Next Article in Journal
The Role of Curcumin in Oral Health and Diseases: A Systematic Review
Previous Article in Journal
Nanoemulsions Based on Soluble Chenopodin/Alginate Complex for Colonic Delivery of Quercetin
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advance in Iron Metabolism, Oxidative Stress and Cellular Dysfunction in Experimental and Human Kidney Diseases

1
Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
2
Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
3
Department of Nephrology, The First Hospital of China Medical University, Shenyang 110001, China
*
Author to whom correspondence should be addressed.
Antioxidants 2024, 13(6), 659; https://doi.org/10.3390/antiox13060659
Submission received: 6 May 2024 / Revised: 22 May 2024 / Accepted: 24 May 2024 / Published: 27 May 2024

Abstract

:
Kidney diseases pose a significant global health issue, frequently resulting in the gradual decline of renal function and eventually leading to end-stage renal failure. Abnormal iron metabolism and oxidative stress-mediated cellular dysfunction facilitates the advancement of kidney diseases. Iron homeostasis is strictly regulated in the body, and disturbance in this regulatory system results in abnormal iron accumulation or deficiency, both of which are associated with the pathogenesis of kidney diseases. Iron overload promotes the production of reactive oxygen species (ROS) through the Fenton reaction, resulting in oxidative damage to cellular molecules and impaired cellular function. Increased oxidative stress can also influence iron metabolism through upregulation of iron regulatory proteins and altering the expression and activity of key iron transport and storage proteins. This creates a harmful cycle in which abnormal iron metabolism and oxidative stress perpetuate each other, ultimately contributing to the advancement of kidney diseases. The crosstalk of iron metabolism and oxidative stress involves multiple signaling pathways, such as hypoxia-inducible factor (HIF) and nuclear factor erythroid 2-related factor 2 (Nrf2) pathways. This review delves into the functions and mechanisms of iron metabolism and oxidative stress, along with the intricate relationship between these two factors in the context of kidney diseases. Understanding the underlying mechanisms should help to identify potential therapeutic targets and develop novel and effective therapeutic strategies to combat the burden of kidney diseases.

1. Introduction

Kidney disease, another name for renal diseases, is a condition in which the kidney is injured and unable to function correctly. The kidneys function by filtering waste and excess fluids from the blood to form urine. When the kidneys are not functioning properly, waste products will accumulate in our body and cause various health problems [1]. There are different types of kidney diseases, including acute kidney injury (AKI) and chronic kidney disease (CKD) and diabetic kidney disease (DKD). AKI occurs suddenly and is often reversible with prompt treatment, while CKD develops over time and can lead to permanent kidney damage [2,3]. DKD is a kidney damage caused by diabetes, and is a specific type of CKD and one of the prevalent etiologies of CKD [4]. Diabetic patients develop DKD due to the effects of long-term high blood sugar and suffer from impaired kidney function. IgA nephropathy is also a type of CKD characterized by immune-mediated deposition of immune complexes in the mesangial cells of the kidneys, leading to chronic inflammation and kidney damage [5].
CKD and AKI are prevalent conditions with significant implications for public health worldwide. The intricate interplay of iron metabolism and oxidative stress is gaining significance in elucidating the pathophysiology and advancement of these renal diseases. Iron, a crucial micronutrient involved in numerous cellular functions, is tightly regulated in the body to maintain homeostasis [6]. A deviation from this fine equilibrium may result in abnormal iron levels, which can aggravate and accelerate CKD and AKI.
In CKD, the dysregulation of iron metabolism can have profound effects on renal function [7]. Iron overload, often observed in CKD patients undergoing hemodialysis, can promote oxidative stress via the overproduction of ROS [8]. ROS, in turn, can induce renal fibrosis and dysfunction, exacerbating the progression of CKD. Conversely, iron deficiency in CKD patients can impair erythropoiesis and exacerbate anemia, further complicating disease management [9].
Iron metabolism and oxidative stress also play a critical role in the pathophysiology of AKI. For example, ischemic AKI, a common form of AKI, is characterized by renal hypoxia and reperfusion injury, also resulting in the overproduction of ROS and oxidative stress [10]. The interaction of iron metabolism and oxidative stress in AKI can exacerbate renal injury and impair recovery, emphasizing the necessity of understanding the underlying mechanisms of acute kidney damage.
In this review, we discuss the roles and molecular mechanisms underlying iron dysregulation and oxidative stress in AKI, CKD and DKD. We also discuss the intricate relationship between iron metabolism and oxidative stress in kidney diseases. Understanding the molecular mechanisms underlying iron dysregulation and oxidative stress can aid in the identification of the prospective treatment targets and novel therapeutic techniques to mitigate renal damage and the process of kidney diseases.

2. The Regulation of Iron Metabolism

2.1. Iron Absorption from Guts to Plasma

After birth, human iron intake depends entirely on dietary iron, which consists mainly of heme iron and non-heme iron [11]. Of these, heme iron is mainly derived from hemoglobin and myoglobin contained in meat and fish. Non-heme iron comes mainly from vegetables and cereals [12]. Heme iron and non-heme iron are absorbed at different rates: heme iron has an absorption rate of approximately 20%, while non-heme iron typically has a lower absorption rate ranging from 2% to 5% [12]. Despite slight variations in the site of absorption, overall, dietary iron is absorbed in the small intestine [13]. Divalent metal transporter 1 (DMT1), located in the apical membrane of enterocytes, is capable of transporting non-heme iron from the intestinal lumen into the enterocyte via proton coupling [14]. However, DMT1 is only responsible for the transport of divalent metal ions; most of the iron in food is in the form of ferric ion. Thus, ferric iron must first be converted into ferrous iron at the brush border of the endocytes by a ferric reductase, duodenal cytochrome b (DCYTB) [15]. Iron is typically stored in ferritin within enterocytes [16]. Animal experiments indicate that in cases of iron deficiency, this iron is released into the plasma through ferroportin (FPN) with the assistance of hephaestin [17,18]. For heme iron, Majid Shayeghi et al. found that a membrane protein named heme carrier protein 1 (HCP1) is capable of its transportation into enterocytes [19].

2.2. Plasma Iron Transportation and Uptake by Other Cells

The transportation of iron throughout the body is facilitated by transferrin (TF), a protein that binds to iron tightly, allowing for uptake by all types of cells. This process requires recognition by transferrin receptor protein 1 (TFR1) [20], a type II dimeric transmembrane receptor [21]. TFR1 is located on all cell membranes and serves as the main receptor for iron entry into most cells. Human transferrin (hTF), a bilobal glycoprotein that is secreted from the liver into the bloodstream, has a high affinity for reversible binding of ferric irons [22]. It has been indicated that a deficiency in TF could lead to the development of anemia, iron overload in specific tissues, and potentially fatal outcomes [23]. TFR1 forms a 2:2 equimolar complex with hTF, with each TFR1 monomer binding to one molecule of hTF [24]. Then, the TF-TFR1 complex is internalized into cells through clathrin-mediated endocytosis [25]. Upon endosomal entry into the cell and subsequent removal of the clathrin proteins, protons flow inward into the endosome, which leads to the acidification of the endosomal environment. This, in turn, induces conformational changes in the complex to mediate the release of ferric iron [26]. The released ferric iron is subsequently reduced to ferrous iron by the six transmembrane epithelial antigen of the prostate family of reductases (STEAP) or by lysosomal cytochrome B (LCYTB), and transported to the cytoplasm by intracellular ferrous iron transporters including DMT1 and zinc transporter (Zip14) [27,28] in in vitro experiments. After releasing the iron, apo-TF/TFR1 complex (without TF-bound iron) returns to the cell surface and dissociates.
TF-bound iron is generally considered as the main source of iron for most cells, whereas it has been reported that mice embryos lacking TF or TFR can still develop normally [29], suggesting that the alternative pathways for iron acquisition may exist. For example, ferritin not only functions during iron storage, but can also be secreted outside the cell to function as an iron donor [30]. During mouse embryonic kidney development, scavenger receptor class A member 5 (SCARA5) interacts with ferritin and promotes its endocytosis [31]. In human cells, TFR1 also interacts with ferritin and promotes its endocytosis [32,33]. In addition, it has been found that Zip14 is also responsible for the transportation of non-transferrin-bound iron (NTBI) into mouse hepatocytes and pancreatic cells [34]. A homologue of ZIP14, ZIP8, functions as an NTBI transporter in human proximal renal epithelial cells [35]. In sum, cellular iron uptake is a complex process and requires multiple molecular synergies. Therefore, further in-depth studies are essential for exploring the mechanism of iron uptake.

2.3. Cellular Iron Transfer to Mitochondria and Its Utilization

Upon entering the cytoplasm, ferrous iron is directed towards the labile iron pool (LIP) [36], which is predominantly utilized by the mitochondria for the synthesis of Fe-S cluster proteins and hemoglobin. To avoid the redox toxicity of LIP, the remaining iron combines with iron chaperones after being released from endosomes via DMT1 [37], and then transfers to ferritin for storage (see below). For example, Vyoral et al. identified a high-molecular-weight intermediate in K562 cells incubated with TF, which appeared to transfer iron from chaperones to ferritin [38]. Furthermore, Shi et al. have shown that poly rC binding protein 1 (PCBP1) also acts as an iron molecular chaperone in yeast cells and Huh7 cells, donating iron to ferritin [39]. Recently, Wang et al. observed a decrease in iron and ferritin levels in the intestinal epithelium of PCBP1-deficient mice, indicating that PCBP1 is also crucial in regulating intestinal iron absorption and maintaining iron homeostasis in the body [40]. Except for transporting iron from the LIP to mitochondria, endosomes containing TF can also directly and temporarily attach to mitochondria to transport iron into them, bypassing the LIP [36]. This idea has been further developed into the “kiss and run” hypothesis [41,42]. Studies have demonstrated that myosin Vb [43], the cytoskeletal regulatory molecule MRCKα [44], and vesicle docking [45,46] all play a role in “kiss and run” hypothesis in in vivo and in vitro experiments. However, the exact mechanism of endosomal and mitochondrial contact is currently unknown. As iron enters the mitochondria, both outer and inner membranes must be crossed to reach the matrix for Fe-S cluster assembly and heme biosynthesis. DMT1 may be involved in iron crossing the outer mitochondrial membrane [47], whereas in vitro experiments clarified that iron flux through the inner mitochondrial membrane is dependent on mitoferrins 1 and 2 (MFRN1 and MFRN2) [48]. MFRN1 and MFRN2 are differentially expressed in tissues. MFRN1 is only highly expressed in erythrocytes whereas MFRN2 is widely expressed in mammalian tissues [49,50].
Within the mitochondria, iron normally has three destinations: the synthesis of hemoglobin [51], the synthesis of Fe-S clusters [52], and storage in mitochondrial ferritin [53]. The multiple steps of heme and Fe-S cluster synthesis occur in mitochondria and cytoplasm. In addition, there is a crosstalk between Fe-S clusters and heme. For example, a Fe-S cluster protein, ferrochelatase (FECH) catalyzes ferrous iron insertion into protoporphyrin (PPIX) macrocycles, which is the last step of heme synthesis [54].

2.4. Cellular Iron Storage

Iron that is temporarily unavailable to cells is stored in ferritin in cytoplasm and mitochondria. The ferritin molecule is composed of a spherical protein shell and an iron-containing core [55]. The protein shell consists of 24 polypeptide chains of different ratios of H and L subunits and its inner surface forms multiple contact cores with up to 4500 ferric iron [56]. The H subunit has redox activity and can catalyze ferrous iron oxidation to storable ferric iron [57], while the L subunits stabilizes the ferritin structure and store ferric iron by mineralization [58]. The ratio of H and L subunits can be varied in different cells, which determines the rate of metal mineralization [59,60]. When required, iron-containing ferritin is degraded by nuclear receptor coactivator 4 (NCOA4)-dependent autophagy (ferritin autophagy), allowing iron to flow back into the LIP [61]. Iron is stored in mitochondrial ferritin in mitochondria. Mitochondrial ferritin, like ferritin in the cytoplasm, has redox activity [53]. However, unlike cytoplasm ferritin, the expression of mitochondrial ferritin varies in different tissues. This suggests that the function of mitochondrial ferritin may be distinct from that of ferritin in cytoplasm.

2.5. Cellular Iron Export

Cellular iron export is a tightly regulated process that maintains intra- and extracellular iron homeostasis. Iron export is mainly divided into two pathways: FPN-dependent and independent pathways. FPN is characterized by two 6-membrane-helix bundles to form a channel for iron exportation [62,63]. Upon acceptance of Fe(II) carried by PCBP2, FPN undergoes a conformational change, leading to a channel opening to delivery of iron outside the cell [64]. To determine the critical role of FPN in iron absorption, it has been found that selective deletion of FPN from intestinal cells results in anemia in mice. In addition, knockout of FPN in macrophages and hepatocytes also developed anemia in mice fed with a low-iron diet but the mice fed with a normal diet had normal erythropoiesis. This result suggests that normal iron absorption can compensate for impaired iron recycling and iron release from storage tissues [65,66]. Aside from transferring iron to plasma, a study in a mouse model showed that FPN is also capable of releasing labile toxic iron from hemoglobin molecules that have been damaged [67].
Although FPN is the main factor for cellular iron export, other factors are also involved in this process. As mentioned above, in addition to functioning as an important molecule for iron storage, FPN also exists in the extracellular space as a carrier of iron ions [68]. Iron-containing ferritin can be secreted by ferritinophagy [30,69] or endosomal microautophagy [70,71] by membrane fusion. Ferritin secretion in exosomes can be stimulated by ferroptosis, iron overload and high lipid activity [68,70,72]. Iron can also be excreted from cells as heme iron. Feline leukemia virus subgroup C cellular receptor 1 (FLVCR1) on the cell membrane is responsible for heme transport [73], and mediates mouse erythroid differentiation [74]. The mutations of FLVCR1 cause abnormal oxidative stress responses in sensory neurons, resulting in sensory nerve-related diseases [75].

2.6. The Regulatory Mechanisms of Iron Hemostasis

2.6.1. The Iron-Regulatory Protein (IRP) and Iron-Responsive Element (IRE) Regulatory System

Iron hemostasis can be regulated post-transcription. Iron-regulatory protein 1/2 (IRP1/2) can bind with cis-regulatory iron-responsive elements (IREs), which are specific stem-loop structures within the untranslated regions (UTRs) of the messenger RNAs (mRNAs) that encodes a variety of iron-related proteins [76]. The combination of IRP1 and IREs is regulated by the concentration and cellular need of iron. When iron concentration is low, IRP1 can bind with the 5′ UTR of the ferritin and FPN mRNAs to block their translation and bind with the 3′ UTR of the TFR1 mRNA to protect it from degradation by endoribonuclease regnases [77,78,79]. Conversely, when cellular iron concentration is high, IRP1 disassociates from IREs, thereby allowing ferritin and FPN for translation and exposing the TFR1 mRNA to degradation. Thus, the activation of the IRP-IRE system allows cells to have lower iron storage and export, but higher iron absorption. IRP2 regulation is crucial for embryonic development and hematopoiesis [80]. In addition to ferritin, FPN and TFR, the mRNAs of other iron-related proteins, such as DMT1 and HIF-2α, also contain IRE structures [76]. The IRP-IRE system is important for embryonic development, intestinal and liver function, immune function of macrophages and T-cells, erythropoiesis and iron management, and the development of neurological disorders [81,82,83,84,85,86].

2.6.2. The Hypoxia-Inducible Factor (HIF) Regulatory System

HIF plays a crucial role in intestinal iron absorption. HIF is a heterodimeric nuclear transcription factor for many iron metabolism-related genes, such as TFRC (encoding TFR), SLC11A2 (encoding DMT1), etc. HIF consists of an oxygen-sensitive α subunit (HIF-1α/HIF-2α) and a common expressed β subunit (HIF-1β) [87,88,89]. Under normal oxygen conditions, two proline residues on HIF-1α and HIF-2α are hydroxylated by iron-dependent prolyl hydroxylase (PHD) and then recognized and degraded by the ubiquitin-proteasome system [90]. The functional integrity of PHD requires the cofactors ferrous iron and oxygen. Hence, under hypoxic or iron-deficiency conditions, the inactivation of PHD causes HIF-1/2α stabilization and combination with HIF-1β. Then, the complex is translocated into the nucleus, in which the complex binds to hypoxia response elements (HREs) to promote the transcription of hypoxia-inducible genes, including EPO, VEGF and GAPDH, to enhance angiogenesis and glycolysis process [91,92,93]. Of note, although HIF-2 overlaps with HIF-1 in some functions (e.g., regulating VEGF and EPO expression), HIF-2 plays a bigger role in iron metabolism. HIF-2 can directly bind to the promoter regions of the TFR, DCYTB and DMT1 genes, thereby promoting their transcription under hypoxic or iron-deficient conditions [94,95]. In addition, there is feedback between HIF-2 and the IRP-IRE regulatory pathway, in that the IRE region of HIF-2α can be bound by IRP to inhibit HIF-2α expression [96,97].

2.6.3. Hepcidin–Ferroportin Regulatory System

It is now known that FPN is highly expressed on the membrane surface of macrophages, enterocytes and hepatocytes, all of which are involved in increased iron fluxes. In contrast, the key iron-regulating hormone hepcidin (HAMP or HEPC), produced by hepatocytes, negatively feedback-regulates plasma membrane FPN levels [98,99]. Hepcidin modifies FPN through post-translational modification, leading to its ubiquitination, endocytosis and degradation by lysosomes [100,101]. Elevated iron levels in the circulation stimulate hepcidin expression, which is mediated by iron-sensing proteins (e.g., HFE, TFR2, HJV and BMP6) that sense changes in iron levels and promote the transcription of the hepcidin gene (HAMP) through the SMAD pathway [102,103,104]. The regulation between systemic iron circulation and cellular iron metabolism is shown in Figure 1.

3. Renal Iron Homeostasis and Cellular Dysfunction in Kidney Diseases

3.1. Renal Iron Homeostasis

In addition to the universal iron transport proteins and regulatory pathways described above, there are kidney-specific iron uptake mechanisms. Due to different expressions of iron transport-related proteins, the ability to process iron varies greatly between different renal segments [105,106]. Under physiological conditions, proximal tubule epithelial cells can take up transferrin-bound iron (TFBI) from the apical membrane via TFR1, or via megalin-dependent and cubilin-mediated endocytosis [107,108]. The expression of TFR1 and megalin–cubilin complex is negatively associated in proximal tubules. Under the iron overloaded condition, the expression of TFR1 is decreased by the IRP-IRE in the kidney, whereas the expression of megalin receptor complex is increased in mouse proximal tubules [109]. In contrast, under iron deficient conditions, such as iron chelation, the expression of TFR1 is increased and the expression of the megalin–cubilin complex is decreased [110]. These results suggest a possibly converse effect between TFR1 and megalin–cubulin. Hemolytic conditions cause the mammalian proximal tubule to overburden with hemoglobin, resulting in the participation of both megalin and the neutrophil gelatinase-associated lipocalin receptor (NGALR) in the uptake of the filtered hemoglobin [111,112]. The level of ferritin and the abundance and activity of IRP1 are also higher in proximal tubules than in distal tubules, indicating that proximal tubules rather than distal tubules are primarily involved in cellular iron flux and renal iron reabsorption [108]. Furthermore, ZIP8, ZIP14 and DMT1 are expressed in the proximal and distal tubules in human kidney biopsy samples [113]. Given that ZIP8 and/or ZIP14 function at a higher pH than DMT1 for maximum iron transport, we can speculate that ZIP8 and/or ZIP14 may facilitate apical uptake of NTBI in mouse proximal tubules, while DMT1 may participate in subcellular uptake of apical NTBI [28,114,115]. In addition to the proximal tubule, the thick ascending limb of Henle’s loop, the distal tubule and collecting duct can also reabsorb TFBI for the expression of TFR1 and NGALR on the apical part of the cell [108,116].
Apart from the tubule’s reabsorption, glomerular epithelial cells also have an iron reabsorption function because of the expression of TFR1, FPN and DMT1 on cultured human glomerular epithelial cells [117]. Alfonso et al. show that human podocytes can take up hemoglobin through macrophage-mediated autophagy and metabolize it via heme oxygenase 1 (HO-1) degradation [118].

3.2. Abnormal Iron Metabolism and Cellular Dysfunction in Renal Injury and Disease

Iron homeostasis plays a vital role in maintaining normal organism function. Disrupted iron homeostasis can significantly impact various organs through both systemic iron overload and iron deficiency. Systemic iron overload leads to elevated plasma TFBI and non-transferrin bound iron (NTBI) levels and increased intracellular LIP, causing structural alternations of diverse proteins, and DNA due to the high redox capacity of iron [119]. An increased urinary iron excretion and renal iron deposition has been found in hemochromatosis (HH) mice [120]. Clinical case reports also indicate that HH can lead to hemosiderin deposition in kidneys and injured kidneys [121,122]. Hemochromatosis is an inherited disorder most commonly caused by mutations in the HFE gene that result in misfolding of the HFE protein, loss of function in sensing iron concentration and defective hepcidin synthesis in hepatocytes, which in turn leads to reduced ubiquitinated degradation of FPN, elevated FPN levels and increased release of cellular iron, resulting in elevated circulating iron levels [123]. In addition, hepcidin deficiency leads to an elevation of plasma TF saturation, inducing the accumulation of toxic NTBI in plasma [124]. Prolonged exposure of the kidney to enhanced levels of non-heme iron was found to potentially lead to renal injury in cultured porcine kidney epithelial cells [125].
Elevated renal injury markers were additionally observed in infants and children with iron deficiency anemia and could be corrected with oral iron therapy [126]. These findings suggest that an iron deficit may cause kidney damage and has a deleterious effect on renal function. In addition, patients with a variety of glomerular and tubular disorders, such as diabetic nephropathy and Fanconi syndrome, also have abnormal content of iron and TF in their urine [127,128,129,130,131]. In addition, patients with glomerular disease often exhibit hypo-transferrinogenemia due to the inability to adequately synthesize TF through the liver. However, there is little clinical evidence regarding whether iron deficiency impairs renal function.

