult patient after multiorgan failure. J. Emerg. Med. 2023, 64, 190–194. [Google Scholar] [CrossRef]
  61. Lo, S.M.; Dal Lin, F.T.; Soares, M.F.; Hauser, A.B.; Pecoits-Filho, R.; Nakao, L.S. Lipoic acid does not improve renal function markers in 5/6 nephrectomy model: Possible role of Nrf2 inactivation. Ren. Fail. 2016, 38, 558–563. [Google Scholar] [CrossRef] [Green Version]
  62. Ghibu, S.; Craciun, C.E.; Rusu, R.; Morgovan, C.; Mogosan, C.; Rochette, L.; Gal, A.F.; Dronca, M. Impact of alpha-lipoic acid chronic discontinuous treatment in cardiometabolic disorders and oxidative stress induced by fructose intake in rats. Antioxidants 2019, 8, 636. [Google Scholar] [CrossRef] [Green Version]
  63. Prabhakar, S.; Starnes, J.; Shi, S.; Lonis, B.; Tran, R. Diabetic nephropathy is associated with oxidative stress and decreased renal nitric oxide production. J. Am. Soc. Nephrol. 2007, 18, 2945–2952. [Google Scholar] [CrossRef] [Green Version]
  64. Yoo, J.Y.; Cha, D.R.; Kim, B.; An, E.J.; Lee, S.R.; Cha, J.J.; Kang, Y.S.; Ghee, J.Y.; Han, J.Y.; Bae, Y.S. Lps-induced acute kidney injury is mediated by nox4-sh3yl1. Cell Rep. 2020, 33, 108245. [Google Scholar] [CrossRef]
  65. Doi, K.; Leelahavanichkul, A.; Yuen, P.S.; Star, R.A. Animal models of sepsis and sepsis-induced kidney injury. J. Clin. Investig. 2009, 119, 2868–2878. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Soni, S.; Martens, M.D.; Takahara, S.; Silver, H.L.; Maayah, Z.H.; Ussher, J.R.; Ferdaoussi, M.; Dyck, J.R.B. Exogenous ketone ester administration attenuates systemic inflammation and reduces organ damage in a lipopolysaccharide model of sepsis. Biochim. Biophys. Acta Mol. Basis Dis. 2022, 1868, 166507. [Google Scholar] [CrossRef] [PubMed]
  67. Wanner, G.A.; Keel, M.; Steckholzer, U.; Beier, W.; Stocker, R.; Ertel, W. Relationship between procalcitonin plasma levels and severity of injury, sepsis, organ failure, and mortality in injured patients. Crit. Care Med. 2000, 28, 950–957. [Google Scholar] [CrossRef] [PubMed]
  68. Branco, R.G.; Garcia, P.C.; Garcia, J.P.; Piva, J.P.; Ross-Russell, R. Interactions of sepsis, organ dysfunction, and outcome of critically ill children. Am. J. Respir. Crit. Care Med. 2005, 172, 1606, author reply 1606–1607. [Google Scholar] [CrossRef]
  69. Chang, Y.M.; Chou, Y.T.; Kan, W.C.; Shiao, C.C. Sepsis and acute kidney injury: A review focusing on the bidirectional interplay. Int. J. Mol. Sci. 2022, 23, 9159. [Google Scholar] [CrossRef] [PubMed]
  70. Petronilho, F.; Florentino, D.; Danielski, L.G.; Vieira, L.C.; Martins, M.M.; Vieira, A.; Bonfante, S.; Goldim, M.P.; Vuolo, F. Alpha-lipoic acid attenuates oxidative damage in organs after sepsis. Inflammation 2016, 39, 357–365. [Google Scholar] [CrossRef]
  71. Jia, J.; Gong, X.; Zhao, Y.; Yang, Z.; Ji, K.; Luan, T.; Zang, B.; Li, G. Autophagy enhancing contributes to the organ protective effect of alpha-lipoic acid in septic rats. Front. Immunol. 2019, 10, 1491. [Google Scholar] [CrossRef]
  72. Li, G.; Fu, J.; Zhao, Y.; Ji, K.; Luan, T.; Zang, B. Alpha-lipoic acid exerts anti-inflammatory effects on lipopolysaccharide-stimulated rat mesangial cells via inhibition of nuclear factor kappa b (nf-kappab) signaling pathway. Inflammation 2015, 38, 510–519. [Google Scholar] [CrossRef]
  73. Suh, S.H.; Lee, K.E.; Kim, I.J.; Kim, O.; Kim, C.S.; Choi, J.S.; Choi, H.I.; Bae, E.H.; Ma, S.K.; Lee, J.U.; et al. Alpha-lipoic acid attenuates lipopolysaccharide-induced kidney injury. Clin. Exp. Nephrol. 2015, 19, 82–91. [Google Scholar] [CrossRef]
  74. Amini, N.; Maleki, M.; Badavi, M. Nephroprotective activity of naringin against chemical-induced toxicity and renal ischemia/reperfusion injury: A review. Avicenna J. Phytomed. 2022, 12, 357–370. [Google Scholar]
  75. Ding, Y.; Zhang, Y.; Zhang, W.; Shang, J.; Xie, Z.; Chen, C. Effects of lipoic acid on ischemia-reperfusion injury. Oxid. Med. Cell. Longev. 2021, 2021, 5093216. [Google Scholar] [CrossRef] [PubMed]
  76. Malek, M.; Nematbakhsh, M. Renal ischemia/reperfusion injury; from pathophysiology to treatment. J. Renal Inj. Prev. 2015, 4, 20–27. [Google Scholar] [PubMed]
  77. Yan, Y.; Bai, J.; Zhou, X.; Tang, J.; Jiang, C.; Tolbert, E.; Bayliss, G.; Gong, R.; Zhao, T.C.; Zhuang, S. P2X7 receptor inhibition protects against ischemic acute kidney injury in mice. Am. J. Physiol. Cell Physiol. 2015, 308, C463–C472. [Google Scholar] [CrossRef] [Green Version]
  78. Bae, E.H.; Lee, K.S.; Lee, J.; Ma, S.K.; Kim, N.H.; Choi, K.C.; Frokiaer, J.; Nielsen, S.; Kim, S.Y.; Kim, S.Z.; et al. Effects of alpha-lipoic acid on ischemia-reperfusion-induced renal dysfunction in rats. Am. J. Physiol. Renal Physiol. 2008, 294, F272–F280. [Google Scholar] [CrossRef] [PubMed]
