Next Article in Journal
A Multisensory Network Drives Nuclear Mechanoadaptation
Next Article in Special Issue
Redox Imbalance and Mitochondrial Abnormalities in Kidney Disease
Previous Article in Journal
Fine Mapping and Characterization of a Major Gene Responsible for Chlorophyll Biosynthesis in Brassica napus L.
Previous Article in Special Issue
Glucose- and Non-Glucose-Induced Mitochondrial Dysfunction in Diabetic Kidney Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mitochondrial Oxidative Stress and Cell Death in Podocytopathies

1
Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
2
5th Medical Department, Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany
*
Author to whom correspondence should be addressed.
Biomolecules 2022, 12(3), 403; https://doi.org/10.3390/biom12030403
Submission received: 14 December 2021 / Revised: 26 February 2022 / Accepted: 1 March 2022 / Published: 4 March 2022
(This article belongs to the Special Issue Redox Imbalance and Mitochondrial Abnormalities in Kidney Disease)

Abstract

:
Podocytopathies are kidney diseases that are driven by podocyte injury with proteinuria and proteinuria-related symptoms as the main clinical presentations. Albeit podocytopathies are the major contributors to end-stage kidney disease, the underlying molecular mechanisms of podocyte injury remain to be elucidated. Mitochondrial oxidative stress is associated with kidney diseases, and increasing evidence suggests that oxidative stress plays a vital role in the pathogenesis of podocytopathies. Accumulating evidence has placed mitochondrial oxidative stress in the focus of cell death research. Excessive generated reactive oxygen species over antioxidant defense under pathological conditions lead to oxidative damage to cellular components and regulate cell death in the podocyte. Conversely, exogenous antioxidants can protect podocyte from cell death. This review provides an overview of the role of mitochondrial oxidative stress in podocytopathies and discusses its role in the cell death of the podocyte, aiming to identify the novel targets to improve the treatment of patients with podocytopathies.

1. Introduction

Podocytopathies are defined as kidney diseases that are driven by podocyte injury with proteinuria and proteinuria-related symptoms as the main clinical features [1]. The incidence of podocytopathies seems to be gradually rising and they are the leading cause of end-stage kidney disease around the world [1,2]. However, efficient therapies for podocytopathies are lacking and current treatment can only retard the progression of diseases. Podocytes are highly specialized epithelial cells that are located in the glomerulus and constitute the filtration barrier with a glomerular basement membrane (GBM) and endothelial cells [2,3]. The interdigitated foot processes and slit diaphragm of podocytes are elementary structures for the selective filtration function of the glomerulus [1]. Excessive stress and harmful stimuli are likely to cause podocyte injury, possibly even death, which is clinically characterized by proteinuria and pathologically characterized by podocyte foot process effacement (FPE), detachment, and loss [1,2]. Considering the poor proliferation capacity of the podocyte, excessive podocyte loss progressively aggravates podocyte damage and eventually leads to global glomerulosclerosis [2]. Understanding how such detrimental stress and stimuli cause podocyte injury can help us to advance our acknowledgement of the mechanisms underlying the occurrence and progression of podocytopathies.
Mitochondrial oxidative stress refers to disrupted redox homeostasis by the elevated generation of reactive oxygen species (ROS) and (or) declined antioxidant defense capacity [4,5]. Increasing evidence suggests that oxidative stress plays a vital role in the pathogenesis of podocytopathies [6,7,8,9]. The excessive accumulation of ROS causes damage to intracellular components and impairs the normal structure and function of podocytes [7,8,10,11,12]. Accumulating evidence has placed mitochondrial oxidative stress in the focus of cell death research [4,13]. Classic cell death includes apoptosis, necrosis, necroptosis, pyroptosis, and ferroptosis [14,15]. Under pathological conditions, when redundant ROS-induced damage is beyond the compensatory capacity of podocytes, cell death occurs [8,16,17,18]. Conversely, the application of exogenous antioxidants can protect podocytes from cell death and improve kidney function [8,19]. Hence, this review provides an overview of the role of mitochondrial oxidative stress in podocytopathies and discusses its role in the cell death of the podocyte, aiming to identify novel targets to improve the treatment of patients with podocytopathies.

2. Spectrum of Podocytopathies

Genetic factors and non-genetic factors, such as immune, infectious, metabolic, and hemodynamic factors, can cause damage to the podocyte [2]. Therefore, podocytopathies can be further divided into genetic and non-genetic podocytopathies on the basis of the causes.
Numerous genetic researches identified many susceptibility genes relevant to podocytopathies, which can be divided into podocyte genes and syndromal non-specific genes in terms of the types of cells that experience genetic variation [1]. For example, genetic variants in PLCE1 and WT1, two podocyte-expressed genes, result in the arrested development of glomeruli and the onset of diffuse mesangial sclerosis [20,21]. APOL1 podocytopathy is the best studied podocytopathy that is associated with genetic variants of susceptibility genes [1]. In Africans carrying a high frequency of APOL1 alleles, the prevalence of chronic kidney disease (CKD) is up to 16% [22].
Non-genetic podocytopathies consist of kidney diseases of many distinct causes. Immune injury to the podocyte can induce the development of podocytopathies, such as IgA nephropathy (IgAN), lupus nephritis (LN), and membranous nephropathy (MN) [2]. Metabolic and hemodynamic abnormalities damage the podocyte as well. Long-term poor glucose control and the hemodynamic changes in diabetics contribute to diabetic nephropathy (DN) [1]. Elevated blood pressure and the accompanying hyperfiltration can also induce podocyte injury, which plays an important role in the pathogenesis of hypertensive nephropathy [23]. Podocytopathies caused by infections and nephrotoxic substances are not negligible, for instance, HIV-associated nephropathy and collapsing glomerulopathy induced by pamidronate [24,25].

3. Oxidative Stress in Podocytopathies

Under physiological conditions, a homeostasis between the production of ROS and the antioxidant defense system exists in the podocyte. ROS are a collection of chemical substances originated from incomplete reduced oxygen, which mainly consist of superoxide anion, hydrogen peroxide (H2O2), singlet oxygen, and hydroxyl radical [13,26]. In the podocyte, ROS mainly come from the mitochondrial respiration chain and NADPH oxidase (NOX) [27,28,29]. The mitochondrial respiration chain is mainly composed of NADH dehydrogenase (complex I), succinate dehydrogenase (complex II), ubiquinol-cytochrome c reductase (complex III), cytochrome c oxidase (complex IV), cytochrome c (Cyt C), and quinone [30,31]. Mitochondrial respiration chain dysfunction contributes to excessive ROS generation. Antioxidant defense systems are developed in the organism to eliminate ROS generated from various sources. Enzymatic defense systems include superoxide dismutase (SOD), glutathione peroxidase (GPX), catalase (CAT), and thioredoxin reductase (TrxR) [4,32]. Additionally, antioxidants are comprised of ascorbic acid (Vitamin C), α-tocopherol (Vitamin E), glutathione (GSH), thioredoxin, peroxiredoxin (Prdx), and carotenoids [4,32]. When the redox homeostasis is disrupted due to external stimuli under pathological conditions or inherent defects of podocyte, oxidative stress occurs and leads to podocyte injury and renal damage (Figure 1). Stimuli that are harmful to the podocyte, such as puromycin aminonucleoside (PA), high glucose (HG), and angiotensin II (Ang II), can all contribute to the intracellular accumulation of ROS [28,33,34].
In this section, we discuss and summarize the mechanisms by which ROS production is elevated in podocytopathies.

3.1. Elevated ROS Production

In focal segmental glomerulosclerosis (FSGS), the level of NOX4, a subunit of NOX complex, is higher in patients with steroid-resistant nephrotic syndrome (SRNS) than those with steroid-sensitive nephrotic syndrome, and the level of ROS is also higher in the glomeruli isolated from patients with SRNS [35]. The induction of NOX4 is observed in the PA-treated podocyte as well [27]. In the lmai rats, a spontaneous FSGS model, the activation of the angiotensin II type 1 receptor (AT1R) by Ang II leads to oxidative stress via upregulating the expression of NOX and downregulating the expression of Nrf2 [36]. In the FSGS phase of puromycin aminonucleoside nephrosis, cytochrome c oxidase subunit 1 (COX1) in the glomerulus is decreased [6]. Mice that lack the cytochrome c oxidase assembly cofactor heme A, farnesyltransferase (COX10), develop severe FSGS at an early age [37]. In the PA-treated podocyte, a decrease in COX1, 2, and 4, and a reduction in complexes I and IV are observed [27]. A mutation in ADCK4, which participates in the biosynthesis of coenzyme Q10 (CoQ10), causes the development of FSGS [38]. Whole-exome sequencing and Sanger sequencing suggests that a coenzyme Q10 mono-oxygenase 6 (COQ6) mutation might increase the production of ROS and associate with the occurrence of FSGS [39]. In PA-induced minimal change disease (MCD), CYP2B1, a cytochrome P450 isozyme, is a source of ROS and catalytic iron formation in the podocyte, with CYP2B1 inhibitors cimetidine and piperine alleviating PA-induced proteinuria and attenuating the increase in H2O2 and catalytic iron [33,40]. Adriamycin can induce the downregulation of complex I subunits both in vitro and in vivo [41]. In patients with a congenital nephrotic syndrome of the Finnish type, the kidney cortex shows the downregulation of complexes II and IV, and the mRNA levels of COX1 and COX2 are reduced [9,42,43].
For patients with IgAN, the copy numbers of COX3 and nicotinamide adenine dinucleotide dehydrogenase subunit 1 (ND1) in the urine are higher than the healthy controls [44]. After treatment, the changes in urinary COX3 and ND1 levels are positively associated with the changes in proteinuria and negatively associated with the changes in eGFR [44]. In the plasma of IgAN patients, the levels of advanced oxidation protein products (AOPPs) and malonaldehyde (MDA) are elevated, which can be attenuated by angiotensin-converting enzyme inhibitors (ACEIs) [7]. In the model of MN, complement activation in the podocyte reinforces the assembly of the C5b-9 membrane attack complex to induce the generation of ROS, which is mainly mediated via the upregulation of NOX [45,46,47]. An overexpression of CYP2B1 markedly elevates the generation of ROS and damages the cytoskeleton of the podocyte in MN, while silencing CYP2B1 ameliorates podocyte injury [48]. In podocytes treated with LN plasma, the production of ROS by mitochondria increases and the level of mitophagy decreases [49]. IgG from LN patients can elicit the generation of ROS as well [50].
HG-induced mitochondrial ROS generation is regarded as the main mechanism of DN [28,51]. Extracellular HG induces the formation of intracellular ROS in the podocyte via NOX and mitochondrial respiration [8,52,53]. In the kidney of Zucker obese rats, the expressions of NOX2, NOX4, and AT1R are upregulated [54]. Silencing NOX4 or pretreatment with NOX inhibitor GKT137831, diminishes high glucose-elicited ROS generation and the upregulation of profibrotic markers of the podocyte [55]. NOX5 is also upregulated in human diabetic kidney biopsies and type I diabetic mice [56,57]. Transgenic mice expressing podocyte-specific NOX5 develop albuminuria, podocyte FPE, and the elevation of systolic BP through the generation of ROS, which are further aggravated under streptozotocin (STZ)-induced diabetes [56]. Hyperglycemia can also activate the mineralocorticoid receptor (MR) to induce podocyte injury and proteinuria via the generation of ROS from NOX [58]. CYP4A, a member of the cytochrome P450 family, is upregulated and 20-hydroxyeicosatetraenoic acid is increased, followed by NOX activation [53]. In db/db mice and the HG-cultured podocyte, the activity of complexes I and III is reduced, while the functional expression of complex I in the cultured podocyte prevents the increase in mitochondrial ROS caused by HG [28,59]. In diabetic mice, the enhanced expression of p66Shc, a redox-regulating protein associated with Cyt C, promotes the generation of ROS from mitochondria [60,61].
A high-salt diet and Ang II both increase NADPH-dependent ROS formation in the kidney cortex [34,62]. Ang II induces an ROS-dependent rearrangement of the cytoskeleton to facilitate podocyte migration and eventual podocyte depletion, which can be inhibited by an NOX4 knockdown [12,63]. MR activation is involved in the podocyte damage in hypertensive kidney damage and metabolic syndrome as well [64,65]. Sprague–Dawley rats administrated with aldosterone and high salt display podocyte injury and proteinuria, resulting from elevated NOX activity and ROS production in the podocyte, while selective aldosterone blocker eplerenone and antioxidant tempol can protect against aldosterone-induced injury and proteinuria [64,65].
Homocysteine-induced podocyte injury and glomerulosclerosis involve NOX activation and endogenously generated superoxide anion and H2O2 that mediate the NLR family pyrin domain containing 3 (NLRP3) inflammasome formation [66]. In spontaneously hypertensive rats, a high level of homocysteine (Hcy) upregulates the mRNA levels of NOX2 and NOX4 [29]. AOPP has been shown to be closely associated with the severity of CKD and proteinuria [67]. It can bind to the receptor of advanced glycation end-products (RAGE) to induce NOX-mediated ROS generation, which further activates the Wnt/β-catenin pathway, resulting in FPE, matrix accumulation, and proteinuria [67,68,69]. Transforming growth factor β (TGFβ) has been shown to accumulate in renal diseases, and it can induce the upregulation of NOX4 via the Smad-dependent pathway and following ERK1/2-mTOR activation [70,71].