4. The Regulation of Redox Homeostasis in the Kidneys

4.1. ROS and Oxidative Stress in the Kidneys

ROS can be categorized into two major classes of free radical- and non-free radical-oxidizing molecules (Table 1). Free radicals are molecules or atoms with unpaired electrons that are highly oxidizable and can directly react with intracellular biomolecules (e.g., proteins, lipids, DNA) in oxidative reactions, leading to cellular damage and oxidative stress [132]. Non-free radical ROS are oxidizing molecules that do not carry unpaired electrons and are less reactive compared to free radical ROS, but they can be converted to free radicals or participate in other oxidative reactions that can also cause cellular damage. ROS act in both directions, either as the “culprits” of oxidative stress, destroying cellular structures and causing damage (see Section 5), or as cellular signaling molecule that regulates various physiological processes. For example, hydrogen peroxide participates in the activation of the epidermal growth factor receptor (EGFR), which in turn activates a number of downstream signaling pathways and promotes cell proliferation and survival [133,134,135]. Mitochondrial ROS also play an important role in T cell activation, and inhibition of mitochondrial ROS production can reduce T cell activation in vivo or in vitro experiments [136]. Recent studies have shown that mitochondrial ROS also contribute to the bactericidal activity of macrophages [137]. In addition, ROS play an important role in the regulation of cellular senescence and stem cell differentiation, which will not be discussed in this article.
The primary producers of endogenous ROS in the kidneys of mammals are mitochondria and NADPH oxidases (NOX) [138,139,140]. As kidneys require large amounts of energy (ATP) for filtration and reabsorption, mitochondria serve as the main source of cellular ATP to power these activities [141]. Mammalian mitochondria can produce superoxide (O2) and/or hydrogen peroxide through substrate catabolism and electron leakage in the electron transfer chain (ETC). During electron transfer, electrons may escape into oxygen to yield O2 and/or hydrogen peroxide. As the substrate of Fenton reaction, hydrogen peroxide can react with ferrous iron from LIP and produce hydroxyl radicals, a ROS with strong oxidization activity [142]. Previous studies suggest ROS are mainly produced from ETC complexes I and III, while mitochondrial pyruvate dehydrogenase and α-ketoglutarate dehydrogenase are involved in this process [143]. Most mitochondrial enzymes near ROS production centers contain thiol groups modified by ROS, implying that mitochondrial ROS production is tightly self-regulated [144]. The NOX family contains seven homologous isoforms differentially distributed among tissues, in which NOX4 is predominant in kidneys [145]. NOX4 is localized mainly to mitochondria, the endoplasmic reticulum and plasma membrane [145,146]. Due to its unique E-loop structure, NOX4 can produce hydrogen peroxide rather than superoxide [147].
Stimulated by oxidative stress, NO synthase (NOS) can be converted from NO-producing enzymes to superoxide-producing enzymes, a process that is known as NOS uncoupling [148]. Uncoupling of NOS leads to NOS dysfunction and contributes to ROS overproduction. Three types of NOS exist in the body: neuronal NOS (nNOS, also called NOS1), inducible NOS (iNOS, also called NOS2) and endothelial NOS (eNOS, also called NOS3). nNOS and eNOS are constitutively expressed, whereas iNOS expression associates with inflammation [148]. It has been found that nNOS is expressed in cortical tubules and eNOS is expressed in glomeruli [149]. NOS mainly mediates endogenous NO synthesis [149]. NO can induce cGMP production, a second messenger which affects cardiovascular, renal and metabolic systems beneficially. However, NOS uncoupling increases ROS production and decreases NO bioavailability, which exacerbates CKD [149,150]. A key eNOS function is to regulate blood flow and maintain endothelial integrity. Mice lacking eNOS exhibited symptoms similar to metabolic syndrome and developed severe kidney disease [151,152]; while supplementing nitrate in the diet could reverse the disease outcome [153]. In addition, eNOS uncoupling and NOX activation were found to generate ROS, leading to endothelial dysfunction in DKD rats [154,155]. When ROS accumulate and cells undergo oxidative stress, the eNOS coenzyme tetrahydrobiopterin (BH4) oxidizes to BH2 by ROS, inducing eNOS uncoupling [156]. At this point, NOS becomes unstable, transferring electrons from L-arginine that should fuel eNOS to oxygen instead, producing O2 [157]. Treatment with BH4 prevents eNOS uncoupling, reduces ROS and improves endothelial function [158,159]. Patients treated with BH4 in clinics showed decreased urinary albumin excretion [154,160].

4.2. Antioxidant Defense Systems in the Kidneys

Like most tissues and organs, the kidney contains an enzymatic and a non-enzymatic antioxidant system (Table 2). The enzymatic system consists of superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPX) and thioredoxin (Trx). The non-enzymatic system is mainly composed of reduced glutathione (GSH) and other molecules.
SOD catalyzes the transformation of superoxide anion into oxygen and hydrogen peroxide, mitigating the effects of ROS. The activity of SOD in mammalian kidneys is largely mediated by SOD1, and reduction of SOD1 has been associated with kidney injury [161,162]. CAT is localized in peroxisomes, which are highly expressed in the liver, lungs and kidneys and reduce SOD-generated hydrogen peroxide to oxygen and water [163]. Antioxidant enzymes like CAT, SOD1 and PRX are found in peroxisomes, which are organelles that control the oxidative state of cells [164]. Targeted phosphorylation of the peroxisomal membrane protein PEX14 shields DNA from ROS by elevating cytoplasmic levels of CAT during mitotic nuclear membrane dissolution in mammalian cells [165]. However, peroxisomes are also involved in fatty acid breakdown through β-oxidation, which produces hydrogen peroxide as a byproduct [166].
GPX can function similarly to CAT to reduce SOD-derived hydrogen peroxide to oxygen and water [167,168]. In addition, GPX specifically reduces lipid peroxides and prevents the lipid peroxidation chain reaction, thereby preventing cell membrane disruption and cell necrosis [167]. Of these, GPX4 is the only known intracellular enzyme that is resistant to phospholipid peroxidation, and GPX4 deficiency is associated with ferroptosis through increased ROS, lipid peroxidation and iron overload [169,170].
Trx primarily accomplishes its antioxidant role by passing electrons to Trx-dependent peroxidases (Prxs), methionine sulfoxide reductases (Msr) and other oxidation-sensitive molecules [171,172]. The Trx system can transfer electrons to Prxs and Msr to remove ROS [173]. Trx then restores redox activity by receiving electrons from TrxR [174]. NADPH, in turn, can transfer electrons to Trx reductase (TrxR) via FAD [175,176]. Furthermore, it was found that GSH can reduce oxidized Trx in vitro, suggesting that there may be crosstalk between the Trx system and GSH [177].
GSH is involved in the scavenging of peroxides as a cofactor of GPX [178,179]. In addition, GSH can reduce two oxidation products, protein thiyl radicals and protein thiolate, back to protein sulfhydryl groups (protein-SH) [180]. GSH can also restore the free radicals of other antioxidants that have lost their antioxidant capacity, such as vitamins C and E, back to their functional, non-free radical state [181].

5. Oxidative Stress and Cellular Dysfunction in Kidney Diseases

Physiological concentration of ROS makes them function as signaling molecules that control a number of critical physiological processes [182]. However, overproduction of ROS disrupts redox homeostasis, leading to oxidative damage to important biomolecules such as DNA, proteins and lipids [183].

5.1. Oxidative Stress and Renal Tubular Cell Dysfunction in Kidney Disease

It is generally estimated that each human cell is exposed to about 105 attacks per day from ROS such as hydroxyl radicals [184]. Damage to DNA by ROS identified so far includes bases modification, deletions, translocations, DNA strand breaks and DNA-protein cross-links and chromosomal rearrangements [185]. Hydroxyl radicals, which can react with all of DNA’s constituents, including the deoxyribose backbone and purine and pyrimidine bases, are primarily responsible for DNA damage [186,187]. These injuries lead to genetic variations, such as mutations and chromosomal rearrangements, which may lead to cell senescence [188]. Renal tubular epithelial cells are commonly associated with renal senescence. Increased tubular epithelial cell senescence was found in animal models of AKI (IRI) and CKD (UUO) as well as in CKD patients [189,190]. Senescent cells are able to secrete various cytokines, such as IL-1β, IL-6, IL-8 and TGFβ1, collectively known as the senescence-associated secretory phenotype (SASP), which significantly affects neighboring cell and tissue functions [191,192,193]. SASP mediates inflammatory and pro-fibrotic responses, which activate renal interstitial fibroblasts, contributing to an overproduction of matrix proteins, which disrupts the physiological structure of renal tissues and promotes the formation of fibrous scars [194].
Apoptosis has been observed in human renal tubular cells of diabetic patients due to high glucose-induced overproduction of ROS and disruption of mitochondrial structure, which leads to decreased mitochondrial respiratory function and ATP production, thus causing energy metabolism disorders and renal tubular cells’ apoptosis [195,196]. In addition, the upregulation of kidney injury molecule-1 (KIM-1) in mouse renal tubular epithelial cells during the course of DKD led to an increase of the internalization of palmitic acid albumin by proximal tubular cells and an activation of NLRP3 inflammatory vesicles by palmitic acid via ROS production, leading to the production of pro-inflammatory factors, such as IL-1β, thereby inducing renal tubular cell death, mitochondrial fragmentation and subsequent fibrosis [197,198,199].
Acute tubular injury is the most frequent pathologic manifestation of AKI. The etiology of prerenal AKI is inadequate perfusion due to decreased renal blood flow, which usually manifests as acute ischemic tubular damage [200]. Under ischemia and hypoxia, renal tubular cells, which are highly metabolically active, are severely injured due to their high oxygen demand [201]. The most common initial manifestations of ischemic AKI are ATP reduction and the alterations of mitochondrial structures [202]. The conditions of hypoxia and ischemia can induce oxidative stress, which leads to strong oxidative damage to mitochondrial lipids and proteins, disrupting energetic metabolism via disrupting ETC function [203]. According to multiphoton imaging reports, the mitochondrion is the major source of ROS in a mouse ischemic AKI model [203]. Subsequently, post-injury renal tubular epithelial cells undergo cell death, such as apoptosis and necrosis, releasing inflammatory mediators that in turn exacerbate tubular injury [204]. In addition, reperfusion after ischemia also causes injury due to mitochondrial ROS bursts [205,206], which can induce infiltration of inflammatory cells, such as neutrophils, early in IRI. ROS and inflammatory factors originating from neutrophils can further aggravate renal tubular injury in rats [207,208]. Furthermore, uncoupling protein 1 (UCP1) in renal tubular epithelial cells is downregulated in a time-dependent manner during renal ischemia–reperfusion, resulting in elevated renal oxidative stress and exacerbating ischemia-induced AKI in mice [209].
ROS are also involved in several aspects of renal tubular cell injury in septic AKI (SAKI). Pathogen invasion activates the innate immune system, and neutrophils and macrophages protect the organism with oxidants such as hydroxyl radicals and peroxides [210]. The generation of ROS during defense can activate the inflammatory response to increased iNOS, resulting in excess NO, which in turn leads to uncoupling of eNOS and production of highly reactive peroxynitrite radicals [211,212]. In addition, excess NO reacts with peroxo-anion radicals to generate peroxynitrite to directly damage renal tubular cells [213,214].
Nephrotoxic drugs such as antibiotics and chemotherapeutic drugs can directly damage kidney tubular cells, leading to AKI. For example, cisplatin increases ROS production and depletes antioxidant molecules such as GSH in vivo, leading to endogenous ROS accumulation in renal tubular cells and then cell death [215]. In addition, cisplatin may induce mitochondrial dysfunction, leading to increased mtROS production in microsomes via cytochrome P450 enzymes, further exacerbating oxidative stress injury in porcine kidneys [216].

5.2. Oxidative Stress and Podocyte Dysfunction in Kidney Diseases

Loss of podocytes is a feature of early diabetic nephropathy and is involved in the progression of DKD. Increased extracellular glucose has been found to stimulate intracellular ROS production via NOX and mitochondrial pathways, which leads to the activation of p38 mitogen-activated protein kinase and caspase 3, ultimately resulting in podocyte apoptosis [217]. Further studies revealed that Ras-related C3 botulinum toxin substrate 1 (Rac1) plays a role in podocyte injury by regulating multiple signaling pathways [218,219]. Rac1 can be activated by various stimuli, including ROS, inflammatory cytokines, angiotensin II and high glucose concentration [220,221,222]. The activation of Rac1 activates various downstream effector molecules and signaling proteins, such as WAVE complex, mDia2, PAK and LIM kinase, which are involved in actin cytoskeleton remodeling, leading to increased mouse podocyte motility [223]. Normal podocytes have a stationary phenotype, whereas damaged podocytes are transformed into a Rac-dependent motile phenotype. The motile phenotype is related to the increase of podocyte motility, loss of podocyte process and proteinuria [224]. Motile podocytes may eventually be depleted, leading to podocytopenia and focal segmental glomerulosclerosis (FSGS) [225]. In addition, angiotensin II activates the Rac1-NOX-ROS cascade reaction by binding to the type I angiotensin II receptor, leading to rearrangement of the actin cytoskeleton of podocytes and increased motility [226]. Similar to renal tubular epithelial cells, podocytes can internalize plasma proteins via free fatty acid receptor-mediated endocytosis, leading to protein accumulation in endoplasmic reticulum (ER) and dysregulation of unfolded protein reaction pathways, ultimately causing ER stress and mitochondrial damage [227,228]. ER stress is considered to be induced by ROS, whereas mitochondrial damage generates ROS, creating a vicious cycle that is detrimental to podocytes.

5.3. Oxidative Stress and Mesangial Cell and Interstitial Fibroblast Dysfunction in Kidney Disease

Glomerular mesangial cells and interstitial fibroblasts are thought to be involved in excessive extracellular matrix synthesis during the progression of CKD. Both cell types respond very similarly to pro-fibrotic stimuli during disease progression, such as by transforming to a myofibroblast phenotype upon activation and synthesizing matrix proteins, including fibronectin, laminin and collagen types I, III and IV [229,230,231,232,233]. It has been reported that a variety of stimuli including TGF-β, angiotensin II and hyperglycemia have been shown to alter the activity of NOX in both mesangial cells and fibroblasts, and ultimately the amount of ROS production [234,235]. More specifically, NOX4 is a major mediator of the activation of thylakoids and the conversion of fibroblasts to myofibroblasts caused by high glucose or angiotensin II [145,236,237]. Increased ROS synthesis in activated mesangial cells and myofibroblasts also activates downstream signaling pathways, leading to increased extracellular matrix synthesis and renal fibrosis.

5.4. Oxidative Stress and Endothelial Cell Dysfunction in Kidney Disease

As discussed above, endothelial cells’ main function is the synthesis and secretion of NO via eNOS. NO is involved in various life processes through cGMP-mediated vasodilation, inflammation and immune responses. Under normal conditions, low NO levels in the endothelium can induce antioxidant gene expression and inhibit cytochrome C oxidase, reducing ROS production [238]. However, elevated ROS levels reduce endothelial NO production by inhibiting and/or uncoupling NOS [239,240]. For example, in early CKD patients, plasma accumulation of ADMA (an L-arginine analogue) and uncoupling of NOS lead to inhibition of NO synthesis and increased ROS production [239,241]. Subsequently, renal endothelial dysfunction and increased vasoconstrictive blood flow resistance lead to glomerular ischemia. In addition, the reaction of NO with superoxide produces peroxynitrite, which further leads to tissue damage by reacting with thiols, lipids and proteins containing aromatic amino acids [241,242].

6. Oxidative Stress-Related Molecules in Kidney Disease

6.1. Nrf2

Nrf2 is a transcription factor that responds to intracellular oxidative stress and is involved in the regulation of cellular redox homeostasis. Normally, Nrf2 interacts with kelch-like ECH-associated protein 1 (KEAP1) to be sequestered in the cytoplasm. Subsequently, Nrf2 and E3 ubiquitin ligase are ligated, causing Nrf2 degradation via the proteasome [243]. During oxidative stress, ROS modify critical cysteine residues within KEAP1, inducing a conformational change that disrupts the KEAP1-Nrf2 interaction and allows Nrf2 to evade degradation [244]. Nrf2 then translocates to the nucleus and binds to antioxidant response elements (AREs) within promoter regions of various antioxidant and cytoprotective genes, including GSH S-transferases, HO-1, CAT, γ-glutamylcysteine synthetase and NAD(P)H quinone oxidoreductase, to increase their expression [245]. Nrf2 is also responsible for the regulation of GSH and Trx systems. Nrf2 regulates the expression of GSH synthases, such as glutamate–cysteine ligase (GCL) enzyme complex, and the amount of the limiting substrate of GSH, cysteine [246,247], to regulate GSH expression. Nrf2 also regulates the expression of Trx through the induction of NADPH, which is crucial for electron transfer in the Trx system [248].
The renoprotective functions of Nrf2 have been demonstrated in various CKD models, and serves to attenuate oxidative stress induced by pathological conditions, such as ischemia-reperfusion injury (IRI), unilateral ureteral obstruction (UUO), nephrotoxicants and diabetes mellitus (DM), to slow disease progression [249]. Nrf2 deficiency in a UUO mice model decreased the expression of antioxidant genes glutamate-cysteine ligase catalytic (Gclc) and HO-1 and increased the expression of genes associated with inflammation and fibrosis such as TGF-β, TNF, IL-6, IL-1b, etc., suggesting that the protective function of Nrf2 for renal tubulointerstitial fibrosis may be facilitated by both anti-inflammatory and antioxidant pathways [250]. DKD is a disease that develops and progresses due to a number of interconnected pathogenic mechanisms, such as altered renal metabolism, mitochondrial dysfunction and oxidative stress. Dandona P. et al. demonstrated that the levels of 8-hydroxy-2′-deoxyguanosine, a biomarker of oxidative stress, were elevated in the urine of diabetic patients relative to controls and correlated positively with other indicators of complications such as proteinuria [251]. Furthermore, in a streptozotocin-induced mice model of diabetes, knockout of Nrf2 led to more intense DNA damage and more ROS production, resulting in severe proteinuria and glomerulosclerosis compared to wild-type controls [252,253]. Nrf2 also confers protective effects on pancreatic beta cells and mitigates insulin resistance in individuals with diabetes [254]. In addition, Nrf2-deficient mice are more susceptible to cisplatin-induced kidney injury than wild-type mice [255]. Pretreatment of wild-type mice with the Nrf2 activator CDDO-Im prevents cisplatin-induced nephrotoxicity [256]. With the in-depth study of Nrf2 activators, a variety of compounds such as astragaloside IV, bardoxolone methy, etc., can attenuate cisplatin-induced renal injury via the activation of the Nrf2/HO-1 signaling pathway [257,258]. Nrf2 activators significantly improved ischemia/reperfusion- and LPS-induced AKI [259]. Qiu et al. have demonstrated that treatment with obacunone, an activator of the Nrf2 pathway, significantly decreased mouse renal cyst growth in autosomal dominant polycystic kidney disease (ADPKD) mice models by suppressing lipid peroxidation through upregulation of glutathione peroxidase 4 (GPX4), and inhibition of lipid peroxidation ultimately led to a decrease of hypercellular proliferation through decreasing the activation of the mechanistic target of rapamycin (mTOR) and mitogen-activated protein kinase (MAPK) signaling cascades [260].

