Next Article in Journal
Uncovering the Genetic and Molecular Features of Huntington’s Disease in Northern Colombia
Next Article in Special Issue
Acute Kidney Injury and BK Polyomavirus in Urine Sediment Cells
Previous Article in Journal
Synergic Action of Systemic Risedronate and Local Rutherpy in Peri-implantar Repair of Ovariectomized Rats: Biomechanical and Molecular Analysis
Previous Article in Special Issue
Metalloporphyrins Reduce Proteinuria in Podocyte Immune Injury: The Role of Metal and Porphyrin Moieties
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The AKI-to-CKD Transition: The Role of Uremic Toxins

1
Department of Clinical Pharmacology, Amiens Medical Center, 80000 Amiens, France
2
GRAP Laboratory, INSERM UMR 1247, University of Picardy Jules Verne, 80000 Amiens, France
3
MP3CV Laboratory, UR UPJV 7517, University of Picardy Jules Verne, 80000 Amiens, France
4
Department of Clinical Biochemistry, Amiens Medical Center, 80000 Amiens, France
5
Department of Nephrology, Dialysis and Transplantation, Amiens Medical Center, 80000 Amiens, France
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(22), 16152; https://doi.org/10.3390/ijms242216152
Submission received: 14 September 2023 / Revised: 31 October 2023 / Accepted: 6 November 2023 / Published: 10 November 2023
(This article belongs to the Special Issue Together and Apart: Acute Kidney Injury and Chronic Kidney Disease)

Abstract

:
After acute kidney injury (AKI), renal function continues to deteriorate in some patients. In a pro-inflammatory and profibrotic environment, the proximal tubules are subject to maladaptive repair. In the AKI-to-CKD transition, impaired recovery from AKI reduces tubular and glomerular filtration and leads to chronic kidney disease (CKD). Reduced kidney secretion capacity is characterized by the plasma accumulation of biologically active molecules, referred to as uremic toxins (UTs). These toxins have a role in the development of neurological, cardiovascular, bone, and renal complications of CKD. However, UTs might also cause CKD as well as be the consequence. Recent studies have shown that these molecules accumulate early in AKI and contribute to the establishment of this pro-inflammatory and profibrotic environment in the kidney. The objective of the present work was to review the mechanisms of UT toxicity that potentially contribute to the AKI-to-CKD transition in each renal compartment.

1. Introduction

Acute kidney injury (AKI) and chronic kidney disease (CKD) are increasingly more frequent and are therefore becoming major public health concerns. In a recent study, the incidence rate for AKI and acute kidney disease (AKD) (standardized by age and sex) ranged from 134 to 162 events/10,000 person-years [1]. CKD affects around one person in ten [2] and mostly results from two cardiovascular disease risk factors: arterial hypertension and diabetes. CKD and AKI are interconnected, and each is a risk factor for the other [3,4]. Patients who experience an episode of AKI—even a nonserious one—have a ~25% greater risk of progression to CKD [5]. Both CKD and AKI are associated with higher morbidity and mortality rates in general and cardiovascular morbidity and mortality rates in particular [6,7,8]. CKD is known to accelerate the arterial and valvular calcification process and promote cardiovascular events, such as acute coronary syndrome and stroke [6,9]. A number of recent studies have demonstrated the link between AKI, cardiovascular events, and mortality independent of CKD [10,11]. Patients are more likely to develop acute heart failure and hypertension in the years following an episode of AKI [8,10,11,12,13]. Given that both CKD and AKI are associated with a higher mortality rate, it is important to limit their occurrence and, notably, to intervene early with regard to the AKI-to-CKD transition.
With a view to better understanding the risk of CKD progression after AKI and to identify potential new therapeutic targets, several research groups have explored the underlying cellular and molecular mechanisms [14]. These mechanisms involve all the compartments of the kidney (i.e., tubular, interstitial, vascular, and glomerular components) and a variety of pathways [14]. It is also thought that various patient-related and AKI-related factors (including uremic toxins (UTs)) influence the AKI-to-CKD transition [15,16]. Indeed, UTs (particularly indoxyl sulfate (IS) and para-cresyl sulfate (pCS)) are known to accumulate in the serum during CKD and AKI and are associated with CKD-related elevation in cardiovascular risk [17,18,19]. The objective of the present study was to review (i) the mechanisms involved in the AKI-to-CKD transition and (ii) UTs’ potential role in this transition. Figure 1 provides a schematic view of the potential impact of UTs on the AKI-to-CKD transition.

2. Acute and Chronic Kidney Diseases and Uremic Toxins: Definitions

2.1. AKI as a Risk Factor for CKD

2.1.1. Definition and Epidemiology of AKI

AKI is defined as a sudden loss of kidney function, as characterized by an elevation in the serum creatinine level and/or a decrease in urine output. These two criteria are used to classify AKI as being mild, moderate, or severe, according to the Kidney Disease Improving Global Outcomes classification [20]. The incidence of AKI is increasing, and the condition is frequently encountered in hospitals in general and in intensive care units (ICUs) in particular [21,22]. More than 50% of ICU patients experience an episode of AKI [23,24]. AKI is associated with a high mortality rate; this can be as high as 50% in patients who require dialysis [23,25]. There are several risk factors for AKI, including patient-related factors, such as older age, pre-existing chronic organ failure (especially kidney, heart, or liver failure), cancer or auto-immune disease, and AKI etiology [26,27,28,29]. Pre-existing CKD is the most significant risk factor for AKI; it increases the risk more than 10-fold [30].
Except in certain specific cases of glomerulus injury, the main histological findings of AKI are acute tubular lesions and interstitial infiltrates. The tubule is very sensitive to injury by ischemia (induced mainly by septic or cardiogenic shock) and drug toxicity [31,32,33]. The pathogenesis of acute tubular injury is a step-wise process [34,35]. The first phase of acute tubular injury is characterized by tubular epithelial cell damage, tubular luminal dilatation, proximal loss of the brush border, and loss of nuclei. Next, ischemic damage, endothelial and tubular epithelial cell necrosis, and apoptosis are observed. Tubular cell detachment can also obstruct the tubular lumen. In the third step, the release of pro-inflammatory molecules worsens the existing damage. The last steps are marked by cell repair, cell regeneration, and the reestablishment of normal tubular function; this phase has a central role in the transition from AKI to recovery or CKD.

2.1.2. Changes in Kidney Function after an Episode of AKI

AKI is also a risk factor for CKD, and there is a continuum between the episode of AKI and the development of CKD. In addition to its severity, AKI is defined by its duration (less than 7 days) [36] and CKD by a persistent loss of kidney function over 3 months. Acute kidney disease corresponds to the AKI-to-CKD transition, i.e., between 7 days and 3 months post-onset. Histological analyses show that CKD damages all four compartments of the kidney [37,38]. Interstitial fibrosis is the most frequent early-onset histological change and is correlated with the progression of CKD [39]. Tubular atrophy is the second most frequent change observed in CKD and is also associated with the degree of renal impairment [40]. Several cellular mechanisms (myofibroblast activation, a tubular epithelial-to-mesenchymal transition (EMT), inflammatory molecule production, immune cell recruitment, and matrix expansion and remodeling) and pathways (Notch and Wnt) have been considered fibrosis markers and targets for slowing down the progression of CKD [41].
Various patient-related and AKI-related risk factors are associated with the course of CKD [15,16]. Thus, gender (men), older age, comorbidities (such as uncontrolled diabetes and hypertension and cardiovascular disease), sociodemographic disparities and genetic factors contribute to the development and progression of CKD after AKI [42,43,44,45,46,47]. Factors linked to AKI (the severity, etiology, and number of episodes) all contribute to the AKI-to-CKD progression [48,49,50]. Thus, the risk of end-stage renal disease increases with AKI severity; in a meta-analysis encompassing 2,017,437 patients, the hazard ratio for CKD was 2.7 in AKI stage 1, 3.4 in AKI stage 2, and 8.9 in AKI stage 3 [51,52,53]. Moreover, each episode of AKI doubles the likelihood of progression to CKD after AKI [54].
It is noteworthy that many of the risk factors for the AKI-to-CKD transition are also risk factors for AKI onset [55]. Based on those findings in general and histological kidney lesions observed during AKI in particular, some researchers have looked for potential biomarkers of CKD risk. In a longitudinal study of 656 patients, increases in the urine concentrations of kidney injury molecule-1 (KIM-1), monocyte chemoattractant protein-1 (MCP-1), and plasma tumor necrosis factor receptor (TNFR)-1 during the 12 months after AKI were associated with a two- to three-fold greater risk of developing CKD [56]. Furthermore, preclinical and clinical studies suggest that neutrophil gelatinase-associated lipocalin (NGAL) is a marker of non-recovery from AKI, whereas urine uromodulin is associated with a reduction in CKD risk [56,57]. The accumulation of metabolites (and especially uremic toxins) might also have a role in the AKI-to-CKD transition.