3.2. Deficient Antioxidant Defense Systems

Patients diagnosed as FSGS show markedly lower plasma and urinary GPX levels compared with MCD and healthy controls [72]. The glomerular GPX immunostaining score in FSGS is also lower than MCD and the normal controls both in patients and rats [72]. CoQ10 acts not only as an electron carrier, but also as an important antioxidant. In patients with idiopathic FSGS, CoQ10 partial deficiency might disturb podocyte biological function and induce renal lesions [73]. Proteinuria and podocyte FPE, resulting from podocyte lesions caused by a superoxide anion formed upon PA via xanthine oxidase, can be attenuated by intravenous SOD treatment [74]. Mitotempo, a SOD mimetic, reduces urinary protein excretion and lipid peroxidation via inhibiting oxidative stress and tissue damage in the MCD model [75]. In adriamycin-induced nephropathy, a model of FSGS, decreased activities of CAT, GPX, and SOD have a vital role in the occurrence of tubulointerstitial injury and glomerulosclerosis [27,76]. Mice lacking CAT are more vulnerable to the renal toxicity of adriamycin than wild mice, and manifest severe proteinuria and pathological lesions [77]. The supplementation of vitamin E in the diet of rats ameliorates adriamycin-induced renal injury via decreasing the activity of SOD1, SOD2, CAT, and GPX, both in the cortex and glomeruli [78].
In MN, anti-SOD2 IgG4 is detected in the plasma of patients, and it is co-localized with C5b-9 in the immune deposits, suggesting that SOD2 has an important role in the pathogenesis of MN [79]. A lower level of SOD in the plasma of IgAN patients is observed as well, which can be significantly reversed by treatment with ACEI [7]. IgAN patients treated with vitamin E show significantly lower proteinuria [80]. In the kidney of pristane-induced LN, the expressions of SOD1 and CAT are significantly reduced [81].
In the DN murine model and HG-treated podocyte, the expression levels of SOD2 and total SOD are significantly lower than controls [59,82,83]. Additionally, HG-induced oxidative stress-dependent podocyte loss and proteinuria can be attenuated by mitotempol [84]. The activities of CAT and GPX are lower at an early age of Zucker obese rats compared with controls, resulting in the accumulation of H2O2 [85]. HG treatment decreases the expression of Prdx6, an isoform of the Prdx family of antioxidant enzymes, which catalyze the reduction in H2O2 and ROS, but increases the expression of the thioredoxin-interacting protein (TxNIP) to inhibit the antioxidative function of Prdx and thioredoxin, leading to the accumulation of ROS and oxidative stress [17,86,87]. However, the overexpression of Prdx6 in the mouse podocyte restrains HG-induced ROS and MDA generation and recovers the activities of SOD and GSH [17]. In streptozotocin-induced TxNIP KO mice, albuminuria, serum creatinine, podocyte FPE and loss, GBM thickening, mesangial matrix expansion, and glomerulosclerosis are effectively attenuated, as well as oxidative stress and inflammation [86].
A high-salt diet and Ang II both downregulate the expression level of SOD in the kidney cortex [34]. Ang II stimulates the downregulation of Prdx2 in the podocyte, which results in elevated ROS release and protein peroxidation [88]. In spontaneously hypertensive rats administrated with high Hcy, a significant decrease in SOD and increase in MDA are observed [29].
Intracellular ROS excessive accumulation can induce damage to cellular macromolecules, such as DNA, proteins, and lipids, ultimately leading to podocyte injury, renal lesions, and the occurrence and progression of podocytopathies [7,10,11,32]. ROS-mediated nuclear and mitochondrial DNA damage and the upregulation of cell-cycle checkpoint proteins result in a poor proliferation of podocytes, which can be mitigated by radical scavenger 1,3-dimethyl-2-thiourea (DMTU) [10,89]. The generation and deposition of lipoperoxides, such as MDA and 4-hydroxynonenal (4-HNE) caused by ROS from damaged mitochondrion, are observed [7,43]. Additionally, elevated ROS also disturb the balance among key regulators of actin cytoskeletons, including RhoA and Rac1, resulting in actin cytoskeleton reorganization and subsequent podocyte FPE, depletion, and glomerulosclerosis [12,47,48]. Deglycosilation of alpha-dystroglycan in the glomeruli mediated by ROS leads to podocyte detachment from the GBM [11]. Moreover, ROS mediate the upregulation of profibrotic markers in the podocyte and promotes the inflammatory responses resulting in podocyte injury [55]. Pathological and ultrastructural features of podocytopathies, such as podocyte FPE, detachment, and loss, GBM thickening, mesangial matrix expansion, tubulointerstitial fibrosis, and glomerulosclerosis, as well as renal dysfunction indicators, such as urinary protein excretion and eGFR, are closely related to oxidative stress in the podocyte [8,44,56,67,68,69,90].

4. Oxidative Stress and Cell Death in Podocytopathies

Cell death is a highly conserved process that not only participates in the morphogenesis and development of organisms, but also the pathophysiological process. Different patterns of cell death manifest with discriminating morphological alterations and mediate the pathogenesis of various diseases [14]. Apoptosis is perhaps the most widely recognized pattern of cell death, which involves the activation of caspases (CASPs), especially CASP3, with morphological manifestations of cytoplasmic shrinkage, chromatin condensation, nuclear fragmentation, and ultimate apoptotic body formations [4,14,91]. Necrosis is another form of cell death characterized by the swelling of organelles and the whole cell coupling with negative morphological features of apoptosis and autophagy, and triggered by cyclophilin D-dependent permeability transition pore complex formation [14,92]. Necroptosis is generally manifested as a necrotic morphology and depends on the sequential activation of receptor interacting with serine/threonine kinase 3 (RIPK3) and a mixed lineage kinase domain, such as pseudokinase (MLKL) [14]. Pyroptosis is an inflammatory response depending on the activation of CASP1 and the formation of the plasma membrane pore by gasdermin-D (GSDMD), and manifests chromatin condensation that is different from apoptosis, cellular swelling, and plasma membrane permeabilization [14,91]. Ferroptosis is a newly discovered iron-dependent cell death that has gained significant attention, and is initiated by lipid peroxidation and manifests as necrotic morphology [14,93].
The death of the podocyte contributes to the progressive loss of the podocyte and subsequent glomerular filtration barrier destruction, and podocyte loss is the primary feature for the progression of podocytopathies [18,90,94,95]. In patients with DN, the number of podocytes in the kidney is decreased, and this is the strongest predictor of the progression of DN [96]. A large amount of research has confirmed that ROS play a significant role in regulating cell death [4,13,14]. Thus, in this section, we summarize the mechanisms by which the accumulation of intracellular ROS can regulate podocyte death.
Apoptosis has been the focus of extensive researches and is widely confirmed to be mediated mainly by two primary signaling pathways: the intrinsic apoptosis pathway and the extrinsic apoptosis pathway [14,97]. The intrinsic apoptosis pathway, or mitochondria-mediated pathway, can be activated by DNA damage, endoplasmic reticulum (ER) stress, oxidative stress, and other stimuli [14,97]. Proapoptotic members of the B-cell lymphoma-2 (BCL2) protein family, such as the BCL2-interacting mediator of cell death (BIM), BH3 interacting domain death agonist (BID), and BCL2 binding component 3 (BBC3), are activated transcriptionally or post-transcriptionally to induce the oligomerization of BCL2 associated X (BAX) and BCL2 antagonist/killer 1 (BAK) and the formation of mitochondrial outer-membrane permeabilization (MOMP) [14,97]. However, pro-survival members of the BCL2 protein family, such as BCL2 and BCL2-like 1 (BCL2L1), can antagonize MOMP to inhibit the occurrence of apoptosis [14,98]. MOMP facilitates the release of Cyt C, which can bind apoptotic peptidase activating factor 1 (APAF1) to mediate the activation of pro-CASP9 [14,97]. The extrinsic apoptosis pathway, or death receptor-mediated pathway, can be triggered by the Fas ligand (FasL), TNF-α, and the TNF-related apoptosis-inducing ligand (TRAIL) [97]. The death receptor executes the assembly of the death-inducing signaling complex (DISC), which regulates the activation of pro-CASP8 [97,99]. CASP9 from the intrinsic apoptosis pathway and CASP8 from the extrinsic apoptosis pathway can both cleave pro-CASP3 to activate CASP3, which cleaves cellular proteins leading to morphological damage and cell death [14,97].
Plenty of studies have shown that excessive ROS accumulation triggers podocyte apoptosis both in vivo or in vitro [8,68,100,101,102]. In type 1 and type 2 diabetic models, podocyte apoptosis is correlated with urinary albumin excretion and precedes the loss of the podocyte [8]. The molecular mechanisms for oxidative stress-triggered podocyte apoptosis are summarized in Figure 2.
The ROS-mediated intrinsic apoptosis pathway has been shown to participate in palmitic acid and HG-induced podocyte apoptosis, which can be markedly alleviated by mitotempo and NOX inhibitor apocynin [18,27,84]. Smad3 is activated to upregulate NOX4 after PA treatment [27]. However, extrinsic apoptosis pathway-associated indicators, including the Fas-associated protein with the death domain (FADD) and CASP8, are not changed by an intervention of palmitic acid [18]. Meanwhile, decreased BCL2 expression, increased BAX expression, and the translocation to mitochondria are observed [18,84]. Additionally, more Cyt C is released from the mitochondria and the levels of CASP9 and CASP3 are increased [18,27,84,103].
The ROS-triggered extrinsic apoptosis pathway also has a vital role in the pathogenesis of podocytopathies. AOPP increases the protein level of FOXO3 in the cultured podocyte in a ROS/mTOR-dependent pathway to induce proapoptotic FasL and BIM expression and the sequential upregulation of CASP3, leading to death receptor mediated apoptosis [104].
Mitogen-activated protein kinases (MAPKs) belong to the serine/threonine kinase superfamily that can regulate the activities of organisms, including growth, differentiation, and apoptosis [13]. Among MAPKs, extracellular signal-regulated kinases 1 or 2 (ERK1/2), Jun N-terminal kinase or stress-activated kinases (JNKs/SAPKs), and p38 MAPKs have been widely investigated [13,105]. HG and PA-mediated oxidative stress induces podocyte apoptosis through the activation of the p38 MAPK pathway, which can be attenuated by NOX inhibitor apocynin and antioxidant N-acetylcysteine (NAC) [8,19,106]. In type 1 diabetes and HG treated podocytes, protein kinase A is excited via dopamine 1 receptor activation to upregulate the expression of NOX5, which contributes to oxidative stress and induces the phosphorylation of p38 MAPK and downstream signaling cascades [57]. Moreover, HG also upregulates the expression of NOX4 to induce the phosphorylation of p38 MAPK [83]. The expression of BCL2 is lower in HG-treated podocytes, while BAX, cleaved caspases 3 and 9, are higher when compared with controls, indicating that the mitochondria-mediated apoptosis pathway is activated [57,83,106]. Advanced glycation end-products (AGEs) binding to RAGE can trigger ROS generation to induce podocyte apoptosis through the activation of Forkhead transcription factor 4 (FOXO4) and the p38 MAPK pathway, whereas the inhibition of p38 MAPK or siRNA for FOXO4 abolishes AGE-induced podocyte apoptosis [101].
The phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathway can be activated by various cellular stimuli and regulate cellular functions, such as growth, proliferation, cell cycle, and survival [107]. Previous studies demonstrated that HG increases the level of ROS in the podocyte and induces oxidative stress [8,28]. Meanwhile, ROS downregulate the expression of PI3K and suppress the phosphorylation of Akt [101,108]. Downregulated Akt phosphorylation activates BAX and CASP3 to trigger podocyte apoptosis [108].
p53 is a tumor suppressor transcriptive factor, which responds to various stresses to maintain homeostasis via triggering cell cycle arrest, apoptosis, senescence, and DNA repair [109]. The HG stimulus downregulates AMP-activated protein kinase (AMPK) to upregulate NOX4 expression and activity, which further activates p53 [110,111]. AOPP can also activate NOX to induce the expression of p53 [69,112]. p53 can regulate downstream proapoptotic genes, including the p53-upregulated modulator of apoptosis (PUMA) and BAX [69,110,111,112]. Downregulated BCL2, upregulated CASP3 and CASP9, and DNA fragmentation are consistent with intrinsic apoptosis pathway activation [110,111,112].
ER stress is caused by misfolded or unfolded protein accumulation and an intracellular calcium imbalance, with the unfolded protein response (UPR) determining the cell fate under ER stress [113]. Palmitic acid and AOPP trigger ROS-mediated ER stress to induce podocyte apoptosis [68,100]. Three UPR pathways, including protein kinase-like ER kinase (PERK), activating transcription factor 6 (ATF6), and inositol requiring 1 (IRE1) pathways, are activated to trigger CCAAT/enhancer-binding protein-homologous protein (CHOP)- and CASP12-dependent podocyte apoptosis with BCL2 downregulation involved in CHOP-dependent apoptosis [68,100].
Moreover, the cytosolic apoptosis-inducing factor (AIF) is also increased in the HG-intervened podocyte, which can activate caspase-independent apoptosis [103,114].
Other forms of cell death also take part in ROS-mediated podocyte damage. Pyroptosis has been shown to participate in HIV-associated nephropathy [16]. HIV-transgenic mice exhibit increased mRNA levels and the expression of the NLR family pyrin domain containing 3 (NLRP3), CARD domain containing adaptor protein (ASC), and CASP1 and IL-1β in the kidney, which can be partially inhibited by tempo, suggesting that ROS are crucial to HIV-induced podocyte pyroptosis [16]. Ferroptosis is involved in ROS-mediated podocytopathies as well. Solute carrier family 7 member 11 (SLC7A11) is a member of Xc-system, which transports cysteine and glutamate, and GPX4 is the main endogenous inhibitor of ferroptosis, which reduces the production of phospholipid hydroperoxide [115]. In the HG-stimulated podocyte, the expressions of SLC7A11 and GPX4 are reduced, which can be eliminated by Prdx6 overexpression [17]. Additionally, the protective effect of Prdx6 overexpression on the podocyte can be suppressed by the ferroptosis-inducing agent erastin [17].
Both necrosis and necroptosis are found to participate in the pathogenesis of podocytopathies, such as LN and DN [116,117,118]. However, the evidence for the relationship between oxidative stress, necrosis, and necroptosis in podocytopathies is still lacking, possibly owing to technical restrictions in detecting and validating necrosis and necroptosis in vivo [15].
Since the major researches focus on oxidative stress-mediated apoptosis in DN, further research focusing on the underlying mechanisms of ROS mediating other types of cell death in podocytes exposed to different stimuli under other pathophysiologic conditions is desirable. In conclusion, oxidative stress can trigger podocyte death through a variety of molecular mechanisms. Targeting oxidative stress-mediated podocyte death may be a potential novel therapy for patients with podocytopathies.