6.2. NF-κB

Inflammation is a major contributor to CKD progression. The renal inflammatory microenvironment consists of inflammatory factors and immune cells. Macrophages, in particular, play a key role in renal inflammation [261]. Macrophages exhibit either M1 or M2 phenotypes. M1 macrophages secrete proinflammatory mediators like ROS, TNF-α, and IL-1β, thereby promoting inflammation and injury. Conversely, M2 macrophages exhibit an anti-inflammatory phenotype through IL-10 secretion, which counteracts acute inflammation caused by M1 cells [262]. However, sustained M2 responses can also result in excessive extracellular matrix (ECM) deposition and exacerbate renal fibrosis [263].
Nuclear factor kappa B (NF-κB) is a ubiquitous transcription factor that normally exists as an inactive cytoplasmic complex through combination with inhibitory κB (IκB) proteins [264]. The activation of the toll-like receptor by TNF-α and IL-1β induces the activation of the IκB kinase complex (IKK), leading to IκB phosphorylation and degradation [264]. This allows NF-κB translocation to the nuclear region, to increase the transcription of proinflammatory genes such as IL-1, IL-6 and TNF-α. Persistent NF-κB activation is associated with chronic inflammation in CKD [265,266]. ROS can activate NF-κB to drive renal fibrotic progression, while NF-κB also upregulates NOX and NOS to exacerbate oxidative stress [267,268]. Nrf2 can inhibit NF-κB signaling and decrease the expression of genes stimulated by NF-κB [245].
Renal oxidative stress and inflammation, which are symbols of DKD, are considered to be regulated by NF-κB. ROS can activate NF-κB signaling, thereby modulating the expression of various adhesion molecules, pro-inflammatory cytokines, and chemokines involved in chronic renal inflammation in DKD [269]. Prolonged activation of NF-κB-mediated inflammation exacerbates oxidative injury to renal cells. Additionally, oxidative stress and inflammation disrupt the normal balance between ECM synthesis and degradation, resulting in excessive ECM accumulation in the kidneys [270,271]. The cumulative effects of aberrant NF-κB signaling, oxidative stress and inflammation ultimately promote renal fibrosis, a key pathological feature of advanced DKD [272]. Administration of sappanone A (SA) mitigated systemic inflammation and cortical renal inflammatory responses as well as renal injury in a murine model of DKD by modulating the NF-κB signaling cascade [273].
It is well known that systemic inhibition of NF-κB can influence the severity of AKI. Inhibition of NF-κB attenuated kidney injury in a folic acid-induced AKI model [274]. Inhibition of IκB kinase at 24 h after the onset of AKI also effectively suppressed NF-κB activity, which in turn improved renal function and attenuated fibrosis [275]. Further studies found that NF-κB plays a role in AKI through immune cells and epithelial cells. Inhibition of NF-κB in macrophages by blocking CD38 can alleviate LPS-induced AKI in mice [276]. Additionally, NF-κB activation may augment M1 macrophage infiltration in AKI through the C-type lectin receptor Mincle [277]. In an ischemia/cisplatin-induced mice AKI model, Yang et al. proposed that KIM-1 mediates phagocytosis of apoptotic epithelial cells to attenuate the inflammatory response, which may be achieved by decreasing the activation of NF-κB [278]. Reduced apoptosis, lowered chemokine expression and attenuated AKI were also observed in mice with renal tubular epithelial cell-specific NF-κB knockout [279].

6.3. Sirtuin 1

The seven mammalian sirtuins (SIRT1-7) regulate various processes related to antioxidant capacity, oxidative stress and metabolism. SIRT1, the most extensively characterized isoform, localizes to the cytoplasm and nucleus [280]. As a NAD+-dependent deacetylase, SIRT1 helps maintain redox homeostasis by modulating the ratio of NAD+/NADH [281,282,283]. SIRT1 inhibits inflammatory responses and oxidative stress by deacetylating the p65 subunit of NF-κB [284]. In human renal tubular epithelial cells (HK-2) cultured with high glucose, SIRT1 deacetylation activity is decreased, resulting in a decrease of p65 deacetylation and then an increase of NF-κB activity, which decrease the expression of miR-29 through directly binding to its promoter. MiR-29 has an ability to target and suppress Keap1 gene expression. The decrease of miR-29 increases Keap1 expression and inhibits the Nrf2/ARE (antioxidant response element) pathway [285]. SIRT1 also promotes the activation of endothelial NOS in rats, leading to more NO production and preventing oxidative stress [286]. FOXO1 and FOXO3 regulate antioxidant genes like SOD2 and CAT through SIRT1-mediated deacetylation [287]. By modifying the transcription of the FoxO target gene, SIRT1 protects diabetic kidneys and blood vessels from oxidative stress and tissue damage [287]. In addition, SIRT1 inhibits p53-dependent apoptosis arising from DNA damage and oxidative stress through p53 deacetylation, promoting human renal cell survival [288]. SIRT1 also negatively regulates the mitochondrial ROS producer p66Shc by promoting its deacetylation, reducing oxidative stress and maintaining mammalian endothelial function [289].
The activation of SIRT1 prevents CKD by mechanisms such as increasing COX-2 expression and attenuating renal fibrosis and inflammation [290]. In a rat model of CKD, SIRT1 activation attenuated inflammation and tubular fibrosis via inhibition of the TGF-β/SMAD pathway [290,291,292]. In DKD, activation of SIRT1 confers renoprotective effects across various models of renal injury. SIRT1 predominantly exerts protection in proximal tubular cells and podocytes. Activation of SIRT1 can reduce the expression of the pro-apoptotic gene, Bcl2-like 11 (Bcl2l11), through deacetylation of FOXO4, thereby preventing podocyte loss in diabetics [293]. SIRT1 also inactivates NF-κB and the signal transducer and activator of transcription 3 (STAT3) to attenuate proteinuria and podocyte damage in db/db mice [294]. Increased proteinuria and progression of DKD were observed in a mouse model with podocyte-specific SIRT1 knockout [295]. In addition, Hasegawa et al. proposed that knockout of SIRT1 increased the expression of the tight junction protein claudin-1 in proximal renal tubules, exacerbating albuminuria and impairing renal function in in vivo and vitro experiments [296].
In a rat model of IRI-induced AKI, upregulation of SIRT1 effectively recovered renal function and attenuated apoptosis compared to controls. SIRT1 expression is observed to increase in HK-2 cells cultured under hypoxic conditions to reduce ROS production [297] (Figure 2).

7. The Crosstalk of Abnormal Iron Metabolism and Oxidative Stress in Kidney Diseases

7.1. How Oxidative Stress Affects Iron Metabolism

It has been found that ROS can alter the activity of Fe-S clusters in IRP1 and influences the activity of IRP1 binding with mRNA and subsequent cellular iron metabolism [298,299,300]. Intracellular superoxide anion inhibits cellular iron uptake capacity by inactivating IRP1, resulting in its inability to bind to the IRE segment of mRNA [301]. Treatment with exogenous hydrogen peroxide stimulates Fe-S clusters’ catabolism, which promotes IRP1 binding to mRNA and increases iron endocytosis in cultured mouse fibroblasts cells [302]. In addition, an IRP-IRE-independent mechanism of iron regulation was identified. The transcriptional level of cellular TFR was elevated upon treatment with low concentrations of hydrogen peroxide, and the ability of IRP to bind to mRNA was unchanged [303]. In contrast, treatment with high concentrations of hydrogen peroxide increased ferritin proteasome degradation in microglial cells [304]. In addition, treatment with hydrogen peroxide may rapidly inactivate PHD by oxidizing divalent iron in the PHD active site and lead to transient activation of HIF-1α in human osteosarcoma cells. However, prolonged hydrogen peroxide treatment results in the activation of ferrireductase and restores enzymatic activity of PHD, resulting in a decrease in HIF-1α [305].

7.2. Abnormal Iron Metabolism Leads to Oxidative Stress and Ferroptosis

In patients with CKD, intracellular iron efflux is usually blocked, leading to intracellular accumulation of iron. Elevated serum hepcidin levels have been found in patients with CKD, which subsequently binds to FPN on the cell membrane and leads to FPN degradation and intracellular iron accumulation [306,307]. Excess iron subsequently produces hydroxyl radicals via the Fenton reaction, which can react with PUFAs on cell membranes and oxygen to form lipid ROS or lipid peroxyl radicals. This results in ferroptosis, a genetically and biochemically unique kind of programmed cell death that is different from others [308]. Ferroptosis is an iron-dependent programmed cell death that is characterized by the accumulation of lipid peroxides and regulates the progression of certain renal disorders [309] (Figure 3).
Recent studies have indicated that ferroptosis significantly leads to the pathogenesis of renal tubular injury progressing to renal fibrosis [310]. Recently, it has been found that repressor element 1-silencing transcription factor (REST) is upregulated in renal tubular epithelial cells of AKI patients and mice and leads to renal injury through the regulation of ferroptosis, and that tubular-specific knockdown of REST significantly attenuates the transition from AKI to CKD and ameliorates renal fibrosis [311]. Treatment with ferroptosis inducers, such as erastin, decreases GPX 4 activity and enhances intracellular lipid peroxidation to aggravate renal fibrosis [312]. Pretreatment with ferroptosis inhibitors stopped the progression of renal fibrosis by preventing ferroptosis-related lipid peroxidation and GSH depletion. In addition, vitexin attenuates CKD by inhibiting ferroptosis in renal tubular epithelial cells through activation of Nrf2 [312], and formononetin and tectorigenin attenuate renal fibrosis by inhibiting Smad3 [313,314].
Iron-dependent lipid peroxidation also exists in db/db and STZ-induced mouse or rat models of DKD and in DKD patients [315,316,317,318]. It has been found that under the condition of high glucose, HK-2 cells were characterized with an iron overload, a reduction of antioxidant ability, an accumulation of ROS and a lipid peroxidation [315,319], whereas all of these could be alleviated by the treatment with ferrostatin-1 (Fer-1), a ferroptosis inhibitor2, suggesting a role of ferroptosis in DKD [315,319]. ZIP14, a cellular iron importer, was upregulated while GPX4 and GSH levels were decreased in rat DKD models and Fer-1 treatment could normalize iron and ZIP14 levels [318]. Increasing mitochondrial GSH content with N-acetylcysteine (NAC) also significantly ameliorated high glucose-induced ferroptosis in renal cells [320].
Ferroptosis also participates in the process of ADPKD [321]. Schreiber et al. discovered that in the renal tissues of ADPKD patients as well as in a mouse model, lipid peroxidation enhanced the activation of the chloride channel TMEM16A, which is closely linked to renal cyst enlargement [322]. In contrast, treatment with Fer-1 effectively inhibited TMEM16A activation and cyst enlargement. Mutations of polycystin 1 predisposed cells to ferroptosis by dysregulating iron and lipid metabolism [323]. In addition, high iron levels, low GPX 4 activity and increased lipid peroxide accumulation were verified in both ADPKD cells and mouse models [323].
A kidney biopsy report of a patient with IgA nephropathy shows a significant deposition of iron-containing hemosiderin in the renal tubules [324]. A cohort study revealed that compared to the healthy control group, IgA patients had significantly lower levels of the antioxidant enzymes SOD and vitamin E in their serum, while the levels of the intermediate product of lipid peroxidation, malondialdehyde (MDA), were elevated [325]. Furthermore, Wu et al. confirmed that GPX4 levels were significantly decreased in kidneys of IgA patients, and treatment with the ferroptosis inhibitor Fer-1 resulted in a decrease of MDA and ROS, an increase of GPX4, an inhibition of ferroptosis in mesangial cells and a delay of the progression of IgA nephropathy [326]. These findings suggest that ferroptosis plays a crucial promoting role in the progression of IgA nephropathy.
In the mouse IRI model of AKI, single-cell sequencing showed that ferroptosis-related genes are predominantly expressed in the renal tubular epithelial cells [327]. Conditional knockout of ferroptosis suppressor protein 1 (FSP1) or GPX4 increased cellular sensitivity to iron death and exacerbated acute tubular necrosis in a mouse model of IRI-AKI [328]. Another study found that knockout of ACSL4 (a promoter of ferroptosis) effectively reduced pathological injury in AKI mice [329]. In addition, treatment with ferroptosis inhibitors has been shown to effectively alleviate IRI-AKI in mice model [327,330]. Ferroptosis is also increased in sepsis-induced AKI mouse models [331]. Recent studies have demonstrated that treatment with melatonin prevents ferroptosis and ameliorates sepsis-associated AKI by upregulating Nrf2/HO-1 cytoprotective pathway [332]. Furthermore, ferroptosis is also involved in the injury response in rhabdomyolysis and cisplatin-induced AKI [333,334]

8. Conclusions and Perspectives

Abnormal iron metabolism and oxidative stress play crucial roles in the progression of kidney diseases, ultimately resulting in end-stage renal disease. Iron homeostasis is necessary to maintain normal life activities of cells, tissues and organs. Abnormal iron accumulation disrupts iron homeostasis, leading to the overproduction of ROS through the Fenton reaction, which in turn exacerbates tissue injury through inflammation, ferroptosis, tissue scar repair and other unfavorable reactions. In addition, ROS acts as signaling molecules to regulate iron homeostasis by affecting the activity of regulators of iron metabolism, which may result in a new form of cell death, ferroptosis. This may be one of the mechanisms of how abnormal iron metabolism and oxidative stress cause renal injury.
Understanding the underlying mechanisms of iron metabolism and oxidative stress in kidney diseases is crucial to discovering possible therapeutic targets and devising effective methods to reduce the burden of kidney diseases. The existence of a vicious cycle of abnormal iron metabolism and oxidative stress perpetuate each other. Further investigation into the correlation between iron metabolism and oxidative stress in the setting of kidney disease is crucial, as it can offer valuable perspectives on potential therapeutic approaches. Future studies need to delve into the following concerns, including verifying (1) besides the Fenton reaction, whether there are any other ways for iron ions to regulate ROS production, and (2) in addition to ROS leading to extreme oxidative stress, whether there are any other mechanisms in ROS to alter cellular phenotypes. Because the mechanisms related to ferroptosis are poorly defined, it is necessary further explore novel molecular pathways and regulatory mechanisms related to ferroptosis. Although there is a possible association between iron deposition and kidney injury in hemochromatosis, more in-depth studies are needed to explore whether and how dysregulation of iron metabolism (iron deposition or iron deficiency) directly leads to kidney injury and impaired renal function. In addition, thus far, almost all mechanistic studies are based on experimental animals. The individual differences between humans and experimental animals should be verified in a clinical setting. In sum, an in-depth understanding of the iron metabolic pathways and the regulatory mechanism of oxidative homeostasis is of great interest in determining their specific roles in various kidney diseases and discovering the corresponding safe therapeutic targets for treatment.

Author Contributions

T.X. prepared most of the figures and manuscript; L.Y. edited the manuscript. X.L. supervised and edited the whole manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