2.2. Uremic Toxins

During CKD, the kidneys’ reduced filtration capacity results in the retention of numerous metabolites called uremic toxins (UTs). A UT is defined as a molecule that accumulates in the blood and organs of CKD patients, where it exerts deleterious effects on biological functions [58]. According to this definition, some 150 molecules are considered to be UTs and are classified by the EUTOX (European Uremic Toxins Work Group) into three groups depending on their physico-chemical properties [59,60]: water-soluble UTs (molecular weight (MW) < 500 Dalton) with urea and trimethylamine-N-oxide (TMAO) (Figure 2); water-soluble UTs (MW > 500 Dalton), including peptides and small proteins (β2-microglobulin or parathyroid hormone); and protein-bound UTs (PBUTs), which are strongly bound to plasma proteins, preventing them from renal filtration elimination and therefore from hemodialysis, including indoxyl sulfate (IS), para-cresyl sulfate (pCS), and indole-3-acetic acid (IAA).
UT precursor compounds are derived from the metabolism of certain dietary amino acids by the gut microbiota (GM). They pass the intestinal barrier and reach the liver through portal circulation, where they are metabolized by hepatic enzymes (sulfotransferase family 1A member 1 (SULT1A1) and flavin-containing mono-oxygenase 3 (FMO3)) into UTs. Once the UTs reach the systemic compartment, protein-binding UTs (including IS and pCS) are strongly bound to plasma proteins (mainly on albumin’s Sudlow sites) [61]. Lastly, UTs are secreted by the kidney after being taken up by the basolateral organic anion transporters 1/3 (OAT1/OAT3) expressed by proximal tubular cells [62,63].
Protein-bound UT (PBUTs) accumulation was first described in CKD patients. Indeed, in CKD patients, plasma PBUT concentrations increase from 27 times for IAA to 68 times for IS compared to healthy patients [64]. The clinical symptoms caused by UT toxicity are referred to collectively as “uremic syndrome.” Several preclinical studies describe an impairment of cardiac, neuron, and kidney cell damage involving the activation of the aryl hydrocarbon receptor (AhR) and nuclear factor-kappa B (NF-κB), reactive oxygen species (ROS) production, and the mitogen-activated protein kinase (MAPK), extracellular signal-regulated kinase (ERK), and transforming growth factor β (TGF-β) signaling pathways in endothelial cells (ECs) [65,66], astrocytes [67], the blood–brain barrier [68], and podocytes [69]. This newly acquired pro-inflammatory phenotype leads to fibrosis and increased apoptosis, together with renal dysfunction [70], ventricular dysfunction [71], and a poor cognitive state [72,73]. Several clinical studies in CKD patients have shown that UTs promote cardiovascular morbimortality [18,74,75], poor post-stroke recovery [76,77], cognitive impairment [78], and a greater risk of fractures [79].
Plasma UT concentrations in AKI are lower than plasma UT concentrations in CKD [17,80,81]. However, studies of the accumulation kinetics of these UTs in AKI showed contradictory data. In contrast to CKD, some UTs do not accumulate (p-cresyl glucuronide (pCG) and 3-carboxy-4-methyl-5-propyl-2-furanpropionate (CMPF)) [17,80,82]. In vitro and ex vivo studies have already observed UTs’ (IS, pCS, p-cresol) deleterious effects (oxidative stress, apoptosis, wound repair) after short exposure (30 min–1 h) in human umbilical vein endothelial cells (HUVECs) and vascular smooth muscle cells (VSMCs) [83,84,85]. Although several clinical studies shift towards a link between transient accumulation of PBUTs and cardiovascular complications during AKI [86], the role of PBUTs in the progression of renal damage remains to be determined, and several mechanisms directly implicate these molecules in the AKI-to-CKD transition.

3. Uremic Toxins in the AKI-to-CKD Transition

3.1. Gut Microbiota Dysbiosis and AKI-to-CKD Transition

The GM plays a central role in the genesis of UTs, is directly linked to AKI and CKD [87], and is located at the center of the bidirectional intestine–kidney axis [88,89]. Both AKI and CKD are responsible for alterations in the GM. In the case of CKD, several factors (a low fiber intake or specific treatment with phosphate binders and iron supplements) are responsible for this dysbiosis favoring the accumulation of UTs [90,91]. In addition to these factors, AKI is specifically associated with reduced levels of short-chain fatty acids produced by commensal bacteria in the microbiota [92]. This phenomenon is linked to systemic inflammation, which also promotes the formation of UTs [93,94].
On the other hand, GM dysbiosis is also associated with the progression of renal disease. Under physiological conditions, the microbiota functions with a low O2 supply [95]. However, GM dysbiosis alters these hypoxic conditions, leading to inflammation and apoptosis and contributing to the AKI-to-CKD transition [96]. GM dysbiosis also influences mitochondrial dysfunction, leading to ROS increase and AhR signaling through UT synthesis (IS, pCS, TMAO) and therefore to the progression of CKD [97,98,99].

3.2. The Tubulo-Interstitial Compartment

3.2.1. Maladaptive Repair and the EMT

In the AKI-to-CKD Transition

After injury, tubular repair allows the patient to recover renal function and is achieved through (i) the regeneration of the lost tubular epithelial cells by dedifferentiation–proliferation–redifferentiation of surviving tubular epithelial cells and (ii) temporary, circumscribed, renal fibrosis as a short-term, adaptive tissue repair mechanism. However, the persistence of fibrosis after severe, repetitive injury leads to maladaptive repair, with replacement of the renal parenchyma by connective tissue, preventing the total recovery of the estimated glomerular filtration rate (eGFR) present before the AKI.
Persistent fibrosis is maintained by cells having undergone partial EMT, although the origin of these cells is still subject to debate [100]. EMT is a pathological mechanism in which tubular epithelial cells transdifferentiate into a fibroblast-like phenotype during tubulo-interstitial dialogue. The loss of epithelial cell junctions and polarity is followed by a change in protein expression (under-expression of the epithelial proteins zonula occludens-1 (ZO-1) and E-cadherin) and over-expression of mesenchymal markers (e.g., alpha-smooth muscle actin (α-SMA), vimentin, and fibronectin). This leads to the acquisition of a pathological extra-cellular matrix (ECM) secretory phenotype and to migratory capacity [101,102,103]. Maladaptive transdifferentiation sustains persistent, uncontrolled fibrosis, which drives the AKI-to-CKD transition through several phenotypic changes: cell cycle arrest, reactivation of proliferation pathways, organelle stress, and epigenetic alterations.

The Involvement of UTs

The results of several preclinical studies have shown that UTs accumulate in tubular cells [104] and contribute to EMT by promoting epithelial marker loss and mesenchymal marker acquisition via induction through TGF-β (Table 1). Exposure of proximal renal tubular NRK-52E cells to IS alters the typical cubic aspect of epithelial cells and turns them into fibroblast-like cells. The epithelial phenotype (ZO-1 and E-cadherin) is replaced by a mesenchymal phenotype (with overexpression of α-SMA) associated with induction of the ERK1/2 and MAPK apoptotic pathways. Pretreatment with the OAT3 inhibitor probenecid and inhibitors of the ERK1/2 and MAPK pathways reduced IS-induced EMT [105]. Similarly, exposure of murine proximal tubule cells and an in vivo mouse model to the UTs IS and pCS activated TGF-β excretion and initiated EMT via the Smad and Snail pathways [106,107]. Fibroblasts are also activated in mice exposed per os to IS [108]. Administration of AST-120 (Kremezin®) to mice allows epithelial cells to undergo EMT [109,110,111]. Senescence and endoplasmic reticulum (ER) stress promoted by IS are cellular adaptations for progression to a mesenchymal phenotype and are attenuated by the administration of a chelating resin [111]. To date, the influence of UTs on the expression of EMT markers has not been investigated in patients with renal disease.

3.2.2. Hypoxia

In the AKI-to-CKD Transition

During AKI, an initial acute injury may expose tubular cells to hypoxia. The kidneys are particularly sensitive to hypoxia [147,148,149]. Under hypoxic conditions, stimulation of oxidative stress, an imbalance between the mitochondrial production and the elimination of ROS (principally the peroxide ion O22− and the superoxide ion O2•−), activates the hypoxia-inducible factor (HIF) heterodimer, which enables the transcription of genes that compensate for oxygen scarcity (angiogenesis, erythropoiesis, anaerobic glycolysis) [150,151]. Hypoxia contributes to the AKI-to-CKD transition by promoting a mesenchymal phenotype via tubulointerstitial fibrosis [152,153]. The involvement of TGF-β and tumor protein 53 (p53) as intermediates between ROS secretion and tubular injury has also been demonstrated [154]. Hypoxia-triggered EMT can be inhibited using HIF siRNA [155]. In a mouse model of ischemia/reperfusion (I/R), exposure to a molecule mimicking the action of the superoxide dismutase (SOD) or the nuclear factor erythroid 2-related factor 2 (Nrf2) reduced ROS and peroxidized lipid production, apoptosis, renal tubular damage, and the number of CD68+ cells, promoting macrophage infiltration [156,157].

The Involvement of UTs

The literature data show that UTs are capable of inducing hypoxia and ROS secretion—both of which are involved in the AKI-to-CKD transition [158,159]. Nakagawa et al. showed that exposure of human proximal tubular (HK2) cells to IS and IAA led to intracellular ROS production [120]. This effect was suppressed by probenecid (OAT3 inhibitor) and antioxidants (NAC or probucol) [112]. Shimizu et al. demonstrated that ROS activation by IS is achieved through NF-κB. Exposure to NF-κB inhibitors (siRNA, pyrrolidine dithiocarbamate, and isohelenin), an antioxidant (NAC), and an NADPH oxidase inhibitor (diphenyleneiodonium (DPI)) reduces the effect of IS on angiotensinogen production by tubular cells and lessens fibrosis and inflammation [118]. Bolati et al. confirmed the involvement of oxidative stress in an in vivo model; the inhibition of transcription factor Nrf2 by IS via NF-κB led to a decrease in levels of the antioxidant enzymes heme oxygenase 1 (HO-1) and NAD(P)H quinone dehydrogenase 1 (NQO1) [119]. Furthermore, the antioxidant action of HO-1 and NQO1 was restored by administration of AST-120 to the animals. Animals exposed per os to indole (an IS precursor) developed kidney hypoxia and damage at 16 weeks [121]. Administration of AST-120 restored physiological oxygen consumption and reduced hypoxia and kidney damage. An improvement was also observed in vitro with the use of the Na/K/ATPase inhibitor ouabain and the NADPH oxidase inhibitor apocynin. Lastly, in a cohort of CKD patients, a negative correlation was identified between plasma concentrations of the UTs N-methyl-2-pyridone-5-carboxamide and N-methyl-4-pyridone-5-carboxamide (2PY and 4PY) and the plasma concentration of reduced glutathione (GSH), the most abundant antioxidant and a major detoxification agent in cells [142]. Inhibition of this protective molecule increases the risk of oxidative cells damages and can lead to CKD [160].