5. Therapeutic Implications

Since ROS-mediated cell death is of great importance for the pathogenesis of podocytopathies, antioxidants that can protect the podocyte from death are promising for the treatment of patients diagnosed with podocytopathies.
Mitoquinone (MitoQ) is a mitochondria-targeted antioxidant that consists of CoQ10 and triphenyl phosphate. It has been shown to mitigate oxidative stress in various diseases, such as hypoxia-induced pulmonary hypertension and renal ischemia-reperfusion injury [119,120]. In Ang II-treated podocytes, MitoQ suppresses Ang II-induced mitochondrial dysfunction and oxidative stress and reduces podocyte apoptosis [121].
Taurine is an amino sulfonic acid that has been used as an oral supplement for the treatment of disorders, such as congestive heart failure, hypertension, and diabetes mellitus [122,123,124]. Antioxidant activity plays an essential role in the protective effect of taurine [125]. In DN and PA-induced podocytopathies, taurine supply alleviates urinary protein excretion and histopathological lesions [126,127]. Mitochondria ROS generation and podocyte apoptosis induced by HG are attenuated by taurine [127].
Melatonin is a hormone secreted by the brain to maintain the circadian rhythm. Some studies revealed that melatonin can mitigate DN via the inhibition of oxidative stress [128,129]. In Ang II-induced podocyte injury, melatonin reduces podocyte apoptosis and enhances the proliferation capacity of the podocyte [130]. Additionally, the protective effect is associated with inhibited oxidative stress and recovered mitochondrial function [130].
Folic acid is a B vitamin that can reduce plasma Hcy and antagonize the harmful effects generated by it [29,131]. In a spontaneously hypertensive model of rats, a high level of Hcy induces mitochondrial oxidative stress and glomerular damage, whereas even a low level of folic acid could attenuate the glomerular damage caused by Hcy via inhibiting oxidative stress and podocyte apoptosis [29]. Additionally, the antioxidant effect of folic acid may be independent of Hcy [29].
Herbal medicine has been applied in the clinic for thousands of years and is regarded as an alternative treatment for a lot of diseases. A great deal of herbs and herb-derived components are found to have an antioxidative capacity, and have been investigated for the therapeutic effectiveness in podocytopathies. Since oxidative stress-regulated cell death has a significant role in the pathogenesis of podocytopathies, those antioxidative herbs and herb-derived components might improve renal injury via the suppression of the death of the podocyte. Herbs and herb-derived components decrease intracellular ROS via downregulating pro-oxidative enzymes, such as NOX, and elevating mitochondrial respiration chain complex activities [59,83]. The levels of antioxidative enzymes, such as CAT and SOD, and antioxidants, such as GSH, are augmented as well [81,132]. ROS-mediated apoptotic pathways, including PI3K/Akt, p38 MAPK, and p53 pathways are found to be regulated by herbs and herb-derived components [83,108,111]. Except for anti-apoptotic ability, geniposide, an ingredient of Gardenia jasminoides Ellis, can alleviate podocyte injury and inhibit the development of DN via the suppression of pyroptosis [133]. In Table 1, we summarize the antioxidative herbs and herb-derived components that were verified to alleviate podocytopathies via the suppression of cell death.
The therapeutic targets of the antioxidants described above are summarized in Table 2.
Considering that oxidative stress plays a detrimental role in podocytopathies, drugs targeting oxidative stress may have potential value for clinical application in patients with podocytopathies. Hence, we summarize those oxidative stress-targeted drugs in clinical trials from ClinicalTrials.gov in Table 3 [160].

6. Conclusions

The evidence supports the determination that mitochondrial oxidative stress, driven by excessive stress and harmful stimuli to the podocyte, plays an essential role in the pathogenesis of podocytopathies. Plenty of studies revealed that oxidative stress can mediate the cell death of the podocyte, especially podocyte apoptosis, via many signaling pathways. Remarkably, oxidative stress can also contribute to the development of podocytopathies via other mechanisms, such as cell cycle arrest. Further researches focusing on the relationship between oxidative stress and other types of cell death in the podocyte are needed. Many potential antioxidants are under investigation for the effectiveness in treating podocytopathies. Considering that podocyte injury is the initiation factor of podocytopathies, and oxidative stress and oxidative stress-mediated cell death are consistent mechanisms among podocyte injury under different pathophysiologic conditions, targeted podocyte oxidative stress therapy that can ameliorate cell death might be promising to slow and even halt the progression of podocytopathies.

Author Contributions

Y.-T.Z.: writing—review and editing, visualization; C.W.: writing, funding acquisition; J.-H.L.: writing—review and editing; H.-P.H.: visualization; C.Z.: conceptualization, funding acquisition. All authors have read and agreed to the published version of the manuscript.