X.L. acknowledges support from National Institutes of Health grant R01 DK129241 and DK126662 and DOD (Focused Program Award: PR221810).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The figures were drawn with Figdraw 2.0.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Xie, H.; Yang, N.; Yu, C.; Lu, L. Uremic toxins mediate kidney diseases: The role of aryl hydrocarbon receptor. Cell. Mol. Biol. Lett. 2024, 29, 38. [Google Scholar] [CrossRef]
  2. Chawla, L.S.; Bellomo, R.; Bihorac, A.; Goldstein, S.L.; Siew, E.D.; Bagshaw, S.M.; Bittleman, D.; Cruz, D.; Endre, Z.; Fitzgerald, R.L.; et al. Acute kidney disease and renal recovery: Consensus report of the Acute Disease Quality Initiative (ADQI) 16 Workgroup. Nat. Rev. Nephrol. 2017, 13, 241–257. [Google Scholar] [CrossRef]
  3. André, C.; Bodeau, S.; Kamel, S.; Bennis, Y.; Caillard, P. The AKI-to-CKD Transition: The Role of Uremic Toxins. Int. J. Mol. Sci. 2023, 24, 16152. [Google Scholar] [CrossRef] [PubMed]
  4. Wang, N.; Zhang, C. Oxidative Stress: A Culprit in the Progression of Diabetic Kidney Disease. Antioxidants 2024, 13, 455. [Google Scholar] [CrossRef]
  5. Hassler, J.R. IgA nephropathy: A brief review. Semin. Diagn. Pathol. 2020, 37, 143–147. [Google Scholar] [CrossRef]
  6. Muchowska, K.B.; Varma, S.J.; Moran, J. Synthesis and breakdown of universal metabolic precursors promoted by iron. Nature 2019, 569, 104–107. [Google Scholar] [CrossRef] [PubMed]
  7. Matsuoka, T.; Abe, M.; Kobayashi, H. Iron Metabolism and Inflammatory Mediators in Patients with Renal Dysfunction. Int. J. Mol. Sci. 2024, 25, 3745. [Google Scholar] [CrossRef] [PubMed]
  8. Nakanishi, T.; Hasuike, Y.; Otaki, Y.; Nanami, M.; Kuragano, T. Dysregulated iron metabolism in patients on hemodialysis. Contrib. Nephrol. 2015, 185, 22–31. [Google Scholar] [CrossRef] [PubMed]
  9. Babitt, J.L.; Lin, H.Y. Mechanisms of anemia in CKD. J. Am. Soc. Nephrol. JASN 2012, 23, 1631–1634. [Google Scholar] [CrossRef]
  10. Chen, Y.; Li, Z.; Zhang, H.; Chen, H.; Hao, J.; Liu, H.; Li, X. Mitochondrial metabolism and targeted treatment strategies in ischemic-induced acute kidney injury. Cell Death Discov. 2024, 10, 69. [Google Scholar] [CrossRef]
  11. Kimura, T.; Suzuki, K. Components of the electron transport system in adrenal steroid hydroxylase. Isolation and properties of non-heme iron protein (adrenodoxin). J. Biol. Chem. 1967, 242, 485–491. [Google Scholar] [CrossRef] [PubMed]
  12. Monsen, E.R.; Hallberg, L.; Layrisse, M.; Hegsted, D.M.; Cook, J.D.; Mertz, W.; Finch, C.A. Estimation of available dietary iron. Am. J. Clin. Nutr. 1978, 31, 134–141. [Google Scholar] [CrossRef] [PubMed]
  13. Zimmermann, M.B.; Hurrell, R.F. Nutritional iron deficiency. Lancet 2007, 370, 511–520. [Google Scholar] [CrossRef] [PubMed]
  14. Gunshin, H.; Mackenzie, B.; Berger, U.V.; Gunshin, Y.; Romero, M.F.; Boron, W.F.; Nussberger, S.; Gollan, J.L.; Hediger, M.A. Cloning and characterization of a mammalian proton-coupled metal-ion transporter. Nature 1997, 388, 482–488. [Google Scholar] [CrossRef] [PubMed]
  15. Riedel, H.D.; Remus, A.J.; Fitscher, B.A.; Stremmel, W. Characterization and partial purification of a ferrireductase from human duodenal microvillus membranes. Biochem. J. 1995, 309 Pt 3, 745–748. [Google Scholar] [CrossRef] [PubMed]
  16. Hentze, M.W.; Muckenthaler, M.U.; Galy, B.; Camaschella, C. Two to tango: Regulation of Mammalian iron metabolism. Cell 2010, 142, 24–38. [Google Scholar] [CrossRef] [PubMed]
  17. Donovan, A.; Lima, C.A.; Pinkus, J.L.; Pinkus, G.S.; Zon, L.I.; Robine, S.; Andrews, N.C. The iron exporter ferroportin/Slc40a1 is essential for iron homeostasis. Cell Metab. 2005, 1, 191–200. [Google Scholar] [CrossRef]
  18. Vulpe, C.D.; Kuo, Y.M.; Murphy, T.L.; Cowley, L.; Askwith, C.; Libina, N.; Gitschier, J.; Anderson, G.J. Hephaestin, a ceruloplasmin homologue implicated in intestinal iron transport, is defective in the sla mouse. Nat. Genet. 1999, 21, 195–199. [Google Scholar] [CrossRef] [PubMed]
  19. Shayeghi, M.; Latunde-Dada, G.O.; Oakhill, J.S.; Laftah, A.H.; Takeuchi, K.; Halliday, N.; Khan, Y.; Warley, A.; McCann, F.E.; Hider, R.C.; et al. Identification of an intestinal heme transporter. Cell 2005, 122, 789–801. [Google Scholar] [CrossRef]
  20. Dunn, L.L.; Suryo Rahmanto, Y.; Richardson, D.R. Iron uptake and metabolism in the new millennium. Trends Cell Biol. 2007, 17, 93–100. [Google Scholar] [CrossRef]
  21. Schneider, C.; Owen, M.J.; Banville, D.; Williams, J.G. Primary structure of human transferrin receptor deduced from the mRNA sequence. Nature 1984, 311, 675–678. [Google Scholar] [CrossRef] [PubMed]
  22. Aisen, P.; Leibman, A.; Zweier, J. Stoichiometric and site characteristics of the binding of iron to human transferrin. J. Biol. Chem. 1978, 253, 1930–1937. [Google Scholar] [CrossRef] [PubMed]
  23. Bartnikas, T.B. Known and potential roles of transferrin in iron biology. Biometals Int. J. Role Met. Ions Biol. Biochem. Med. 2012, 25, 677–686. [Google Scholar] [CrossRef] [PubMed]
  24. Enns, C.A.; Sussman, H.H. Physical characterization of the transferrin receptor in human placentae. J. Biol. Chem. 1981, 256, 9820–9823. [Google Scholar] [CrossRef] [PubMed]
  25. Ponka, P.; Beaumont, C.; Richardson, D.R. Function and regulation of transferrin and ferritin. Semin. Hematol. 1998, 35, 35–54. [Google Scholar] [PubMed]
  26. Andrews, N.C. Forging a field: The golden age of iron biology. Blood 2008, 112, 219–230. [Google Scholar] [CrossRef]
  27. Ohgami, R.S.; Campagna, D.R.; Greer, E.L.; Antiochos, B.; McDonald, A.; Chen, J.; Sharp, J.J.; Fujiwara, Y.; Barker, J.E.; Fleming, M.D. Identification of a ferrireductase required for efficient transferrin-dependent iron uptake in erythroid cells. Nat. Genet. 2005, 37, 1264–1269. [Google Scholar] [CrossRef]
  28. Zhao, N.; Gao, J.; Enns, C.A.; Knutson, M.D. ZRT/IRT-like protein 14 (ZIP14) promotes the cellular assimilation of iron from transferrin. J. Biol. Chem. 2010, 285, 32141–32150. [Google Scholar] [CrossRef]
  29. Levy, J.E.; Jin, O.; Fujiwara, Y.; Kuo, F.; Andrews, N.C. Transferrin receptor is necessary for development of erythrocytes and the nervous system. Nat. Genet. 1999, 21, 396–399. [Google Scholar] [CrossRef] [PubMed]
  30. Truman-Rosentsvit, M.; Berenbaum, D.; Spektor, L.; Cohen, L.A.; Belizowsky-Moshe, S.; Lifshitz, L.; Ma, J.; Li, W.; Kesselman, E.; Abutbul-Ionita, I.; et al. Ferritin is secreted via 2 distinct nonclassical vesicular pathways. Blood 2018, 131, 342–352. [Google Scholar] [CrossRef]
  31. Li, J.Y.; Paragas, N.; Ned, R.M.; Qiu, A.; Viltard, M.; Leete, T.; Drexler, I.R.; Chen, X.; Sanna-Cherchi, S.; Mohammed, F.; et al. Scara5 is a ferritin receptor mediating non-transferrin iron delivery. Dev. Cell 2009, 16, 35–46. [Google Scholar] [CrossRef] [PubMed]
  32. Montemiglio, L.C.; Testi, C.; Ceci, P.; Falvo, E.; Pitea, M.; Savino, C.; Arcovito, A.; Peruzzi, G.; Baiocco, P.; Mancia, F.; et al. Cryo-EM structure of the human ferritin-transferrin receptor 1 complex. Nat. Commun. 2019, 10, 1121. [Google Scholar] [CrossRef]
  33. Li, L.; Fang, C.J.; Ryan, J.C.; Niemi, E.C.; Lebrón, J.A.; Björkman, P.J.; Arase, H.; Torti, F.M.; Torti, S.V.; Nakamura, M.C.; et al. Binding and uptake of H-ferritin are mediated by human transferrin receptor-1. Proc. Natl. Acad. Sci. USA 2010, 107, 3505–3510. [Google Scholar] [CrossRef]
  34. Liuzzi, J.P.; Aydemir, F.; Nam, H.; Knutson, M.D.; Cousins, R.J. Zip14 (Slc39a14) mediates non-transferrin-bound iron uptake into cells. Proc. Natl. Acad. Sci. USA 2006, 103, 13612–13617. [Google Scholar] [CrossRef] [PubMed]
  35. van Raaij, S.E.G.; Srai, S.K.S.; Swinkels, D.W.; van Swelm, R.P.L. Iron uptake by ZIP8 and ZIP14 in human proximal tubular epithelial cells. Biometals Int. J. Role Met. Ions Biol. Biochem. Med. 2019, 32, 211–226. [Google Scholar] [CrossRef] [PubMed]
  36. Das, A.; Nag, S.; Mason, A.B.; Barroso, M.M. Endosome-mitochondria interactions are modulated by iron release from transferrin. J. Cell Biol. 2016, 214, 831–845. [Google Scholar] [CrossRef] [PubMed]
  37. Jacobs, A. Low molecular weight intracellular iron transport compounds. Blood 1977, 50, 433–439. [Google Scholar] [CrossRef]
  38. Vyoral, D.; Hradilek, A.; Neuwirt, J. Transferrin and iron distribution in subcellular fractions of K562 cells in the early stages of transferrin endocytosis. Biochim. Biophys. Acta 1992, 1137, 148–154. [Google Scholar] [CrossRef] [PubMed]
  39. Shi, H.; Bencze, K.Z.; Stemmler, T.L.; Philpott, C.C. A cytosolic iron chaperone that delivers iron to ferritin. Science 2008, 320, 1207–1210. [Google Scholar] [CrossRef]
  40. Jain, C.; Shah, Y.M. PCBP1 is essential for proper iron absorption. Blood 2023, 142, 1585–1587. [Google Scholar] [CrossRef]
  41. Ponka, P. Tissue-specific regulation of iron metabolism and heme synthesis: Distinct control mechanisms in erythroid cells. Blood 1997, 89, 1–25. [Google Scholar] [CrossRef] [PubMed]
  42. Zhang, A.S.; Sheftel, A.D.; Ponka, P. Intracellular kinetics of iron in reticulocytes: Evidence for endosome involvement in iron targeting to mitochondria. Blood 2005, 105, 368–375. [Google Scholar] [CrossRef] [PubMed]
  43. Provance, D.W., Jr.; Gourley, C.R.; Silan, C.M.; Cameron, L.C.; Shokat, K.M.; Goldenring, J.R.; Shah, K.; Gillespie, P.G.; Mercer, J.A. Chemical-genetic inhibition of a sensitized mutant myosin Vb demonstrates a role in peripheral-pericentriolar membrane traffic. Proc. Natl. Acad. Sci. USA 2004, 101, 1868–1873. [Google Scholar] [CrossRef] [PubMed]
  44. Iacopetta, B.J.; Morgan, E.H. The kinetics of transferrin endocytosis and iron uptake from transferrin in rabbit reticulocytes. J. Biol. Chem. 1983, 258, 9108–9115. [Google Scholar] [CrossRef] [PubMed]
  45. Lim, J.E.; Jin, O.; Bennett, C.; Morgan, K.; Wang, F.; Trenor, C.C., 3rd; Fleming, M.D.; Andrews, N.C. A mutation in Sec15l1 causes anemia in hemoglobin deficit (hbd) mice. Nat. Genet. 2005, 37, 1270–1273. [Google Scholar] [CrossRef] [PubMed]
  46. Zhang, A.S.; Sheftel, A.D.; Ponka, P. The anemia of “haemoglobin-deficit” (hbd/hbd) mice is caused by a defect in transferrin cycling. Exp. Hematol. 2006, 34, 593–598. [Google Scholar] [CrossRef]
  47. Wolff, N.A.; Garrick, M.D.; Zhao, L.; Garrick, L.M.; Ghio, A.J.; Thévenod, F. A role for divalent metal transporter (DMT1) in mitochondrial uptake of iron and manganese. Sci. Rep. 2018, 8, 211. [Google Scholar] [CrossRef] [PubMed]
  48. Paradkar, P.N.; Zumbrennen, K.B.; Paw, B.H.; Ward, D.M.; Kaplan, J. Regulation of mitochondrial iron import through differential turnover of mitoferrin 1 and mitoferrin 2. Mol. Cell. Biol. 2009, 29, 1007–1016. [Google Scholar] [CrossRef] [PubMed]
  49. Seguin, A.; Jia, X.; Earl, A.M.; Li, L.; Wallace, J.; Qiu, A.; Bradley, T.; Shrestha, R.; Troadec, M.B.; Hockin, M.; et al. The mitochondrial metal transporters mitoferrin1 and mitoferrin2 are required for liver regeneration and cell proliferation in mice. J. Biol. Chem. 2020, 295, 11002–11020. [Google Scholar] [CrossRef]
  50. Troadec, M.B.; Warner, D.; Wallace, J.; Thomas, K.; Spangrude, G.J.; Phillips, J.; Khalimonchuk, O.; Paw, B.H.; Ward, D.M.; Kaplan, J. Targeted deletion of the mouse Mitoferrin1 gene: From anemia to protoporphyria. Blood 2011, 117, 5494–5502. [Google Scholar] [CrossRef]
  51. Napier, I.; Ponka, P.; Richardson, D.R. Iron trafficking in the mitochondrion: Novel pathways revealed by disease. Blood 2005, 105, 1867–1874. [Google Scholar] [CrossRef] [PubMed]
  52. Lill, R.; Mühlenhoff, U. Maturation of iron-sulfur proteins in eukaryotes: Mechanisms, connected processes, and diseases. Annu. Rev. Biochem. 2008, 77, 669–700. [Google Scholar] [CrossRef] [PubMed]
  53. Levi, S.; Corsi, B.; Bosisio, M.; Invernizzi, R.; Volz, A.; Sanford, D.; Arosio, P.; Drysdale, J. A human mitochondrial ferritin encoded by an intronless gene. J. Biol. Chem. 2001, 276, 24437–24440. [Google Scholar] [CrossRef] [PubMed]
  54. Liu, G.; Sil, D.; Maio, N.; Tong, W.H.; Bollinger, J.M., Jr.; Krebs, C.; Rouault, T.A. Heme biosynthesis depends on previously unrecognized acquisition of iron-sulfur cofactors in human amino-levulinic acid dehydratase. Nat. Commun. 2020, 11, 6310. [Google Scholar] [CrossRef] [PubMed]
  55. Harrison, P.M.; Fischbach, F.A.; Hoy, T.G.; Haggis, G.H. Ferric oxyhydroxide core of ferritin. Nature 1967, 216, 1188–1190. [Google Scholar] [CrossRef] [PubMed]
  56. Harrison, P.M.; Arosio, P. The ferritins: Molecular properties, iron storage function and cellular regulation. Biochim. Biophys. Acta 1996, 1275, 161–203. [Google Scholar] [CrossRef]
  57. Koorts, A.M.; Viljoen, M. Ferritin and ferritin isoforms I: Structure-function relationships, synthesis, degradation and secretion. Arch. Physiol. Biochem. 2007, 113, 30–54. [Google Scholar] [CrossRef] [PubMed]
  58. Arosio, P.; Levi, S. Ferritin, iron homeostasis, and oxidative damage. Free Radic. Biol. Med. 2002, 33, 457–463. [Google Scholar] [CrossRef] [PubMed]
  59. Li, W.; Garringer, H.J.; Goodwin, C.B.; Richine, B.; Acton, A.; VanDuyn, N.; Muhoberac, B.B.; Irimia-Dominguez, J.; Chan, R.J.; Peacock, M.; et al. Systemic and cerebral iron homeostasis in ferritin knock-out mice. PLoS ONE 2015, 10, e0117435. [Google Scholar] [CrossRef]
  60. Arosio, P.; Yokota, M.; Drysdale, J.W. Structural and immunological relationships of isoferritins in normal and malignant cells. Cancer Res. 1976, 36, 1735–1739. [Google Scholar]
  61. Li, J.Y.; Feng, Y.H.; Li, Y.X.; He, P.Y.; Zhou, Q.Y.; Tian, Y.P.; Yao, R.Q.; Yao, Y.M. Ferritinophagy: A novel insight into the double-edged sword in ferritinophagy-ferroptosis axis and human diseases. Cell Prolif. 2024, e13621. [Google Scholar]
  62. Pan, Y.; Ren, Z.; Gao, S.; Shen, J.; Wang, L.; Xu, Z.; Yu, Y.; Bachina, P.; Zhang, H.; Fan, X.; et al. Structural basis of ion transport and inhibition in ferroportin. Nat. Commun. 2020, 11, 5686. [Google Scholar] [CrossRef] [PubMed]
  63. Billesbølle, C.B.; Azumaya, C.M.; Kretsch, R.C.; Powers, A.S.; Gonen, S.; Schneider, S.; Arvedson, T.; Dror, R.O.; Cheng, Y.; Manglik, A. Structure of hepcidin-bound ferroportin reveals iron homeostatic mechanisms. Nature 2020, 586, 807–811. [Google Scholar] [CrossRef] [PubMed]
  64. Yanatori, I.; Richardson, D.R.; Imada, K.; Kishi, F. Iron Export through the Transporter Ferroportin 1 Is Modulated by the Iron Chaperone PCBP2. J. Biol. Chem. 2016, 291, 17303–17318. [Google Scholar] [CrossRef] [PubMed]
  65. Zhang, Z.; Zhang, F.; An, P.; Guo, X.; Shen, Y.; Tao, Y.; Wu, Q.; Zhang, Y.; Yu, Y.; Ning, B.; et al. Ferroportin1 deficiency in mouse macrophages impairs iron homeostasis and inflammatory responses. Blood 2011, 118, 1912–1922. [Google Scholar] [CrossRef] [PubMed]
  66. Zhang, Z.; Zhang, F.; Guo, X.; An, P.; Tao, Y.; Wang, F. Ferroportin1 in hepatocytes and macrophages is required for the efficient mobilization of body iron stores in mice. Hepatology 2012, 56, 961–971. [Google Scholar] [CrossRef] [PubMed]
  67. Zhang, D.L.; Ghosh, M.C.; Ollivierre, H.; Li, Y.; Rouault, T.A. Ferroportin deficiency in erythroid cells causes serum iron deficiency and promotes hemolysis due to oxidative stress. Blood 2018, 132, 2078–2087. [Google Scholar] [CrossRef] [PubMed]
  68. Ito, F.; Kato, K.; Yanatori, I.; Murohara, T.; Toyokuni, S. Ferroptosis-dependent extracellular vesicles from macrophage contribute to asbestos-induced mesothelial carcinogenesis through loading ferritin. Redox Biol. 2021, 47, 102174. [Google Scholar] [CrossRef]
  69. Kimura, T.; Jia, J.; Kumar, S.; Choi, S.W.; Gu, Y.; Mudd, M.; Dupont, N.; Jiang, S.; Peters, R.; Farzam, F.; et al. Dedicated SNAREs and specialized TRIM cargo receptors mediate secretory autophagy. EMBO J. 2017, 36, 42–60. [Google Scholar] [CrossRef]
  70. Brown, C.W.; Amante, J.J.; Chhoy, P.; Elaimy, A.L.; Liu, H.; Zhu, L.J.; Baer, C.E.; Dixon, S.J.; Mercurio, A.M. Prominin2 Drives Ferroptosis Resistance by Stimulating Iron Export. Dev. Cell 2019, 51, 575–586.e574. [Google Scholar] [CrossRef]
  71. Yanatori, I.; Richardson, D.R.; Dhekne, H.S.; Toyokuni, S.; Kishi, F. CD63 is regulated by iron via the IRE-IRP system and is important for ferritin secretion by extracellular vesicles. Blood 2021, 138, 1490–1503. [Google Scholar] [CrossRef] [PubMed]
  72. Gao, H.; Jin, Z.; Bandyopadhyay, G.; Wang, G.; Zhang, D.; Rocha, K.C.E.; Liu, X.; Zhao, H.; Kisseleva, T.; Brenner, D.A.; et al. Aberrant iron distribution via hepatocyte-stellate cell axis drives liver lipogenesis and fibrosis. Cell Metab. 2022, 34, 1201–1213.e1205. [Google Scholar] [CrossRef]
  73. Kenny, T.C.; Khan, A.; Son, Y.; Yue, L.; Heissel, S.; Sharma, A.; Pasolli, H.A.; Liu, Y.; Gamazon, E.R.; Alwaseem, H.; et al. Integrative genetic analysis identifies FLVCR1 as a plasma-membrane choline transporter in mammals. Cell Metab. 2023, 35, 1057–1071.e1012. [Google Scholar] [CrossRef] [PubMed]
  74. Chiabrando, D.; Marro, S.; Mercurio, S.; Giorgi, C.; Petrillo, S.; Vinchi, F.; Fiorito, V.; Fagoonee, S.; Camporeale, A.; Turco, E.; et al. The mitochondrial heme exporter FLVCR1b mediates erythroid differentiation. J. Clin. Investig. 2012, 122, 4569–4579. [Google Scholar] [CrossRef] [PubMed]
  75. Chiabrando, D.; Castori, M.; di Rocco, M.; Ungelenk, M.; Gießelmann, S.; Di Capua, M.; Madeo, A.; Grammatico, P.; Bartsch, S.; Hübner, C.A.; et al. Mutations in the Heme Exporter FLVCR1 Cause Sensory Neurodegeneration with Loss of Pain Perception. PLoS Genet. 2016, 12, e1006461. [Google Scholar] [CrossRef] [PubMed]
  76. Wilkinson, N.; Pantopoulos, K. The IRP/IRE system in vivo: Insights from mouse models. Front. Pharmacol. 2014, 5, 176. [Google Scholar] [CrossRef] [PubMed]
  77. Muckenthaler, M.; Gray, N.K.; Hentze, M.W. IRP-1 binding to ferritin mRNA prevents the recruitment of the small ribosomal subunit by the cap-binding complex eIF4F. Mol. Cell 1998, 2, 383–388. [Google Scholar] [CrossRef] [PubMed]