3.2.3. Organelle Stress

In the AKI-to-CKD Transition

The mitochondria is the powerhouse of the cell, and ROS-dependent mitochondrial stress is associated with many kidney diseases [161]—including AKI and the AKI-to-CKD transition [162]. Several in vivo studies have confirmed the involvement of mitochondrial stress in renal tubules in the progression from AKI to CKD through various mechanisms: redox alterations (ROS production, decreased β-oxidation, depolarization of membrane potential) [163], activation of mitochondrial fission [164], and decreased mitochondrial ATP synthesis [165]. Clinical studies have corroborated these data, as shown by Hu et al.’s report of a correlation between the urine mitochondrial DNA concentration and markers in patients with severe AKI relative to patients with mild AKI [166].
Permanent triggering of the unfolded protein response (UPR) pathway during ER stress activates apoptotic pathways via the CEBP-homologous protein (CHOP, a molecular intermediate triggered by ER stress and a mediator of apoptosis) and promotes the AKI-to-CKD transition [167,168,169]. Lastly, clinical studies have corroborated the preclinical results by highlighting a correlation between ER stress marker activation (CHOP, X-box binding protein 1, and ER-associated protein reticulon 1A) and a decrease in the eGFR [170]. These markers were also overexpressed in AKI patients with progression relative to AKI patients without progression [171].

The Involvement of UTs

The AKI-to-CKD transition might also involve stressed organelles, including mitochondria. Some UTs (notably, hippuric acid (HA), IAA, and uric acid) can be produced by mitochondria via physiological or pathological pathways [172]. UTs (such as IS, pCS, HA, and CMPF) may be responsible for mitochondrial stress in renal tubular cells, as has already been shown in muscle [173,174] and the endothelium [84]. Mutsaers et al. showed that exposure of CiPTEC kidney cells to UTs inhibited mitochondrial succinate dehydrogenase (complex II) activity by over 20% compared with controls. There was also an 18% reduction in the capacity of the electron transport system [122]. Aerobic and anaerobic metabolic pathways were impaired in a model of nephrectomized B-6 mice treated with IS and pCS; the increased mitochondrial fission and autophagy were similar to those observed in senescent cells and in AKI [123]. Lastly, a study of a cohort of hemodialysis patients showed that CMPF reduced mitochondrial respiration by inhibiting ADP-dependent oxidation of NADH-dependent substrates [143].
A few studies have investigated the induction of ER stress by UTs in kidney. For example, Jeon et al. found that apigenin treatment in an HK-2 cell model attenuated the ER stress induced by IS and led to a decrease in interleukin (IL)-6 and cyclin-dependent kinase inhibitor 1 (p21) levels [124]. Similar results were found in vivo. Indeed, administration of AST-120 in a nephrectomized rat model reduced the plasma concentrations of IS and CHOP. Inhibition of activating transcription factor-(ATF)-4 (the endoplasmic receptor activating the UPR pathway) by siRNA reduced the expression of IL-6, a proinflammatory cytokine involved in fibrosis [125].

3.2.4. Metabolic Reprogramming

In the AKI-to-CKD Transition

Metabolic reprogramming is the ability of cells to adapt their metabolic response to changes in their environment. Tubulointerstitial cells need a lot of ATP to carry out their excretion and reabsorption functions. These functions depend mainly on the β-oxidation of fatty acids, since glucose is reabsorbed by sodium glucose cotransporters (SGLTs), making glycolysis less available. During AKI, β-oxidation decreases and ATP synthesis is compensated through aerobic glycolysis (insufficient to meet NADH and FADH2 cofactor requirements), followed by anaerobic glycolysis (leading to lactate synthesis). Activation of the HIF-1α factor and activation of AhR [175] and mitochondrial dysfunction [176] are both causes and consequences of the transition from normoxic to anaerobic metabolism. In addition, the processes involved in the progression of renal pathology (collagen deposition, chronic inflammation, etc.) require large amounts of energy [177,178,179].

The Involvement of UTs

Some UTs arise directly from metabolic reprogramming, notably, kynurenine [177] and lipid intermediates [180]. However, they are as much the result of kidney damage as the driving force behind its progression. Hopp et al. showed a link between kidney damage, metabolic reprogramming, and the accumulation of UTs, where an alteration in tryptophan metabolism was associated with a deterioration in kidney function and the accumulation of UTs in an in vivo model of autosomal dominant polycystic kidney disease [127]. UTs also stimulate inflammation via an AhR-dependent reprogramming of arachidonic acid metabolism [126].

3.2.5. Epigenetic Alterations and Cell Cycle Arrest

In the AKI-to-CKD Transition

Epigenetic alterations triggered by hypoxia via the HIF (histone modifications, DNA methylation, non-coding RNAs, and chromatin conformational changes) form the pathophysiological “hypoxic memory” of the primary injury [181]. These epigenetic changes are conserved in kidney cells after AKI and predispose the cell to progression to CKD [182]. Several in vivo studies have shown that epigenetic alterations aggravate AKI [183,184,185,186]. A number of clinical studies have confirmed these data: methylation-enriched areas in renal biopsies [187], hypermethylation of the kallikrein gene [188], and overexpression of circulating RNAs [189,190].
In response to severe or repeated injury, tubular cells exhibit G2/M cell cycle arrest (as opposed to G1/0 arrest under normal conditions); this leads to premature renal tubular cell senescence in AKI [191] and CKD [192]. Several in vitro models have evidenced a link between G2/M phase arrest-triggering senescence (p21) and transdifferentiation towards a pro-inflammatory phenotype (α-SMA, fibronectins, and fibroblast size) [193,194]. In vivo studies have confirmed these data [195,196]. Furthermore, NAC reduced cisplatin-induced renal senescence in a C57BL/6 mouse model [197]. Several clinical studies have found that two urine biomarkers of cell cycle arrest (tissue inhibitor metalloproteinase (TIMP)-2 and insulin-like growth factor-binding protein (IGFBP)-7) predict the imminent risk of AKI in critical care patients [198].

The Involvement of UTs

The results of a few preclinical studies have illustrated the effects of UTs on epigenetic alterations in renal tubular cells and thus their contribution to the AKI-to-CKD transition. In a study of HK-2 cells, for example, IS increased the methylation of secreted frizzled-related protein (sFRP) CpG islands (inhibiting its expression) and led to Wnt/β-catenin pathway activation and to aberrant kidney fibrosis. Administration of recombinant sFRP5 in vivo alleviated IS-induced fibrosis [128]. In a nephrectomized B-6 mouse model, exposure to IS and pCS induced expression of the DNA methytransferase involved in the hypermethylation of Klotho; this abrogated Klotho expression in renal tubules [129], as also seen in AKI [199]. The impact of UTs on renal cell cycle arrest initiates molecular pathways involved in the AKI-to-CKD transition. In a renal tubular cell model, D-serine (a novel UT) induced senescence via activation of general control nonderepressible 2 (GCN2) [145]. In the same manner, IS promoted senescence in HK-2 cells through activation of NF-κB, leading to TGF-β overexpression, p53 and p21 activation, and senescence. These effects were blocked by the NF-κB inhibitors pyrrolidine dithiocarbamate and isohelenin. Similar results were found in a rat CKD model, in which AST-120 administration decreased the upregulation of phosphorylated p65, p53, p21, β-galactosidase, TGF-β, and α-SMA [130,131,132].

3.2.6. The TGF-β Signaling Pathway

In the AKI-to-CKD Transition

TGF-β is produced by dedifferentiated epithelial tubular cells after renal injury; it promotes epithelial dedifferentiation and cell cycle arrest, sensitizes cells to apoptosis, shortens peritubular capillaries, aggravates hypoxia, and favors progression from AKI to CKD. Furthermore, TGF-β stimulates EMT and aggravating fibrosis. Lastly, TGF-β acts as a chemoattractant for macrophages and promotes their infiltration of the tubules. Knock-out of the gene for TGF-β receptor II (TGF-βRII) in a mouse model attenuated the symptoms of AKI [200]. Furthermore, control mice showed greater TGF-β expression, fibrosis, and macrophage infiltration than TGF-βRII-/- mice [201,202], prevented by the use of genetic or pharmacologic TGF-β inhibitors [203]. Finally, a review of nephrodiabetic patients showed that TGF-β induced EMT in both the proximal and the distal region of the nephron [204].

The Involvement of UTs

Several studies have demonstrated that UTs stimulate the production of TGF-β, a central mediator in the progression of tubules towards fibrosis. A growing body of evidence from preclinical studies points in this direction. In an HK-2 cell model, for example, IS induced TGF-β expression and thus expression of the EMT marker α-SMA [113]. Another UT (IAA) is also capable of inducing TGF-β [112]. Several in vivo studies have demonstrated that administration to animals of asymmetric dimethylarginine (a water-soluble UT), IS or pCS is associated with oxidative stress, elevated TGF-β, EMT, and renal fibrosis [106,107,146]. Lastly, administration of AST-120 decreased UT and TGF-β levels and thus reduced renal fibrosis—confirming UTs’ involvement in renal EMT [114,115,116,117].

3.2.7. Inflammation

In the AKI-to-CKD Transition

During the activation of the adaptive immune system, the balance between pro-inflammatory phenotypes (M1 macrophages and interferon gamma-secreting lymphocytes) and anti-inflammatory phenotypes (M2 macrophages and IL-10-secreting lymphocytes) favors (or not) tubular repair and fibroblast overactivation and thus determines the resolution of AKI or its progression to CKD. Several studies of preclinical models have shown that the hypersecretion of pro-inflammatory cytokines by macrophages is involved in the progression of AKI to CKD [205]. Mitochondrial injury can also induce inflammation [206,207]. The results of a number of clinical studies have confirmed these findings. Histological analysis of post-mortem renal biopsies from patients with AKI after septic shock showed intense leukocytic infiltration and marked apoptosis [208]. In a study of a cohort of post-surgical AKI patients, elevated C-reactive protein (CRP) levels were associated with higher mortality [209].