  79. Sehirli, O.; Sener, E.; Cetinel, S.; Yuksel, M.; Gedik, N.; Sener, G. Alpha-lipoic acid protects against renal ischaemia-reperfusion injury in rats. Clin. Exp. Pharmacol. Physiol. 2008, 35, 249–255. [Google Scholar] [CrossRef] [PubMed]
  80. Othman, M.A.; Mubarak, H.A.; Sayed, M.M. Ameliorative role of alpha-lipoic acid in renal cortical structural damage, induced by limb ischemia-reperfusion injury in the rat. Ultrastruct. Pathol. 2022, 46, 110–121. [Google Scholar] [CrossRef]
  81. Farag, M.M.; Ahmed, S.M.; Elhadidy, W.F.; Rashad, R.M. Superior protective effects of febuxostat plus alpha-lipoic acid on renal ischemia/reperfusion-induced hepatorenal injury in rats. Saudi J. Kidney Dis. Transpl. 2019, 30, 1364–1374. [Google Scholar] [CrossRef]
  82. Takaoka, M.; Ohkita, M.; Kobayashi, Y.; Yuba, M.; Matsumura, Y. Protective effect of alpha-lipoic acid against ischaemic acute renal failure in rats. Clin. Exp. Pharmacol. Physiol. 2002, 29, 189–194. [Google Scholar] [CrossRef]
  83. Mao, Y.; Yu, J.; Da, J.; Yu, F.; Zha, Y. Acteoside alleviates uuo-induced inflammation and fibrosis by regulating the hmgn1/tlr4/trem1 signaling pathway. PeerJ 2023, 11, e14765. [Google Scholar] [CrossRef]
  84. Sun, D.; Guo, J.; Liang, W.; Chen, Y.; Chen, X.; Wang, L. Anlotinib alleviates renal fibrosis via inhibition of the erk and akt signaling pathways. Oxid. Med. Cell. Longev. 2023, 2023, 1686804. [Google Scholar] [CrossRef]
  85. Wu, R.; Li, J.; Tu, G.; Su, Y.; Zhang, X.; Luo, Z.; Rong, R.; Zhang, Y. Comprehensive molecular and cellular characterization of acute kidney injury progression to renal fibrosis. Front. Immunol. 2021, 12, 699192. [Google Scholar] [CrossRef] [PubMed]
  86. Yu, H.X.; Lin, W.; Yang, K.; Wei, L.J.; Chen, J.L.; Liu, X.Y.; Zhong, K.; Chen, X.; Pei, M.; Yang, H.T. Transcriptome-based network analysis reveals hirudin potentiates anti-renal fibrosis efficacy in uuo rats. Front. Pharmacol. 2021, 12, 741801. [Google Scholar] [CrossRef] [PubMed]
  87. Zhang, Y.; Hao, J.; Ma, X.; Zhao, Q.; Gao, X.; Wang, X.; Xu, Q. Huoxue jiedu huayu recipe ameliorates mesangial cell pyroptosis in contralateral kidney of uuo rats. Evid. Based Complement. Altern. Med. 2020, 2020, 2530431. [Google Scholar] [CrossRef] [PubMed]
  88. Wongmekiat, O.; Leelarungrayub, D.; Thamprasert, K. Alpha-lipoic acid attenuates renal injury in rats with obstructive nephropathy. Biomed Res. Int. 2013, 2013, 138719. [Google Scholar] [CrossRef] [Green Version]
  89. Sun, Y.; Zhang, Y.; Zhao, D.; Ding, G.; Huang, S.; Zhang, A.; Jia, Z. Rotenone remarkably attenuates oxidative stress, inflammation, and fibrosis in chronic obstructive uropathy. Mediat. Inflamm. 2014, 2014, 670106. [Google Scholar] [CrossRef] [Green Version]
  90. Huang, J.; Zhang, Z.; Liu, B.; Gao, Y.; Nie, J.; Wen, S.; Lai, X.; Liang, H. Identification of circular rna expression profiles in renal fibrosis induced by obstructive injury. Ren. Fail. 2021, 43, 1368–1377. [Google Scholar] [CrossRef] [PubMed]
  91. Li, R.; Guo, Y.; Zhang, Y.; Zhang, X.; Zhu, L.; Yan, T. Salidroside ameliorates renal interstitial fibrosis by inhibiting the TLR4/NF-kappab and MAPK signaling pathways. Int. J. Mol. Sci. 2019, 20, 1103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Liu, F.; Wang, L.; Qi, H.; Wang, J.; Wang, Y.; Jiang, W.; Xu, L.; Liu, N.; Zhuang, S. Nintedanib, a triple tyrosine kinase inhibitor, attenuates renal fibrosis in chronic kidney disease. Clin. Sci. 2017, 131, 2125–2143. [Google Scholar] [CrossRef]
  93. Miguel, V.; Ramos, R.; Garcia-Bermejo, L.; Rodriguez-Puyol, D.; Lamas, S. The program of renal fibrogenesis is controlled by micrornas regulating oxidative metabolism. Redox Biol. 2021, 40, 101851. [Google Scholar] [CrossRef]
  94. Dendooven, A.; Ishola, D.A., Jr.; Nguyen, T.Q.; Van der Giezen, D.M.; Kok, R.J.; Goldschmeding, R.; Joles, J.A. Oxidative stress in obstructive nephropathy. Int. J. Exp. Pathol. 2011, 92, 202–210. [Google Scholar] [CrossRef]
  95. Grande, M.T.; Perez-Barriocanal, F.; Lopez-Novoa, J.M. Role of inflammation in tubulo-interstitial damage associated to obstructive nephropathy. J. Inflamm. 2010, 7, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Cho, H.S.; Kim, J.H.; Jang, H.N.; Lee, T.W.; Jung, M.H.; Kim, T.H.; Chang, S.H.; Park, D.J. Alpha-lipoic acid ameliorates the epithelial mesenchymal transition induced by unilateral ureteral obstruction in mice. Sci. Rep. 2017, 7, 46065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Iskander, A.; Yan, L.J. Cisplatin-induced kidney toxicity: Potential roles of major NAD+-dependent enzymes and plant-derived natural products. Biomolecules 2022, 12, 1078. [Google Scholar] [CrossRef] [PubMed]