Funding

This work was financially supported by the National Natural Science Foundation of China (81961138007, 81974096, 81770711, 81873602), the National Key Research and Development Program (2018YFC1314000), and the Program for HUST Academic Frontier Youth Team (2017QYTD20).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kopp, J.B.; Anders, H.J.; Susztak, K.; Podesta, M.A.; Remuzzi, G.; Hildebrandt, F.; Romagnani, P. Podocytopathies. Nat. Rev. Dis. Primers 2020, 6, 68. [Google Scholar] [CrossRef] [PubMed]
  2. Wiggins, R.C. The Spectrum of Podocytopathies: A Unifying View of Glomerular Diseases. Kidney Int. 2007, 71, 1205–1214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Griffin, S.V.; Petermann, A.T.; Durvasula, R.V.; Shankland, S.J. Podocyte Proliferation and Differentiation in Glomerular Disease: Role of Cell-Cycle Regulatory Proteins. Nephrol. Dial. Transplant. 2003, 18 (Suppl. 6), vi8–vi13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Orrenius, S.; Gogvadze, V.; Zhivotovsky, B. Mitochondrial Oxidative Stress: Implications for Cell Death. Annu. Rev. Pharmacol. Toxicol. 2007, 47, 143–183. [Google Scholar] [CrossRef] [PubMed]
  5. Sies, H. Oxidative Stress: A Concept in Redox Biology and Medicine. Redox Biol. 2015, 4, 180–183. [Google Scholar] [CrossRef] [Green Version]
  6. Hagiwara, M.; Yamagata, K.; Capaldi, R.A.; Koyama, A. Mitochondrial Dysfunction in Focal Segmental Glomerulosclerosis of Puromycin Aminonucleoside Nephrosis. Kidney Int. 2006, 69, 1146–1152. [Google Scholar] [CrossRef] [Green Version]
  7. Pei, Y.; Xu, Y.; Ruan, J.; Rong, L.; Jiang, M.; Mo, Y.; Jiang, X. Plasma Oxidative Stress Level of IgA Nephropathy in Children and the Effect of Early Intervention with Angiotensin-Converting Enzyme Inhibitors. J. Renin Angiotensin Aldosterone Syst. 2016, 17, 1470320316647240. [Google Scholar] [CrossRef] [Green Version]
  8. Susztak, K.; Raff, A.C.; Schiffer, M.; Bottinger, 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]
  9. Solin, M.L.; Pitkanen, S.; Taanman, J.W.; Holthofer, H. Mitochondrial Dysfunction in Congenital Nephrotic Syndrome. Lab Investig. 2000, 80, 1227–1232. [Google Scholar] [CrossRef]
  10. Marshall, C.B.; Pippin, J.W.; Krofft, R.D.; Shankland, S.J. Puromycin Aminonucleoside Induces Oxidant-Dependent DNA Damage in Podocytes in Vitro and in Vivo. Kidney Int. 2006, 70, 1962–1973. [Google Scholar] [CrossRef] [Green Version]
  11. Vogtlander, N.P.; Tamboer, W.P.; Bakker, M.A.; Campbell, K.P.; van der Vlag, J.; Berden, J.H. Reactive Oxygen Species Deglycosilate Glomerular Alpha-Dystroglycan. Kidney Int. 2006, 69, 1526–1534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Hsu, H.H.; Hoffmann, S.; Endlich, N.; Velic, A.; Schwab, A.; Weide, T.; Schlatter, E.; Pavenstadt, H. Mechanisms of Angiotensin II Signaling on Cytoskeleton of Podocytes. J. Mol. Med. 2008, 86, 1379–1394. [Google Scholar] [CrossRef] [Green Version]
  13. Ryter, S.W.; Kim, H.P.; Hoetzel, A.; Park, J.W.; Nakahira, K.; Wang, X.; Choi, A.M. Mechanisms of Cell Death in Oxidative Stress. Antioxid. Redox Signal. 2007, 9, 49–89. [Google Scholar] [CrossRef] [PubMed]
  14. Galluzzi, L.; Vitale, I.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; Amelio, I.; Andrews, D.W.; et al. Molecular Mechanisms of Cell Death: Recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018, 25, 486–541. [Google Scholar] [CrossRef] [PubMed]
  15. Braun, F.; Becker, J.U.; Brinkkoetter, P.T. Live or Let Die: Is There Any Cell Death in Podocytes? Semin. Nephrol. 2016, 36, 208–219. [Google Scholar] [CrossRef] [PubMed]
  16. Haque, S.; Lan, X.; Wen, H.; Lederman, R.; Chawla, A.; Attia, M.; Bongu, R.P.; Husain, M.; Mikulak, J.; Saleem, M.A.; et al. HIV Promotes NLRP3 Inflammasome Complex Activation in Murine HIV-Associated Nephropathy. Am. J. Pathol. 2016, 186, 347–358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Zhang, Q.; Hu, Y.; Hu, J.E.; Ding, Y.; Shen, Y.; Xu, H.; Chen, H.; Wu, N. Sp1-Mediated Upregulation of Prdx6 Expression Prevents Podocyte Injury in Diabetic Nephropathy via Mitigation of Oxidative Stress and Ferroptosis. Life Sci. 2021, 278, 119529. [Google Scholar] [CrossRef]
  18. Liu, T.; Chen, X.M.; Sun, J.Y.; Jiang, X.S.; Wu, Y.; Yang, S.; Huang, H.Z.; Ruan, X.Z.; Du, X.G. Palmitic Acid-Induced Podocyte Apoptosis via the Reactive Oxygen Species-Dependent Mitochondrial Pathway. Kidney Blood Press Res. 2018, 43, 206–219. [Google Scholar] [CrossRef]
  19. Chen, Z.; Wan, X.; Hou, Q.; Shi, S.; Wang, L.; Chen, P.; Zhu, X.; Zeng, C.; Qin, W.; Zhou, W.; et al. GADD45B Mediates Podocyte Injury in Zebrafish by Activating the ROS-GADD45B-P38 Pathway. Cell Death Dis. 2016, 7, e2068. [Google Scholar] [CrossRef] [Green Version]
  20. Hinkes, B.; Wiggins, R.C.; Gbadegesin, R.; Vlangos, C.N.; Seelow, D.; Nürnberg, G.; Garg, P.; Verma, R.; Chaib, H.; Hoskins, B.E.; et al. Positional Cloning Uncovers Mutations in PLCE1 Responsible for A Nephrotic Syndrome Variant that May Be Reversible. Nat. Genet. 2006, 38, 1397–1405. [Google Scholar] [CrossRef]
  21. Yang, Y.; Jeanpierre, C.; Dressler, G.R.; Lacoste, M.; Niaudet, P.; Gubler, M.C. WT1 and PAX-2 Podocyte Expression in Denys-Drash Syndrome and Isolated Diffuse Mesangial Sclerosis. Am. J. Pathol. 1999, 154, 181–192. [Google Scholar] [CrossRef] [Green Version]
  22. Abd ElHafeez, S.; Bolignano, D.; D’Arrigo, G.; Dounousi, E.; Tripepi, G.; Zoccali, C. Prevalence and Burden of Chronic Kidney Disease among the General Population and High-Risk Groups in Africa: A Systematic Review. BMJ Open 2018, 8, e015069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Udani, S.; Lazich, I.; Bakris, G.L. Epidemiology of Hypertensive Kidney Disease. Nat. Rev. Nephrol. 2011, 7, 11–21. [Google Scholar] [CrossRef] [PubMed]
  24. Yang, Y.; Gubler, M.C.; Beaufils, H. Dysregulation of Podocyte Phenotype in Idiopathic Collapsing Glomerulopathy and HIV-Associated Nephropathy. Nephron 2002, 91, 416–423. [Google Scholar] [CrossRef]
  25. Markowitz, G.S.; Appel, G.B.; Fine, P.L.; Fenves, A.Z.; Loon, N.R.; Jagannath, S.; Kuhn, J.A.; Dratch, A.D.; D’Agati, V.D. Collapsing Focal Segmental Glomerulosclerosis Following Treatment with High-Dose Pamidronate. J. Am. Soc. Nephrol. 2001, 12, 1164–1172. [Google Scholar] [CrossRef]
  26. Yang, B.; Chen, Y.; Shi, J. Reactive Oxygen Species (ROS)-Based Nanomedicine. Chem. Rev. 2019, 119, 4881–4985. [Google Scholar] [CrossRef]
  27. Yu, L.; Liu, Y.; Wu, Y.; Liu, Q.; Feng, J.; Gu, X.; Xiong, Y.; Fan, Q.; Ye, J. Smad3/Nox4-Mediated Mitochondrial Dysfunction Plays A Crucial Role in Puromycin Aminonucleoside-Induced Podocyte Damage. Cell Signal. 2014, 26, 2979–2991. [Google Scholar] [CrossRef]
  28. Galvan, D.L.; Badal, S.S.; Long, J.; Chang, B.H.; Schumacker, P.T.; Overbeek, P.A.; Danesh, F.R. Real-Time in Vivo Mitochondrial Redox Assessment Confirms Enhanced Mitochondrial Reactive Oxygen Species in Diabetic Nephropathy. Kidney Int. 2017, 92, 1282–1287. [Google Scholar] [CrossRef]
  29. Gao, N.; Zhang, Y.; Lei, L.; Li, L.; Cao, P.; Zhao, X.; Lin, L.; Xu, R. Low Doses of Folic Acid Can Reduce Hyperhomocysteinemia-Induced Glomerular Injury in Spontaneously Hypertensive Rats. Hypertens. Res. 2020, 43, 1182–1191. [Google Scholar] [CrossRef]
  30. Turrens, J.F. Mitochondrial Formation of Reactive Oxygen Species. J. Physiol. 2003, 552, 335–344. [Google Scholar] [CrossRef]
  31. Sarewicz, M.; Osyczka, A. Electronic Connection Between the Quinone and Cytochrome C Redox Pools and Its Role in Regulation of Mitochondrial Electron Transport and Redox Signaling. Physiol. Rev. 2015, 95, 219–243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. 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] [PubMed]
  33. Liu, H.; Bigler, S.A.; Henegar, J.R.; Baliga, R. Cytochrome P450 2B1 Mediates Oxidant Injury in Puromycin-Induced Nephrotic Syndrome. Kidney Int. 2002, 62, 868–876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Kitiyakara, C.; Chabrashvili, T.; Chen, Y.; Blau, J.; Karber, A.; Aslam, S.; Welch, W.J.; Wilcox, C.S. Salt Intake, Oxidative Stress, and Renal Expression of NADPH Oxidase and Superoxide Dismutase. J. Am. Soc. Nephrol. 2003, 14, 2775–2782. [Google Scholar] [CrossRef] [Green Version]
  35. Tong, J.; Jin, Y.; Weng, Q.; Yu, S.; Hussain, H.M.J.; Ren, H.; Xu, J.; Zhang, W.; Li, X.; Wang, W.; et al. Glomerular Transcriptome Profiles in Focal Glomerulosclerosis: New Genes and Pathways for Steroid Resistance. Am. J. Nephrol. 2020, 51, 442–452. [Google Scholar] [CrossRef] [PubMed]
  36. Kim, H.J.; Sato, T.; Rodriguez-Iturbe, B.; Vaziri, N.D. Role of Intrarenal Angiotensin System Activation, Oxidative Stress, Inflammation, and Impaired Nuclear Factor-Erythroid-2-Related Factor 2 Activity in The Progression of Focal Glomerulosclerosis. J. Pharmacol. Exp. Ther. 2011, 337, 583–590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Baek, J.H.; Gomez, I.G.; Wada, Y.; Roach, A.; Mahad, D.; Duffield, J.S. Deletion of The Mitochondrial Complex-IV Cofactor Heme A:Farnesyltransferase Causes Focal Segmental Glomerulosclerosis and Interferon Response. Am. J. Pathol. 2018, 188, 2745–2762. [Google Scholar] [CrossRef] [Green Version]
  38. Ashraf, S.; Gee, H.Y.; Woerner, S.; Xie, L.X.; Vega-Warner, V.; Lovric, S.; Fang, H.; Song, X.; Cattran, D.C.; Avila-Casado, C.; et al. ADCK4 Mutations Promote Steroid-Resistant Nephrotic Syndrome through CoQ10 Biosynthesis Disruption. J. Clin. Investig. 2013, 123, 5179–5189. [Google Scholar] [CrossRef] [Green Version]
  39. Song, C.C.; Hong, Q.; Geng, X.D.; Wang, X.; Wang, S.Q.; Cui, S.Y.; Guo, M.D.; Li, O.; Cai, G.Y.; Chen, X.M.; et al. New Mutation of Coenzyme Q10 Monooxygenase 6 Causing Podocyte Injury in A Focal Segmental Glomerulosclerosis Patient. Chin. Med. J. 2018, 131, 2666–2675. [Google Scholar] [CrossRef]
  40. Liu, H.; Baliga, M.; Bigler, S.A.; Baliga, R. Role of Cytochrome P450 2B1 in Puromycin Aminonucleoside-Induced Cytotoxicity to Glomerular Epithelial Cells. Nephron Exp. Nephrol. 2003, 94, e17–e24. [Google Scholar] [CrossRef]
  41. Xie, K.; Zhu, M.; Xiang, P.; Chen, X.; Kasimumali, A.; Lu, R.; Wang, Q.; Mou, S.; Ni, Z.; Gu, L.; et al. Protein Kinase A/Creb Signaling Prevents Adriamycin-Induced Podocyte Apoptosis via Upregulation of Mitochondrial Respiratory Chain Complexes. Mol. Cell Biol. 2018, 38, e00181-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Holthofer, H.; Kretzler, M.; Haltia, A.; Solin, M.L.; Taanman, J.W.; Schagger, H.; Kriz, W.; Kerjaschki, D.; Schlondorff, D. Altered Gene Expression and Functions of Mitochondria in Human Nephrotic Syndrome. FASEB J. 1999, 13, 523–532. [Google Scholar] [CrossRef] [PubMed]