The Involvement of UTs

UTs stimulate the production of proinflammatory molecules by immune cells. This was demonstrated by Sun et al.’s study of inflammation genes expressed by renal tubular cells after exposure to IS and pCS. The cytokines IL-6, IL-15, and TGF-β were overexpressed, as were the NF-κB, Smad, Stat, and apoptosis pathway components (B2m, Bax, and Bcl2) [133]. This effect was attenuated by treatment with anti-Stat3 siRNA [132], indole propionic acid antioxidant [134], and AST-120 [135]. In a cohort of patients with abdominal aortic aneurysm (AAA) and matched control patients, there was a positive correlation between the plasma IS concentration and the proportion of proinflammatory CD14+/CD16+ monocytes. After exposure of THP1 monocytes to increasing concentrations of IS or serum from AAA patients, the cells differentiated into macrophages secreting proinflammatory cytokines (IL-6, MCP-1, and cyclooxygenase 2 (COX2)) [136]. The induction of MCP-1 expression by IS was also observed in vivo and was associated with renal monocyte/macrophage infiltration [137].

3.2.8. Iron Death Pathways

In the AKI-to-CKD Transition

Ferroptosis is a type of cell death that is different than apoptosis and necrosis, playing a role in the AKI-to-CKD transition [210]. Iron metabolism is a key factor in initiating the ferroptosis process via the accumulation of products of lipid peroxidation and overproduction of ROS [211,212]. Several cellular pathways involved in the AKI-to-CKD transition depend on ferroptosis (mitochondrial dysfunction, fibrosis) [213,214,215]. Inflammation is closely linked to ferroptosis, and they favor each other. On the one hand, multiple metabolites produced during the inflammatory state increase the susceptibility of the kidney to ferroptosis [216,217], and on the other hand, ferroptosis is able to promote the production of damage-associated molecular patterns (DAMPs) and inflammatory mediators [218,219]. In a mouse model of AKI, pre-treatment with an anti-oxidant, FG-4592, reduces the progression of fibrosis by blocking ferroptosis [220]. Finally, ferroptosis is also interconnected with other forms of cell death (necrosis, pyroptosis, eryptosis), which accelerates the destruction of renal tubular cells [221,222]. Eryptosis (programmed erythrocytes death) is involved in anemia during CKD [223] and depends on iron homeostasis and oxidative stress [224,225].

The Involvement of UTs

Only a few studies have examined the role of UTs in ferroptosis during AKI-to-CKD transition. In a recent in vivo study, the authors observed a higher level of lipid peroxidation and a higher iron concentration in the kidneys of AKI mice, in association with overexpression of ferroptosis proteins (such as transferring receptor 1 (TfR1)) [144]. PBUTs have also been implicated in ferroptosis in vitro. CMPF showed the greatest effect, inhibiting the expression of ferritin and GPX4, a protein decreasing lipid peroxidation. In another study, the authors demonstrated the involvement of IS in iron metabolism during CKD [138]. Indeed, IS increases the gene and protein expression of hepcidin through AhR. Moreover, IS and IAA promote eryptosis via cytosolic Ca2+ and ROS accumulation, aggravating anemia and CKD [139,140,141,226].

3.3. Endothelial Dysfunction

3.3.1. In the AKI-to-CKD Transition

Renal microcirculation in general and peritubular capillaries in particular are very susceptible to injury [14]. Vascular lesion begins with poor repair and results in functional and structural lesions, with vasoreactivity alteration, greater inflammation, altered endothelial permeability, and capillary rarefaction [227]. In preclinical models, an imbalance between EC destruction and regeneration contributes to the transition to chronic vascular lesions [228]. ECs exhibit an expression of caspase-3, an abnormal cytoskeleton, and a disruption to adherens junctions, with increased vessel permeability and apoptosis [229,230,231]. Although dialogue between ECs and pericytes allows for the remodeling and repair of injured vessels [232,233], the loss of pericytes provokes permanent EC damage in peritubular capillaries and is associated with tubular injury in AKI mice [234]. Furthermore, the expression of angiogenic factors (such as vascular endothelial growth factor (VEGF)) are markedly lower [235,236] after kidney injury and inhibited EC proliferation [237,238], whereas the anti-angiogenic factors (such as endostatin) are overexpressed [239,240]. The angiogenic factor silent information regulator two protein (Sirtuin)-1 protects against capillary rarefaction by blocking premature EC senescence secondary to p53 and Notch1 signaling downregulation [241,242,243]. Both ECs and pericytes are able to differentiate into myofibroblasts that also promote kidney fibrosis and progression to CKD [100,244]. Overall, all these factors promote the AKI-to-CKD transition.

3.3.2. Role of UTs in Endothelial Dysfunction during the AKI-to-CKD Transition

UTs (especially IS and pCS) are known to have harmful effects on the vascular wall during CKD (Table 2). In clinical studies, IS and pCS have been linked to cardiovascular disease and mortality [245,246,247]. In preclinical studies, IS exerted direct toxicity on ECs by decreasing cell viability and proliferation and by promoting oxidative stress [83,84,248]. Moreover, IS induced the upregulation of vascular adhesion molecules (such as ICAM-1 and MCP-1) associated with leukocyte extravasation and decreased vascular relaxation [249,250,251]. IS also acts on the calcification process by inducing the expression of osteoblast-specific proteins in VSMC and ECs’ secretion of procalcifying IL-8 [252,253]. Furthermore, IS increased tissue factor production and induced a pro-thrombotic state [254]. Recent studies have revealed a novel antiangiogenic action of IS through the activation of AhR associated with a decrease in EC proliferation [255]. Moreover, plasma angiopoietin-1 concentration increased significantly after hemodiafiltration, a technique used to purify UTs in the dialysate of CKD patients [256]. In the kidney vasculature specifically, IS also exerts toxicity on arterioles obstructed by ECs. After 8 weeks of intravenous IS administration, larger arteries presented reduplication of elastic lamina [69]. Like IS, pCS is associated with cardiovascular events in CKD patients [257,258,259]. Several preclinical studies have investigated pCS’s pathological effects on vascular cells. pCS is responsible for the release of microparticles and an increase in endothelial wall permeability [260]. Furthermore, pCS promotes atherogenesis and plaque instability via its action on leukocyte recruitment and on VSMC migration and proliferation [261,262].
There are now some preclinical data on the pathological vascular role of UTs during AKI. Short-term exposure of ECs to increasing concentrations of IS induced ROS production and ICAM-1 expression and reduced cell viability and NO bioavailability [84,248,249]. Furthermore, IS inhibits EC regeneration via its toxic effect on endothelial progenitor cells (EPCs), with a decrease in their proliferation and an increase in their senescence. In another preclinical study of cultured human EPCs, VEGF expression (usually induced by HIF-1α and IL-10/STAT3 pathways) was suppressed by IS pretreatment [263]. In an animal model of AKI, IS induced a pro-oxidant state with the downregulation of endothelial nitric oxide synthase (eNOs) expression in kidney arteries after I/R and a decrease in EPC egress from the bone marrow [264]. IS also upregulated EC expression of E-selectin in kidney via IL-1β activation, associated with ROS overproduction and greater endothelial monocyte adhesion [265]. In the same manner, ex vivo aorta exposure to IS induced a reduction in vasorelaxation [266,267]. A few studies have demonstrated the harmful action of pCS during AKI. pCS induced ROS production by ECs and VSMCs after only 30 min of exposure in vitro [85]. This effect was associated with vascular leukocyte adhesion and pathological constriction of the aorta in vivo. It was also responsible for an alteration in vascular permeability [268]. Given IS’s ability to block angiogenesis during AKI, this UT probably contributes to the AKI-to-CKD transition. Moreover, IS is able to promote pro-inflammatory cytokines (such as IL-1β) that block the expression of angiogenic factors [235,236,265]. Furthermore, IS is harmful to the EPCs involved in vascular repair [269]. Notably, through its action on vascular wall permeability, pCS might contribute to the AKI-to-CKD transition [260,268].
Table 2. Mechanisms of UT toxicity involved in the AKI-to-CKD transition in the endothelial compartment.
Table 2. Mechanisms of UT toxicity involved in the AKI-to-CKD transition in the endothelial compartment.
UTsMechanisms Underlying the AKI-to-CKD TransitionEndothelium
ModelsMain ResultsReferences
ISOxidative stressIn vitro- ↑ ICAM-1, ↑ MCP-1, ↑ NF-κB, ↑ ROS, ↑ E-selectin, ↑ IL-1β
- NADPH oxidase inhibitors decreased IS-induced oxidative stress
- ↓ Hypoxia-induced migration and tube formation
- ↓ Vasorelaxation
[84,248,249,263,265,266,267]
In vivo- ↓ eNOs
- AST-120 ↑ neovascularization
[263,264]
pCSOxidative stress
Inflammation
In vivo- ↑ ROS[85]
In vivo- ↑ Leukocyte adhesion, ↑ vascular permeability[268]
Abbreviations: eNOs, endothelial nitric oxide synthase; ICAM-1, intercellular adhesion molecule-1; IL-1β, interleukin 1β; IS, indoxyl sulfate; MCP-1, monocyte chemoattractant protein 1; NF-κB, nuclear factor-κ B; pCS, para-cresyl sulfate; ROS, reactive oxygen species; UT, uremic toxin.