  98. Kim, K.H.; Lee, B.; Kim, Y.R.; Kim, M.A.; Ryu, N.; Jung, D.J.; Kim, U.K.; Baek, J.I.; Lee, K.Y. Evaluating protective and therapeutic effects of alpha-lipoic acid on cisplatin-induced ototoxicity. Cell Death Dis. 2018, 9, 827. [Google Scholar] [CrossRef] [Green Version]
  99. Kang, K.P.; Kim, D.H.; Jung, Y.J.; Lee, A.S.; Lee, S.; Lee, S.Y.; Jang, K.Y.; Sung, M.J.; Park, S.K.; Kim, W. Alpha-lipoic acid attenuates cisplatin-induced acute kidney injury in mice by suppressing renal inflammation. Nephrol. Dial. Transplant. 2009, 24, 3012–3020. [Google Scholar] [CrossRef]
  100. El-Beshbishy, H.A.; Bahashwan, S.A.; Aly, H.A.; Fakher, H.A. Abrogation of cisplatin-induced nephrotoxicity in mice by alpha lipoic acid through ameliorating oxidative stress and enhancing gene expression of antioxidant enzymes. Eur. J. Pharmacol. 2011, 668, 278–284. [Google Scholar] [CrossRef]
  101. Bae, E.H.; Lee, J.; Ma, S.K.; Kim, I.J.; Frokiaer, J.; Nielsen, S.; Kim, S.Y.; Kim, S.W. Alpha-lipoic acid prevents cisplatin-induced acute kidney injury in rats. Nephrol. Dial. Transplant. 2009, 24, 2692–2700. [Google Scholar] [CrossRef] [Green Version]
  102. Hussein, A.; Ahmed, A.A.; Shouman, S.A.; Sharawy, S. Ameliorating effect of dl-α-lipoic acid against cisplatin-induced nephrotoxicity and cardiotoxicity in experimental animals. Drug Discov. Ther. 2012, 6, 147–156. [Google Scholar] [CrossRef]
  103. Pianta, T.J.; Succar, L.; Davidson, T.; Buckley, N.A.; Endre, Z.H. Monitoring treatment of acute kidney injury with damage biomarkers. Toxicol. Lett. 2017, 268, 63–70. [Google Scholar] [CrossRef]
  104. Yan, L.J.; Allen, D.C. Cadmium-induced kidney injury: Oxidative damage as a unifying mechanism. Biomolecules 2021, 11, 1575. [Google Scholar] [CrossRef]
  105. Li, X.; Zou, Y.; Fu, Y.Y.; Xing, J.; Wang, K.Y.; Wan, P.Z.; Zhai, X.Y. A-lipoic acid alleviates folic acid-induced renal damage through inhibition of ferroptosis. Front. Physiol. 2021, 12, 680544. [Google Scholar] [CrossRef] [PubMed]
  106. Yan, L.J. Folic acid-induced animal model of kidney disease. Anim. Model. Exp. Med. 2021, 4, 329–342. [Google Scholar] [CrossRef] [PubMed]
  107. Almeer, R.S.; AlBasher, G.I.; Alarifi, S.; Alkahtani, S.; Ali, D.; Abdel Moneim, A.E. Royal jelly attenuates cadmium-induced nephrotoxicity in male mice. Sci. Rep. 2019, 9, 5825. [Google Scholar] [CrossRef] [Green Version]
  108. Chen, C.; Han, X.; Wang, G.; Liu, D.; Bao, L.; Jiao, C.; Luan, J.; Hou, Y.; Xu, Y.; Wang, H.; et al. Nrf2 deficiency aggravates the kidney injury induced by subacute cadmium exposure in mice. Arch. Toxicol. 2021, 95, 883–893. [Google Scholar] [CrossRef] [PubMed]
  109. Almeer, R.S.; Kassab, R.B.; AlBasher, G.I.; Alarifi, S.; Alkahtani, S.; Ali, D.; Abdel Moneim, A.E. Royal jelly mitigates cadmium-induced neuronal damage in mouse cortex. Mol. Biol. Rep. 2019, 46, 119–131. [Google Scholar] [CrossRef] [PubMed]
  110. Chen, S.; Liu, G.; Long, M.; Zou, H.; Cui, H. Alpha lipoic acid attenuates cadmium-induced nephrotoxicity via the mitochondrial apoptotic pathways in rat. J. Inorg. Biochem. 2018, 184, 19–26. [Google Scholar] [CrossRef]
  111. Luo, T.; Liu, G.; Long, M.; Yang, J.; Song, R.; Wang, Y.; Yuan, Y.; Bian, J.; Liu, X.; Gu, J.; et al. Treatment of cadmium-induced renal oxidative damage in rats by administration of alpha-lipoic acid. Environ. Sci. Pollut. Res. Int. 2017, 24, 1832–1844. [Google Scholar] [CrossRef]
  112. Veljkovic, A.R.; Nikolic, R.S.; Kocic, G.M.; Pavlovic, D.D.; Cvetkovic, T.P.; Sokolovic, D.T.; Jevtovic, T.M.; Basic, J.T.; Laketic, D.M.; Marinkovic, M.R.; et al. Protective effects of glutathione and lipoic acid against cadmium-induced oxidative stress in rat’s kidney. Ren. Fail. 2012, 34, 1281–1287. [Google Scholar] [CrossRef]
  113. Fukuda, A.; Osawa, T.; Oda, H.; Tanaka, T.; Toyokuni, S.; Uchida, K. Oxidative stress response in iron-induced acute nephrotoxicity: Enhanced expression of heat shock protein 90. Biochem. Biophys. Res. Commun. 1996, 219, 76–81. [Google Scholar] [CrossRef]
  114. Leaf, D.E.; Swinkels, D.W. Catalytic iron and acute kidney injury. Am. J. Physiol. Renal Physiol. 2016, 311, F871–F876. [Google Scholar] [CrossRef] [Green Version]
  115. Amarapurkar, P.; Roberts, L.; Navarrete, J.; El Rassi, F. Sickle cell disease and kidney. Adv. Chronic Kidney Dis. 2022, 29, 141–148e1. [Google Scholar] [CrossRef] [PubMed]
  116. Chawla, L.S.; Beers-Mulroy, B.; Tidmarsh, G.F. Therapeutic opportunities for hepcidin in acute care medicine. Crit. Care Clin. 2019, 35, 357–374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Martines, A.M.; Masereeuw, R.; Tjalsma, H.; Hoenderop, J.G.; Wetzels, J.F.; Swinkels, D.W. Iron metabolism in the pathogenesis of iron-induced kidney injury. Nat. Rev. Nephrol. 2013, 9, 385–398. [Google Scholar] [CrossRef] [PubMed]