  78. Yoshinaga, M.; Nakatsuka, Y.; Vandenbon, A.; Ori, D.; Uehata, T.; Tsujimura, T.; Suzuki, Y.; Mino, T.; Takeuchi, O. Regnase-1 Maintains Iron Homeostasis via the Degradation of Transferrin Receptor 1 and Prolyl-Hydroxylase-Domain-Containing Protein 3 mRNAs. Cell Rep. 2017, 19, 1614–1630. [Google Scholar] [CrossRef] [PubMed]
  79. Corral, V.M.; Schultz, E.R.; Eisenstein, R.S.; Connell, G.J. Roquin is a major mediator of iron-regulated changes to transferrin receptor-1 mRNA stability. iScience 2021, 24, 102360. [Google Scholar] [CrossRef]
  80. Muto, Y.; Nishiyama, M.; Nita, A.; Moroishi, T.; Nakayama, K.I. Essential role of FBXL5-mediated cellular iron homeostasis in maintenance of hematopoietic stem cells. Nat. Commun. 2017, 8, 16114. [Google Scholar] [CrossRef]
  81. Smith, S.R.; Ghosh, M.C.; Ollivierre-Wilson, H.; Hang Tong, W.; Rouault, T.A. Complete loss of iron regulatory proteins 1 and 2 prevents viability of murine zygotes beyond the blastocyst stage of embryonic development. Blood Cells Mol. Dis. 2006, 36, 283–287. [Google Scholar] [CrossRef] [PubMed]
  82. Galy, B.; Ferring-Appel, D.; Kaden, S.; Gröne, H.J.; Hentze, M.W. Iron regulatory proteins are essential for intestinal function and control key iron absorption molecules in the duodenum. Cell Metab. 2008, 7, 79–85. [Google Scholar] [CrossRef] [PubMed]
  83. Galy, B.; Ferring-Appel, D.; Becker, C.; Gretz, N.; Gröne, H.J.; Schümann, K.; Hentze, M.W. Iron regulatory proteins control a mucosal block to intestinal iron absorption. Cell Rep. 2013, 3, 844–857. [Google Scholar] [CrossRef]
  84. Nairz, M.; Ferring-Appel, D.; Casarrubea, D.; Sonnweber, T.; Viatte, L.; Schroll, A.; Haschka, D.; Fang, F.C.; Hentze, M.W.; Weiss, G.; et al. Iron Regulatory Proteins Mediate Host Resistance to Salmonella Infection. Cell Host Microbe 2015, 18, 254–261. [Google Scholar] [CrossRef] [PubMed]
  85. Wilkinson, N.; Pantopoulos, K. IRP1 regulates erythropoiesis and systemic iron homeostasis by controlling HIF2α mRNA translation. Blood 2013, 122, 1658–1668. [Google Scholar] [CrossRef] [PubMed]
  86. Cooperman, S.S.; Meyron-Holtz, E.G.; Olivierre-Wilson, H.; Ghosh, M.C.; McConnell, J.P.; Rouault, T.A. Microcytic anemia, erythropoietic protoporphyria, and neurodegeneration in mice with targeted deletion of iron-regulatory protein 2. Blood 2005, 106, 1084–1091. [Google Scholar] [CrossRef] [PubMed]
  87. Semenza, G.L.; Wang, G.L. A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation. Mol. Cell. Biol. 1992, 12, 5447–5454. [Google Scholar] [CrossRef] [PubMed]
  88. Wang, G.L.; Jiang, B.H.; Rue, E.A.; Semenza, G.L. Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc. Natl. Acad. Sci. USA 1995, 92, 5510–5514. [Google Scholar] [CrossRef]
  89. Wang, G.L.; Semenza, G.L. Characterization of hypoxia-inducible factor 1 and regulation of DNA binding activity by hypoxia. J. Biol. Chem. 1993, 268, 21513–21518. [Google Scholar] [CrossRef]
  90. Jaakkola, P.; Mole, D.R.; Tian, Y.M.; Wilson, M.I.; Gielbert, J.; Gaskell, S.J.; von Kriegsheim, A.; Hebestreit, H.F.; Mukherji, M.; Schofield, C.J.; et al. Targeting of HIF-alpha to the von Hippel-Lindau ubiquitylation complex by O2-regulated prolyl hydroxylation. Science 2001, 292, 468–472. [Google Scholar] [CrossRef]
  91. Tsuzuki, Y.; Fukumura, D.; Oosthuyse, B.; Koike, C.; Carmeliet, P.; Jain, R.K. Vascular endothelial growth factor (VEGF) modulation by targeting hypoxia-inducible factor-1alpha--> hypoxia response element--> VEGF cascade differentially regulates vascular response and growth rate in tumors. Cancer Res. 2000, 60, 6248–6252. [Google Scholar] [PubMed]
  92. Firth, J.D.; Ebert, B.L.; Pugh, C.W.; Ratcliffe, P.J. Oxygen-regulated control elements in the phosphoglycerate kinase 1 and lactate dehydrogenase A genes: Similarities with the erythropoietin 3’ enhancer. Proc. Natl. Acad. Sci. USA 1994, 91, 6496–6500. [Google Scholar] [CrossRef] [PubMed]
  93. Lu, S.; Gu, X.; Hoestje, S.; Epner, D.E. Identification of an additional hypoxia responsive element in the glyceraldehyde-3-phosphate dehydrogenase gene promoter. Biochim. Biophys. Acta 2002, 1574, 152–156. [Google Scholar] [CrossRef] [PubMed]
  94. Shah, Y.M.; Matsubara, T.; Ito, S.; Yim, S.H.; Gonzalez, F.J. Intestinal hypoxia-inducible transcription factors are essential for iron absorption following iron deficiency. Cell Metab. 2009, 9, 152–164. [Google Scholar] [CrossRef] [PubMed]
  95. Mastrogiannaki, M.; Matak, P.; Keith, B.; Simon, M.C.; Vaulont, S.; Peyssonnaux, C. HIF-2alpha, but not HIF-1alpha, promotes iron absorption in mice. J. Clin. Investig. 2009, 119, 1159–1166. [Google Scholar] [CrossRef]
  96. Davis, M.R.; Shawron, K.M.; Rendina, E.; Peterson, S.K.; Lucas, E.A.; Smith, B.J.; Clarke, S.L. Hypoxia inducible factor-2 α is translationally repressed in response to dietary iron deficiency in Sprague-Dawley rats. J. Nutr. 2011, 141, 1590–1596. [Google Scholar] [CrossRef] [PubMed]
  97. Sanchez, M.; Galy, B.; Muckenthaler, M.U.; Hentze, M.W. Iron-regulatory proteins limit hypoxia-inducible factor-2alpha expression in iron deficiency. Nat. Struct. Mol. Biol. 2007, 14, 420–426. [Google Scholar] [CrossRef]
  98. Ganz, T. Systemic iron homeostasis. Physiol. Rev. 2013, 93, 1721–1741. [Google Scholar] [CrossRef]
  99. Park, C.H.; Valore, E.V.; Waring, A.J.; Ganz, T. Hepcidin, a urinary antimicrobial peptide synthesized in the liver. J. Biol. Chem. 2001, 276, 7806–7810. [Google Scholar] [CrossRef]
  100. Abboud, S.; Haile, D.J. A novel mammalian iron-regulated protein involved in intracellular iron metabolism. J. Biol. Chem. 2000, 275, 19906–19912. [Google Scholar] [CrossRef]
  101. Qiao, B.; Sugianto, P.; Fung, E.; Del-Castillo-Rueda, A.; Moran-Jimenez, M.J.; Ganz, T.; Nemeth, E. Hepcidin-induced endocytosis of ferroportin is dependent on ferroportin ubiquitination. Cell Metab. 2012, 15, 918–924. [Google Scholar] [CrossRef]
  102. D’Alessio, F.; Hentze, M.W.; Muckenthaler, M.U. The hemochromatosis proteins HFE, TfR2, and HJV form a membrane-associated protein complex for hepcidin regulation. J. Hepatol. 2012, 57, 1052–1060. [Google Scholar] [CrossRef]
  103. Varga, E.; Pap, R.; Jánosa, G.; Sipos, K.; Pandur, E. IL-6 Regulates Hepcidin Expression Via the BMP/SMAD Pathway by Altering BMP6, TMPRSS6 and TfR2 Expressions at Normal and Inflammatory Conditions in BV2 Microglia. Neurochem. Res. 2021, 46, 1224–1238. [Google Scholar] [CrossRef] [PubMed]
  104. Kowdley, K.V.; Gochanour, E.M.; Sundaram, V.; Shah, R.A.; Handa, P. Hepcidin Signaling in Health and Disease: Ironing Out the Details. Hepatol. Commun. 2021, 5, 723–735. [Google Scholar] [CrossRef] [PubMed]
  105. Thévenod, F.; Wolff, N.A. Iron transport in the kidney: Implications for physiology and cadmium nephrotoxicity. Met. Integr. Biometal Sci. 2016, 8, 17–42. [Google Scholar] [CrossRef] [PubMed]
  106. 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]
  107. Kozyraki, R.; Fyfe, J.; Verroust, P.J.; Jacobsen, C.; Dautry-Varsat, A.; Gburek, J.; Willnow, T.E.; Christensen, E.I.; Moestrup, S.K. Megalin-dependent cubilin-mediated endocytosis is a major pathway for the apical uptake of transferrin in polarized epithelia. Proc. Natl. Acad. Sci. USA 2001, 98, 12491–12496. [Google Scholar] [CrossRef] [PubMed]
  108. Zhang, D.; Meyron-Holtz, E.; Rouault, T.A. Renal iron metabolism: Transferrin iron delivery and the role of iron regulatory proteins. J. Am. Soc. Nephrol. JASN 2007, 18, 401–406. [Google Scholar] [CrossRef] [PubMed]
  109. Weiss, A.; Spektor, L.; Cohen, L.A.; Lifshitz, L.; Magid Gold, I.; Zhang, D.L.; Truman-Rosentsvit, M.; Leichtmann-Bardoogo, Y.; Nyska, A.; Addadi, S.; et al. Orchestrated regulation of iron trafficking proteins in the kidney during iron overload facilitates systemic iron retention. PLoS ONE 2018, 13, e0204471. [Google Scholar] [CrossRef]
  110. Smith, C.P.; Lee, W.K.; Haley, M.; Poulsen, S.B.; Thévenod, F.; Fenton, R.A. Proximal tubule transferrin uptake is modulated by cellular iron and mediated by apical membrane megalin-cubilin complex and transferrin receptor 1. J. Biol. Chem. 2019, 294, 7025–7036. [Google Scholar] [CrossRef]
  111. Gburek, J.; Verroust, P.J.; Willnow, T.E.; Fyfe, J.C.; Nowacki, W.; Jacobsen, C.; Moestrup, S.K.; Christensen, E.I. Megalin and cubilin are endocytic receptors involved in renal clearance of hemoglobin. J. Am. Soc. Nephrol. JASN 2002, 13, 423–430. [Google Scholar] [CrossRef]
  112. van Swelm, R.P.L.; Vos, M.; Verhoeven, F.; Thévenod, F.; Swinkels, D.W. Endogenous hepcidin synthesis protects the distal nephron against hemin and hemoglobin mediated necroptosis. Cell Death Dis. 2018, 9, 550. [Google Scholar] [CrossRef]
  113. van Raaij, S.; van Swelm, R.; Bouman, K.; Cliteur, M.; van den Heuvel, M.C.; Pertijs, J.; Patel, D.; Bass, P.; van Goor, H.; Unwin, R.; et al. Publisher Correction: Tubular iron deposition and iron handling proteins in human healthy kidney and chronic kidney disease. Sci. Rep. 2018, 8, 13390. [Google Scholar] [CrossRef] [PubMed]
  114. He, L.; Girijashanker, K.; Dalton, T.P.; Reed, J.; Li, H.; Soleimani, M.; Nebert, D.W. ZIP8, member of the solute-carrier-39 (SLC39) metal-transporter family: Characterization of transporter properties. Mol. Pharmacol. 2006, 70, 171–180. [Google Scholar] [CrossRef]
  115. Pinilla-Tenas, J.J.; Sparkman, B.K.; Shawki, A.; Illing, A.C.; Mitchell, C.J.; Zhao, N.; Liuzzi, J.P.; Cousins, R.J.; Knutson, M.D.; Mackenzie, B. Zip14 is a complex broad-scope metal-ion transporter whose functional properties support roles in the cellular uptake of zinc and nontransferrin-bound iron. Am. J. Physiol. Cell Physiol. 2011, 301, C862–C871. [Google Scholar] [CrossRef] [PubMed]
  116. Langelueddecke, C.; Roussa, E.; Fenton, R.A.; Wolff, N.A.; Lee, W.K.; Thévenod, F. Lipocalin-2 (24p3/neutrophil gelatinase-associated lipocalin (NGAL)) receptor is expressed in distal nephron and mediates protein endocytosis. J. Biol. Chem. 2012, 287, 159–169. [Google Scholar] [CrossRef] [PubMed]
  117. Chen, A.C.; Donovan, A.; Ned-Sykes, R.; Andrews, N.C. Noncanonical role of transferrin receptor 1 is essential for intestinal homeostasis. Proc. Natl. Acad. Sci. USA 2015, 112, 11714–11719. [Google Scholar] [CrossRef]
  118. Rubio-Navarro, A.; Sanchez-Niño, M.D.; Guerrero-Hue, M.; García-Caballero, C.; Gutiérrez, E.; Yuste, C.; Sevillano, Á.; Praga, M.; Egea, J.; Román, E.; et al. Podocytes are new cellular targets of haemoglobin-mediated renal damage. J. Pathol. 2018, 244, 296–310. [Google Scholar] [CrossRef]
  119. Slotki, I.; Cabantchik, Z.I. The Labile Side of Iron Supplementation in CKD. J. Am. Soc. Nephrol. JASN 2015, 26, 2612–2619. [Google Scholar] [CrossRef] [PubMed]
  120. Moulouel, B.; Houamel, D.; Delaby, C.; Tchernitchko, D.; Vaulont, S.; Letteron, P.; Thibaudeau, O.; Puy, H.; Gouya, L.; Beaumont, C.; et al. Hepcidin regulates intrarenal iron handling at the distal nephron. Kidney Int. 2013, 84, 756–766. [Google Scholar] [CrossRef]
  121. Nakayama, M.; Kaizu, Y.; Uesugi, N.; Nakashita, S.; Suehiro, T. A case of IgA nephropathy and renal hemosiderosis associated with primary hemochromatosis. Ren. Fail. 2008, 30, 813–817. [Google Scholar] [CrossRef] [PubMed]
  122. Ozkurt, S.; Acikalin, M.F.; Temiz, G.; Akay, O.M.; Soydan, M. Renal hemosiderosis and rapidly progressive glomerulonephritis associated with primary hemochromatosis. Ren. Fail. 2014, 36, 814–816. [Google Scholar] [CrossRef] [PubMed]
  123. Adams, P.C.; Jeffrey, G.; Ryan, J. Haemochromatosis. Lancet 2023, 401, 1811–1821. [Google Scholar] [CrossRef] [PubMed]
  124. Olynyk, J.K.; Ramm, G.A. Hemochromatosis. N. Engl. J. Med. 2022, 387, 2159–2170. [Google Scholar] [CrossRef]
  125. Sponsel, H.T.; Alfrey, A.C.; Hammond, W.S.; Durr, J.A.; Ray, C.; Anderson, R.J. Effect of iron on renal tubular epithelial cells. Kidney Int. 1996, 50, 436–444. [Google Scholar] [CrossRef]
  126. Hassan, R.H.; Kandil, S.M.; Zeid, M.S.; Zaki, M.E.; Fouda, A.E. Kidney injury in infants and children with iron-deficiency anemia before and after iron treatment. Hematology 2017, 22, 565–570. [Google Scholar] [CrossRef] [PubMed]
  127. Norden, A.G.; Lapsley, M.; Lee, P.J.; Pusey, C.D.; Scheinman, S.J.; Tam, F.W.; Thakker, R.V.; Unwin, R.J.; Wrong, O. Glomerular protein sieving and implications for renal failure in Fanconi syndrome. Kidney Int. 2001, 60, 1885–1892. [Google Scholar] [CrossRef] [PubMed]
  128. Brown, E.A.; Sampson, B.; Muller, B.R.; Curtis, J.R. Urinary iron loss in the nephrotic syndrome--an unusual cause of iron deficiency with a note on urinary copper losses. Postgrad. Med. J. 1984, 60, 125–128. [Google Scholar] [CrossRef]
  129. Ellis, D. Anemia in the course of the nephrotic syndrome secondary to transferrin depletion. J. Pediatr. 1977, 90, 953–955. [Google Scholar] [CrossRef]
  130. Howard, R.L.; Buddington, B.; Alfrey, A.C. Urinary albumin, transferrin and iron excretion in diabetic patients. Kidney Int. 1991, 40, 923–926. [Google Scholar] [CrossRef]
  131. Prinsen, B.; Velden, M.; Kaysen, G.A.; Straver, H.; Rijn, H.; Stellaard, F.; Berger, R.; Rabelink, T.J. Transferrin synthesis is increased in nephrotic patients insufficiently to replace urinary losses. J. Am. Soc. Nephrol. JASN 2001, 12, 1017–1025. [Google Scholar] [CrossRef]
  132. Valko, M.; Leibfritz, D.; Moncol, J.; Cronin, M.T.; Mazur, M.; Telser, J. Free radicals and antioxidants in normal physiological functions and human disease. Int. J. Biochem. Cell Biol. 2007, 39, 44–84. [Google Scholar] [CrossRef]
  133. Sundaresan, M.; Yu, Z.X.; Ferrans, V.J.; Irani, K.; Finkel, T. Requirement for generation of H2O2 for platelet-derived growth factor signal transduction. Science 1995, 270, 296–299. [Google Scholar] [CrossRef]
  134. Lemmon, M.A.; Schlessinger, J. Cell signaling by receptor tyrosine kinases. Cell 2010, 141, 1117–1134. [Google Scholar] [CrossRef]
  135. Cantley, L.C. The phosphoinositide 3-kinase pathway. Science 2002, 296, 1655–1657. [Google Scholar] [CrossRef] [PubMed]
  136. Sena, L.A.; Li, S.; Jairaman, A.; Prakriya, M.; Ezponda, T.; Hildeman, D.A.; Wang, C.R.; Schumacker, P.T.; Licht, J.D.; Perlman, H.; et al. Mitochondria are required for antigen-specific T cell activation through reactive oxygen species signaling. Immunity 2013, 38, 225–236. [Google Scholar] [CrossRef] [PubMed]
  137. West, A.P.; Brodsky, I.E.; Rahner, C.; Woo, D.K.; Erdjument-Bromage, H.; Tempst, P.; Walsh, M.C.; Choi, Y.; Shadel, G.S.; Ghosh, S. TLR signalling augments macrophage bactericidal activity through mitochondrial ROS. Nature 2011, 472, 476–480. [Google Scholar] [CrossRef] [PubMed]
  138. Baud, L.; Hagege, J.; Sraer, J.; Rondeau, E.; Perez, J.; Ardaillou, R. Reactive oxygen production by cultured rat glomerular mesangial cells during phagocytosis is associated with stimulation of lipoxygenase activity. J. Exp. Med. 1983, 158, 1836–1852. [Google Scholar] [CrossRef]
  139. Ohsaki, Y.; O’Connor, P.; Mori, T.; Ryan, R.P.; Dickinson, B.C.; Chang, C.J.; Lu, Y.; Ito, S.; Cowley, A.W., Jr. Increase of sodium delivery stimulates the mitochondrial respiratory chain H2O2 production in rat renal medullary thick ascending limb. Am. J. Physiol. Ren. Physiol. 2012, 302, F95–F102. [Google Scholar] [CrossRef]
  140. Radeke, H.H.; Cross, A.R.; Hancock, J.T.; Jones, O.T.; Nakamura, M.; Kaever, V.; Resch, K. Functional expression of NADPH oxidase components (alpha- and beta-subunits of cytochrome b558 and 45-kDa flavoprotein) by intrinsic human glomerular mesangial cells. J. Biol. Chem. 1991, 266, 21025–21029. [Google Scholar] [CrossRef]
  141. Pfaller, W.; Rittinger, M. Quantitative morphology of the rat kidney. Int. J. Biochem. 1980, 12, 17–22. [Google Scholar] [CrossRef] [PubMed]
  142. Zhao, Z. Hydroxyl radical generations form the physiologically relevant Fenton-like reactions. Free Radic. Biol. Med. 2023, 208, 510–515. [Google Scholar] [CrossRef] [PubMed]
  143. Starkov, A.A.; Fiskum, G.; Chinopoulos, C.; Lorenzo, B.J.; Browne, S.E.; Patel, M.S.; Beal, M.F. Mitochondrial alpha-ketoglutarate dehydrogenase complex generates reactive oxygen species. J. Neurosci. Off. J. Soc. Neurosci. 2004, 24, 7779–7788. [Google Scholar] [CrossRef]
  144. Mailloux, R.J.; Jin, X.; Willmore, W.G. Redox regulation of mitochondrial function with emphasis on cysteine oxidation reactions. Redox Biol. 2014, 2, 123–139. [Google Scholar] [CrossRef] [PubMed]
  145. Bedard, K.; Krause, K.H. The NOX family of ROS-generating NADPH oxidases: Physiology and pathophysiology. Physiol. Rev. 2007, 87, 245–313. [Google Scholar] [CrossRef] [PubMed]
  146. Block, K.; Gorin, Y.; Abboud, H.E. Subcellular localization of Nox4 and regulation in diabetes. Proc. Natl. Acad. Sci. USA 2009, 106, 14385–14390. [Google Scholar] [CrossRef] [PubMed]
  147. Takac, I.; Schröder, K.; Zhang, L.; Lardy, B.; Anilkumar, N.; Lambeth, J.D.; Shah, A.M.; Morel, F.; Brandes, R.P. The E-loop is involved in hydrogen peroxide formation by the NADPH oxidase Nox4. J. Biol. Chem. 2011, 286, 13304–13313. [Google Scholar] [CrossRef] [PubMed]
  148. Förstermann, U.; Sessa, W.C. Nitric oxide synthases: Regulation and function. Eur. Heart J. 2012, 33, 829–837, 837a–837d. [Google Scholar] [CrossRef] [PubMed]
  149. Carlström, M. Nitric oxide signalling in kidney regulation and cardiometabolic health. Nat. Rev. Nephrol. 2021, 17, 575–590. [Google Scholar] [CrossRef]
  150. Baylis, C. Arginine, arginine analogs and nitric oxide production in chronic kidney disease. Nat. Clin. Pract. Nephrol. 2006, 2, 209–220. [Google Scholar] [CrossRef]
  151. Nishimura, K.; Taguchi, K.; Kishi, S.; Brooks, C.R.; Ochi, A.; Kadoya, H.; Ikeda, Y.; Miyoshi, M.; Tamaki, M.; Abe, H.; et al. Dual disruption of eNOS and ApoE gene accelerates kidney fibrosis and senescence after injury. Biochem. Biophys. Res. Commun. 2021, 556, 142–148. [Google Scholar] [CrossRef] [PubMed]
  152. Knowles, J.W.; Reddick, R.L.; Jennette, J.C.; Shesely, E.G.; Smithies, O.; Maeda, N. Enhanced atherosclerosis and kidney dysfunction in eNOS(-/-)Apoe(-/-) mice are ameliorated by enalapril treatment. J. Clin. Investig. 2000, 105, 451–458. [Google Scholar] [CrossRef] [PubMed]