3.4. Glomerular Injury

3.4.1. Glomerular Injury in the AKI-to-CKD Transition

During AKI and CKD, immune activation includes the recruitment of different inflammatory cells (predominantly neutrophils and monocytes/macrophages), leading to tubular inflammation. Macrophages are also able to promote the injury of other cells [270]. The podocyte is particularly susceptible to injury and is involved in the AKI-to-CKD transition. In a murine model, podocyte lesions were more prominent than tubular lesions 28 days after I/R injury and characterized by excessive foot process fusion, glomerular basement membrane exposure, and downregulation of proteins with an important role in podocyte integrity (especially synaptopodin and nephrin) [271]. In addition, pro-inflammatory cytokines and angiotensin II cause podocyte lesions and loss, which lead to ROS production and Notch activation [272,273,274]. Notch signaling induced podocyte foot process effacement and EMT, which also promote glomerulosclerosis [273,275]. Furthermore, the senescence of glomerular cells (including podocytes and ECs) leads to senescence-associated secretory phenotypes (SASPs) as part of a switch to a permanent senescence process linked to CKD progression [276]. Glomerular parietal epithelial cells are also involved in the AKI-CKD transition via the formation of crescents [277,278] and the development of glomerulosclerosis [279,280].

3.4.2. The Role of UTs in Glomerular Injury during the AKI-to-CKD Transition

During CKD progression, IS also has harmful effects on glomerular function (Table 3). IS induces podocyte injury both in vitro and in vivo. Mice exposed to intravenous IS for 8 weeks presented elevated albuminuria and histological damage (ischemic damage to the glomerular basement membranes and mesangiolysis) [69]. A specific analysis of podocytes also revealed foot process effacement and cytoplasmic vacuoles. Furthermore, the expression of podocyte proteins (such as synaptopodin) was lower, whereas AhR expression was higher. In vitro, mouse podocytes showed an altered actin cytoskeleton and a switch to a pro-inflammatory phenotype. IS also induced oxidative stress (particularly in mesangial cells) with the production of ROS in a dose-dependent manner [281,282]. Moreover, IS activated heat shock protein 90 (HSp90) through the AhR pathway and promoted renal fibroblast activation [108]. In uremic rats orally supplemented with IS, a histological analysis of the kidney demonstrated higher levels of glomerular sclerosis; these were reversed by AST-120, an oral sorbent that reduced the serum IS level [283,284].
Although pCS is known to have specific toxic effects on interstitial and tubular cells, nothing is known about its potential toxic effects on glomerular cells [285]. Furthermore, there are no data on the potential acute glomerular toxicity of UTs. However, we can assume that IS is also involved in the AKI-to-CKD transition through its harmful glomerular effects. Thus, podocyte and mesangial cell alterations observed after IS exposure are known to contribute to features of the AKI-to-CKD transition, such as ROS production, the upregulation of pro-inflammatory cytokines, cell senescence, fibrosis, and the downregulation of proteins required for podocyte integrity (synaptopodin, nephrin, and CD2AP) [69,271,272,276,281].

4. Therapies and Future Research Directions

Today, the only effective way of temporarily reducing plasma concentrations of PBUTs and their complications is renal transplantation. Because of their binding to plasma proteins, PBUTs are poorly eliminated by the various dialysis techniques. Carbon chelating agents (AST-120 (Kremezin®), sevelamer carbonate (Renvela®)) make it possible to limit PBUTs upstream of their accumulation in plasma, for example, by reducing the passage of their amino acid precursors through the intestinal barrier. However, there is no consensus on their use [286,287,288,289], and there is a lack of data on the long-term effects on improving renal function and reducing cardiovascular complications.
During CKD and AKI, the GM modifies its metabolism, favoring proteolytic fermentation, to the detriment of saccharolytic fermentation [93,290]. Several clinical studies in patients with CKD have shown that a high-fiber, low-protein diet reduces plasma concentrations of UTs [291,292,293,294,295]. However, further long-term studies of nutritional intervention are needed to assess the contribution of single or repeated administration of specific foods or nutrients to plasma retention of UTs in patients. SGLT inhibitors also restore the proteolytic/saccharolytic fermentation balance [296,297,298]. Another strategy targeting the absorption phase is the use of prebiotics, probiotics, and postbiotics. Their use is also controversial, with in vivo data showing efficacy [299] and clinical studies showing mixed results [300,301]. A randomized double-blind clinical trial in 29 patients with chronic renal failure showed that administration of enzobiotic therapy (synbiotics with proteolytic enzymes) for 3 months reduced plasma pCS concentrations with an eGFR that remained stable compared with the placebo group [302]. Fecal microbiota transplantation is another avenue worth exploring to limit the endosynthesis of UTs [303,304,305]. However, preclinical studies show contradictory results in terms of the benefit in animals with AKI [306,307]. To our knowledge, there are currently no clinical studies looking at fecal transplantation in the context of renal disease, and a standardization before setting up such studies is needed (pre-treatment with ATB, galenic form, frequency of administration, inter-individual variability, and management of adverse effects). Certain classes of drugs, such as vancomycin, can be used to restore an altered GM [308]. Genetic modification of the Bacteroides strain no longer expressing tryptophanase may decrease the plasma accumulation of UTs [309].
Another approach is to intervene in the hepatic metabolism of UTs by inhibiting the hepatic enzymes that synthesize them. Thus, in a mouse model of acute renal failure, administration of molecules that inhibit the hepatic enzyme SULT1A1 induces nephroprotection against the renal toxicity of IS [310,311]. Similarly, the study of the inhibition of the FMO3 responsible for the activation of trimethylamine to TMAO may be a therapeutic pathway [312].
PBUTs are very poorly eliminated by conventional dialysis methods. In order to optimize PBUT elimination, various alternative techniques are being studied, such as the combination of diffusive and convective techniques in pre- or post-hemodiafiltration [313,314,315]. The use of an adsorbent during hemodialysis (albumin, nanoporous monolith, activated charcoal, highly porous microparticles, graphene nanoplatelets) has also shown interesting results [316,317]. Another method is to use molecules with a higher affinity for albumin than UTs (sodium octaonate, furosemide, ibuprofen) [318,319,320,321]. Finally, it is likely that limiting the deleterious effects of UTs will require a combination of all of these therapies.

5. Conclusions

Many studies of preclinical models of AKI and CKD have shown that post-injury cellular defense and adaptation mechanisms induce the progression from AKI to CKD regardless of the injured renal compartment (the tubule, glomerulus, or endothelium). After AKI, the induction of an immune response leads to the secretion of proinflammatory and profibrotic factors. This harmful environment modifies the metabolism of tubular cells, glomerular cells, and ECs, which leads to ROS secretion, organelle stress, induction of NF-κB, cellular senescence, EMT, ECM synthesis, and fibrosis. UTs are involved in the same cellular adaptation mechanisms in each kidney compartment, which ultimately lead to chronic inflammation, tubular fibrosis, and progression to CKD. The various cellular and molecular mechanisms described here are interrelated. For example, ER stress and mitochondrial stress are strongly activated under hypoxia. It is therefore difficult to study each mechanism individually. To date, only one preclinical study has specifically investigated the impact of these UTs on the AKI-to-CKD transition. Further studies are therefore needed to investigate the link between UTs and AKI/CKD in order to determine whether these toxins are risk factors and therapeutic targets for the AKI-to-CKD transition.

Author Contributions

Writing—original draft preparation, C.A. and P.C.; writing—review and editing, C.A., S.B., S.K., Y.B. and P.C. All authors have read and agreed to the published version of the manuscript.