  118. Gwozdzinski, K.; Pieniazek, A.; Gwozdzinski, L. Reactive oxygen species and their involvement in red blood cell damage in chronic kidney disease. Oxid. Med. Cell. Longev. 2021, 2021, 6639199. [Google Scholar] [CrossRef] [PubMed]
  119. Walker, V.J.; Agarwal, A. Targeting iron homeostasis in acute kidney injury. Semin. Nephrol. 2016, 36, 62–70. [Google Scholar] [CrossRef] [Green Version]
  120. Chaudhary, K.; Chilakala, A.; Ananth, S.; Mandala, A.; Veeranan-Karmegam, R.; Powell, F.L.; Ganapathy, V.; Gnana-Prakasam, J.P. Renal iron accelerates the progression of diabetic nephropathy in the hfe gene knockout mouse model of iron overload. Am. J. Physiol. Renal Physiol. 2019, 317, F512–F517. [Google Scholar] [CrossRef]
  121. Gao, W.; Li, X.; Gao, Z.; Li, H. Iron increases diabetes-induced kidney injury and oxidative stress in rats. Biol. Trace Elem. Res. 2014, 160, 368–375. [Google Scholar] [CrossRef]
  122. Guan, P.; Sun, Z.M.; Luo, L.F.; Zhao, Y.S.; Yang, S.C.; Yu, F.Y.; Wang, N.; Ji, E.S. Hydrogen gas alleviates chronic intermittent hypoxia-induced renal injury through reducing iron overload. Molecules 2019, 24, 1184. [Google Scholar] [CrossRef] [Green Version]
  123. Annamalai, C.; Ganesh, R.N.; Viswanathan, P. Ferrotoxicity and its amelioration by endogenous vitamin d in experimental acute kidney injury. Exp. Biol. Med. 2020, 245, 1474–1489. [Google Scholar] [CrossRef]
  124. Li, Y.H.; Chien, S.P.; Chu, P.Y.; Liu, M.Y. Prophylactic and therapeutic effects of a subcutaneous injection of sesame oil against iron-induced acute renal injury in mice. JPEN J. Parenter. Enteral. Nutr. 2012, 36, 344–348. [Google Scholar] [CrossRef]
  125. Gupta, A.; Sharma, S.; Chopra, K. Reversal of iron-induced nephrotoxicity in rats by molsidomine, a nitric oxide donor. Food Chem. Toxicol. 2008, 46, 537–543. [Google Scholar] [CrossRef] [PubMed]
  126. Cavdar, Z.; Oktan, M.A.; Ural, C.; Calisir, M.; Kocak, A.; Heybeli, C.; Yildiz, S.; Arici, A.; Ellidokuz, H.; Celik, A.; et al. Renoprotective effects of alpha lipoic acid on iron overload-induced kidney injury in rats by suppressing nadph oxidase 4 and p38 mapk signaling. Biol. Trace Elem. Res. 2020, 193, 483–493. [Google Scholar] [CrossRef] [PubMed]
  127. Cavdar, Z.; Oktan, M.A.; Ural, C.; Kocak, A.; Calisir, M.; Heybeli, C.; Yildiz, S.; Ozbal, S.; Arslan, S.; Ergur, B.U.; et al. Alpha lipoic acid attenuates iron induced oxidative acute kidney injury in rats. Biotech. Histochem. 2021, 96, 409–417. [Google Scholar] [CrossRef]
  128. Di Stefano, A.; Sozio, P.; Cocco, A.; Iannitelli, A.; Santucci, E.; Costa, M.; Pecci, L.; Nasuti, C.; Cantalamessa, F.; Pinnen, F. L-dopa- and dopamine-(r)-alpha-lipoic acid conjugates as multifunctional codrugs with antioxidant properties. J. Med. Chem. 2006, 49, 1486–1493. [Google Scholar] [CrossRef]
  129. Persson, H.L.; Svensson, A.I.; Brunk, U.T. Alpha-lipoic acid and alpha-lipoamide prevent oxidant-induced lysosomal rupture and apoptosis. Redox Rep. 2001, 6, 327–334. [Google Scholar] [CrossRef] [PubMed]
  130. Camiolo, G.; Tibullo, D.; Giallongo, C.; Romano, A.; Parrinello, N.L.; Musumeci, G.; Di Rosa, M.; Vicario, N.; Brundo, M.V.; Amenta, F.; et al. Alpha-lipoic acid reduces iron-induced toxicity and oxidative stress in a model of iron overload. Int. J. Mol. Sci. 2019, 20, 609. [Google Scholar] [CrossRef] [Green Version]
  131. Sharifi-Zahabi, E.; Abdollahzad, H.; Mostafa Nachvak, S.; Moloudi, J.; Golpayegani, M.R.; Asiaei, S.; Rezavand, L.; Iraji, Z.; Jamshidi, K. Effects of alpha lipoic acid on iron overload, lipid profile and oxidative stress indices in beta-thalassemia major patients: A cross-over randomised controlled clinical trial. Int. J. Clin. Pract. 2021, 75, e14062. [Google Scholar] [CrossRef] [PubMed]
  132. Chang-Panesso, M. Acute kidney injury and aging. Pediatr. Nephrol. 2021, 36, 2997–3006. [Google Scholar] [CrossRef]
  133. Zhou, X.J.; Rakheja, D.; Yu, X.; Saxena, R.; Vaziri, N.D.; Silva, F.G. The aging kidney. Kidney Int. 2008, 74, 710–720. [Google Scholar] [CrossRef] [Green Version]
  134. Kotob, M.; Hussein, A.; Abd-Elkareem, M. Histopathological changes of kidney tissue during aging. SVU-Int. J. Vet. Sci. 2021, 4, 54–65. [Google Scholar] [CrossRef]
  135. Khafaga, A.F.; Elewa, Y.H.A.; Atta, M.S.; Noreldin, A.E. Aging-related functional and structural changes in renal tissues: Lesson from a camel model. Microsc. Microanal. 2021, 27, 566–578. [Google Scholar] [CrossRef]
  136. Arivazhagan, P.; Ramanathan, K.; Panneerselvam, C. Effect of dl-alpha-lipoic acid on glutathione metabolic enzymes in aged rats. Exp. Gerontol. 2001, 37, 81–87. [Google Scholar] [CrossRef]