  43. Solin, M.L.; Ahola, H.; Haltia, A.; Ursini, F.; Montine, T.; Roveri, A.; Kerjaschki, D.; Holthofer, H. Lipid Peroxidation in Human Proteinuric Disease. Kidney Int. 2001, 59, 481–487. [Google Scholar] [CrossRef] [Green Version]
  44. Yu, B.C.; Cho, N.-J.; Park, S.; Kim, H.; Choi, S.J.; Kim, J.K.; Hwang, S.D.; Gil, H.-W.; Lee, E.Y.; Jeon, J.S.; et al. IgA Nephropathy Is Associated with Elevated Urinary Mitochondrial DNA Copy Numbers. Sci. Rep. 2019, 9, 16068. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Takano, T.; Elimam, H.; Cybulsky, A.V. Complement-Mediated Cellular Injury. Semin. Nephrol. 2013, 33, 586–601. [Google Scholar] [CrossRef] [PubMed]
  46. Nangaku, M.; Shankland, S.J.; Couser, W.G. Cellular Response to Injury in Membranous Nephropathy. J. Am. Soc. Nephrol. 2005, 16, 1195–1204. [Google Scholar] [CrossRef] [PubMed]
  47. Chen, Z.H.; Qin, W.S.; Zeng, C.H.; Zheng, C.X.; Hong, Y.M.; Lu, Y.Z.; Li, L.S.; Liu, Z.H. Triptolide Reduces Proteinuria in Experimental Membranous Nephropathy and Protects Against C5b-9-Induced Podocyte Injury In Vitro. Kidney Int. 2010, 77, 974–988. [Google Scholar] [CrossRef] [Green Version]
  48. Liu, H.; Tian, N.; Arany, I.; Bigler, S.A.; Waxman, D.J.; Shah, S.V.; Baliga, R. Cytochrome P450 2B1 Mediates Complement-Dependent Sublytic Injury in A Model of Membranous Nephropathy. J. Biol. Chem. 2010, 285, 40901–40910. [Google Scholar] [CrossRef] [Green Version]
  49. Tian, Y.; Guo, H.; Miao, X.; Xu, J.; Yang, R.; Zhao, L.; Liu, J.; Yang, L.; Gao, F.; Zhang, W.; et al. Nestin Protects Podocyte from Injury in Lupus Nephritis by Mitophagy and Oxidative Stress. Cell Death Dis. 2020, 11, 319. [Google Scholar] [CrossRef]
  50. Qi, Y.Y.; Zhou, X.J.; Cheng, F.J.; Hou, P.; Ren, Y.L.; Wang, S.X.; Zhao, M.H.; Yang, L.; Martinez, J.; Zhang, H. Increased Autophagy Is Cytoprotective Against Podocyte Injury Induced by Antibody and Interferon-Alpha in Lupus Nephritis. Ann. Rheum. Dis. 2018, 77, 1799–1809. [Google Scholar] [CrossRef]
  51. Brownlee, M. Biochemistry and Molecular Cell Biology of Diabetic Complications. Nature 2001, 414, 813–820. [Google Scholar] [CrossRef] [PubMed]
  52. Eid, S.; Boutary, S.; Braych, K.; Sabra, R.; Massaad, C.; Hamdy, A.; Rashid, A.; Moodad, S.; Block, K.; Gorin, Y.; et al. mTORC2 Signaling Regulates Nox4-Induced Podocyte Depletion in Diabetes. Antioxid. Redox Signal. 2016, 25, 703–719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Eid, A.A.; Gorin, Y.; Fagg, B.M.; Maalouf, R.; Barnes, J.L.; Block, K.; Abboud, H.E. Mechanisms of Podocyte Injury in Diabetes: Role of Cytochrome P450 and NADPH Oxidases. Diabetes 2009, 58, 1201–1211. [Google Scholar] [CrossRef] [Green Version]
  54. Habibi, J.; Aroor, A.R.; Das, N.A.; Manrique-Acevedo, C.M.; Johnson, M.S.; Hayden, M.R.; Nistala, R.; Wiedmeyer, C.; Chandrasekar, B.; DeMarco, V.G. The Combination of A Neprilysin Inhibitor (Sacubitril) and Angiotensin-II Receptor Blocker (Valsartan) Attenuates Glomerular and Tubular Injury in the Zucker Obese Rat. Cardiovasc Diabetol. 2019, 18, 40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Jha, J.C.; Gray, S.P.; Barit, D.; Okabe, J.; El-Osta, A.; Namikoshi, T.; Thallas-Bonke, V.; Wingler, K.; Szyndralewiez, C.; Heitz, F.; et al. Genetic Targeting or Pharmacologic Inhibition of NADPH Oxidase Nox4 Provides Renoprotection in Long-Term Diabetic Nephropathy. J. Am. Soc. Nephrol. 2014, 25, 1237–1254. [Google Scholar] [CrossRef] [Green Version]
  56. Holterman, C.E.; Thibodeau, J.F.; Towaij, C.; Gutsol, A.; Montezano, A.C.; Parks, R.J.; Cooper, M.E.; Touyz, R.M.; Kennedy, C.R. Nephropathy and Elevated BP in Mice with Podocyte-Specific NADPH Oxidase 5 Expression. J. Am. Soc. Nephrol. 2014, 25, 784–797. [Google Scholar] [CrossRef] [Green Version]
  57. Shao, X.; Zhang, X.; Hu, J.; Gao, T.; Chen, J.; Xu, C.; Wei, C. Dopamine 1 Receptor Activation Protects Mouse Diabetic Podocytes Injury via Regulating the PKA/NOX-5/P38 MAPK Axis. Exp. Cell Res. 2020, 388, 111849. [Google Scholar] [CrossRef]
  58. Toyonaga, J.; Tsuruya, K.; Ikeda, H.; Noguchi, H.; Yotsueda, H.; Fujisaki, K.; Hirakawa, M.; Taniguchi, M.; Masutani, K.; Iida, M. Spironolactone Inhibits Hyperglycemia-Induced Podocyte Injury by Attenuating ROS Production. Nephrol. Dial. Transplant. 2011, 26, 2475–2484. [Google Scholar] [CrossRef] [Green Version]
  59. Zhang, T.; Chi, Y.; Kang, Y.; Lu, H.; Niu, H.; Liu, W.; Li, Y. Resveratrol Ameliorates Podocyte Damage in Diabetic Mice via SIRT1/PGC-1alpha Mediated Attenuation of Mitochondrial Oxidative Stress. J. Cell Physiol. 2019, 234, 5033–5043. [Google Scholar] [CrossRef]
  60. Bock, F.; Shahzad, K.; Wang, H.; Stoyanov, S.; Wolter, J.; Dong, W.; Pelicci, P.G.; Kashif, M.; Ranjan, S.; Schmidt, S.; et al. Activated Protein C Ameliorates Diabetic Nephropathy by Epigenetically Inhibiting the Redox Enzyme P66shc. Proc. Natl. Acad. Sci. USA 2013, 110, 648–653. [Google Scholar] [CrossRef] [Green Version]
  61. Zheng, D.; Tao, M.; Liang, X.; Li, Y.; Jin, J.; He, Q. P66shc Regulates Podocyte Autophagy in High Glucose Environment Through the Notch-PTEN-PI3k/Akt/mTOR Pathway. Histol. Histopathol. 2020, 35, 405–415. [Google Scholar] [CrossRef]
  62. Lambeth, J.D. Nox Enzymes and the Biology of Reactive Oxygen. Nat. Rev. Immunol. 2004, 4, 181–189. [Google Scholar] [CrossRef] [PubMed]
  63. Che, G.; Gao, H.; Hu, Q.; Xie, H.; Zhang, Y. Angiotensin II Promotes Podocyte Injury by Activating Arf6-Erk1/2-Nox4 Signaling Pathway. PLoS ONE 2020, 15, e0229747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Shibata, S.; Nagase, M.; Yoshida, S.; Kawachi, H.; Fujita, T. Podocyte as the Target for Aldosterone: Roles of Oxidative Stress and Sgk1. Hypertension 2007, 49, 355–364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Nagase, M.; Fujita, T. Aldosterone and Glomerular Podocyte Injury. Clin. Exp. Nephrol. 2008, 12, 233–242. [Google Scholar] [CrossRef] [PubMed]
  66. Abais, J.M.; Xia, M.; Li, G.; Gehr, T.W.; Boini, K.M.; Li, P.L. Contribution of Endogenously Produced Reactive Oxygen Species to the Activation of Podocyte NLRP3 Inflammasomes in Hyperhomocysteinemia. Free Radic. Biol. Med. 2014, 67, 211–220. [Google Scholar] [CrossRef] [Green Version]
  67. Zhou, L.; Chen, X.; Lu, M.; Wu, Q.; Yuan, Q.; Hu, C.; Miao, J.; Zhang, Y.; Li, H.; Hou, F.F.; et al. Wnt/Beta-Catenin Links Oxidative Stress to Podocyte Injury and Proteinuria. Kidney Int. 2019, 95, 830–845. [Google Scholar] [CrossRef]
  68. Rong, G.; Tang, X.; Guo, T.; Duan, N.; Wang, Y.; Yang, L.; Zhang, J.; Liang, X. Advanced Oxidation Protein Products Induce Apoptosis in Podocytes Through Induction of Endoplasmic Reticulum Stress. J. Physiol. Biochem 2015, 71, 455–470. [Google Scholar] [CrossRef]
  69. Zhou, L.L.; Hou, F.F.; Wang, G.B.; Yang, F.; Xie, D.; Wang, Y.P.; Tian, J.W. Accumulation of Advanced Oxidation Protein Products Induces Podocyte Apoptosis and Deletion Through Nadph-Dependent Mechanisms. Kidney Int. 2009, 76, 1148–1160. [Google Scholar] [CrossRef] [Green Version]
  70. Das, R.; Xu, S.; Nguyen, T.T.; Quan, X.; Choi, S.K.; Kim, S.J.; Lee, E.Y.; Cha, S.K.; Park, K.S. Transforming Growth Factor Beta1-Induced Apoptosis in Podocytes via the Extracellular Signal-Regulated Kinase-Mammalian Target of Rapamycin Complex 1-NADPH Oxidase 4 Axis. J. Biol. Chem. 2015, 290, 30830–30842. [Google Scholar] [CrossRef] [Green Version]
  71. Das, R.; Xu, S.; Quan, X.; Nguyen, T.T.; Kong, I.D.; Chung, C.H.; Lee, E.Y.; Cha, S.K.; Park, K.S. Upregulation of Mitochondrial Nox4 Mediates TGF-Beta-Induced Apoptosis in Cultured Mouse Podocytes. Am. J. Physiol. Renal Physiol. 2014, 306, F155–F167. [Google Scholar] [CrossRef] [PubMed]
  72. Chen, H.C.; Guh, J.Y.; Lai, Y.H. Alterations of Glomerular and Extracellular Glutathione Peroxidase Levels in Patients and Rats with Focal Segmental Glomerulosclerosis. J. Lab Clin. Med. 2001, 137, 279–283. [Google Scholar] [CrossRef] [PubMed]
  73. Gasser, D.L.; Winkler, C.A.; Peng, M.; An, P.; McKenzie, L.M.; Kirk, G.D.; Shi, Y.; Xie, L.X.; Marbois, B.N.; Clarke, C.F.; et al. Focal Segmental Glomerulosclerosis Is Associated with A PDSS2 Haplotype and, Independently, with A Decreased Content of Coenzyme Q10. Am. J. Physiol. Renal Physiol. 2013, 305, F1228–F1238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Diamond, J.R.; Bonventre, J.V.; Karnovsky, M.J. A Role for Oxygen Free Radicals in Aminonucleoside Nephrosis. Kidney Int. 1986, 29, 478–483. [Google Scholar] [CrossRef] [Green Version]
  75. Fujii, Y.; Matsumura, H.; Yamazaki, S.; Shirasu, A.; Nakakura, H.; Ogihara, T.; Ashida, A. Efficacy of A Mitochondrion-Targeting Agent for Reducing the Level of Urinary Protein in Rats with Puromycin Aminonucleoside-Induced Minimal-Change Nephrotic Syndrome. PLoS ONE 2020, 15, e0227414. [Google Scholar] [CrossRef]
  76. Deman, A.; Ceyssens, B.; Pauwels, M.; Zhang, J.; Houte, K.V.; Verbeelen, D.; Van den Branden, C. Altered Antioxidant Defence in A Mouse Adriamycin Model of Glomerulosclerosis. Nephrol. Dial. Transplant. 2001, 16, 147–150. [Google Scholar] [CrossRef] [Green Version]
  77. Takiue, K.; Sugiyama, H.; Inoue, T.; Morinaga, H.; Kikumoto, Y.; Kitagawa, M.; Kitamura, S.; Maeshima, Y.; Wang, D.-H.; Masuoka, N.; et al. Acatalasemic Mice Are Mildly Susceptible to Adriamycin Nephropathy and Exhibit Increased Albuminuria and Glomerulosclerosis. BMC Nephrol. 2012, 13, 14. [Google Scholar] [CrossRef]
  78. Van den Branden, C.; Deman, A.; Ceyssens, B.; Pauwels, M.; Empsen, C.; Verbeelen, D. Vitamin E Protects Renal Antioxidant Enzymes and Attenuates Glomerulosclerosis in Adriamycin-Treated Rats. Nephron 2002, 91, 129–133. [Google Scholar] [CrossRef]
  79. Prunotto, M.; Carnevali, M.L.; Candiano, G.; Murtas, C.; Bruschi, M.; Corradini, E.; Trivelli, A.; Magnasco, A.; Petretto, A.; Santucci, L.; et al. Autoimmunity in Membranous Nephropathy Targets Aldose Reductase and SOD2. J. Am. Soc. Nephrol. 2010, 21, 507–519. [Google Scholar] [CrossRef] [Green Version]
  80. Chan, J.C.; Mahan, J.D.; Trachtman, H.; Scheinman, J.; Flynn, J.T.; Alon, U.S.; Lande, M.B.; Weiss, R.A.; Norkus, E.P. Vitamin E Therapy in IgA Nephropathy: A Double-Blind, Placebo-Controlled Study. Pediatr. Nephrol. 2003, 18, 1015–1019. [Google Scholar] [CrossRef]