Funding

This research did not receive any specific funding from agencies or organizations in the public, commercial, or not-for-profit sectors. The Open Access and Article Processing Charge were funded by Amiens Medical Center (Amiens, France).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Sawhney, S.; Bell, S.; Black, C.; Christiansen, C.F.; Heide-Jørgensen, U.; Jensen, S.K.; Ronksley, P.E.; Tan, Z.; Tonelli, M.; Walker, H.; et al. Harmonization of Epidemiology of Acute Kidney Injury and Acute Kidney Disease Produces Comparable Findings across Four Geographic Populations. Kidney Int. 2022, 101, 1271–1281. [Google Scholar] [CrossRef]
  2. Kovesdy, C.P. Epidemiology of Chronic Kidney Disease: An Update 2022. Kidney Int. Suppl. 2022, 12, 7–11. [Google Scholar] [CrossRef] [PubMed]
  3. Hsu, C.Y.; Ordoñez, J.D.; Chertow, G.M.; Fan, D.; McCulloch, C.E.; Go, A.S. The Risk of Acute Renal Failure in Patients with Chronic Kidney Disease. Kidney Int. 2008, 74, 101–107. [Google Scholar] [CrossRef] [PubMed]
  4. Hsu, R.K.; Hsu, C. The Role of Acute Kidney Injury in Chronic Kidney Disease. Semin. Nephrol. 2016, 36, 283–292. [Google Scholar] [CrossRef] [PubMed]
  5. Rubin, S.; Orieux, A.; Clouzeau, B.; Rigothier, C.; Combe, C.; Gruson, D.; Boyer, A. The Incidence of Chronic Kidney Disease Three Years after Non-Severe Acute Kidney Injury in Critically Ill Patients: A Single-Center Cohort Study. J. Clin. Med. 2019, 8, 2215. [Google Scholar] [CrossRef]
  6. Jankowski, J.; Floege, J.; Fliser, D.; Böhm, M.; Marx, N. Cardiovascular Disease in Chronic Kidney Disease: Pathophysiological Insights and Therapeutic Options. Circulation 2021, 143, 1157–1172. [Google Scholar] [CrossRef]
  7. Silver, S.A.; Harel, Z.; McArthur, E.; Nash, D.M.; Acedillo, R.; Kitchlu, A.; Garg, A.X.; Chertow, G.M.; Bell, C.M.; Wald, R. Causes of Death after a Hospitalization with AKI. J. Am. Soc. Nephrol. 2018, 29, 1001–1010. [Google Scholar] [CrossRef]
  8. Hansen, M.K.; Gammelager, H.; Jacobsen, C.-J.; Hjortdal, V.E.; Layton, J.B.; Rasmussen, B.S.; Andreasen, J.J.; Johnsen, S.P.; Christiansen, C.F. Acute Kidney Injury and Long-Term Risk of Cardiovascular Events After Cardiac Surgery: A Population-Based Cohort Study. J. Cardiothorac. Vasc. Anesth. 2015, 29, 617–625. [Google Scholar] [CrossRef]
  9. Carracedo, J.; Alique, M.; Vida, C.; Bodega, G.; Ceprián, N.; Morales, E.; Praga, M.; De Sequera, P.; Ramírez, R. Mechanisms of Cardiovascular Disorders in Patients with Chronic Kidney Disease: A Process Related to Accelerated Senescence. Front. Cell Dev. Biol. 2020, 8, 185. [Google Scholar] [CrossRef]
  10. Wu, V.-C.; Wu, C.-H.; Huang, T.-M.; Wang, C.-Y.; Lai, C.-F.; Shiao, C.-C.; Chang, C.-H.; Lin, S.-L.; Chen, Y.-Y.; Chen, Y.-M.; et al. Long-Term Risk of Coronary Events after AKI. J. Am. Soc. Nephrol. 2014, 25, 595–605. [Google Scholar] [CrossRef]
  11. Lee, S.; Park, S.; Kang, M.W.; Yoo, H.-W.; Han, K.; Kim, Y.; Lee, J.P.; Joo, K.W.; Lim, C.S.; Kim, Y.S.; et al. Postdischarge Long-Term Cardiovascular Outcomes of Intensive Care Unit Survivors Who Developed Dialysis-Requiring Acute Kidney Injury after Cardiac Surgery. J. Crit. Care 2019, 50, 92–98. [Google Scholar] [CrossRef] [PubMed]
  12. Hsu, C.; Hsu, R.K.; Yang, J.; Ordonez, J.D.; Zheng, S.; Go, A.S. Elevated BP after AKI. J. Am. Soc. Nephrol. 2016, 27, 914–923. [Google Scholar] [CrossRef] [PubMed]
  13. Go, A.S.; Hsu, C.; Yang, J.; Tan, T.C.; Zheng, S.; Ordonez, J.D.; Liu, K.D. Acute Kidney Injury and Risk of Heart Failure and Atherosclerotic Events. Clin. J. Am. Soc. Nephrol. 2018, 13, 833–841. [Google Scholar] [CrossRef]
  14. Guzzi, F.; Cirillo, L.; Roperto, R.M.; Romagnani, P.; Lazzeri, E. Molecular Mechanisms of the Acute Kidney Injury to Chronic Kidney Disease Transition: An Updated View. Int. J. Mol. Sci. 2019, 20, 4941. [Google Scholar] [CrossRef]
  15. Kurzhagen, J.T.; Dellepiane, S.; Cantaluppi, V.; Rabb, H. AKI: An Increasingly Recognized Risk Factor for CKD Development and Progression. J. Nephrol. 2020, 33, 1171–1187. [Google Scholar] [CrossRef] [PubMed]
  16. Belayev, L.Y.; Palevsky, P.M. The Link between Acute Kidney Injury and Chronic Kidney Disease. Curr. Opin. Nephrol. Hypertens. 2014, 23, 149–154. [Google Scholar] [CrossRef] [PubMed]
  17. André, C.; Bennis, Y.; Titeca-Beauport, D.; Caillard, P.; Cluet, Y.; Kamel, S.; Choukroun, G.; Maizel, J.; Liabeuf, S.; Bodeau, S. Two Rapid, Accurate Liquid Chromatography Tandem Mass Spectrometry Methods for the Quantification of Seven Uremic Toxins: An Application for Describing Their Accumulation Kinetic Profile in a Context of Acute Kidney Injury. J. Chromatogr. B 2020, 1152, 122234. [Google Scholar] [CrossRef] [PubMed]
  18. Barreto, F.C.; Barreto, D.V.; Liabeuf, S.; Meert, N.; Glorieux, G.; Temmar, M.; Choukroun, G.; Vanholder, R.; Massy, Z.A. Serum Indoxyl Sulfate Is Associated with Vascular Disease and Mortality in Chronic Kidney Disease Patients. Clin. J. Am. Soc. Nephrol. 2009, 4, 1551–1558. [Google Scholar] [CrossRef]
  19. Meijers, B.K.I.; Claes, K.; Bammens, B.; De Loor, H.; Viaene, L.; Verbeke, K.; Kuypers, D.; Vanrenterghem, Y.; Evenepoel, P. P-Cresol and Cardiovascular Risk in Mild-to-Moderate Kidney Disease. Clin. J. Am. Soc. Nephrol. 2010, 5, 1182–1189. [Google Scholar] [CrossRef]
  20. Khwaja, A. KDIGO Clinical Practice Guidelines for Acute Kidney Injury. Nephron Clin. Pr. 2012, 120, c179–c184. [Google Scholar] [CrossRef]
  21. Hsu, R.K.; McCulloch, C.E.; Dudley, R.A.; Lo, L.J.; Hsu, C. Temporal Changes in Incidence of Dialysis-Requiring AKI. J. Am. Soc. Nephrol. 2013, 24, 37–42. [Google Scholar] [CrossRef]
  22. Hoste, E.A.J.; Kellum, J.A.; Selby, N.M.; Zarbock, A.; Palevsky, P.M.; Bagshaw, S.M.; Goldstein, S.L.; Cerdá, J.; Chawla, L.S. Global Epidemiology and Outcomes of Acute Kidney Injury. Nat. Rev. Nephrol. 2018, 14, 607–625. [Google Scholar] [CrossRef] [PubMed]
  23. Susantitaphong, P.; Cruz, D.N.; Cerda, J.; Abulfaraj, M.; Alqahtani, F.; Koulouridis, I.; Jaber, B.L. World Incidence of AKI: A Meta-Analysis. Clin. J. Am. Soc. Nephrol. 2013, 8, 1482–1493. [Google Scholar] [CrossRef] [PubMed]
  24. Hoste, E.A.J.; Bagshaw, S.M.; Bellomo, R.; Cely, C.M.; Colman, R.; Cruz, D.N.; Edipidis, K.; Forni, L.G.; Gomersall, C.D.; Govil, D.; et al. Epidemiology of Acute Kidney Injury in Critically Ill Patients: The Multinational AKI-EPI Study. Intensive Care Med. 2015, 41, 1411–1423. [Google Scholar] [CrossRef] [PubMed]
  25. Abebe, A.; Kumela, K.; Belay, M.; Kebede, B.; Wobie, Y. Mortality and Predictors of Acute Kidney Injury in Adults: A Hospital-Based Prospective Observational Study. Sci. Rep. 2021, 11, 15672. [Google Scholar] [CrossRef] [PubMed]
  26. Mehta, R.L.; Cerdá, J.; Burdmann, E.A.; Tonelli, M.; García-García, G.; Jha, V.; Susantitaphong, P.; Rocco, M.; Vanholder, R.; Sever, M.S.; et al. International Society of Nephrology’s 0by25 Initiative for Acute Kidney Injury (Zero Preventable Deaths by 2025): A Human Rights Case for Nephrology. Lancet 2015, 385, 2616–2643. [Google Scholar] [CrossRef]
  27. Mehta, R.L.; Burdmann, E.A.; Cerdá, J.; Feehally, J.; Finkelstein, F.; García-García, G.; Godin, M.; Jha, V.; Lameire, N.H.; Levin, N.W.; et al. Recognition and Management of Acute Kidney Injury in the International Society of Nephrology 0by25 Global Snapshot: A Multinational Cross-Sectional Study. Lancet 2016, 387, 2017–2025. [Google Scholar] [CrossRef]
  28. Finlay, S.; Bray, B.; Lewington, A.; Hunter-Rowe, C.; Banerjee, A.; Atkinson, J.; Jones, M. Identification of Risk Factors Associated with Acute Kidney Injury in Patients Admitted to Acute Medical Units. Clin. Med. 2013, 13, 233–238. [Google Scholar] [CrossRef]