  137. Arivazhagan, P.; Ramanathan, K.; Panneerselvam, C. Effect of dl-alpha-lipoic acid on mitochondrial enzymes in aged rats. Chem. Biol. Interact. 2001, 138, 189–198. [Google Scholar] [CrossRef]
  138. Arivazhagan, P.; Ramanathan, K.; Panneerselvam, C. Effect of dl-alpha-lipoic acid on the status of lipid peroxidation and antioxidants in mitochondria of aged rats. J. Nutr. Biochem. 2001, 12, 2–6. [Google Scholar] [CrossRef]
  139. Zhang, Y.; Chen, X.; Yang, L.; Zu, Y.; Lu, Q. Effects of rosmarinic acid on liver and kidney antioxidant enzymes, lipid peroxidation and tissue ultrastructure in aging mice. Food Funct. 2015, 6, 927–931. [Google Scholar] [CrossRef] [PubMed]
  140. Hall, J.A.; Yerramilli, M.; Obare, E.; Yerramilli, M.; Panickar, K.S.; Bobe, G.; Jewell, D.E. Nutritional interventions that slow the age-associated decline in renal function in a canine geriatric model for elderly humans. J. Nutr. Health Aging 2016, 20, 1010–1023. [Google Scholar] [CrossRef]
  141. Zhang, C.J.; Li, H.; Xiong, Y.Z.; Chang, Y.; Yang, F.; Ma, X.L.; Wang, X.T.; Shimosawa, T.; Ji, E.S.; Xu, Q.Y. Chronic intermittent hypoxia induces renal fibrosis through mr activation. Exp. Gerontol. 2022, 163, 111780. [Google Scholar] [CrossRef]
  142. Su, Y.; Li, C.; Liu, W.; Liu, Y.; Li, L.; Chen, Q. Comprehensive analysis of differentially expressed mirnas in mice with kidney injury induced by chronic intermittent hypoxia. Front. Genet. 2022, 13, 918728. [Google Scholar] [CrossRef] [PubMed]
  143. Chen, Q.; Hong, Z.; Chen, Z.; Chen, Y.; Liu, D. Circrna expression profiles and functional analysis in a mouse model of chronic intermittent hypoxia-induced renal injury: New insight into pathogenesis. PeerJ 2023, 11, e14957. [Google Scholar] [CrossRef] [PubMed]
  144. Badran, M.; Abuyassin, B.; Golbidi, S.; Ayas, N.; Laher, I. Alpha lipoic acid improves endothelial function and oxidative stress in mice exposed to chronic intermittent hypoxia. Oxid. Med. Cell. Longev. 2019, 2019, 4093018. [Google Scholar] [CrossRef] [Green Version]
  145. Abuyassin, B.; Badran, M.; Ayas, N.T.; Laher, I. The antioxidant alpha-lipoic acid attenuates intermittent hypoxia-related renal injury in a mouse model of sleep apnea. Sleep 2019, 42, zsz066. [Google Scholar] [CrossRef] [PubMed]
  146. Mennuni, S.; Rubattu, S.; Pierelli, G.; Tocci, G.; Fofi, C.; Volpe, M. Hypertension and kidneys: Unraveling complex molecular mechanisms underlying hypertensive renal damage. J. Hum. Hypertens. 2014, 28, 74–79. [Google Scholar] [CrossRef] [PubMed]
  147. Chu, Y.; Zhou, Y.; Lu, S.; Lu, F.; Hu, Y. Pathogenesis of higher blood pressure and worse renal function in salt-sensitive hypertension. Kidney Blood Press. Res. 2021, 46, 236–244. [Google Scholar] [CrossRef]
  148. Ameer, O.Z. Hypertension in chronic kidney disease: What lies behind the scene. Front. Pharmacol. 2022, 13, 949260. [Google Scholar] [CrossRef] [PubMed]
  149. Martinelli, I.; Tomassoni, D.; Roy, P.; Di Cesare Mannelli, L.; Amenta, F.; Tayebati, S.K. Antioxidant properties of alpha-lipoic (thioctic) acid treatment on renal and heart parenchyma in a rat model of hypertension. Antioxidants 2021, 10, 1006. [Google Scholar] [CrossRef] [PubMed]
  150. Diwan, V.; Brown, L.; Gobe, G.C. Adenine-induced chronic kidney disease in rats. Nephrology 2018, 23, 5–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Yuan, H.; Zheng, C.; Zhu, L.; Song, Z.; Dai, L.; Hu, Q.; Wang, L.; Chen, Y.; Xiong, J. Contribution of tfeb-mediated autophagy to tubulointerstitial fibrosis in mice with adenine-induced chronic kidney disease. Biomed. Pharm. 2021, 133, 110949. [Google Scholar] [CrossRef]
  152. Ho, H.J.; Shirakawa, H. Oxidative stress and mitochondrial dysfunction in chronic kidney disease. Cells 2022, 12, 88. [Google Scholar] [CrossRef]
  153. Ali, B.H.; Inuwa, I.; Al Za’abi, M.; Al Bahlani, S.; Al Issaei, H.; Ramkumar, A.; Madanagopal, T.; Nemmar, A.; Malheiros, D.M.; Zatz, R. Renal and myocardial histopathology and morphometry in rats with adenine—induced chronic renal failure: Influence of gum acacia. Cell. Physiol. Biochem. 2014, 34, 818–828. [Google Scholar] [CrossRef]
  154. Liu, X.; Zhang, B.; Huang, S.; Wang, F.; Zheng, L.; Lu, J.; Zeng, Y.; Chen, J.; Li, S. Metabolomics analysis reveals the protection mechanism of huangqi-danshen decoction on adenine-induced chronic kidney disease in rats. Front. Pharmacol. 2019, 10, 992. [Google Scholar] [CrossRef]
  155. Kumaran, M.; Lingaraju, M.C.; Srivastava, V.; Mathesh, K.; Manickam, K.; Parida, S.; Singh, T.U.; Kumar, D. Pre-084 ameliorated kidney injury by reducing endoplasmic reticulum stress in the rat model of adenine-induced chronic kidney disease. Mol. Biol. Rep. 2023, 50, 3681–3691. [Google Scholar] [CrossRef] [PubMed]