  153. Carlström, M.; Larsen, F.J.; Nyström, T.; Hezel, M.; Borniquel, S.; Weitzberg, E.; Lundberg, J.O. Dietary inorganic nitrate reverses features of metabolic syndrome in endothelial nitric oxide synthase-deficient mice. Proc. Natl. Acad. Sci. USA 2010, 107, 17716–17720. [Google Scholar] [CrossRef]
  154. Satoh, M.; Fujimoto, S.; Haruna, Y.; Arakawa, S.; Horike, H.; Komai, N.; Sasaki, T.; Tsujioka, K.; Makino, H.; Kashihara, N. NAD(P)H oxidase and uncoupled nitric oxide synthase are major sources of glomerular superoxide in rats with experimental diabetic nephropathy. Am. J. Physiol. Ren. Physiol. 2005, 288, F1144–F1152. [Google Scholar] [CrossRef] [PubMed]
  155. Gil, C.L.; Hooker, E.; Larrivée, B. Diabetic Kidney Disease, Endothelial Damage, and Podocyte-Endothelial Crosstalk. Kidney Med. 2021, 3, 105–115. [Google Scholar] [CrossRef]
  156. Janaszak-Jasiecka, A.; Płoska, A.; Wierońska, J.M.; Dobrucki, L.W.; Kalinowski, L. Endothelial dysfunction due to eNOS uncoupling: Molecular mechanisms as potential therapeutic targets. Cell. Mol. Biol. Lett. 2023, 28, 21. [Google Scholar] [CrossRef] [PubMed]
  157. Forrester, S.J.; Kikuchi, D.S.; Hernandes, M.S.; Xu, Q.; Griendling, K.K. Reactive Oxygen Species in Metabolic and Inflammatory Signaling. Circ. Res. 2018, 122, 877–902. [Google Scholar] [CrossRef] [PubMed]
  158. Shinozaki, K.; Nishio, Y.; Okamura, T.; Yoshida, Y.; Maegawa, H.; Kojima, H.; Masada, M.; Toda, N.; Kikkawa, R.; Kashiwagi, A. Oral administration of tetrahydrobiopterin prevents endothelial dysfunction and vascular oxidative stress in the aortas of insulin-resistant rats. Circ. Res. 2000, 87, 566–573. [Google Scholar] [CrossRef]
  159. Yamamoto, E.; Hirata, Y.; Tokitsu, T.; Kusaka, H.; Sakamoto, K.; Yamamuro, M.; Kaikita, K.; Watanabe, H.; Hokimoto, S.; Sugiyama, S.; et al. The pivotal role of eNOS uncoupling in vascular endothelial dysfunction in patients with heart failure with preserved ejection fraction. Int. J. Cardiol. 2015, 190, 335–337. [Google Scholar] [CrossRef]
  160. Satoh, M.; Fujimoto, S.; Arakawa, S.; Yada, T.; Namikoshi, T.; Haruna, Y.; Horike, H.; Sasaki, T.; Kashihara, N. Angiotensin II type 1 receptor blocker ameliorates uncoupled endothelial nitric oxide synthase in rats with experimental diabetic nephropathy. Nephrol. Dial. Transpl. 2008, 23, 3806–3813. [Google Scholar] [CrossRef]
  161. Fujita, H.; Fujishima, H.; Chida, S.; Takahashi, K.; Qi, Z.; Kanetsuna, Y.; Breyer, M.D.; Harris, R.C.; Yamada, Y.; Takahashi, T. Reduction of renal superoxide dismutase in progressive diabetic nephropathy. J. Am. Soc. Nephrol. JASN 2009, 20, 1303–1313. [Google Scholar] [CrossRef] [PubMed]
  162. Davies, S.J.; Reichardt-Pascal, S.Y.; Vaughan, D.; Russell, G.I. Differential effect of ischaemia-reperfusion injury on anti-oxidant enzyme activity in the rat kidney. Exp. Nephrol. 1995, 3, 348–354. [Google Scholar]
  163. Ho, Y.S.; Xiong, Y.; Ma, W.; Spector, A.; Ho, D.S. Mice lacking catalase develop normally but show differential sensitivity to oxidant tissue injury. J. Biol. Chem. 2004, 279, 32804–32812. [Google Scholar] [CrossRef] [PubMed]
  164. Lee, Y.M.; He, W.; Liou, Y.C. The redox language in neurodegenerative diseases: Oxidative post-translational modifications by hydrogen peroxide. Cell Death Dis. 2021, 12, 58. [Google Scholar] [CrossRef] [PubMed]
  165. Yamashita, K.; Tamura, S.; Honsho, M.; Yada, H.; Yagita, Y.; Kosako, H.; Fujiki, Y. Mitotic phosphorylation of Pex14p regulates peroxisomal import machinery. J. Cell Biol. 2020, 219, e202001003. [Google Scholar] [CrossRef] [PubMed]
  166. Antonenkov, V.D.; Grunau, S.; Ohlmeier, S.; Hiltunen, J.K. Peroxisomes are oxidative organelles. Antioxid. Redox Signal. 2010, 13, 525–537. [Google Scholar] [CrossRef] [PubMed]
  167. Lubos, E.; Loscalzo, J.; Handy, D.E. Glutathione peroxidase-1 in health and disease: From molecular mechanisms to therapeutic opportunities. Antioxid. Redox Signal. 2011, 15, 1957–1997. [Google Scholar] [CrossRef] [PubMed]
  168. Jomova, K.; Raptova, R.; Alomar, S.Y.; Alwasel, S.H.; Nepovimova, E.; Kuca, K.; Valko, M. Reactive oxygen species, toxicity, oxidative stress, and antioxidants: Chronic diseases and aging. Arch. Toxicol. 2023, 97, 2499–2574. [Google Scholar] [CrossRef] [PubMed]
  169. Cao, J.Y.; Dixon, S.J. Mechanisms of ferroptosis. Cell. Mol. Life Sci. CMLS 2016, 73, 2195–2209. [Google Scholar] [CrossRef]
  170. Wang, H.; Liu, C.; Zhao, Y.; Gao, G. Mitochondria regulation in ferroptosis. Eur. J. Cell Biol. 2020, 99, 151058. [Google Scholar] [CrossRef]
  171. Chae, H.Z.; Chung, S.J.; Rhee, S.G. Thioredoxin-dependent peroxide reductase from yeast. J. Biol. Chem. 1994, 269, 27670–27678. [Google Scholar] [CrossRef] [PubMed]
  172. Wood, Z.A.; Schröder, E.; Robin Harris, J.; Poole, L.B. Structure, mechanism and regulation of peroxiredoxins. Trends Biochem. Sci. 2003, 28, 32–40. [Google Scholar] [CrossRef] [PubMed]
  173. Rhee, S.G.; Woo, H.A. Multiple functions of peroxiredoxins: Peroxidases, sensors and regulators of the intracellular messenger H₂O₂, and protein chaperones. Antioxid. Redox Signal. 2011, 15, 781–794. [Google Scholar] [CrossRef] [PubMed]
  174. Lu, J.; Holmgren, A. The thioredoxin antioxidant system. Free Radic. Biol. Med. 2014, 66, 75–87. [Google Scholar] [CrossRef] [PubMed]
  175. Cheng, Q.; Sandalova, T.; Lindqvist, Y.; Arnér, E.S. Crystal structure and catalysis of the selenoprotein thioredoxin reductase 1. J. Biol. Chem. 2009, 284, 3998–4008. [Google Scholar] [CrossRef] [PubMed]
  176. Zhong, L.; Holmgren, A. Essential role of selenium in the catalytic activities of mammalian thioredoxin reductase revealed by characterization of recombinant enzymes with selenocysteine mutations. J. Biol. Chem. 2000, 275, 18121–18128. [Google Scholar] [CrossRef] [PubMed]
  177. Du, Y.; Zhang, H.; Lu, J.; Holmgren, A. Glutathione and glutaredoxin act as a backup of human thioredoxin reductase 1 to reduce thioredoxin 1 preventing cell death by aurothioglucose. J. Biol. Chem. 2012, 287, 38210–38219. [Google Scholar] [CrossRef] [PubMed]
  178. Couto, N.; Wood, J.; Barber, J. The role of glutathione reductase and related enzymes on cellular redox homoeostasis network. Free Radic. Biol. Med. 2016, 95, 27–42. [Google Scholar] [CrossRef]
  179. Lennicke, C.; Rahn, J.; Lichtenfels, R.; Wessjohann, L.A.; Seliger, B. Hydrogen peroxide—Production, fate and role in redox signaling of tumor cells. Cell Commun. Signal. CCS 2015, 13, 39. [Google Scholar] [CrossRef]
  180. Valko, M.; Rhodes, C.J.; Moncol, J.; Izakovic, M.; Mazur, M. Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chem.-Biol. Interact. 2006, 160, 1–40. [Google Scholar] [CrossRef]
  181. Pham-Huy, L.A.; He, H.; Pham-Huy, C. Free radicals, antioxidants in disease and health. Int. J. Biomed. Sci. IJBS 2008, 4, 89–96. [Google Scholar] [CrossRef] [PubMed]
  182. Finkel, T. Signal transduction by reactive oxygen species. J. Cell Biol. 2011, 194, 7–15. [Google Scholar] [CrossRef] [PubMed]
  183. Liguori, I.; Russo, G.; Curcio, F.; Bulli, G.; Aran, L.; Della-Morte, D.; Gargiulo, G.; Testa, G.; Cacciatore, F.; Bonaduce, D.; et al. Oxidative stress, aging, and diseases. Clin. Interv. Aging 2018, 13, 757–772. [Google Scholar] [CrossRef] [PubMed]
  184. Valko, M.; Izakovic, M.; Mazur, M.; Rhodes, C.J.; Telser, J. Role of oxygen radicals in DNA damage and cancer incidence. Mol. Cell. Biochem. 2004, 266, 37–56. [Google Scholar] [CrossRef] [PubMed]
  185. Cooke, M.S.; Evans, M.D.; Dizdaroglu, M.; Lunec, J. Oxidative DNA damage: Mechanisms, mutation, and disease. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2003, 17, 1195–1214. [Google Scholar] [CrossRef] [PubMed]
  186. Lombardi, V.; Valko, L.; Stolc, S.; Valko, M.; Ondrejicková, O.; Horáková, L.; Placek, J.; Troncone, A. Free radicals in rabbit spinal cord ischemia: Electron spin resonance spectroscopy and correlation with SOD activity. Cell. Mol. Neurobiol. 1998, 18, 399–412. [Google Scholar] [CrossRef] [PubMed]
  187. Stolc, S.; Valko, L.; Valko, M.; Lombardi, V. A technique for the fast sampling of biological tissues for electron paramagnetic resonance spectroscopy. Free Radic. Biol. Med. 1996, 20, 89–91. [Google Scholar] [CrossRef]
  188. Guyton, K.Z.; Kensler, T.W. Oxidative mechanisms in carcinogenesis. Br. Med. Bull. 1993, 49, 523–544. [Google Scholar] [CrossRef] [PubMed]
  189. Luo, C.; Zhou, S.; Zhou, Z.; Liu, Y.; Yang, L.; Liu, J.; Zhang, Y.; Li, H.; Liu, Y.; Hou, F.F.; et al. Wnt9a Promotes Renal Fibrosis by Accelerating Cellular Senescence in Tubular Epithelial Cells. J. Am. Soc. Nephrol. JASN 2018, 29, 1238–1256. [Google Scholar] [CrossRef]
  190. Kim, S.R.; Puranik, A.S.; Jiang, K.; Chen, X.; Zhu, X.Y.; Taylor, I.; Khodadadi-Jamayran, A.; Lerman, A.; Hickson, L.J.; Childs, B.G.; et al. Progressive Cellular Senescence Mediates Renal Dysfunction in Ischemic Nephropathy. J. Am. Soc. Nephrol. JASN 2021, 32, 1987–2004. [Google Scholar] [CrossRef]
  191. van Deursen, J.M. The role of senescent cells in ageing. Nature 2014, 509, 439–446. [Google Scholar] [CrossRef] [PubMed]
  192. Takahashi, A.; Loo, T.M.; Okada, R.; Kamachi, F.; Watanabe, Y.; Wakita, M.; Watanabe, S.; Kawamoto, S.; Miyata, K.; Barber, G.N.; et al. Downregulation of cytoplasmic DNases is implicated in cytoplasmic DNA accumulation and SASP in senescent cells. Nat. Commun. 2018, 9, 1249. [Google Scholar] [CrossRef]
  193. Freund, A.; Orjalo, A.V.; Desprez, P.Y.; Campisi, J. Inflammatory networks during cellular senescence: Causes and consequences. Trends Mol. Med. 2010, 16, 238–246. [Google Scholar] [CrossRef]
  194. Xu, J.; Zhou, L.; Liu, Y. Cellular Senescence in Kidney Fibrosis: Pathologic Significance and Therapeutic Strategies. Front. Pharmacol. 2020, 11, 601325. [Google Scholar] [CrossRef]
  195. Kanwar, Y.S.; Sun, L.; Xie, P.; Liu, F.Y.; Chen, S. A glimpse of various pathogenetic mechanisms of diabetic nephropathy. Annu. Rev. Pathol. 2011, 6, 395–423. [Google Scholar] [CrossRef]
  196. Kumar, D.; Robertson, S.; Burns, K.D. Evidence of apoptosis in human diabetic kidney. Mol. Cell. Biochem. 2004, 259, 67–70. [Google Scholar] [CrossRef]
  197. Abais, J.M.; Xia, M.; Zhang, Y.; Boini, K.M.; Li, P.L. Redox regulation of NLRP3 inflammasomes: ROS as trigger or effector? Antioxid. Redox Signal. 2015, 22, 1111–1129. [Google Scholar] [CrossRef] [PubMed]
  198. Pan, J.; Ou, Z.; Cai, C.; Li, P.; Gong, J.; Ruan, X.Z.; He, K. Fatty acid activates NLRP3 inflammasomes in mouse Kupffer cells through mitochondrial DNA release. Cell. Immunol. 2018, 332, 111–120. [Google Scholar] [CrossRef] [PubMed]
  199. Mori, Y.; Ajay, A.K.; Chang, J.H.; Mou, S.; Zhao, H.; Kishi, S.; Li, J.; Brooks, C.R.; Xiao, S.; Woo, H.M.; et al. KIM-1 mediates fatty acid uptake by renal tubular cells to promote progressive diabetic kidney disease. Cell Metab. 2021, 33, 1042–1061.e1047. [Google Scholar] [CrossRef]
  200. Makris, K.; Spanou, L. Acute Kidney Injury: Definition, Pathophysiology and Clinical Phenotypes. Clin. Biochemist. Rev. 2016, 37, 85–98. [Google Scholar]
  201. Munshi, R.; Hsu, C.; Himmelfarb, J. Advances in understanding ischemic acute kidney injury. BMC Med. 2011, 9, 11. [Google Scholar] [CrossRef]
  202. Raimondi, V.; Ciccarese, F.; Ciminale, V. Oncogenic pathways and the electron transport chain: A dangeROS liaison. Br. J. Cancer 2020, 122, 168–181. [Google Scholar] [CrossRef]
  203. Hall, A.M.; Rhodes, G.J.; Sandoval, R.M.; Corridon, P.R.; Molitoris, B.A. In vivo multiphoton imaging of mitochondrial structure and function during acute kidney injury. Kidney Int. 2013, 83, 72–83. [Google Scholar] [CrossRef]
  204. Sharfuddin, A.A.; Molitoris, B.A. Pathophysiology of ischemic acute kidney injury. Nat. Rev. Nephrol. 2011, 7, 189–200. [Google Scholar] [CrossRef]
  205. Zweier, J.L.; Flaherty, J.T.; Weisfeldt, M.L. Direct measurement of free radical generation following reperfusion of ischemic myocardium. Proc. Natl. Acad. Sci. USA 1987, 84, 1404–1407. [Google Scholar] [CrossRef]
  206. Chouchani, E.T.; Pell, V.R.; James, A.M.; Work, L.M.; Saeb-Parsy, K.; Frezza, C.; Krieg, T.; Murphy, M.P. A Unifying Mechanism for Mitochondrial Superoxide Production during Ischemia-Reperfusion Injury. Cell Metab. 2016, 23, 254–263. [Google Scholar] [CrossRef] [PubMed]
  207. Shanley, P.F.; Rosen, M.D.; Brezis, M.; Silva, P.; Epstein, F.H.; Rosen, S. Topography of focal proximal tubular necrosis after ischemia with reflow in the rat kidney. Am. J. Pathol. 1986, 122, 462–468. [Google Scholar]
  208. Friedewald, J.J.; Rabb, H. Inflammatory cells in ischemic acute renal failure. Kidney Int. 2004, 66, 486–491. [Google Scholar] [CrossRef] [PubMed]
  209. Jia, P.; Wu, X.; Pan, T.; Xu, S.; Hu, J.; Ding, X. Uncoupling protein 1 inhibits mitochondrial reactive oxygen species generation and alleviates acute kidney injury. eBioMedicine 2019, 49, 331–340. [Google Scholar] [CrossRef] [PubMed]
  210. Herb, M.; Schramm, M. Functions of ROS in Macrophages and Antimicrobial Immunity. Antioxidants 2021, 10, 313. [Google Scholar] [CrossRef]
  211. Mittal, M.; Siddiqui, M.R.; Tran, K.; Reddy, S.P.; Malik, A.B. Reactive oxygen species in inflammation and tissue injury. Antioxid. Redox Signal. 2014, 20, 1126–1167. [Google Scholar] [CrossRef] [PubMed]
  212. Pavlakou, P.; Liakopoulos, V.; Eleftheriadis, T.; Mitsis, M.; Dounousi, E. Oxidative Stress and Acute Kidney Injury in Critical Illness: Pathophysiologic Mechanisms-Biomarkers-Interventions, and Future Perspectives. Oxid. Med. Cell. Longev. 2017, 2017, 6193694. [Google Scholar] [CrossRef] [PubMed]
  213. Radi, R. Peroxynitrite, a stealthy biological oxidant. J. Biol. Chem. 2013, 288, 26464–26472. [Google Scholar] [CrossRef] [PubMed]
  214. Zarbock, A.; Gomez, H.; Kellum, J.A. Sepsis-induced acute kidney injury revisited: Pathophysiology, prevention and future therapies. Curr. Opin. Crit. Care 2014, 20, 588–595. [Google Scholar] [CrossRef] [PubMed]
  215. Arany, I.; Safirstein, R.L. Cisplatin nephrotoxicity. Semin. Nephrol. 2003, 23, 460–464. [Google Scholar] [CrossRef] [PubMed]
  216. Kruidering, M.; Van de Water, B.; de Heer, E.; Mulder, G.J.; Nagelkerke, J.F. Cisplatin-induced nephrotoxicity in porcine proximal tubular cells: Mitochondrial dysfunction by inhibition of complexes I to IV of the respiratory chain. J. Pharmacol. Exp. Ther. 1997, 280, 638–649. [Google Scholar] [PubMed]
  217. Susztak, K.; Raff, A.C.; Schiffer, M.; Böttinger, E.P. Glucose-induced reactive oxygen species cause apoptosis of podocytes and podocyte depletion at the onset of diabetic nephropathy. Diabetes 2006, 55, 225–233. [Google Scholar] [CrossRef] [PubMed]
  218. Takai, Y.; Sasaki, T.; Matozaki, T. Small GTP-binding proteins. Physiol. Rev. 2001, 81, 153–208. [Google Scholar] [CrossRef] [PubMed]
  219. Heasman, S.J.; Ridley, A.J. Mammalian Rho GTPases: New insights into their functions from in vivo studies. Nat. Rev. Mol. Cell Biol. 2008, 9, 690–701. [Google Scholar] [CrossRef]
  220. Nagase, M.; Ayuzawa, N.; Kawarazaki, W.; Ishizawa, K.; Ueda, K.; Yoshida, S.; Fujita, T. Oxidative stress causes mineralocorticoid receptor activation in rat cardiomyocytes: Role of small GTPase Rac1. Hypertension 2012, 59, 500–506. [Google Scholar] [CrossRef]
  221. Papaharalambus, C.; Sajjad, W.; Syed, A.; Zhang, C.; Bergo, M.O.; Alexander, R.W.; Ahmad, M. Tumor necrosis factor alpha stimulation of Rac1 activity. Role of isoprenylcysteine carboxylmethyltransferase. J. Biol. Chem. 2005, 280, 18790–18796. [Google Scholar] [CrossRef] [PubMed]
  222. Shen, E.; Li, Y.; Li, Y.; Shan, L.; Zhu, H.; Feng, Q.; Arnold, J.M.; Peng, T. Rac1 is required for cardiomyocyte apoptosis during hyperglycemia. Diabetes 2009, 58, 2386–2395. [Google Scholar] [CrossRef] [PubMed]
  223. Nagase, M.; Fujita, T. Role of Rac1-mineralocorticoid-receptor signalling in renal and cardiac disease. Nat. Rev. Nephrol. 2013, 9, 86–98. [Google Scholar] [CrossRef] [PubMed]
  224. Mundel, P.; Reiser, J. Proteinuria: An enzymatic disease of the podocyte? Kidney Int. 2010, 77, 571–580. [Google Scholar] [CrossRef] [PubMed]
  225. D’Agati, V.D.; Kaskel, F.J.; Falk, R.J. Focal segmental glomerulosclerosis. N. Engl. J. Med. 2011, 365, 2398–2411. [Google Scholar] [CrossRef] [PubMed]
  226. Hsu, H.H.; Hoffmann, S.; Endlich, N.; Velic, A.; Schwab, A.; Weide, T.; Schlatter, E.; Pavenstädt, H. Mechanisms of angiotensin II signaling on cytoskeleton of podocytes. J. Mol. Med. 2008, 86, 1379–1394. [Google Scholar] [CrossRef] [PubMed]
  227. Inagi, R.; Ishimoto, Y.; Nangaku, M. Proteostasis in endoplasmic reticulum--new mechanisms in kidney disease. Nat. Rev. Nephrol. 2014, 10, 369–378. [Google Scholar] [CrossRef]
  228. Inagi, R.; Nangaku, M.; Onogi, H.; Ueyama, H.; Kitao, Y.; Nakazato, K.; Ogawa, S.; Kurokawa, K.; Couser, W.G.; Miyata, T. Involvement of endoplasmic reticulum (ER) stress in podocyte injury induced by excessive protein accumulation. Kidney Int. 2005, 68, 2639–2650. [Google Scholar] [CrossRef] [PubMed]
  229. Alpers, C.E.; Pichler, R.; Johnson, R.J. Phenotypic features of cortical interstitial cells potentially important in fibrosis. Kidney Int. Suppl. 1996, 54, S28–S31. [Google Scholar]
  230. Kliem, V.; Johnson, R.J.; Alpers, C.E.; Yoshimura, A.; Couser, W.G.; Koch, K.M.; Floege, J. Mechanisms involved in the pathogenesis of tubulointerstitial fibrosis in 5/6-nephrectomized rats. Kidney Int. 1996, 49, 666–678. [Google Scholar] [CrossRef]
  231. Chai, Q.; Krag, S.; Chai, S.; Ledet, T.; Wogensen, L. Localisation and phenotypical characterisation of collagen-producing cells in TGF-beta 1-induced renal interstitial fibrosis. Histochem. Cell Biol. 2003, 119, 267–280. [Google Scholar] [CrossRef] [PubMed]
  232. Barnes, J.L.; Hastings, R.R.; De la Garza, M.A. Sequential expression of cellular fibronectin by platelets, macrophages, and mesangial cells in proliferative glomerulonephritis. Am. J. Pathol. 1994, 145, 585–597. [Google Scholar] [PubMed]