  81. Dos Santos, M.; Poletti, P.T.; Favero, G.; Stacchiotti, A.; Bonomini, F.; Montanari, C.C.; Bona, S.R.; Marroni, N.P.; Rezzani, R.; Veronese, F.V. Protective Effects of Quercetin Treatment in A Pristane-Induced Mouse Model of Lupus Nephritis. Autoimmunity 2018, 51, 69–80. [Google Scholar] [CrossRef] [PubMed]
  82. Morigi, M.; Perico, L.; Corna, D.; Locatelli, M.; Cassis, P.; Carminati, C.E.; Bolognini, S.; Zoja, C.; Remuzzi, G.; Benigni, A.; et al. C3a Receptor Blockade Protects Podocytes from Injury in Diabetic Nephropathy. JCI Insight 2020, 5, e131849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Cui, F.Q.; Tang, L.; Gao, Y.B.; Wang, Y.F.; Meng, Y.; Shen, C.; Shen, Z.L.; Liu, Z.Q.; Zhao, W.J.; Liu, W.J. Effect of Baoshenfang Formula on Podocyte Injury via Inhibiting the NOX-4/ROS/P38 Pathway in Diabetic Nephropathy. J. Diabetes Res. 2019, 2019, 2981705. [Google Scholar] [CrossRef]
  84. Chen, J.; Chen, J.K.; Harris, R.C. EGR Receptor Deletion in Podocytes Attenuates Diabetic Nephropathy. J. Am. Soc. Nephrol. 2015, 26, 1115–1125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Poirier, B.; Lannaud-Bournoville, M.; Conti, M.; Bazin, R.; Michel, O.; Bariety, J.; Chevalier, J.; Myara, I. Oxidative Stress Occurs in Absence of Hyperglycaemia and Inflammation in the Onset of Kidney Lesions in Normotensive Obese Rats. Nephrol. Dial. Transplant. 2000, 15, 467–476. [Google Scholar] [CrossRef] [Green Version]
  86. Shah, A.; Xia, L.; Masson, E.A.; Gui, C.; Momen, A.; Shikatani, E.A.; Husain, M.; Quaggin, S.; John, R.; Fantus, I.G. Thioredoxin-Interacting Protein Deficiency Protects Against Diabetic Nephropathy. J. Am. Soc. Nephrol. 2015, 26, 2963–2977. [Google Scholar] [CrossRef]
  87. Wood, Z.A.; Poole, L.B.; Karplus, P.A. Peroxiredoxin Evolution and the Regulation of Hydrogen Peroxide Signaling. Science 2003, 300, 650–653. [Google Scholar] [CrossRef]
  88. Hsu, H.H.; Hoffmann, S.; Di Marco, G.S.; Endlich, N.; Peter-Katalinic, J.; Weide, T.; Pavenstadt, H. Downregulation of the Antioxidant Protein Peroxiredoxin 2 Contributes to Angiotensin II-Mediated Podocyte Apoptosis. Kidney Int. 2011, 80, 959–969. [Google Scholar] [CrossRef] [Green Version]
  89. Liu, X.; Liu, Y.; Yang, Y.; Xu, J.; Dai, D.; Yan, C.; Li, X.; Tang, R.; Yu, C.; Ren, H. Antioxidative Stress Effects of Salvia Przewalskii Extract in Experimentally Injured Podocytes. Nephron 2016, 134, 253–271. [Google Scholar] [CrossRef]
  90. Zheng, S.; Carlson, E.C.; Yang, L.; Kralik, P.M.; Huang, Y.; Epstein, P.N. Podocyte-Specific Overexpression of the Antioxidant Metallothionein Reduces Diabetic Nephropathy. J. Am. Soc. Nephrol. 2008, 19, 2077–2085. [Google Scholar] [CrossRef] [Green Version]
  91. Bergsbaken, T.; Fink, S.L.; Cookson, B.T. Pyroptosis: Host Cell Death and Inflammation. Nat. Rev. Microbiol. 2009, 7, 99–109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Orrenius, S.; Zhivotovsky, B.; Nicotera, P. Regulation of Cell Death: The Calcium-Apoptosis Link. Nat. Rev. Mol. Cell Biol. 2003, 4, 552–565. [Google Scholar] [CrossRef] [PubMed]
  93. Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An Iron-Dependent Form of Nonapoptotic Cell Death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Tharaux, P.L.; Huber, T.B. How Many Ways Can A Podocyte Die? Semin. Nephrol. 2012, 32, 394–404. [Google Scholar] [CrossRef] [Green Version]
  95. Kim, S.Y.; Park, S.; Lee, S.W.; Lee, J.H.; Lee, E.S.; Kim, M.; Kim, Y.; Kang, J.S.; Chung, C.H.; Moon, J.S.; et al. RIPK3 Contributes to Lyso-Gb3-Induced Podocyte Death. Cells 2021, 10, 245. [Google Scholar] [CrossRef]
  96. Meyer, T.W.; Bennett, P.H.; Nelson, R.G. Podocyte Number Predicts Long-Term Urinary Albumin Excretion in Pima Indians with Type II Diabetes and Microalbuminuria. Diabetologia 1999, 42, 1341–1344. [Google Scholar] [CrossRef] [Green Version]
  97. Nagata, S. Apoptosis and Clearance of Apoptotic Cells. Annu. Rev. Immunol. 2018, 36, 489–517. [Google Scholar] [CrossRef]
  98. Czabotar, P.E.; Lessene, G.; Strasser, A.; Adams, J.M. Control of Apoptosis by the Bcl-2 Protein Family: Implications for Physiology and Therapy. Nat. Rev. Mol. Cell Biol. 2014, 15, 49–63. [Google Scholar] [CrossRef]
  99. Dickens, L.S.; Powley, I.R.; Hughes, M.A.; MacFarlane, M. The ‘Complexities’ of Life and Death: Death Receptor Signalling Platforms. Experimental. Cell Res. 2012, 318, 1269–1277. [Google Scholar] [CrossRef] [Green Version]
  100. Xiang, X.Y.; Liu, T.; Wu, Y.; Jiang, X.S.; He, J.L.; Chen, X.M.; Du, X.G. Berberine Alleviates Palmitic Acid-Induced Podocyte Apoptosis by Reducing Reactive Oxygen Species-Mediated Endoplasmic Reticulum Stress. Mol. Med. Rep. 2021, 23. [Google Scholar] [CrossRef]
  101. Chuang, P.Y.; Yu, Q.; Fang, W.; Uribarri, J.; He, J.C. Advanced Glycation Endproducts Induce Podocyte Apoptosis by Activation of the FOXO4 Transcription Factor. Kidney Int. 2007, 72, 965–976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Liu, Y.; Hitomi, H.; Diah, S.; Deguchi, K.; Mori, H.; Masaki, T.; Nakano, D.; Kobori, H.; Nishiyama, A. Roles of Na(+)/H(+) Exchanger Type 1 and Intracellular pH in Angiotensin II-Induced Reactive Oxygen Species Generation and Podocyte Apoptosis. J. Pharmacol. Sci. 2013, 122, 176–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Qiao, C.; Ye, W.; Li, S.; Wang, H.; Ding, X. Icariin Modulates Mitochondrial Function and Apoptosis in High Glucose-Induced Glomerular Podocytes Through G Protein-Coupled Estrogen Receptors. Mol. Cell Endocrinol. 2018, 473, 146–155. [Google Scholar] [CrossRef] [PubMed]
  104. Chen, X.; Liu, W.; Xiao, J.; Zhang, Y.; Chen, Y.; Luo, C.; Huang, Q.; Peng, F.; Gong, W.; Li, S.; et al. FOXO3a Accumulation and Activation Accelerate Oxidative Stress-Induced Podocyte Injury. FASEB J. 2020, 34, 13300–13316. [Google Scholar] [CrossRef] [PubMed]
  105. Arthur, J.S.; Ley, S.C. Mitogen-Activated Protein Kinases in Innate Immunity. Nat. Rev. Immunol. 2013, 13, 679–692. [Google Scholar] [CrossRef] [PubMed]
  106. Cui, F.; Gao, Y.; Zhao, W.; Zou, D.; Zhu, Z.; Wu, X.; Tian, N.; Wang, X.; Liu, J.; Tong, Y. Effect of Tongxinluo on Podocyte Apoptosis via Inhibition of Oxidative Stress and P38 Pathway in Diabetic Rats. Evid. Based Complement. Altern. Med. 2016, 2016, 5957423. [Google Scholar] [CrossRef] [Green Version]
  107. Cantley, L.C. The Phosphoinositide 3-Kinase Pathway. Science 2002, 296, 1655–1657. [Google Scholar] [CrossRef]
  108. Cui, F.Q.; Wang, Y.F.; Gao, Y.B.; Meng, Y.; Cai, Z.; Shen, C.; Liu, Z.Q.; Jiang, X.C.; Zhao, W.J. Effects of BSF on Podocyte Apoptosis via Regulating the Ros-Mediated PI3K/AKT Pathway in DN. J. Diabetes Res. 2019, 2019, 9512406. [Google Scholar] [CrossRef]
  109. Vousden, K.H.; Prives, C. Blinded by the Light: The Growing Complexity of P53. Cell 2009, 137, 413–431. [Google Scholar] [CrossRef] [Green Version]
  110. Eid, A.A.; Ford, B.M.; Block, K.; Kasinath, B.S.; Gorin, Y.; Ghosh-Choudhury, G.; Barnes, J.L.; Abboud, H.E. AMP-Activated Protein Kinase (AMPK) Negatively Regulates Nox4-Dependent Activation of P53 and Epithelial Cell Apoptosis in Diabetes. J. Biol. Chem. 2010, 285, 37503–37512. [Google Scholar] [CrossRef] [Green Version]
  111. Li, Z.; Deng, W.; Cao, A.; Zang, Y.; Wang, Y.; Wang, H.; Wang, L.; Peng, W. Huangqi Decoction Inhibits Hyperglycemia-Induced Podocyte Apoptosis by Down-Regulated Nox4/P53/Bax Signaling In Vitro and In Vivo. Am. J. Transl. Res. 2019, 11, 3195–3212. [Google Scholar] [PubMed]
  112. Zhou, L.L.; Cao, W.; Xie, C.; Tian, J.; Zhou, Z.; Zhou, Q.; Zhu, P.; Li, A.; Liu, Y.; Miyata, T.; et al. The Receptor of Advanced Glycation End Products Plays A Central Role in Advanced Oxidation Protein Products-Induced Podocyte Apoptosis. Kidney Int. 2012, 82, 759–770. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Oakes, S.A.; Papa, F.R. The Role of Endoplasmic Reticulum Stress in Human Pathology. Annu. Rev. Pathol. 2015, 10, 173–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Susin, S.A.; Lorenzo, H.K.; Zamzami, N.; Marzo, I.; Snow, B.E.; Brothers, G.M.; Mangion, J.; Jacotot, E.; Costantini, P.; Loeffler, M.; et al. Molecular Characterization of Mitochondrial Apoptosis-Inducing Factor. Nature 1999, 397, 441–446. [Google Scholar] [CrossRef] [PubMed]
  115. Tang, D.; Chen, X.; Kang, R.; Kroemer, G. Ferroptosis: Molecular Mechanisms and Health Implications. Cell Res. 2020, 32, 107–125. [Google Scholar] [CrossRef]
  116. Guo, C.; Fu, R.; Zhou, M.; Wang, S.; Huang, Y.; Hu, H.; Zhao, J.; Gaskin, F.; Yang, N.; Fu, S.M. Pathogenesis of Lupus Nephritis: RIP3 Dependent Necroptosis and NLRP3 Inflammasome Activation. J. Autoimmun. 2019, 103, 102286. [Google Scholar] [CrossRef]
  117. Xu, Y.; Gao, H.; Hu, Y.; Fang, Y.; Qi, C.; Huang, J.; Cai, X.; Wu, H.; Ding, X.; Zhang, Z. High Glucose-Induced Apoptosis and Necroptosis in Podocytes Is Regulated by UCHL1 via RIPK1/RIPK3 Pathway. Exp. Cell Res. 2019, 382, 111463. [Google Scholar] [CrossRef]
  118. Lan, X.; Jhaveri, A.; Cheng, K.; Wen, H.; Saleem, M.A.; Mathieson, P.W.; Mikulak, J.; Aviram, S.; Malhotra, A.; Skorecki, K.; et al. APOL1 Risk Variants Enhance Podocyte Necrosis through Compromising Lysosomal Membrane Permeability. Am. J. Physiol. Renal. Physiol. 2014, 307, F326–F336. [Google Scholar] [CrossRef] [Green Version]
  119. Pak, O.; Scheibe, S.; Esfandiary, A.; Gierhardt, M.; Sydykov, A.; Logan, A.; Fysikopoulos, A.; Veit, F.; Hecker, M.; Kroschel, F.; et al. Impact of the Mitochondria-Targeted Antioxidant MitoQ on Hypoxia-Induced Pulmonary Hypertension. Eur. Respir. J. 2018, 51, 1701024. [Google Scholar] [CrossRef]
  120. Dare, A.J.; Bolton, E.A.; Pettigrew, G.J.; Bradley, J.A.; Saeb-Parsy, K.; Murphy, M.P. Protection Against Renal Ischemia-Reperfusion Injury in Vivo by the Mitochondria Targeted Antioxidant MitoQ. Redox. Biol. 2015, 5, 163–168. [Google Scholar] [CrossRef] [Green Version]
  121. Zhu, Z.; Liang, W.; Chen, Z.; Hu, J.; Feng, J.; Cao, Y.; Ma, Y.; Ding, G. Mitoquinone Protects Podocytes from Angiotensin II-Induced Mitochondrial Dysfunction and Injury via the Keap1-Nrf2 Signaling Pathway. Oxid. Med. Cell Longev. 2021, 2021, 1394486. [Google Scholar] [CrossRef] [PubMed]
  122. Azuma, J.; Sawamura, A.; Awata, N. Usefulness of Taurine in Chronic Congestive Heart Failure and Its Prospective Application. Jpn. Circ. J. 1992, 56, 95–99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Katakawa, M.; Fukuda, N.; Tsunemi, A.; Mori, M.; Maruyama, T.; Matsumoto, T.; Abe, M.; Yamori, Y. Taurine and Magnesium Supplementation Enhances the Function of Endothelial Progenitor Cells Through Antioxidation in Healthy Men and Spontaneously Hypertensive Rats. Hypertens. Res. 2016, 39, 848–856. [Google Scholar] [CrossRef] [PubMed]