  156. Natesan, V.; Kim, S.J. Diabetic nephropathy—A review of risk factors, progression, mechanism, and dietary management. Biomol. Ther. 2021, 29, 365–372. [Google Scholar] [CrossRef] [PubMed]
  157. Agarwal, R. Pathogenesis of diabetic nephropathy. In Chronic Kidney Disease and Type 2 Diabetes; American Diabetes Association: Arlington, VA, USA, 2021; pp. 2–7. [Google Scholar]
  158. Alkhatib, L.; Velez Diaz, L.A.; Varma, S.; Chowdhary, A.; Bapat, P.; Pan, H.; Kukreja, G.; Palabindela, P.; Selvam, S.A.; Kalra, K. Lifestyle modifications and nutritional and therapeutic interventions in delaying the progression of chronic kidney disease: A review. Cureus 2023, 15, e34572. [Google Scholar] [CrossRef]
  159. Floege, J.; Daha, M.R. Iga nephropathy: New insights into the role of complement. Kidney Int. 2018, 94, 16–18. [Google Scholar] [CrossRef]
  160. Wu, X.; Zhao, L.; Li, K.; Yang, J. The role of NLRP3 inflammasome in iga nephropathy. Medicina 2022, 59, 82. [Google Scholar] [CrossRef] [PubMed]
  161. Wu, C.Y.; Hua, K.F.; Yang, S.R.; Tsai, Y.S.; Yang, S.M.; Hsieh, C.Y.; Wu, C.C.; Chang, J.F.; Arbiser, J.L.; Chang, C.T.; et al. Tris dba ameliorates iga nephropathy by blunting the activating signal of NLRP3 inflammasome through sirt1- and sirt3-mediated autophagy induction. J. Cell. Mol. Med. 2020, 24, 13609–13622. [Google Scholar] [CrossRef]
  162. Selvaskandan, H.; Gonzalez-Martin, G.; Barratt, J.; Cheung, C.K. Iga nephropathy: An overview of drug treatments in clinical trials. Expert Opin. Investig. Drugs 2022, 31, 1321–1338. [Google Scholar] [CrossRef] [PubMed]
  163. Liao, J.; Zhou, Y.; Xu, X.; Huang, K.; Chen, P.; Wu, Y.; Jin, B.; Hu, Q.; Chen, G.; Zhao, S. Current knowledge of targeted-release budesonide in immunoglobulin a nephropathy: A comprehensive review. Front. Immunol. 2022, 13, 926517. [Google Scholar] [CrossRef]
  164. Taherkhani, A.; Farrokhi Yekta, R.; Mohseni, M.; Saidijam, M.; Arefi Oskouie, A. Chronic kidney disease: A review of proteomic and metabolomic approaches to membranous glomerulonephritis, focal segmental glomerulosclerosis, and iga nephropathy biomarkers. Proteome Sci. 2019, 17, 7. [Google Scholar] [CrossRef] [Green Version]
  165. Cao, W.; Xu, J.; Zhou, Z.M.; Wang, G.B.; Hou, F.F.; Nie, J. Advanced oxidation protein products activate intrarenal renin-angiotensin system via a CD36-mediated, redox-dependent pathway. Antioxid. Redox Signal. 2013, 18, 19–35. [Google Scholar] [CrossRef] [Green Version]
  166. Tian, J.; Chen, J.H.; Li, Q.; He, Q.; Lin, W.Q. Lipid peroxidation in iga nephropathy and the effect of lipo-prostaglandin e1. J. Nephrol. 2005, 18, 243–248. [Google Scholar] [PubMed]
  167. Parinyasiri, U.; Ong-Ajyooth, L.; Parichatikanond, P.; Ong-Ajyooth, S.; Liammongkolkul, S.; Kanyog, S. Effect of fish oil on oxidative stress, lipid profile and renal function in iga nephropathy. J. Med. Assoc. Thai. 2004, 87, 143–149. [Google Scholar] [PubMed]
  168. Chaudhary, A.; Mishra, A.; Sethi, S. Oxidized omega-3 fatty acids inhibit pro-inflammatory responses in glomerular endothelial cells. Nephron Exp. Nephrol. 2004, 97, e136–e145. [Google Scholar] [CrossRef] [PubMed]
  169. Kasahara, J.; Kobayashi, K.; Maeshima, Y.; Yamasaki, Y.; Yasuda, T.; Matsuura, E.; Makino, H. Clinical significance of serum oxidized low-density lipoprotein/beta2-glycoprotein i complexes in patients with chronic renal diseases. Nephron Clin. Pract. 2004, 98, c15–c24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Wehbi, B.; Pascal, V.; Zawil, L.; Cogne, M.; Aldigier, J.C. History of iga nephropathy mouse models. J. Clin. Med. 2021, 10, 3142. [Google Scholar] [CrossRef]
  171. Monteiro, R.C.; Suzuki, Y. Are there animal models of iga nephropathy? Semin. Immunopathol. 2021, 43, 639–648. [Google Scholar] [CrossRef]
  172. Mao, Z.M.; Shen, S.M.; Wan, Y.G.; Sun, W.; Chen, H.L.; Huang, M.M.; Yang, J.J.; Wu, W.; Tang, H.T.; Tang, R.M. Huangkui capsule attenuates renal fibrosis in diabetic nephropathy rats through regulating oxidative stress and p38mapk/akt pathways, compared to alpha-lipoic acid. J. Ethnopharmacol. 2015, 173, 256–265. [Google Scholar] [CrossRef]
  173. Ensergueix, G.; Pallet, N.; Joly, D.; Levi, C.; Chauvet, S.; Trivin, C.; Augusto, J.F.; Boudet, R.; Aboudagga, H.; Touchard, G.; et al. Ifosfamide nephrotoxicity in adult patients. Clin. Kidney J. 2020, 13, 660–665. [Google Scholar] [CrossRef] [Green Version]
  174. Boskabadi, J.; Yousefi-Mazhin, E.; Salehifar, E. Ifosfamide-induced acute kidney injury in a patient with leiomyosarcoma: A case report. Cancer Rep. 2022, 5, e1666. [Google Scholar] [CrossRef]