  29. Jiang, Y.-J.; Xi, X.-M.; Jia, H.-M.; Zheng, X.; Wang, M.-P.; Li, W.; Li, W.-X. Risk Factors, Clinical Features and Outcome of New-Onset Acute Kidney Injury among Critically Ill Patients: A Database Analysis Based on Prospective Cohort Study. BMC Nephrol. 2021, 22, 289. [Google Scholar] [CrossRef]
  30. Chawla, L.S.; Eggers, P.W.; Star, R.A.; Kimmel, P.L. Acute Kidney Injury and Chronic Kidney Disease as Interconnected Syndromes. N. Engl. J. Med. 2014, 371, 58–66. [Google Scholar] [CrossRef]
  31. Santos, W.J.; Zanetta, D.M.; Pires, A.C.; Lobo, S.M.; Lima, E.Q.; Burdmann, E.A. Patients with ischaemic, mixed and nephrotoxic acute tubular necrosis in the intensive care unit—A homogeneous population? Crit. Care 2006, 10, R68. [Google Scholar] [CrossRef] [PubMed]
  32. Filippone, E.; Kraft, W.; Farber, J. The Nephrotoxicity of Vancomycin. Clin. Pharmacol. Ther. 2017, 102, 459–469. [Google Scholar] [CrossRef] [PubMed]
  33. Mulay, S.R.; Anders, H.-J. Crystal Nephropathies: Mechanisms of Crystal-Induced Kidney Injury. Nat. Rev. Nephrol. 2017, 13, 226–240. [Google Scholar] [CrossRef] [PubMed]
  34. Turgut, F.; Awad, A.S.; Abdel-Rahman, E.M. Acute Kidney Injury: Medical Causes and Pathogenesis. J. Clin. Med. 2023, 12, 375. [Google Scholar] [CrossRef]
  35. Gaut, J.P.; Liapis, H. Acute Kidney Injury Pathology and Pathophysiology: A Retrospective Review. Clin. Kidney J. 2021, 14, 526–536. [Google Scholar] [CrossRef]
  36. Kellum, J.A.; Romagnani, P.; Ashuntantang, G.; Ronco, C.; Zarbock, A.; Anders, H.-J. Acute Kidney Injury. Nat. Rev. Dis. Primers 2021, 7, 52. [Google Scholar] [CrossRef]
  37. Eadon, M.T.; Schwantes-An, T.-H.; Phillips, C.L.; Roberts, A.R.; Greene, C.V.; Hallab, A.; Hart, K.J.; Lipp, S.N.; Perez-Ledezma, C.; Omar, K.O.; et al. Kidney Histopathology and Prediction of Kidney Failure: A Retrospective Cohort Study. Am. J. Kidney Dis. 2020, 76, 350–360. [Google Scholar] [CrossRef]
  38. Gunawardena, S.; Dayaratne, M.; Wijesinghe, H.; Wijewickrama, E. A Systematic Review of Renal Pathology in Chronic Kidney Disease of Uncertain Etiology. Kidney Int. Rep. 2021, 6, 1711–1728. [Google Scholar] [CrossRef]
  39. Selvarajah, M.; Weeratunga, P.; Sivayoganthan, S.; Rathnatunga, N.; Rajapakse, S. Clinicopathological Correlates of Chronic Kidney Disease of Unknown Etiology in Sri Lanka. Indian. J. Nephrol. 2016, 26, 357. [Google Scholar] [CrossRef]
  40. Schelling, J.R. Tubular Atrophy in the Pathogenesis of Chronic Kidney Disease Progression. Pediatr. Nephrol. 2016, 31, 693–706. [Google Scholar] [CrossRef]
  41. Huang, R.; Fu, P.; Ma, L. Kidney Fibrosis: From Mechanisms to Therapeutic Medicines. Signal. Transduct. Target. Ther. 2023, 8, 129. [Google Scholar] [CrossRef] [PubMed]
  42. Pereira, B.J.; Barreto, S.; Gentil, T.; Assis, L.S.; Soeiro, E.M.; Castro, I.D.; Laranja, S.M. Risk Factors for the Progression of Chronic Kidney Disease after Acute Kidney Injury. Bras. J. Nefrol. 2017, 39, 239–245. [Google Scholar] [CrossRef]
  43. Xue, J.L.; Daniels, F.; Star, R.A.; Kimmel, P.L.; Eggers, P.W.; Molitoris, B.A.; Himmelfarb, J.; Collins, A.J. Incidence and Mortality of Acute Renal Failure in Medicare Beneficiaries, 1992 to 2001. J. Am. Soc. Nephrol. 2006, 17, 1135–1142. [Google Scholar] [CrossRef] [PubMed]
  44. Fischer, M.J.; Hsu, J.Y.; Lora, C.M.; Ricardo, A.C.; Anderson, A.H.; Bazzano, L.; Cuevas, M.M.; Hsu, C.; Kusek, J.W.; Renteria, A.; et al. CKD Progression and Mortality among Hispanics and Non-Hispanics. J. Am. Soc. Nephrol. 2016, 27, 3488–3497. [Google Scholar] [CrossRef] [PubMed]
  45. Parsa, A.; Kao, W.H.L.; Xie, D.; Astor, B.C.; Li, M.; Hsu, C.; Feldman, H.I.; Parekh, R.S.; Kusek, J.W.; Greene, T.H.; et al. APOL1 Risk Variants, Race, and Progression of Chronic Kidney Disease. N. Engl. J. Med. 2013, 369, 2183–2196. [Google Scholar] [CrossRef]
  46. Hannan, M.; Ansari, S.; Meza, N.; Anderson, A.H.; Srivastava, A.; Waikar, S.; Charleston, J.; Weir, M.R.; Taliercio, J.; Horwitz, E.; et al. Risk Factors for CKD Progression: Overview of Findings from the CRIC Study. Clin. J. Am. Soc. Nephrolog. 2021, 16, 648–659. [Google Scholar] [CrossRef]
  47. Taal, M.W.; Brenner, B.M. Predicting Initiation and Progression of Chronic Kidney Disease: Developing Renal Risk Scores. Kidney Int. 2006, 70, 1694–1705. [Google Scholar] [CrossRef]
  48. Vaara, S.T.; Bhatraju, P.K.; Stanski, N.L.; McMahon, B.A.; Liu, K.; Joannidis, M.; Bagshaw, S.M. Subphenotypes in Acute Kidney Injury: A Narrative Review. Crit. Care 2022, 26, 251. [Google Scholar] [CrossRef]
  49. Amdur, R.L.; Chawla, L.S.; Amodeo, S.; Kimmel, P.L.; Palant, C.E. Outcomes Following Diagnosis of Acute Renal Failure in U.S. Veterans: Focus on Acute Tubular Necrosis. Kidney Int. 2009, 76, 1089–1097. [Google Scholar] [CrossRef]
  50. Vikse, B.E.; Irgens, L.M.; Leivestad, T.; Skjærven, R.; Iversen, B.M. Preeclampsia and the Risk of End-Stage Renal Disease. N. Engl. J. Med. 2008, 359, 800–809. [Google Scholar] [CrossRef]
  51. See, E.J.; Jayasinghe, K.; Glassford, N.; Bailey, M.; Johnson, D.W.; Polkinghorne, K.R.; Toussaint, N.D.; Bellomo, R. Long-Term Risk of Adverse Outcomes after Acute Kidney Injury: A Systematic Review and Meta-Analysis of Cohort Studies Using Consensus Definitions of Exposure. Kidney Int. 2019, 95, 160–172. [Google Scholar] [CrossRef] [PubMed]
  52. Coca, S.G.; Singanamala, S.; Parikh, C.R. Chronic Kidney Disease after Acute Kidney Injury: A Systematic Review and Meta-Analysis. Kidney Int. 2012, 81, 442–448. [Google Scholar] [CrossRef] [PubMed]
  53. Ishani, A.; Nelson, D.; Clothier, B.; Schult, T.; Nugent, S.; Greer, N.; Slinin, Y.; Ensrud, K.E. The Magnitude of Acute Serum Creatinine Increase After Cardiac Surgery and the Risk of Chronic Kidney Disease, Progression of Kidney Disease, and Death. Arch. Intern. Med. 2011, 171, 226. [Google Scholar] [CrossRef]
  54. Thakar, C.V.; Christianson, A.; Himmelfarb, J.; Leonard, A.C. Acute Kidney Injury Episodes and Chronic Kidney Disease Risk in Diabetes Mellitus. Clin. J. Am. Soc. Nephrol. 2011, 6, 2567–2572. [Google Scholar] [CrossRef] [PubMed]
  55. Rifkin, D.E.; Coca, S.G.; Kalantar-Zadeh, K. Does AKI Truly Lead to CKD? J. Am. Soc. Nephrol. 2012, 23, 979–984. [Google Scholar] [CrossRef]
  56. Wen, Y.; Xu, L.; Melchinger, I.; Thiessen-Philbrook, H.; Moledina, D.G.; Coca, S.G.; Hsu, C.; Go, A.S.; Liu, K.D.; Siew, E.D.; et al. Longitudinal Biomarkers and Kidney Disease Progression after Acute Kidney Injury. JCI Insight 2023, 8, e167731. [Google Scholar] [CrossRef]
  57. Ko, G.J.; Grigoryev, D.N.; Linfert, D.; Jang, H.R.; Watkins, T.; Cheadle, C.; Racusen, L.; Rabb, H. Transcriptional Analysis of Kidneys during Repair from AKI Reveals Possible Roles for NGAL and KIM-1 as Biomarkers of AKI-to-CKD Transition. Am. J. Physiol. Ren. Physiol. 2010, 298, F1472–F1483. [Google Scholar] [CrossRef]
  58. Glassock, R.J. Uremic Toxins: What Are They? An Integrated Overview of Pathobiology and Classification. J. Ren. Nutr. 2008, 18, 2–6. [Google Scholar] [CrossRef]
  59. Rosner, M.H.; Reis, T.; Husain-Syed, F.; Vanholder, R.; Hutchison, C.; Stenvinkel, P.; Blankestijn, P.J.; Cozzolino, M.; Juillard, L.; Kashani, K.; et al. Classification of Uremic Toxins and Their Role in Kidney Failure. Clin. J. Am. Soc. Nephrol. 2021, 16, 1918–1928. [Google Scholar] [CrossRef]