  233. Tang, W.W.; Van, G.Y.; Qi, M. Myofibroblast and alpha 1 (III) collagen expression in experimental tubulointerstitial nephritis. Kidney Int. 1997, 51, 926–931. [Google Scholar] [CrossRef] [PubMed]
  234. Gorin, Y.; Block, K.; Hernandez, J.; Bhandari, B.; Wagner, B.; Barnes, J.L.; Abboud, H.E. Nox4 NAD(P)H oxidase mediates hypertrophy and fibronectin expression in the diabetic kidney. J. Biol. Chem. 2005, 280, 39616–39626. [Google Scholar] [CrossRef] [PubMed]
  235. Bondi, C.D.; Manickam, N.; Lee, D.Y.; Block, K.; Gorin, Y.; Abboud, H.E.; Barnes, J.L. NAD(P)H oxidase mediates TGF-beta1-induced activation of kidney myofibroblasts. J. Am. Soc. Nephrol. JASN 2010, 21, 93–102. [Google Scholar] [CrossRef] [PubMed]
  236. Cucoranu, I.; Clempus, R.; Dikalova, A.; Phelan, P.J.; Ariyan, S.; Dikalov, S.; Sorescu, D. NAD(P)H oxidase 4 mediates transforming growth factor-beta1-induced differentiation of cardiac fibroblasts into myofibroblasts. Circ. Res. 2005, 97, 900–907. [Google Scholar] [CrossRef] [PubMed]
  237. Block, K.; Eid, A.; Griendling, K.K.; Lee, D.Y.; Wittrant, Y.; Gorin, Y. Nox4 NAD(P)H oxidase mediates Src-dependent tyrosine phosphorylation of PDK-1 in response to angiotensin II: Role in mesangial cell hypertrophy and fibronectin expression. J. Biol. Chem. 2008, 283, 24061–24076. [Google Scholar] [CrossRef] [PubMed]
  238. Modlinger, P.S.; Wilcox, C.S.; Aslam, S. Nitric oxide, oxidative stress, and progression of chronic renal failure. Semin. Nephrol. 2004, 24, 354–365. [Google Scholar] [CrossRef] [PubMed]
  239. Yilmaz, M.I.; Saglam, M.; Caglar, K.; Cakir, E.; Sonmez, A.; Ozgurtas, T.; Aydin, A.; Eyileten, T.; Ozcan, O.; Acikel, C.; et al. The determinants of endothelial dysfunction in CKD: Oxidative stress and asymmetric dimethylarginine. Am. J. Kidney Dis. Off. J. Natl. Kidney Found. 2006, 47, 42–50. [Google Scholar] [CrossRef]
  240. Sullivan, J.C.; Pollock, J.S. Coupled and uncoupled NOS: Separate but equal? Uncoupled NOS in endothelial cells is a critical pathway for intracellular signaling. Circ. Res. 2006, 98, 717–719. [Google Scholar] [CrossRef]
  241. Aldámiz-Echevarría, L.; Andrade, F. Asymmetric dimethylarginine, endothelial dysfunction and renal disease. Int. J. Mol. Sci. 2012, 13, 11288–11311. [Google Scholar] [CrossRef] [PubMed]
  242. Chu, S.; Mao, X.; Guo, H.; Wang, L.; Li, Z.; Zhang, Y.; Wang, Y.; Wang, H.; Zhang, X.; Peng, W. Indoxyl sulfate potentiates endothelial dysfunction via reciprocal role for reactive oxygen species and RhoA/ROCK signaling in 5/6 nephrectomized rats. Free Radic. Res. 2017, 51, 237–252. [Google Scholar] [CrossRef] [PubMed]
  243. Canning, P.; Sorrell, F.J.; Bullock, A.N. Structural basis of Keap1 interactions with Nrf2. Free Radic. Biol. Med. 2015, 88, 101–107. [Google Scholar] [CrossRef] [PubMed]
  244. 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] [PubMed]
  245. Guerrero-Hue, M.; Rayego-Mateos, S.; Vázquez-Carballo, C.; Palomino-Antolín, A.; García-Caballero, C.; Opazo-Rios, L.; Morgado-Pascual, J.L.; Herencia, C.; Mas, S.; Ortiz, A.; et al. Protective Role of Nrf2 in Renal Disease. Antioxidants 2020, 10, 39. [Google Scholar] [CrossRef] [PubMed]
  246. Cuadrado, A.; Rojo, A.I.; Wells, G.; Hayes, J.D.; Cousin, S.P.; Rumsey, W.L.; Attucks, O.C.; Franklin, S.; Levonen, A.L.; Kensler, T.W.; et al. Therapeutic targeting of the NRF2 and KEAP1 partnership in chronic diseases. Nat. Rev. Drug Discov. 2019, 18, 295–317. [Google Scholar] [CrossRef] [PubMed]
  247. Gorrini, C.; Harris, I.S.; Mak, T.W. Modulation of oxidative stress as an anticancer strategy. Nat. Rev. Drug Discov. 2013, 12, 931–947. [Google Scholar] [CrossRef] [PubMed]
  248. Benarroch, E.E. Nrf2, cellular redox regulation, and neurologic implications. Neurology 2017, 88, 1942–1950. [Google Scholar] [CrossRef] [PubMed]
  249. Lin, D.W.; Hsu, Y.C.; Chang, C.C.; Hsieh, C.C.; Lin, C.L. Insights into the Molecular Mechanisms of NRF2 in Kidney Injury and Diseases. Int. J. Mol. Sci. 2023, 24, 6053. [Google Scholar] [CrossRef]
  250. Kong, W.; Fu, J.; Liu, N.; Jiao, C.; Guo, G.; Luan, J.; Wang, H.; Yao, L.; Wang, L.; Yamamoto, M.; et al. Nrf2 deficiency promotes the progression from acute tubular damage to chronic renal fibrosis following unilateral ureteral obstruction. Nephrol. Dial. Transplant. Off. Publ. Eur. Dial. Transpl. Assoc.-Eur. Ren. Assoc. 2018, 33, 771–783. [Google Scholar] [CrossRef]
  251. Dandona, P.; Thusu, K.; Cook, S.; Snyder, B.; Makowski, J.; Armstrong, D.; Nicotera, T. Oxidative damage to DNA in diabetes mellitus. Lancet 1996, 347, 444–445. [Google Scholar] [CrossRef] [PubMed]
  252. Jiang, T.; Huang, Z.; Lin, Y.; Zhang, Z.; Fang, D.; Zhang, D.D. The protective role of Nrf2 in streptozotocin-induced diabetic nephropathy. Diabetes 2010, 59, 850–860. [Google Scholar] [CrossRef] [PubMed]
  253. Yoh, K.; Hirayama, A.; Ishizaki, K.; Yamada, A.; Takeuchi, M.; Yamagishi, S.; Morito, N.; Nakano, T.; Ojima, M.; Shimohata, H.; et al. Hyperglycemia induces oxidative and nitrosative stress and increases renal functional impairment in Nrf2-deficient mice. Genes Cells Devoted Mol. Cell. Mech. 2008, 13, 1159–1170. [Google Scholar] [CrossRef] [PubMed]
  254. Yagishita, Y.; Fukutomi, T.; Sugawara, A.; Kawamura, H.; Takahashi, T.; Pi, J.; Uruno, A.; Yamamoto, M. Nrf2 protects pancreatic β-cells from oxidative and nitrosative stress in diabetic model mice. Diabetes 2014, 63, 605–618. [Google Scholar] [CrossRef] [PubMed]
  255. Liu, M.; Grigoryev, D.N.; Crow, M.T.; Haas, M.; Yamamoto, M.; Reddy, S.P.; Rabb, H. Transcription factor Nrf2 is protective during ischemic and nephrotoxic acute kidney injury in mice. Kidney Int. 2009, 76, 277–285. [Google Scholar] [CrossRef] [PubMed]
  256. Aleksunes, L.M.; Goedken, M.J.; Rockwell, C.E.; Thomale, J.; Manautou, J.E.; Klaassen, C.D. Transcriptional regulation of renal cytoprotective genes by Nrf2 and its potential use as a therapeutic target to mitigate cisplatin-induced nephrotoxicity. J. Pharmacol. Exp. Ther. 2010, 335, 2–12. [Google Scholar] [CrossRef] [PubMed]
  257. Yan, W.; Xu, Y.; Yuan, Y.; Tian, L.; Wang, Q.; Xie, Y.; Shao, X.; Zhang, M.; Ni, Z.; Mou, S. Renoprotective mechanisms of Astragaloside IV in cisplatin-induced acute kidney injury. Free Radic. Res. 2017, 51, 669–683. [Google Scholar] [CrossRef] [PubMed]
  258. Atilano-Roque, A.; Aleksunes, L.M.; Joy, M.S. Bardoxolone methyl modulates efflux transporter and detoxifying enzyme expression in cisplatin-induced kidney cell injury. Toxicol. Lett. 2016, 259, 52–59. [Google Scholar] [CrossRef] [PubMed]
  259. Wei, W.; Ma, N.; Fan, X.; Yu, Q.; Ci, X. The role of Nrf2 in acute kidney injury: Novel molecular mechanisms and therapeutic approaches. Free Radic. Biol. Med. 2020, 158, 1–12. [Google Scholar] [CrossRef]
  260. Qiu, Z.; He, J.; Shao, G.; Hu, J.; Li, X.; Zhou, H.; Li, M.; Yang, B. Obacunone Retards Renal Cyst Development in Autosomal Dominant Polycystic Kidney Disease by Activating NRF2. Antioxidants 2021, 11, 38. [Google Scholar] [CrossRef]
  261. Tang, P.M.; Nikolic-Paterson, D.J.; Lan, H.Y. Macrophages: Versatile players in renal inflammation and fibrosis. Nat. Rev. Nephrol. 2019, 15, 144–158. [Google Scholar] [CrossRef]
  262. Meng, X.M.; Nikolic-Paterson, D.J.; Lan, H.Y. Inflammatory processes in renal fibrosis. Nat. Rev. Nephrol. 2014, 10, 493–503. [Google Scholar] [CrossRef] [PubMed]
  263. Panizo, S.; Martínez-Arias, L.; Alonso-Montes, C.; Cannata, P.; Martín-Carro, B.; Fernández-Martín, J.L.; Naves-Díaz, M.; Carrillo-López, N.; Cannata-Andía, J.B. Fibrosis in Chronic Kidney Disease: Pathogenesis and Consequences. Int. J. Mol. Sci. 2021, 22, 408. [Google Scholar] [CrossRef] [PubMed]
  264. Sanz, A.B.; Sanchez-Niño, M.D.; Ramos, A.M.; Moreno, J.A.; Santamaria, B.; Ruiz-Ortega, M.; Egido, J.; Ortiz, A. NF-kappaB in renal inflammation. J. Am. Soc. Nephrol. JASN 2010, 21, 1254–1262. [Google Scholar] [CrossRef] [PubMed]
  265. Jin, Q.; Liu, T.; Qiao, Y.; Liu, D.; Yang, L.; Mao, H.; Ma, F.; Wang, Y.; Peng, L.; Zhan, Y. Oxidative stress and inflammation in diabetic nephropathy: Role of polyphenols. Front. Immunol. 2023, 14, 1185317. [Google Scholar] [CrossRef] [PubMed]
  266. Yuan, Q.; Tang, B.; Zhang, C. Signaling pathways of chronic kidney diseases, implications for therapeutics. Signal Transduct. Target. Ther. 2022, 7, 182. [Google Scholar] [CrossRef] [PubMed]
  267. Jin, H.; Wang, Y.; Wang, D.; Zhang, L. Effects of Qingshen Granules on the Oxidative Stress-NF/kB Signal Pathway in Unilateral Ureteral Obstruction Rats. Evid.-Based Complement. Altern. Med. Ecam 2018, 2018, 4761925. [Google Scholar] [CrossRef] [PubMed]
  268. Morgan, M.J.; Liu, Z.G. Crosstalk of reactive oxygen species and NF-κB signaling. Cell Res. 2011, 21, 103–115. [Google Scholar] [CrossRef]
  269. Kim, Y.; Park, C.W. Adenosine monophosphate-activated protein kinase in diabetic nephropathy. Kidney Res. Clin. Pract. 2016, 35, 69–77. [Google Scholar] [CrossRef]
  270. Yao, L.; Liang, X.; Qiao, Y.; Chen, B.; Wang, P.; Liu, Z. Mitochondrial dysfunction in diabetic tubulopathy. Metab. Clin. Exp. 2022, 131, 155195. [Google Scholar] [CrossRef]
  271. Yuan, T.; Yang, T.; Chen, H.; Fu, D.; Hu, Y.; Wang, J.; Yuan, Q.; Yu, H.; Xu, W.; Xie, X. New insights into oxidative stress and inflammation during diabetes mellitus-accelerated atherosclerosis. Redox Biol. 2019, 20, 247–260. [Google Scholar] [CrossRef] [PubMed]
  272. Aboolian, A.; Urner, S.; Roden, M.; Jha, J.C.; Jandeleit-Dahm, K. Diabetic Kidney Disease: From Pathogenesis to Novel Treatment Possibilities. Handb. Exp. Pharmacol. 2022, 274, 269–307. [Google Scholar] [CrossRef] [PubMed]
  273. Wang, Z.; Chen, Z.; Wang, X.; Hu, Y.; Kong, J.; Lai, J.; Li, T.; Hu, B.; Zhang, Y.; Zheng, X.; et al. Sappanone a prevents diabetic kidney disease by inhibiting kidney inflammation and fibrosis via the NF-κB signaling pathway. Front. Pharmacol. 2022, 13, 953004. [Google Scholar] [CrossRef] [PubMed]
  274. Kumar, D.; Singla, S.K.; Puri, V.; Puri, S. The restrained expression of NF-kB in renal tissue ameliorates folic acid induced acute kidney injury in mice. PLoS ONE 2015, 10, e115947. [Google Scholar] [CrossRef] [PubMed]
  275. Johnson, F.L.; Patel, N.S.A.; Purvis, G.S.D.; Chiazza, F.; Chen, J.; Sordi, R.; Hache, G.; Merezhko, V.V.; Collino, M.; Yaqoob, M.M.; et al. Inhibition of IκB Kinase at 24 Hours After Acute Kidney Injury Improves Recovery of Renal Function and Attenuates Fibrosis. J. Am. Heart Assoc. 2017, 6, e005092. [Google Scholar] [CrossRef] [PubMed]
  276. Shu, B.; Feng, Y.; Gui, Y.; Lu, Q.; Wei, W.; Xue, X.; Sun, X.; He, W.; Yang, J.; Dai, C. Blockade of CD38 diminishes lipopolysaccharide-induced macrophage classical activation and acute kidney injury involving NF-κB signaling suppression. Cell. Signal. 2018, 42, 249–258. [Google Scholar] [CrossRef]
  277. Lv, L.L.; Tang, P.M.; Li, C.J.; You, Y.K.; Li, J.; Huang, X.R.; Ni, J.; Feng, M.; Liu, B.C.; Lan, H.Y. The pattern recognition receptor, Mincle, is essential for maintaining the M1 macrophage phenotype in acute renal inflammation. Kidney Int. 2017, 91, 587–602. [Google Scholar] [CrossRef]
  278. Yang, L.; Brooks, C.R.; Xiao, S.; Sabbisetti, V.; Yeung, M.Y.; Hsiao, L.L.; Ichimura, T.; Kuchroo, V.; Bonventre, J.V. KIM-1-mediated phagocytosis reduces acute injury to the kidney. J. Clin. Investig. 2015, 125, 1620–1636. [Google Scholar] [CrossRef] [PubMed]
  279. Markó, L.; Vigolo, E.; Hinze, C.; Park, J.K.; Roël, G.; Balogh, A.; Choi, M.; Wübken, A.; Cording, J.; Blasig, I.E.; et al. Tubular Epithelial NF-κB Activity Regulates Ischemic AKI. J. Am. Soc. Nephrol. JASN 2016, 27, 2658–2669. [Google Scholar] [CrossRef]
  280. Guarente, L. Calorie restriction and sirtuins revisited. Genes Dev. 2013, 27, 2072–2085. [Google Scholar] [CrossRef]
  281. Cohen, H.Y.; Miller, C.; Bitterman, K.J.; Wall, N.R.; Hekking, B.; Kessler, B.; Howitz, K.T.; Gorospe, M.; de Cabo, R.; Sinclair, D.A. Calorie restriction promotes mammalian cell survival by inducing the SIRT1 deacetylase. Science 2004, 305, 390–392. [Google Scholar] [CrossRef]
  282. Haigis, M.C.; Sinclair, D.A. Mammalian sirtuins: Biological insights and disease relevance. Annu. Rev. Pathol. 2010, 5, 253–295. [Google Scholar] [CrossRef] [PubMed]
  283. Kitada, M.; Ogura, Y.; Monno, I.; Koya, D. Sirtuins and Type 2 Diabetes: Role in Inflammation, Oxidative Stress, and Mitochondrial Function. Front. Endocrinol. 2019, 10, 187. [Google Scholar] [CrossRef] [PubMed]
  284. Kitada, M.; Kume, S.; Kanasaki, K.; Takeda-Watanabe, A.; Koya, D. Sirtuins as possible drug targets in type 2 diabetes. Curr. Drug Targets 2013, 14, 622–636. [Google Scholar] [CrossRef]
  285. Zhou, L.; Xu, D.Y.; Sha, W.G.; Shen, L.; Lu, G.Y.; Yin, X.; Wang, M.J. High glucose induces renal tubular epithelial injury via Sirt1/NF-kappaB/microR-29/Keap1 signal pathway. J. Transl. Med. 2015, 13, 352. [Google Scholar] [CrossRef]
  286. Lempiäinen, J.; Finckenberg, P.; Mervaala, E.E.; Sankari, S.; Levijoki, J.; Mervaala, E.M. Caloric restriction ameliorates kidney ischaemia/reperfusion injury through PGC-1α-eNOS pathway and enhanced autophagy. Acta Physiol. 2013, 208, 410–421. [Google Scholar] [CrossRef]
  287. Klotz, L.O.; Sánchez-Ramos, C.; Prieto-Arroyo, I.; Urbánek, P.; Steinbrenner, H.; Monsalve, M. Redox regulation of FoxO transcription factors. Redox Biol. 2015, 6, 51–72. [Google Scholar] [CrossRef]
  288. Kao, C.L.; Chen, L.K.; Chang, Y.L.; Yung, M.C.; Hsu, C.C.; Chen, Y.C.; Lo, W.L.; Chen, S.J.; Ku, H.H.; Hwang, S.J. Resveratrol protects human endothelium from H2O2-induced oxidative stress and senescence via SirT1 activation. J. Atheroscler. Thromb. 2010, 17, 970–979. [Google Scholar] [CrossRef] [PubMed]
  289. Nemoto, S.; Finkel, T. Redox regulation of forkhead proteins through a p66shc-dependent signaling pathway. Science 2002, 295, 2450–2452. [Google Scholar] [CrossRef] [PubMed]
  290. He, W.; Wang, Y.; Zhang, M.Z.; You, L.; Davis, L.S.; Fan, H.; Yang, H.C.; Fogo, A.B.; Zent, R.; Harris, R.C.; et al. Sirt1 activation protects the mouse renal medulla from oxidative injury. J. Clin. Investig. 2010, 120, 1056–1068. [Google Scholar] [CrossRef]
  291. Li, J.; Qu, X.; Ricardo, S.D.; Bertram, J.F.; Nikolic-Paterson, D.J. Resveratrol inhibits renal fibrosis in the obstructed kidney: Potential role in deacetylation of Smad3. Am. J. Pathol. 2010, 177, 1065–1071. [Google Scholar] [CrossRef] [PubMed]
  292. Zhang, Y.; Connelly, K.A.; Thai, K.; Wu, X.; Kapus, A.; Kepecs, D.; Gilbert, R.E. Sirtuin 1 Activation Reduces Transforming Growth Factor-β1-Induced Fibrogenesis and Affords Organ Protection in a Model of Progressive, Experimental Kidney and Associated Cardiac Disease. Am. J. Pathol. 2017, 187, 80–90. [Google Scholar] [CrossRef] [PubMed]
  293. Chuang, P.Y.; Dai, Y.; Liu, R.; He, H.; Kretzler, M.; Jim, B.; Cohen, C.D.; He, J.C. Alteration of forkhead box O (foxo4) acetylation mediates apoptosis of podocytes in diabetes mellitus. PLoS ONE 2011, 6, e23566. [Google Scholar] [CrossRef] [PubMed]
  294. Liu, R.; Zhong, Y.; Li, X.; Chen, H.; Jim, B.; Zhou, M.M.; Chuang, P.Y.; He, J.C. Role of transcription factor acetylation in diabetic kidney disease. Diabetes 2014, 63, 2440–2453. [Google Scholar] [CrossRef] [PubMed]
  295. Chuang, P.Y.; Cai, W.; Li, X.; Fang, L.; Xu, J.; Yacoub, R.; He, J.C.; Lee, K. Reduction in podocyte SIRT1 accelerates kidney injury in aging mice. Am. J. Physiol. Ren. Physiol. 2017, 313, F621–F628. [Google Scholar] [CrossRef] [PubMed]
  296. Hasegawa, K.; Wakino, S.; Simic, P.; Sakamaki, Y.; Minakuchi, H.; Fujimura, K.; Hosoya, K.; Komatsu, M.; Kaneko, Y.; Kanda, T.; et al. Renal tubular Sirt1 attenuates diabetic albuminuria by epigenetically suppressing Claudin-1 overexpression in podocytes. Nat. Med. 2013, 19, 1496–1504. [Google Scholar] [CrossRef] [PubMed]
  297. Gong, L.; He, J.; Sun, X.; Li, L.; Zhang, X.; Gan, H. Activation of sirtuin1 protects against ischemia/reperfusion-induced acute kidney injury. Biomed. Pharmacother. 2020, 125, 110021. [Google Scholar] [CrossRef] [PubMed]
  298. Gardner, P.R.; Raineri, I.; Epstein, L.B.; White, C.W. Superoxide radical and iron modulate aconitase activity in mammalian cells. J. Biol. Chem. 1995, 270, 13399–13405. [Google Scholar] [CrossRef] [PubMed]
  299. Rouault, T.A.; Klausner, R.D. Iron-sulfur clusters as biosensors of oxidants and iron. Trends Biochem. Sci. 1996, 21, 174–177. [Google Scholar] [CrossRef]
  300. Cairo, G.; Recalcati, S.; Pietrangelo, A.; Minotti, G. The iron regulatory proteins: Targets and modulators of free radical reactions and oxidative damage. Free Radic. Biol. Med. 2002, 32, 1237–1243. [Google Scholar] [CrossRef]
  301. Cairo, G.; Castrusini, E.; Minotti, G.; Bernelli-Zazzera, A. Superoxide and hydrogen peroxide-dependent inhibition of iron regulatory protein activity: A protective stratagem against oxidative injury. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 1996, 10, 1326–1335. [Google Scholar] [CrossRef]
  302. Pantopoulos, K.; Mueller, S.; Atzberger, A.; Ansorge, W.; Stremmel, W.; Hentze, M.W. Differences in the regulation of iron regulatory protein-1 (IRP-1) by extra- and intracellular oxidative stress. J. Biol. Chem. 1997, 272, 9802–9808. [Google Scholar] [CrossRef] [PubMed]
  303. Andriopoulos, B.; Hegedüsch, S.; Mangin, J.; Riedel, H.D.; Hebling, U.; Wang, J.; Pantopoulos, K.; Mueller, S. Sustained hydrogen peroxide induces iron uptake by transferrin receptor-1 independent of the iron regulatory protein/iron-responsive element network. J. Biol. Chem. 2007, 282, 20301–20308. [Google Scholar] [CrossRef] [PubMed]
  304. Mehlhase, J.; Sandig, G.; Pantopoulos, K.; Grune, T. Oxidation-induced ferritin turnover in microglial cells: Role of proteasome. Free Radic. Biol. Med. 2005, 38, 276–285. [Google Scholar] [CrossRef] [PubMed]
  305. Niecknig, H.; Tug, S.; Reyes, B.D.; Kirsch, M.; Fandrey, J.; Berchner-Pfannschmidt, U. Role of reactive oxygen species in the regulation of HIF-1 by prolyl hydroxylase 2 under mild hypoxia. Free Radic. Res. 2012, 46, 705–717. [Google Scholar] [CrossRef]