  175. Arjmand, A.; Mashhadi, M.; Kaveh, A.; Kamranfar, F.; Seydi, E.; Pourahmad, J. Mitochondrial transplantation therapy against ifosfamide induced toxicity on rat renal proximal tubular cells. Drug Res. 2023, 73, 113–120. [Google Scholar] [CrossRef]
  176. El-Sisi Ael, D.; El-Syaad, M.E.; El-Desoky, K.I.; Moussa, E.A. Protective effects of alpha lipoic acid versus n-acetylcysteine on ifosfamide-induced nephrotoxicity. Toxicol. Ind. Health 2015, 31, 97–107. [Google Scholar] [CrossRef] [PubMed]
  177. Alshammari, G.M.; Al-Ayed, M.S.; Abdelhalim, M.A.; Al-Harbi, L.N.; Yahya, M.A. Effects of antioxidant combinations on the renal toxicity induced rats by gold nanoparticles. Molecules 2023, 28, 1879. [Google Scholar] [CrossRef] [PubMed]
  178. Castellani, C.; Radu, C.M.; Morillas-Becerril, L.; Barison, I.; Menato, F.; Do Nascimento, T.M.; Fedrigo, M.; Giarraputo, A.; Virzi, G.M.; Simioni, P.; et al. Poly(lipoic acid)-based nanoparticles as a new therapeutic tool for delivering active molecules. Nanomedicine 2022, 45, 102593. [Google Scholar] [CrossRef] [PubMed]
  179. Tillman, L.; Tabish, T.A.; Kamaly, N.; Moss, P.; El-Briri, A.; Thiemermann, C.; Pranjol, M.Z.I.; Yaqoob, M.M. Advancements in nanomedicines for the detection and treatment of diabetic kidney disease. Biomater. Biosyst. 2022, 6, 100047. [Google Scholar] [CrossRef]
  180. Peres, R.A.S.; Silva-Aguiar, R.P.; Teixeira, D.E.; Peruchetti, D.B.; Alves, S.A.S.; Leal, A.B.C.; Castro, G.F.; Ribeiro, N.B.S.; Guimaraes, F.V.; Pinheiro, A.A.S.; et al. Gold nanoparticles reduce tubule-interstitial injury and proteinuria in a murine model of subclinical acute kidney injury. Biochim. Biophys. Acta Gen. Subj. 2023, 1867, 130314. [Google Scholar] [CrossRef]
  181. Wang, Y.; Jiang, H.; Zhang, L.; Yao, P.; Wang, S.; Yang, Q. Nanosystems for oxidative stress regulation in the anti-inflammatory therapy of acute kidney injury. Front. Bioeng. Biotechnol. 2023, 11, 1120148. [Google Scholar] [CrossRef]
  182. Awadalla, A.; Hamam, E.T.; El-Senduny, F.F.; Omar, N.M.; Mahdi, M.R.; Barakat, N.; Ammar, O.A.; Hussein, A.M.; Shokeir, A.A.; Khirallah, S.M. Zinc oxide nanoparticles and spironolactone-enhanced Nrf2/HO-1 pathway and inhibited wnt/beta-catenin pathway in adenine-induced nephrotoxicity in rats. Redox Rep. 2022, 27, 249–258. [Google Scholar] [CrossRef]
  183. Ortega, M.T.; Riviere, J.E.; Choi, K.; Monteiro-Riviere, N.A. Biocorona formation on gold nanoparticles modulates human proximal tubule kidney cell uptake, cytotoxicity and gene expression. Toxicol. Vitro 2017, 42, 150–160. [Google Scholar] [CrossRef]
  184. Abdelmoneim, D.; Porter, G.; Duncan, W.; Lim, K.; Easingwood, R.; Woodfield, T.; Coates, D. Three-dimensional evaluation of the cytotoxicity and antibacterial properties of alpha lipoic acid-capped silver nanoparticle constructs for oral applications. Nanomaterials 2023, 13, 705. [Google Scholar] [CrossRef]
Figure 1. Animal models of kidney injury discussed in this article. These models include both acute kidney injury and chronic kidney disease.
Figure 1. Animal models of kidney injury discussed in this article. These models include both acute kidney injury and chronic kidney disease.
Nutrients 15 01732 g001
Figure 2. Lipoic acid is a cofactor of mitochondrial 2-ketoacid dehydrogenase complex, including pyruvate dehydrogenase complex, α-ketoglutarate dehydrogenase complex, and branched-chain amino acid dehydrogenase complex. The E1 subunit is 2-ketoacid decarboxylase using TPP as a cofactor; the E2 subunit is a dihydrolipoamide acyltransferase using lipoic acid as a cofactor; the E3 subunit is a dihydrolipoamide dehydrogenase, which uses NAD+ as an electron acceptor for the oxidation of the lipoyl group linked to the E2 subunit. E3 catalyzes the formation of the oxidized form of lipoic acid and generates NADH in the meantime [32,33,34].
Figure 2. Lipoic acid is a cofactor of mitochondrial 2-ketoacid dehydrogenase complex, including pyruvate dehydrogenase complex, α-ketoglutarate dehydrogenase complex, and branched-chain amino acid dehydrogenase complex. The E1 subunit is 2-ketoacid decarboxylase using TPP as a cofactor; the E2 subunit is a dihydrolipoamide acyltransferase using lipoic acid as a cofactor; the E3 subunit is a dihydrolipoamide dehydrogenase, which uses NAD+ as an electron acceptor for the oxidation of the lipoyl group linked to the E2 subunit. E3 catalyzes the formation of the oxidized form of lipoic acid and generates NADH in the meantime [32,33,34].
Nutrients 15 01732 g002
Figure 3. Lipoic acid is involved in thiol-disulfide exchanges that modulate cell’s redox and energy status.
Figure 3. Lipoic acid is involved in thiol-disulfide exchanges that modulate cell’s redox and energy status.