  124. Nakaya, Y.; Minami, A.; Harada, N.; Sakamoto, S.; Niwa, Y.; Ohnaka, M. Taurine Improves Insulin Sensitivity in the Otsuka Long-Evans Tokushima Fatty Rat, A Model of Spontaneous Type 2 Diabetes. Am. J. Clin. Nutr. 2000, 71, 54–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Schaffer, S.; Kim, H.W. Effects and Mechanisms of Taurine as A Therapeutic Agent. Biomol. Ther. 2018, 26, 225–241. [Google Scholar] [CrossRef]
  126. Stacchiotti, A.; Favero, G.; Lavazza, A.; Monsalve, M.; Rodella, L.F.; Rezzani, R. Taurine Supplementation Alleviates Puromycin Aminonucleoside Damage by Modulating Endoplasmic Reticulum Stress and Mitochondrial-Related Apoptosis in Rat Kidney. Nutrients 2018, 10, 689. [Google Scholar] [CrossRef] [Green Version]
  127. Zhang, R.; Wang, X.; Gao, Q.; Jiang, H.; Zhang, S.; Lu, M.; Liu, F.; Xue, X. Taurine Supplementation Reverses Diabetes-Induced Podocytes Injury via Modulation of the Cse/Trpc6 Axis and Improvement of Mitochondrial Function. Nephron 2020, 144, 84–95. [Google Scholar] [CrossRef]
  128. Ha, H.; Yu, M.R.; Kim, K.H. Melatonin and Taurine Reduce Early Glomerulopathy in Diabetic Rats. Free Radic. Biol. Med. 1999, 26, 944–950. [Google Scholar] [CrossRef]
  129. Oktem, F.; Ozguner, F.; Yilmaz, H.R.; Uz, E.; Dundar, B. Melatonin Reduces Urinary Excretion of N-Acetyl-Beta-D-Glucosaminidase, Albumin and Renal Oxidative Markers in Diabetic Rats. Clin. Exp. Pharmacol. Physiol. 2006, 33, 95–101. [Google Scholar] [CrossRef]
  130. Ji, Z.Z.; Xu, Y.C. Melatonin Protects Podocytes from Angiotensin II-Induced Injury in An in Vitro Diabetic Nephropathy Model. Mol. Med. Rep. 2016, 14, 920–926. [Google Scholar] [CrossRef] [Green Version]
  131. Romecin, P.; Atucha, N.M.; Navarro, E.G.; Ortiz, M.C.; Iyu, D.; Rosado, J.A.; Garcia-Estan, J. Role of Homocysteine and Folic Acid on the Altered Calcium Homeostasis of Platelets from Rats with Biliary Cirrhosis. Platelets 2017, 28, 698–705. [Google Scholar] [CrossRef] [PubMed]
  132. Wu, Q.; Li, W.; Zhao, J.; Sun, W.; Yang, Q.; Chen, C.; Xia, P.; Zhu, J.; Zhou, Y.; Huang, G.; et al. Apigenin Ameliorates Doxorubicin-Induced Renal Injury via Inhibition of Oxidative Stress and Inflammation. Biomed Pharm. 2021, 137, 111308. [Google Scholar] [CrossRef] [PubMed]
  133. Li, F.; Chen, Y.; Li, Y.; Huang, M.; Zhao, W. Geniposide Alleviates Diabetic Nephropathy of Mice through AMPK/SIRT1/NF-kappaB Pathway. Eur. J. Pharmacol. 2020, 886, 173449. [Google Scholar] [CrossRef]
  134. Yang, S.M.; Hua, K.F.; Lin, Y.C.; Chen, A.; Chang, J.M.; Kuoping Chao, L.; Ho, C.L.; Ka, S.M. Citral Is Renoprotective for Focal Segmental Glomerulosclerosis by Inhibiting Oxidative Stress and Apoptosis and Activating Nrf2 Pathway in Mice. PLoS ONE 2013, 8, e74871. [Google Scholar] [CrossRef] [PubMed]
  135. Liu, G.; He, L. Epigallocatechin-3-Gallate Attenuates Adriamycin-Induced Focal Segmental Glomerulosclerosis via Suppression of Oxidant Stress and Apoptosis by Targeting Hypoxia-Inducible Factor-1alpha/ Angiopoietin-Like 4 Pathway. Pharmacology 2019, 103, 303–314. [Google Scholar] [CrossRef]
  136. Yang, S.-M.; Chan, Y.-L.; Hua, K.-F.; Chang, J.-M.; Chen, H.-L.; Tsai, Y.-J.; Hsu, Y.-J.; Chao, L.K.; Feng-Ling, Y.; Tsai, Y.-L.; et al. Osthole Improves An Accelerated Focal Segmental Glomerulosclerosis Model in the Early Stage by Activating the Nrf2 Antioxidant Pathway and Subsequently Inhibiting NF-kappaB-Mediated COX-2 Expression and Apoptosis. Free Radic. Biol. Med. 2014, 73, 260–269. [Google Scholar] [CrossRef]
  137. Liu, X.; Cao, W.; Qi, J.; Li, Q.; Zhao, M.; Chen, Z.; Zhu, J.; Huang, Z.; Wu, L.; Zhang, B.; et al. Leonurine Ameliorates Adriamycin-Induced Podocyte Injury via Suppression of Oxidative Stress. Free Radic Res. 2018, 52, 952–960. [Google Scholar] [CrossRef] [PubMed]
  138. Son, S.S.; Kang, J.S.; Lee, E.Y. Paclitaxel Ameliorates Palmitate-Induced Injury in Mouse Podocytes. Med. Sci. Monit. Basic Res. 2020, 26, e928265. [Google Scholar] [CrossRef]
  139. Wang, F.; Li, R.; Zhao, L.; Ma, S.; Qin, G. Resveratrol Ameliorates Renal Damage by Inhibiting Oxidative Stress-Mediated Apoptosis of Podocytes in Diabetic Nephropathy. Eur. J. Pharmacol. 2020, 885, 173387. [Google Scholar] [CrossRef]
  140. Zhang, T.; Chi, Y.; Ren, Y.; Du, C.; Shi, Y.; Li, Y. Resveratrol Reduces Oxidative Stress and Apoptosis in Podocytes via Sir2-Related Enzymes, Sirtuins1 (SIRT1)/Peroxisome Proliferator-Activated Receptor Gamma Co-Activator 1alpha (PGC-1alpha) Axis. Med. Sci. Monit. 2019, 25, 1220–1231. [Google Scholar] [CrossRef]
  141. Yang, K.; Bai, Y.; Yu, N.; Lu, B.; Han, G.; Yin, C.; Pang, Z. Huidouba Improved Podocyte Injury by Down-Regulating Nox4 Expression in Rats with Diabetic Nephropathy. Front. Pharmacol. 2020, 11, 587995. [Google Scholar] [CrossRef] [PubMed]
  142. Lu, H.; Li, Y.; Zhang, T.; Liu, M.; Chi, Y.; Liu, S.; Shi, Y. Salidroside Reduces High-Glucose-Induced Podocyte Apoptosis and Oxidative Stress via Upregulating Heme Oxygenase-1 (HO-1) Expression. Med. Sci. Monit. 2017, 23, 4067–4076. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Chen, Y.; Liu, Q.; Shan, Z.; Zhao, Y.; Li, M.; Wang, B.; Zheng, X.; Feng, W. The Protective Effect and Mechanism of Catalpol on High Glucose-Induced Podocyte Injury. BMC Complement. Altern. Med. 2019, 19, 244. [Google Scholar] [CrossRef] [PubMed]
  144. Chen, Y.; Chen, J.; Jiang, M.; Fu, Y.; Zhu, Y.; Jiao, N.; Liu, L.; Du, Q.; Wu, H.; Xu, H.; et al. Loganin and Catalpol Exert Cooperative Ameliorating Effects on Podocyte Apoptosis upon Diabetic Nephropathy by Targeting Ages-Rage Signaling. Life Sci. 2020, 252, 117653. [Google Scholar] [CrossRef]
  145. Gui, D.; Guo, Y.; Wang, F.; Liu, W.; Chen, J.; Chen, Y.; Huang, J.; Wang, N. Astragaloside IV, A Novel Antioxidant, Prevents Glucose-Induced Podocyte Apoptosis in Vitro and In Vivo. PLoS ONE 2012, 7, e39824. [Google Scholar] [CrossRef] [Green Version]
  146. Yu, Q.; Zhang, M.; Qian, L.; Wen, D.; Wu, G. Luteolin Attenuates High Glucose-Induced Podocyte Injury via Suppressing NLRP3 Inflammasome Pathway. Life Sci. 2019, 225, 1–7. [Google Scholar] [CrossRef] [PubMed]
  147. Kang, M.K.; Park, S.H.; Kim, Y.H.; Lee, E.J.; Antika, L.D.; Kim, D.Y.; Choi, Y.J.; Kang, Y.H. Chrysin Ameliorates Podocyte Injury and Slit Diaphragm Protein Loss via Inhibition of the PERK-eIF2alpha-ATF-CHOP Pathway in Diabetic Mice. Acta Pharmacol. Sin. 2017, 38, 1129–1140. [Google Scholar] [CrossRef]
  148. Zhao, K.; Li, Y.; Wang, Z.; Han, N.; Wang, Y. Carnosine Protects Mouse Podocytes from High Glucose Induced Apoptosis Through PI3K/AKT and Nrf2 Pathways. Biomed. Res. Int. 2019, 2019, 4348973. [Google Scholar] [CrossRef]
  149. Riedl, E.; Pfister, F.; Braunagel, M.; Brinkkötter, P.; Sternik, P.; Deinzer, M.; Bakker, S.J.; Henning, R.H.; Born, J.V.D.; Krämer, B.K.; et al. Carnosine Prevents Apoptosis of Glomerular Cells and Podocyte Loss in STZ Diabetic Rats. Cell Physiol. Biochem. 2011, 28, 279–288. [Google Scholar] [CrossRef]
  150. Quan, X.; Liu, H.; Ye, D.; Ding, X.; Su, X. Forsythoside A Alleviates High Glucose-Induced Oxidative Stress and Inflammation in Podocytes by Inactivating MAPK Signaling via MMP12 Inhibition. Diabetes Metab. Syndr. Obes. 2021, 14, 1885–1895. [Google Scholar] [CrossRef]
  151. He, J.Y.; Hong, Q.; Chen, B.X.; Cui, S.Y.; Liu, R.; Cai, G.Y.; Guo, J.; Chen, X.M. Ginsenoside Rb1 Alleviates Diabetic Kidney Podocyte Injury by Inhibiting Aldose Reductase Activity. Acta Pharmacol. Sin. 2022, 43, 342–353. [Google Scholar] [CrossRef] [PubMed]
  152. Cai, X.; Bao, L.; Ren, J.; Li, Y.; Zhang, Z. Grape Seed Procyanidin B2 Protects Podocytes from High Glucose-Induced Mitochondrial Dysfunction and Apoptosis via the AMPK-SIRT1-PGC-1alpha Axis In Vitro. Food Funct. 2016, 7, 805–815. [Google Scholar] [CrossRef] [PubMed]
  153. Borges, C.M.; Papadimitriou, A.; Duarte, D.A.; Lopes de Faria, J.M.; Lopes de Faria, J.B. The Use of Green Tea Polyphenols for Treating Residual Albuminuria in Diabetic Nephropathy: A Double-Blind Randomised Clinical Trial. Sci. Rep. 2016, 6, 28282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Lee, M.; Zhao, H.; Liu, X.; Liu, D.; Chen, J.; Li, Z.; Chu, S.; Kou, X.; Liao, S.; Deng, Y.; et al. Protective Effect of Hydroxysafflor Yellow A on Nephropathy by Attenuating Oxidative Stress and Inhibiting Apoptosis in Induced Type 2 Diabetes in Rat. Oxid. Med. Cell Longev. 2020, 2020, 7805393. [Google Scholar] [CrossRef] [PubMed]
  155. Li, T.X.; Mao, J.H.; Huang, L.; Fu, H.D.; Chen, S.H.; Liu, A.M.; Liang, Y.Q. Beneficial Effects of Huaiqihuang on Hyperglycemia-Induced MPC5 Podocyte Dysfunction through the Suppression of Mitochondrial Dysfunction and Endoplasmic Reticulum Stress. Mol. Med. Rep. 2017, 16, 1465–1471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Zhang, J.; Yang, S.; Li, H.; Chen, F.; Shi, J. Naringin Ameliorates Diabetic Nephropathy by Inhibiting NADPH Oxidase 4. Eur. J. Pharmacol. 2017, 804, 1–6. [Google Scholar] [CrossRef]
  157. Xue, W.; Mao, J.; Chen, Q.; Ling, W.; Sun, Y. Mogroside IIIE Alleviates High Glucose-Induced Inflammation, Oxidative Stress and Apoptosis of Podocytes by the Activation of AMPK/SIRT1 Signaling Pathway. Diabetes Metab. Syndr. Obes. 2020, 13, 3821–3830. [Google Scholar] [CrossRef]
  158. Sohn, E.; Kim, J.; Kim, C.S.; Jo, K.; Kim, J.S. Extract of Rhizoma Polygonum Cuspidatum Reduces Early Renal Podocyte Injury in Streptozotocin-Induced Diabetic Rats and Its Active Compound Emodin Inhibits Methylglyoxalmediated Glycation of Proteins. Mol. Med. Rep. 2015, 12, 5837–5845. [Google Scholar] [CrossRef] [Green Version]
  159. Li, J.; Wang, B.; Zhou, G.; Yan, X.; Zhang, Y. Tetrahydroxy Stilbene Glucoside Alleviates High Glucose-Induced MPC5 Podocytes Injury through Suppression of NLRP3 Inflammasome. Am. J. Med. Sci. 2018, 355, 588–596. [Google Scholar] [CrossRef]
  160. ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ (accessed on 23 February 2022).