  306. Kuragano, T.; Shimonaka, Y.; Kida, A.; Furuta, M.; Nanami, M.; Otaki, Y.; Hasuike, Y.; Nonoguchi, H.; Nakanishi, T. Determinants of hepcidin in patients on maintenance hemodialysis: Role of inflammation. Am. J. Nephrol. 2010, 31, 534–540. [Google Scholar] [CrossRef]
  307. Ashby, D.R.; Gale, D.P.; Busbridge, M.; Murphy, K.G.; Duncan, N.D.; Cairns, T.D.; Taube, D.H.; Bloom, S.R.; Tam, F.W.; Chapman, R.S.; et al. Plasma hepcidin levels are elevated but responsive to erythropoietin therapy in renal disease. Kidney Int. 2009, 75, 976–981. [Google Scholar] [CrossRef]
  308. Hirschhorn, T.; Stockwell, B.R. The development of the concept of ferroptosis. Free Radic. Biol. Med. 2019, 133, 130–143. [Google Scholar] [CrossRef] [PubMed]
  309. Jiang, X.; Stockwell, B.R.; Conrad, M. Ferroptosis: Mechanisms, biology and role in disease. Nat. Rev. Mol. Cell Biol. 2021, 22, 266–282. [Google Scholar] [CrossRef]
  310. Liu, Y.; Wang, J. Ferroptosis, a Rising Force against Renal Fibrosis. Oxid. Med. Cell. Longev. 2022, 2022, 7686956. [Google Scholar] [CrossRef]
  311. Gong, S.; Zhang, A.; Yao, M.; Xin, W.; Guan, X.; Qin, S.; Liu, Y.; Xiong, J.; Yang, K.; Xiong, L.; et al. REST contributes to AKI-to-CKD transition through inducing ferroptosis in renal tubular epithelial cells. JCI Insight 2023, 8, e166001. [Google Scholar] [CrossRef] [PubMed]
  312. Song, J.; Wang, H.; Sheng, J.; Zhang, W.; Lei, J.; Gan, W.; Cai, F.; Yang, Y. Vitexin attenuates chronic kidney disease by inhibiting renal tubular epithelial cell ferroptosis via NRF2 activation. Mol. Med. 2023, 29, 147. [Google Scholar] [CrossRef] [PubMed]
  313. Li, J.; Yang, J.; Zhu, B.; Fan, J.; Hu, Q.; Wang, L. Tectorigenin protects against unilateral ureteral obstruction by inhibiting Smad3-mediated ferroptosis and fibrosis. Phytother. Res. PTR 2022, 36, 475–487. [Google Scholar] [CrossRef] [PubMed]
  314. Zhu, B.; Ni, Y.; Gong, Y.; Kang, X.; Guo, H.; Liu, X.; Li, J.; Wang, L. Formononetin ameliorates ferroptosis-associated fibrosis in renal tubular epithelial cells and in mice with chronic kidney disease by suppressing the Smad3/ATF3/SLC7A11 signaling. Life Sci. 2023, 315, 121331. [Google Scholar] [CrossRef] [PubMed]
  315. Kim, S.; Kang, S.W.; Joo, J.; Han, S.H.; Shin, H.; Nam, B.Y.; Park, J.; Yoo, T.H.; Kim, G.; Lee, P.; et al. Characterization of ferroptosis in kidney tubular cell death under diabetic conditions. Cell Death Dis. 2021, 12, 160. [Google Scholar] [CrossRef] [PubMed]
  316. Feng, X.; Wang, S.; Sun, Z.; Dong, H.; Yu, H.; Huang, M.; Gao, X. Ferroptosis Enhanced Diabetic Renal Tubular Injury via HIF-1α/HO-1 Pathway in db/db Mice. Front. Endocrinol. 2021, 12, 626390. [Google Scholar] [CrossRef] [PubMed]
  317. Wang, Y.; Bi, R.; Quan, F.; Cao, Q.; Lin, Y.; Yue, C.; Cui, X.; Yang, H.; Gao, X.; Zhang, D. Ferroptosis involves in renal tubular cell death in diabetic nephropathy. Eur. J. Pharmacol. 2020, 888, 173574. [Google Scholar] [CrossRef] [PubMed]
  318. Wu, K.; Fei, L.; Wang, X.; Lei, Y.; Yu, L.; Xu, W.; Chen, J.; Zhu, E.; Zhong, M.; Huang, M.; et al. ZIP14 is involved in iron deposition and triggers ferroptosis in diabetic nephropathy. Met. Integr. Biometal Sci. 2022, 14, mfac034. [Google Scholar] [CrossRef] [PubMed]
  319. Li, S.; Zheng, L.; Zhang, J.; Liu, X.; Wu, Z. Inhibition of ferroptosis by up-regulating Nrf2 delayed the progression of diabetic nephropathy. Free Radic. Biol. Med. 2021, 162, 435–449. [Google Scholar] [CrossRef]
  320. Li, Q.; Liao, J.; Chen, W.; Zhang, K.; Li, H.; Ma, F.; Zhang, H.; Han, Q.; Guo, J.; Li, Y.; et al. NAC alleviative ferroptosis in diabetic nephropathy via maintaining mitochondrial redox homeostasis through activating SIRT3-SOD2/Gpx4 pathway. Free Radic. Biol. Med. 2022, 187, 158–170. [Google Scholar] [CrossRef]
  321. Agborbesong, E.; Li, L.X.; Li, L.; Li, X. Molecular Mechanisms of Epigenetic Regulation, Inflammation, and Cell Death in ADPKD. Front. Mol. Biosci. 2022, 9, 922428. [Google Scholar] [CrossRef] [PubMed]
  322. Schreiber, R.; Buchholz, B.; Kraus, A.; Schley, G.; Scholz, J.; Ousingsawat, J.; Kunzelmann, K. Lipid Peroxidation Drives Renal Cyst Growth In Vitro through Activation of TMEM16A. J. Am. Soc. Nephrol. JASN 2019, 30, 228–242. [Google Scholar] [CrossRef] [PubMed]
  323. Zhang, X.; Li, L.X.; Ding, H.; Torres, V.E.; Yu, C.; Li, X. Ferroptosis Promotes Cyst Growth in Autosomal Dominant Polycystic Kidney Disease Mouse Models. J. Am. Soc. Nephrol. JASN 2021, 32, 2759–2776. [Google Scholar] [CrossRef] [PubMed]
  324. Taguchi, S.; Hidaka, S.; Yanai, M.; Ishioka, K.; Matsui, K.; Mochida, Y.; Moriya, H.; Ohtake, T.; Kobayashi, S. Renal hemosiderosis presenting with acute kidney Injury and macroscopic hematuria in Immunoglobulin A nephropathy: A case report. BMC Nephrol. 2021, 22, 132. [Google Scholar] [CrossRef] [PubMed]
  325. 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]
  326. Wu, J.; Shao, X.; Shen, J.; Lin, Q.; Zhu, X.; Li, S.; Li, J.; Zhou, W.; Qi, C.; Ni, Z. Downregulation of PPARα mediates FABP1 expression, contributing to IgA nephropathy by stimulating ferroptosis in human mesangial cells. Int. J. Biol. Sci. 2022, 18, 5438–5458. [Google Scholar] [CrossRef] [PubMed]
  327. Zhao, Z.; Wu, J.; Xu, H.; Zhou, C.; Han, B.; Zhu, H.; Hu, Z.; Ma, Z.; Ming, Z.; Yao, Y.; et al. XJB-5-131 inhibited ferroptosis in tubular epithelial cells after ischemia-reperfusion injury. Cell Death Dis. 2020, 11, 629. [Google Scholar] [CrossRef] [PubMed]
  328. Tonnus, W.; Meyer, C.; Steinebach, C.; Belavgeni, A.; von Mässenhausen, A.; Gonzalez, N.Z.; Maremonti, F.; Gembardt, F.; Himmerkus, N.; Latk, M.; et al. Dysfunction of the key ferroptosis-surveilling systems hypersensitizes mice to tubular necrosis during acute kidney injury. Nat. Commun. 2021, 12, 4402. [Google Scholar] [CrossRef] [PubMed]
  329. Wang, Y.; Zhang, M.; Bi, R.; Su, Y.; Quan, F.; Lin, Y.; Yue, C.; Cui, X.; Zhao, Q.; Liu, S.; et al. ACSL4 deficiency confers protection against ferroptosis-mediated acute kidney injury. Redox Biol. 2022, 51, 102262. [Google Scholar] [CrossRef]
  330. Qi, Y.; Hu, M.; Qiu, Y.; Zhang, L.; Yan, Y.; Feng, Y.; Feng, C.; Hou, X.; Wang, Z.; Zhang, D.; et al. Mitoglitazone ameliorates renal ischemia/reperfusion injury by inhibiting ferroptosis via targeting mitoNEET. Toxicol. Appl. Pharmacol. 2023, 465, 116440. [Google Scholar] [CrossRef]
  331. Zhou, Z.; Zhang, H. CHAC1 exacerbates LPS-induced ferroptosis and apoptosis in HK-2 cells by promoting oxidative stress. Allergol. Immunopathol. 2023, 51, 99–110. [Google Scholar] [CrossRef] [PubMed]
  332. Qiu, W.; An, S.; Wang, T.; Li, J.; Yu, B.; Zeng, Z.; Chen, Z.; Lin, B.; Lin, X.; Gao, Y. Melatonin suppresses ferroptosis via activation of the Nrf2/HO-1 signaling pathway in the mouse model of sepsis-induced acute kidney injury. Int. Immunopharmacol. 2022, 112, 109162. [Google Scholar] [CrossRef] [PubMed]
  333. Feng, Q.; Yu, X.; Qiao, Y.; Pan, S.; Wang, R.; Zheng, B.; Wang, H.; Ren, K.D.; Liu, H.; Yang, Y. Ferroptosis and Acute Kidney Injury (AKI): Molecular Mechanisms and Therapeutic Potentials. Front. Pharmacol. 2022, 13, 858676. [Google Scholar] [CrossRef] [PubMed]
  334. Zhou, L.; Yu, P.; Wang, T.T.; Du, Y.W.; Chen, Y.; Li, Z.; He, M.L.; Feng, L.; Li, H.R.; Han, X.; et al. Polydatin Attenuates Cisplatin-Induced Acute Kidney Injury by Inhibiting Ferroptosis. Oxid. Med. Cell. Longev. 2022, 2022, 9947191. [Google Scholar] [CrossRef]
Figure 1. The regulation and the association between systemic iron circulation and cellular iron metabolism. Systemic and cellular iron are tightly regulated. Dietary iron is the only source of exogenous iron used to replace iron lost through iron excretion or bleeding. Dietary iron is absorbed into the bloodstream by enterocytes and exists in the circulation mainly as TFBI, which is recognized and taken up for use by cells of various tissues via TFR1. The liver is primarily responsible for systemic iron storage and regulates systemic iron homeostasis by synthesizing hepcidin. In the plasma, macrophages phagocytose senescent erythrocytes, releasing iron from hemoglobin and re-entering the circulation. Intracellularly, iron in the LIP is available for the synthesis of substances, such as the synthesis of heme by red lineage cells and the synthesis of Fe-S clusters by cells of various tissues, while excess iron is stored in ferritin. Ferritin can release the stored iron into the LIP via ferritinophagy, and intracellular iron can also be excreted directly from the cell via the FPN or by secretion of ferritin. When the intracellular iron regulation mechanism fails or the cellular iron is overloaded, the iron in LIP can generate toxic ROS through the Fenton reaction, leading to oxidative stress and cellular damage. Abbreviation: TFBI: transferrin bound iron, FPN: ferroportin, LIP: labile iron pool, HIF: hypoxia-inducible factor, IRP-IRE: iron-regulatory protein and iron-responsive element.
Figure 1. The regulation and the association between systemic iron circulation and cellular iron metabolism. Systemic and cellular iron are tightly regulated. Dietary iron is the only source of exogenous iron used to replace iron lost through iron excretion or bleeding. Dietary iron is absorbed into the bloodstream by enterocytes and exists in the circulation mainly as TFBI, which is recognized and taken up for use by cells of various tissues via TFR1. The liver is primarily responsible for systemic iron storage and regulates systemic iron homeostasis by synthesizing hepcidin. In the plasma, macrophages phagocytose senescent erythrocytes, releasing iron from hemoglobin and re-entering the circulation. Intracellularly, iron in the LIP is available for the synthesis of substances, such as the synthesis of heme by red lineage cells and the synthesis of Fe-S clusters by cells of various tissues, while excess iron is stored in ferritin. Ferritin can release the stored iron into the LIP via ferritinophagy, and intracellular iron can also be excreted directly from the cell via the FPN or by secretion of ferritin. When the intracellular iron regulation mechanism fails or the cellular iron is overloaded, the iron in LIP can generate toxic ROS through the Fenton reaction, leading to oxidative stress and cellular damage. Abbreviation: TFBI: transferrin bound iron, FPN: ferroportin, LIP: labile iron pool, HIF: hypoxia-inducible factor, IRP-IRE: iron-regulatory protein and iron-responsive element.
Antioxidants 13 00659 g001
Figure 2. The main sources of ROS and ROS mediated the activation of Nrf2, NF-κB and SIRT1 in kidneys. Intracellular ROS in renal tissues is mainly derived from the escape of O2 and H2O2 during mitochondrial aerobic respiratory electron transfer and NOX4 transfers electrons directly to oxygen molecules to produce H2O2. When ROS is elevated, the structure of Keap1-Nrf2 is disrupted, resulting in the release and entry of Nrf2 into the nucleus, where it binds to the ARE in DNA and increases the expression of downstream antioxidant genes. In addition, NF-κB is bound under normal conditions by IκB, which exists as an inactive complex in the cytoplasm. When intracellular ROS is elevated, IκB would be dissociated from NF-κB, leading to the activation of NF-κB activation and its translocation into the nucleus, where it binds to IRE in DNA and initiates the expression of downstream pro-inflammatory genes. Furthermore, elevated ROS can also lead to an increase of NAD+ and the activation of SIRT1, a NAD+-dependent deacetylase capable of inhibiting NF-κB activity by deacetylating its p65 subunit, thereby inhibiting the inflammatory response. In addition, SIRT1 promotes FOXO1 activation and nuclear translocation by deacetylating FOXO1, leading to an increase of the expression of various antioxidant genes, such as SOD2 and CAT. Abbreviation: NOX4: NADPH oxidases 4, ETC: electron transfer chain, Keap1: kelch-like ECH-associated protein 1, Nrf2: nuclear factor erythroid 2-related factor 2, ARE: antioxidant response element, IκB: inhibitory κB, NF-κB: nuclear factor kappa B, IRE: iron-responsive element, SIRT1: sirtuin 1, FOXO1: forkhead box O1, HO-1: heme oxygenase 1, CAT: catalase.
Figure 2. The main sources of ROS and ROS mediated the activation of Nrf2, NF-κB and SIRT1 in kidneys. Intracellular ROS in renal tissues is mainly derived from the escape of O2 and H2O2 during mitochondrial aerobic respiratory electron transfer and NOX4 transfers electrons directly to oxygen molecules to produce H2O2. When ROS is elevated, the structure of Keap1-Nrf2 is disrupted, resulting in the release and entry of Nrf2 into the nucleus, where it binds to the ARE in DNA and increases the expression of downstream antioxidant genes. In addition, NF-κB is bound under normal conditions by IκB, which exists as an inactive complex in the cytoplasm. When intracellular ROS is elevated, IκB would be dissociated from NF-κB, leading to the activation of NF-κB activation and its translocation into the nucleus, where it binds to IRE in DNA and initiates the expression of downstream pro-inflammatory genes. Furthermore, elevated ROS can also lead to an increase of NAD+ and the activation of SIRT1, a NAD+-dependent deacetylase capable of inhibiting NF-κB activity by deacetylating its p65 subunit, thereby inhibiting the inflammatory response. In addition, SIRT1 promotes FOXO1 activation and nuclear translocation by deacetylating FOXO1, leading to an increase of the expression of various antioxidant genes, such as SOD2 and CAT. Abbreviation: NOX4: NADPH oxidases 4, ETC: electron transfer chain, Keap1: kelch-like ECH-associated protein 1, Nrf2: nuclear factor erythroid 2-related factor 2, ARE: antioxidant response element, IκB: inhibitory κB, NF-κB: nuclear factor kappa B, IRE: iron-responsive element, SIRT1: sirtuin 1, FOXO1: forkhead box O1, HO-1: heme oxygenase 1, CAT: catalase.
Antioxidants 13 00659 g002
Figure 3. Oxidative stress as a mediating factor connecting abnormal iron metabolism and aggravation of kidney diseases. Abnormal iron metabolism leads to oxidative stress by generating excess ROS through the Fenton reaction. Increased ROS promotes DNA damage, mitochondrial dysfunction, inflammatory response, ER stress and lipid peroxidation (ferroptosis), resulting in an increase of cellular senescence, cell death, ETC dysfunction and ferroptosis. Subsequently, this causes irreversible renal damage because of loss of irreducible cells, such as podocytes and endothelial cells, as well as abnormal tissue repair (nonfunctional scar repair), leading to the increase of the progression of renal disease to further deteriorate renal function. Abbreviation: SASP: senescence-associated secretory phenotype, ER: endoplasmic reticulum, GPX4: glutathione peroxidase 4, AKI: acute kidney disease.
Figure 3. Oxidative stress as a mediating factor connecting abnormal iron metabolism and aggravation of kidney diseases. Abnormal iron metabolism leads to oxidative stress by generating excess ROS through the Fenton reaction. Increased ROS promotes DNA damage, mitochondrial dysfunction, inflammatory response, ER stress and lipid peroxidation (ferroptosis), resulting in an increase of cellular senescence, cell death, ETC dysfunction and ferroptosis. Subsequently, this causes irreversible renal damage because of loss of irreducible cells, such as podocytes and endothelial cells, as well as abnormal tissue repair (nonfunctional scar repair), leading to the increase of the progression of renal disease to further deteriorate renal function. Abbreviation: SASP: senescence-associated secretory phenotype, ER: endoplasmic reticulum, GPX4: glutathione peroxidase 4, AKI: acute kidney disease.
Antioxidants 13 00659 g003
Table 1. The classification of ROS.
Table 1. The classification of ROS.
Reactive Oxygen SpeciesProperties
Non-
radical
Hydrogen peroxide (H2O2)Low oxidative activity, participates in many physiological processes as a signal molecule
Organic hydroperoxides (ROOH)Lipid peroxides derived from polyunsaturated acids (PUFAs) in ferroptosis
Singlet oxygenHigh oxidative activity involved in many biological processes
Electronically excited carbonyl (R–C=O)High oxidative activity
Peroxynitrite (ONOO)Formed by the reaction of superoxide with nitric oxide
Ozone (O3)In atmosphere, but toxic to humans
Free radicalSuperoxide (O2)Relatively low oxidative activity, participates in the synthesis of H2O2
Hydroxyl radicals (HO)High oxidative activity and unstable, react with various cellular proteins, DNA, lipids
Peroxyl radical (ROO)High oxidative activity, involved in the spread of lipid peroxidation
Nitric oxide (NO)Relatively low oxidative activity, participates in the synthesis of H2O2
Table 2. The enzymatic and non-enzymatic renal antioxidant systems.
Table 2. The enzymatic and non-enzymatic renal antioxidant systems.
Enzymatic SystemNon-Enzymatic System
Superoxide dismutase (SOD)Glutathione (GSH)
Catalase (CAT)Antioxidant vitamins: vitamin A/C/E
Glutathione peroxidase (GPX)Antioxidant minerals: copper, zinc, manganese
Thioredoxin (Trx)Hormones: melatonin, flavenoids,
coenzyme Q
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

Xie, T.; Yao, L.; Li, X. Advance in Iron Metabolism, Oxidative Stress and Cellular Dysfunction in Experimental and Human Kidney Diseases. Antioxidants 2024, 13, 659. https://doi.org/10.3390/antiox13060659

AMA Style

Xie T, Yao L, Li X. Advance in Iron Metabolism, Oxidative Stress and Cellular Dysfunction in Experimental and Human Kidney Diseases. Antioxidants. 2024; 13(6):659. https://doi.org/10.3390/antiox13060659

Chicago/Turabian Style

Xie, Tiancheng, Li Yao, and Xiaogang Li. 2024. "Advance in Iron Metabolism, Oxidative Stress and Cellular Dysfunction in Experimental and Human Kidney Diseases" Antioxidants 13, no. 6: 659. https://doi.org/10.3390/antiox13060659

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