Nutrients 15 01732 g003
Figure 4. Biological actions of lipoic acid and its potential mechanisms, which include regeneration of glutathione, vitamins C and E, scavenging ROS, chelating metal ions, and anti-inflammatory power.
Figure 4. Biological actions of lipoic acid and its potential mechanisms, which include regeneration of glutathione, vitamins C and E, scavenging ROS, chelating metal ions, and anti-inflammatory power.
Nutrients 15 01732 g004
Figure 5. Protective effects of lipoic acid on diabetic kidney disease. Histological staining of the kidney tissues derived from control, diabetic, and diabetic + lipoic acid treatment. This figure was reproduced from reference [55]. For all the microscopic images, the amplification magnitude is ×800. (A) normal control group; (B) diabetic control group; (C) diabetic + ALA group. Glomerular hypertrophy, mesangial region expansion, proliferation of mesangial cells, and inflammatory cell filtration could also be observed in H&E staining. Disruption of glomerular basement membrane and mesangial region expansion could also be observed in PAS staining. Additionally, collagen fiber staining shows an increased intensity in the diabetic control group but exhibits a significant decrease in ALA-treated animals. For PAS staining, the red arrow indicates thickening and deformation in the glomerular basement membrane in the diabetic kidney. For Masson’s stained sections, the red arrows indicate the increased intensity of collagen fiber stain in the diabetic kidney.
Figure 5. Protective effects of lipoic acid on diabetic kidney disease. Histological staining of the kidney tissues derived from control, diabetic, and diabetic + lipoic acid treatment. This figure was reproduced from reference [55]. For all the microscopic images, the amplification magnitude is ×800. (A) normal control group; (B) diabetic control group; (C) diabetic + ALA group. Glomerular hypertrophy, mesangial region expansion, proliferation of mesangial cells, and inflammatory cell filtration could also be observed in H&E staining. Disruption of glomerular basement membrane and mesangial region expansion could also be observed in PAS staining. Additionally, collagen fiber staining shows an increased intensity in the diabetic control group but exhibits a significant decrease in ALA-treated animals. For PAS staining, the red arrow indicates thickening and deformation in the glomerular basement membrane in the diabetic kidney. For Masson’s stained sections, the red arrows indicate the increased intensity of collagen fiber stain in the diabetic kidney.
Nutrients 15 01732 g005
Figure 6. Protective effects of lipoic acid on folic acid-induced kidney injury. Histological staining of the kidney tissues derived from control, folic acid (FA), and folic acid + lipoic acid (LA). This figure was reproduced from reference [105]. (A) Representative images of hematoxylin & eosin (H&E) staining reflecting the histological changes in folic acid-induced acute kidney injury and low- and high-ALA-treated animal groups. (B) Renal damage assessed by Periodic acid/Schiff staining in folic acid-induced acute kidney injury and low- and high-ALA-treated animal groups.
Figure 6. Protective effects of lipoic acid on folic acid-induced kidney injury. Histological staining of the kidney tissues derived from control, folic acid (FA), and folic acid + lipoic acid (LA). This figure was reproduced from reference [105]. (A) Representative images of hematoxylin & eosin (H&E) staining reflecting the histological changes in folic acid-induced acute kidney injury and low- and high-ALA-treated animal groups. (B) Renal damage assessed by Periodic acid/Schiff staining in folic acid-induced acute kidney injury and low- and high-ALA-treated animal groups.
Nutrients 15 01732 g006
Figure 7. Mechanisms underlying lipoic acid’s role in various animal models of kidney injury discussed in the text. Lipoic acid alleviates kidney injury and improves kidney function by exerting various biological actions, as depicted in this figure.
Figure 7. Mechanisms underlying lipoic acid’s role in various animal models of kidney injury discussed in the text. Lipoic acid alleviates kidney injury and improves kidney function by exerting various biological actions, as depicted in this figure.
Nutrients 15 01732 g007
Table 1. Mechanisms of ALA’s renal protection in various animal models of kidney injury discussed in this review.
Table 1. Mechanisms of ALA’s renal protection in various animal models of kidney injury discussed in this review.
Model of Kidney InjuryMechanismReference
Diabetic kidney injuryActivating Nrf2, decreasing
oxidative stress, enhancing mitochondrial function, and inhibiting fibrosis
[7,29,50,51]
Sepsis-induced kidney injuryEnhancing autophagy, inhibiting NF-KB,
attenuating mitochondrial oxidative stress,
inhibiting inflammatory cytokine release
[22,71,72,73]
Ischemia/reperfusion injuryCounteracting oxidative stress,
downregulating Na-K-ATPase and NOS,
mitigating neutrophil infiltration,
inhibiting inflammation, and
suppressing endothelin-1 upregulation
[75,78,79,80,81,82]
UUO-induced kidney injuryAttenuating oxidative stress,
decreasing nitric oxide production, decreasing transforming factor-1 expression, and
ameliorating mesenchymal transition
[88,96]
Cisplatin-induced kidney injuryIncreasing glomerular filtration,
lowering plasma creatinine levels,
increasing creatinine clearance, and
attenuating oxidative damage
[98,99,101,103]
FA-induced kidney injuryBlocking p53 from causing ferroptosis[105]
Cadmium-induced kidney injuryChelating cadmium,
decreasing oxidative stress,
elevating glutathione content, and
decreasing apoptosis
[108,110,111,112]
Iron-induced kidney injuryAttenuating oxidative damage,
inhibiting p38 MAPK,
inhibiting NADPH oxidase4, and
chelating iron
[126,127,128,129]
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

Kamt, S.F.; Liu, J.; Yan, L.-J. Renal-Protective Roles of Lipoic Acid in Kidney Disease. Nutrients 2023, 15, 1732. https://doi.org/10.3390/nu15071732

AMA Style

Kamt SF, Liu J, Yan L-J. Renal-Protective Roles of Lipoic Acid in Kidney Disease. Nutrients. 2023; 15(7):1732. https://doi.org/10.3390/nu15071732

Chicago/Turabian Style

Kamt, Sulin F., Jiankang Liu, and Liang-Jun Yan. 2023. "Renal-Protective Roles of Lipoic Acid in Kidney Disease" Nutrients 15, no. 7: 1732. https://doi.org/10.3390/nu15071732

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