Figure 1. Oxidative stress plays a significant role in the pathogenesis of podocytopathies. Various harmful stimuli, such as puromycin aminonucleoside, immune complexes, HG, and Ang II, upregulate NOX, Cyt C, and P450, and downregulate mitochondrial respiration chain complexes (complex I, II, III, and IV), Ub, and antioxidant defense systems, including SOD, GPX, CAT, GSH, Prdx, and Trx. Excessive ROS accumulation in the podocyte causes damage to DNA, lipids, and proteins, and activates downstream signaling pathways, leading to podocyte foot process effacement, loss, and detachment, with clinical presentations of proteinuria and proteinuria-related symptoms. HG, high glucose; Ang II, angiotensin II; AT1R, angiotensin II type 1 receptor; AOPPs, advanced oxidation protein products; AGEs, advanced glycation end-products; RAGE, receptor of advanced glycation end-products; MC, mineralocorticoid; MR, mineralocorticoid receptor; C5b-9, C5b-9 membrane attack complex; NOX, NADPH oxidase; I, complex I; II, complex II; III, complex III; IV, complex IV; Ub, quinone; Cyt c, cytochrome c; SOD, superoxide dismutase; GPX, glutathione peroxidase; CAT, catalase; GSH, glutathione; Prdx, peroxiredoxin; Trx, thioredoxin; P450, cytochrome P450; and ROS, reactive oxygen species.
Figure 1. Oxidative stress plays a significant role in the pathogenesis of podocytopathies. Various harmful stimuli, such as puromycin aminonucleoside, immune complexes, HG, and Ang II, upregulate NOX, Cyt C, and P450, and downregulate mitochondrial respiration chain complexes (complex I, II, III, and IV), Ub, and antioxidant defense systems, including SOD, GPX, CAT, GSH, Prdx, and Trx. Excessive ROS accumulation in the podocyte causes damage to DNA, lipids, and proteins, and activates downstream signaling pathways, leading to podocyte foot process effacement, loss, and detachment, with clinical presentations of proteinuria and proteinuria-related symptoms. HG, high glucose; Ang II, angiotensin II; AT1R, angiotensin II type 1 receptor; AOPPs, advanced oxidation protein products; AGEs, advanced glycation end-products; RAGE, receptor of advanced glycation end-products; MC, mineralocorticoid; MR, mineralocorticoid receptor; C5b-9, C5b-9 membrane attack complex; NOX, NADPH oxidase; I, complex I; II, complex II; III, complex III; IV, complex IV; Ub, quinone; Cyt c, cytochrome c; SOD, superoxide dismutase; GPX, glutathione peroxidase; CAT, catalase; GSH, glutathione; Prdx, peroxiredoxin; Trx, thioredoxin; P450, cytochrome P450; and ROS, reactive oxygen species.
Biomolecules 12 00403 g001
Figure 2. Molecular mechanisms for oxidative stress-triggered podocyte apoptosis. Oxidative stress upregulates FasL to activate the CASP8-mediated extrinsic apoptosis pathway. As for intrinsic apoptosis, p53, PI3K/Akt, and P38 MAPK pathways are activated by oxidative stress, and the expression levels of BAX, PUMA, BIM, and Cyt C are elevated, whereas anti-apoptotic BCL2 is reduced. Whereafter, CASP9 is activated to cleave pro-CASP3 to activate CASP3, which ultimately cleaves cellular components and leads to apoptosis. ER stress is also activated, UPR triggers CHOP- and CASP12-dependent apoptosis, and CHOP activation downregulates BCL2, indicating that intrinsic apoptosis is involved. Moreover, oxidative stress activates CASP-independent apoptosis via upregulating AIF. FasL, Fas ligand; CASP, caspase; Cyt C, cytochrome c; BCL2, B-cell lymphoma-2; BIM, BCL2-interacting mediator of cell death; PUMA, p53-upregulated modulator of apoptosis; BAX, BCL2-associated X; ER, endoplasmic reticulum; UPR, unfolded protein response; CHOP, CCAAT/enhancer-binding protein-homologous protein; and AIF, apoptosis-inducing factor.
Figure 2. Molecular mechanisms for oxidative stress-triggered podocyte apoptosis. Oxidative stress upregulates FasL to activate the CASP8-mediated extrinsic apoptosis pathway. As for intrinsic apoptosis, p53, PI3K/Akt, and P38 MAPK pathways are activated by oxidative stress, and the expression levels of BAX, PUMA, BIM, and Cyt C are elevated, whereas anti-apoptotic BCL2 is reduced. Whereafter, CASP9 is activated to cleave pro-CASP3 to activate CASP3, which ultimately cleaves cellular components and leads to apoptosis. ER stress is also activated, UPR triggers CHOP- and CASP12-dependent apoptosis, and CHOP activation downregulates BCL2, indicating that intrinsic apoptosis is involved. Moreover, oxidative stress activates CASP-independent apoptosis via upregulating AIF. FasL, Fas ligand; CASP, caspase; Cyt C, cytochrome c; BCL2, B-cell lymphoma-2; BIM, BCL2-interacting mediator of cell death; PUMA, p53-upregulated modulator of apoptosis; BAX, BCL2-associated X; ER, endoplasmic reticulum; UPR, unfolded protein response; CHOP, CCAAT/enhancer-binding protein-homologous protein; and AIF, apoptosis-inducing factor.
Biomolecules 12 00403 g002
Table 1. A summary of antioxidative herbs and herb-derived components that can protect the podocyte from death.
Table 1. A summary of antioxidative herbs and herb-derived components that can protect the podocyte from death.
Herbs/ComponentsModels of PodocytopathiesAnti-Death MechanismsRefs.
Salvia przewalskiiPA-induced ratsAnti-apoptosis[89]
BerberinePalmitic acid-cultured podocytesAnti-apoptosis[100]
ApigeninAdriamycin-induced miceAnti-apoptosis[132]
CitralAdriamycin-induced ratsAnti-apoptosis[134]
Epigallocatechin-3-GallateAdriamycin-induced miceAnti-apoptosis[135]
OstholeAdriamycin-induced miceAnti-apoptosis[136]
LeonurineAdriamycin-induced miceAnti-apoptosis[137]
PaclitaxelPalmitate-cultured podocytesAnti-apoptosis[138]
QuercetinPristane-induced miceAnti-apoptosis[81]
Resveratroldb/db mice
STZ-induced mice
Anti-apoptosis[59,139,140]
BaoshenfangKK-Ay mice
STZ-induced rats
Anti-apoptosis[83,108]
IcariinHG-cultured podocytesAnti-apoptosis[103]
TongxinluoSTZ-induced ratsAnti-apoptosis[106]
HuangqiSTZ-induced miceAnti-apoptosis[111]
GeniposideHFD/STZ-induced miceAnti-pyroptosis[133]
HuidoubaUnilateral nephrectomy/STZ-induced ratsAnti-apoptosis[141]
SalidrosideHG-cultured podocytesAnti-apoptosis[142]
CatalpolKK-Ay/HFD induced mice
HG-cultured podocytes
AGEs-cultured podocytes
Anti-apoptosis[143,144]
LoganinKK-Ay/HFD mice
AGEs-cultured podocytes
Anti-apoptosis[144]
Astragaloside IVSTZ-induced ratsAnti-apoptosis[145]
LuteolinHG-cultured podocytesAnti-apoptosis[146]
Chrysindb/db miceAnti-apoptosis[147]
CarnosineSTZ-induced rats
HG-cultured podocytes
Anti-apoptosis[148,149]
Forsythoside AHG-cultured podocytesAnti-apoptosis[150]
Ginsenoside Rb1STZ-induced miceAnti-apoptosis[151]
Grape seed procyanidin B2HG-treated podocytesAnti-apoptosis[152]
Green tea polyphenolsDiabetic patients’ plasma-cultured podocytesAnti-apoptosis[153]
Hydroxysafflor yellow ASTZ-induced ratsAnti-apoptosis[154]
HuaiqihuangHG-treated podocytesAnti-apoptosis[155]
NaringinSTZ-induced ratsAnti-apoptosis[156]
Mogroside IIIEHG-treated podocytesAnti-apoptosis[157]
Rhizoma Polygonum cuspidatumSTZ-induced ratsAnti-apoptosis[158]
Tetrahydroxy stilbene glucosideHG-treated podocytesAnti-apoptosis[159]
Abbreviations: PA, puromycin aminonucleoside; STZ, streptozotocin; HG, high glucose; HFD, high-fat diet; and AGEs, advanced glycation end-products.
Table 2. A summary of the therapeutic targets of antioxidants that can protect the podocyte from death.
Table 2. A summary of the therapeutic targets of antioxidants that can protect the podocyte from death.
Therapeutic TargetsAntioxidants
Mitochondrial respiration chainComplex IResveratrol [59,139,140]
Complex IIIResveratrol [59,139,140]
Complex IVChrysin [147]; Forsythoside A [150]
Cyt CResveratrol [59,139,140]; Carnosine [149]
QuinoneMitoQ [121]
NADPH oxidaseFolic Acid [29]; Resveratrol [59,139,140]; Baoshenfang [83,108]; Huangqi [111]; Epigallocatechin-3-Gallate [135]; Paclitaxel [138]; Tongxinluo [106]; Huidouba [141]; Catalpol [143,144]; Loganin [144]; Forsythoside A [150]; Ginsenoside Rb1 [151]; Naringin [156]
Antioxidant defense systemsSODMelatonin [129]; Folic Acid [29]; Resveratrol [59,139,140]; Quercetin [81]; Apigenin [132]; Leonurine [137]; Paclitaxel [138]; Geniposide [133]; Catalpol [143,144]; Loganin [144]; Astragaloside IV [145]; Forsythoside A [150]; Grape Seed Procyanidin B2 [152]; Hydroxysafflor Yellow A [154]; Mogroside IIIE [157]
CATQuercetin [81]; Astragaloside IV [145]; Forsythoside A [150]; Mogroside IIIE [157]
GPXOsthole [136]; Geniposide [133]; Grape Seed Procyanidin B2 [152]; Naringin [156]
GSHApigenin [132]
Abbreviations: Complex I, NADH dehydrogenase; Complex III, ubiquinol-cytochrome c reductase; Complex IV, cytochrome c oxidase; Cyt C, cytochrome c; SOD, superoxide dismutase; CAT, catalase; GPX, glutathione peroxidase; and GSH, glutathione.
Table 3. A summary of drugs targeting oxidative stress in clinical trials.
Table 3. A summary of drugs targeting oxidative stress in clinical trials.
Therapeutic TargetsDrugs in Clinical Trials
Mitochondrial respiration chainComplex IVS-equol (NCT02142777), Resveratrol (NCT02123121), etc.
QuinoneMitoQ (NCT02364648), Ubiquinol (NCT02847585), Coenzyme Q10 (NCT01163500), etc.
NADPH oxidaseGKT137831 (NCT02010242, NCT03865927), Pioglitazone (NCT03060772), Januvia (NCT00659711), ImmunAge (NCT02332993), Threalose plus polyphenols (NCT04061070), Apocynin (NCT03680404), etc.
Antioxidant defense systemsSODGC4711 (NCT03762031), PC-SOD (NCT03995732), APN201 (NCT01513278), rhSOD (NCT00264186), Glisodin (NCT03878433), Melatonin (NCT02463318), etc.
CATMelatonin (NCT02463318), Oligopin (NCT03260803), etc.
GPXN-acetylcysteine (NCT00493727), Curcumin (NCT03475017), Rutin (NCT04955145), Melatonin (NCT01858909), etc.
GSHGlutathione (NCT02948673, NCT02948673), etc.
Other antioxidantsVitamin E (NCT00384618), Vitamin C (NCT04210453), Vitamin D3 (NCT03931889), L-carnitine (NCT01819701), etc.
Abbreviations: Complex IV, cytochrome c oxidase; SOD, superoxide dismutase; CAT, catalase; GPX, glutathione peroxidase; and GSH, glutathione.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zhu, Y.-T.; Wan, C.; Lin, J.-H.; Hammes, H.-P.; Zhang, C. Mitochondrial Oxidative Stress and Cell Death in Podocytopathies. Biomolecules 2022, 12, 403. https://doi.org/10.3390/biom12030403

AMA Style

Zhu Y-T, Wan C, Lin J-H, Hammes H-P, Zhang C. Mitochondrial Oxidative Stress and Cell Death in Podocytopathies. Biomolecules. 2022; 12(3):403. https://doi.org/10.3390/biom12030403

Chicago/Turabian Style

Zhu, Yu-Ting, Cheng Wan, Ji-Hong Lin, Hans-Peter Hammes, and Chun Zhang. 2022. "Mitochondrial Oxidative Stress and Cell Death in Podocytopathies" Biomolecules 12, no. 3: 403. https://doi.org/10.3390/biom12030403

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