  60. Vanholder, R.; De Smet, R.; Glorieux, G.; Argilés, A.; Baurmeister, U.; Brunet, P.; Clark, W.; Cohen, G.; De Deyn, P.P.; Deppisch, R.; et al. Review on Uremic Toxins: Classification, Concentration, and Interindividual Variability. Kidney Int. 2003, 63, 1934–1943. [Google Scholar] [CrossRef]
  61. Watanabe, H.; Noguchi, T.; Miyamoto, Y.; Kadowaki, D.; Kotani, S.; Nakajima, M.; Miyamura, S.; Ishima, Y.; Otagiri, M.; Maruyama, T. Interaction between Two Sulfate-Conjugated Uremic Toxins, p -Cresyl Sulfate and Indoxyl Sulfate, during Binding with Human Serum Albumin. Drug Metab. Dispos. 2012, 40, 1423–1428. [Google Scholar] [CrossRef] [PubMed]
  62. Wu, W.; Bush, K.T.; Nigam, S.K. Key Role for the Organic Anion Transporters, OAT1 and OAT3, in the in Vivo Handling of Uremic Toxins and Solutes. Sci. Rep. 2017, 7, 4939. [Google Scholar] [CrossRef] [PubMed]
  63. Takada, T.; Yamamoto, T.; Matsuo, H.; Tan, J.K.; Ooyama, K.; Sakiyama, M.; Miyata, H.; Yamanashi, Y.; Toyoda, Y.; Higashino, T.; et al. Identification of ABCG2 as an Exporter of Uremic Toxin Indoxyl Sulfate in Mice and as a Crucial Factor Influencing CKD Progression. Sci. Rep. 2018, 8, 11147. [Google Scholar] [CrossRef] [PubMed]
  64. European Uremic Toxin Work Group (EUTox) —Uremic Solutes Database. List of Uremic Solutes. Available online: https://database.uremic-toxins.org/soluteList.php (accessed on 8 August 2023).
  65. Addi, T.; Poitevin, S.; McKay, N.; El Mecherfi, K.E.; Kheroua, O.; Jourde-Chiche, N.; De Macedo, A.; Gondouin, B.; Cerini, C.; Brunet, P.; et al. Mechanisms of Tissue Factor Induction by the Uremic Toxin Indole-3 Acetic Acid through Aryl Hydrocarbon Receptor/Nuclear Factor-Kappa B Signaling Pathway in Human Endothelial Cells. Arch. Toxicol. 2019, 93, 121–136. [Google Scholar] [CrossRef]
  66. Dou, L.; Poitevin, S.; Sallée, M.; Addi, T.; Gondouin, B.; McKay, N.; Denison, M.S.; Jourde-Chiche, N.; Duval-Sabatier, A.; Cerini, C.; et al. Aryl Hydrocarbon Receptor Is Activated in Patients and Mice with Chronic Kidney Disease. Kidney Int. 2018, 93, 986–999. [Google Scholar] [CrossRef]
  67. Adesso, S.; Magnus, T.; Cuzzocrea, S.; Campolo, M.; Rissiek, B.; Paciello, O.; Autore, G.; Pinto, A.; Marzocco, S. Indoxyl Sulfate Affects Glial Function Increasing Oxidative Stress and Neuroinflammation in Chronic Kidney Disease: Interaction between Astrocytes and Microglia. Front. Pharmacol. 2017, 8, 370. [Google Scholar] [CrossRef]
  68. Bobot, M.; Thomas, L.; Moyon, A.; Fernandez, S.; McKay, N.; Balasse, L.; Garrigue, P.; Brige, P.; Chopinet, S.; Poitevin, S.; et al. Uremic Toxic Blood-Brain Barrier Disruption Mediated by AhR Activation Leads to Cognitive Impairment during Experimental Renal Dysfunction. J. Am. Soc. Nephrol. 2020, 31, 1509–1521. [Google Scholar] [CrossRef]
  69. Ichii, O.; Otsuka-Kanazawa, S.; Nakamura, T.; Ueno, M.; Kon, Y.; Chen, W.; Rosenberg, A.Z.; Kopp, J.B. Podocyte Injury Caused by Indoxyl Sulfate, a Uremic Toxin and Aryl-Hydrocarbon Receptor Ligand. PLoS ONE 2014, 9, e108448. [Google Scholar] [CrossRef]
  70. Satoh, M.; Hayashi, H.; Watanabe, M.; Ueda, K.; Yamato, H.; Yoshioka, T.; Motojima, M. Uremic Toxins Overload Accelerates Renal Damage in a Rat Model of Chronic Renal Failure. Nephron Exp. Nephrol. 2004, 95, e111–e118. [Google Scholar] [CrossRef]
  71. Han, H.; Zhu, J.; Zhu, Z.; Ni, J.; Du, R.; Dai, Y.; Chen, Y.; Wu, Z.; Lu, L.; Zhang, R. p-Cresyl Sulfate Aggravates Cardiac Dysfunction Associated with Chronic Kidney Disease by Enhancing Apoptosis of Cardiomyocytes. J. Am. Heart Assoc. 2015, 4, e001852. [Google Scholar] [CrossRef]
  72. Hénaut, L.; Grissi, M.; Brazier, F.; Assem, M.; Poirot-Leclercq, S.; Lenglet, G.; Boudot, C.; Avondo, C.; Boullier, A.; Choukroun, G.; et al. Cellular and Molecular Mechanisms Associated with Ischemic Stroke Severity in Female Mice with Chronic Kidney Disease. Sci. Rep. 2019, 9, 6432. [Google Scholar] [CrossRef] [PubMed]
  73. Karbowska, M.; Hermanowicz, J.M.; Tankiewicz-Kwedlo, A.; Kalaska, B.; Kaminski, T.W.; Nosek, K.; Wisniewska, R.J.; Pawlak, D. Neurobehavioral Effects of Uremic Toxin–Indoxyl Sulfate in the Rat Model. Sci. Rep. 2020, 10, 9483. [Google Scholar] [CrossRef] [PubMed]
  74. Liabeuf, S.; Barreto, D.V.; Barreto, F.C.; Meert, N.; Glorieux, G.; Schepers, E.; Temmar, M.; Choukroun, G.; Vanholder, R.; Massy, Z.A.; et al. Free P-Cresylsulphate Is a Predictor of Mortality in Patients at Different Stages of Chronic Kidney Disease. Nephrol. Dial. Transplant. 2010, 25, 1183–1191. [Google Scholar] [CrossRef] [PubMed]
  75. Dou, L.; Sallée, M.; Cerini, C.; Poitevin, S.; Gondouin, B.; Jourde-Chiche, N.; Fallague, K.; Brunet, P.; Calaf, R.; Dussol, B.; et al. The Cardiovascular Effect of the Uremic Solute Indole-3 Acetic Acid. J. Am. Soc. Nephrol. 2015, 26, 876–887. [Google Scholar] [CrossRef] [PubMed]
  76. Cherng, Y.-G.; Lin, C.-S.; Shih, C.-C.; Hsu, Y.-H.; Yeh, C.-C.; Hu, C.-J.; Chen, T.-L.; Liao, C.-C. Stroke Risk and Outcomes in Patients with Chronic Kidney Disease or End-Stage Renal Disease: Two Nationwide Studies. PLoS ONE 2018, 13, e0191155. [Google Scholar] [CrossRef]
  77. Tan, X.; Zou, J.; Xiang, F.; Zhang, P.; Shen, B.; Wang, Y.; Ding, X.; Cao, X. P-Cresyl Sulfate Predicts Ischemic Stroke among Patients on Hemodialysis: A Prospective Cohort Study. Dis. Markers 2022, 2022, 1358419. [Google Scholar] [CrossRef]
  78. Lin, Y.-T.; Wu, P.-H.; Liang, S.-S.; Mubanga, M.; Yang, Y.-H.; Hsu, Y.-L.; Kuo, M.-C.; Hwang, S.-J.; Kuo, P.-L. Protein-Bound Uremic Toxins Are Associated with Cognitive Function among Patients Undergoing Maintenance Hemodialysis. Sci. Rep. 2019, 9, 20388. [Google Scholar] [CrossRef]
  79. Desjardins, L.; Liabeuf, S.; Oliveira, R.B.; Louvet, L.; Kamel, S.; Lemke, H.-D.; Vanholder, R.; Choukroun, G.; Massy, Z.A. Uremic Toxicity and Sclerostin in Chronic Kidney Disease Patients. Néphrologie Thérapeutique 2014, 10, 463–470. [Google Scholar] [CrossRef]
  80. Wang, W.; Hao, G.; Pan, Y.; Ma, S.; Yang, T.; Shi, P.; Zhu, Q.; Xie, Y.; Ma, S.; Zhang, Q.; et al. Serum Indoxyl Sulfate Is Associated with Mortality in Hospital-Acquired Acute Kidney Injury: A Prospective Cohort Study. BMC Nephrol. 2019, 20, 57. [Google Scholar] [CrossRef]
  81. Veldeman, L.; Vanmassenhove, J.; Van Biesen, W.; Massy, Z.A.; Liabeuf, S.; Glorieux, G.; Vanholder, R. Evolution of Protein-Bound Uremic Toxins Indoxyl Sulphate and p-Cresyl Sulphate in Acute Kidney Injury. Int. Urol. Nephrol. 2019, 51, 293–302. [Google Scholar] [CrossRef]
  82. Mabuchi, H.; Nakahashi, H. A Major Inhibitor of Phenytoin Binding to Serum Protein in Uremia. Nephron 1988, 48, 310–314. [Google Scholar] [CrossRef] [PubMed]
  83. Dou, L.; Bertrand, E.; Cerini, C.; Faure, V.; Sampol, J.; Vanholder, R.; Berland, Y.; Brunet, P. The Uremic Solutes P-Cresol and Indoxyl Sulfate Inhibit Endothelial Proliferation and Wound Repair. Kidney Int. 2004, 65, 442–451. [Google Scholar] [CrossRef] [PubMed]
  84. Dou, L.; Jourde-Chiche, N.; Faure, V.; Cerini, C.; Berland, Y.; Dignat-George, F.; Brunet, P. The Uremic Solute Indoxyl Sulfate Induces Oxidative Stress in Endothelial Cells. J. Thromb. Haemost. 2007, 5, 1302–1308. [Google Scholar] [CrossRef] [PubMed]
  85. Gross, P.; Massy, Z.A.; Henaut, L.; Boudot, C.; Cagnard, J.; March, C.; Kamel, S.; Drueke, T.B.; Six, I. Para-Cresyl Sulfate Acutely Impairs Vascular Reactivity and Induces Vascular Remodeling: Para-Cresyl Sulfate and Vascular Dysfunction. J. Cell. Physiol. 2015, 230, 2927–2935. [Google Scholar] [CrossRef]
  86. Caillard, P.; Bennis, Y.; Six, I.; Bodeau, S.; Kamel, S.; Choukroun, G.; Maizel, J.; Titeca-Beauport, D. The Role of Gut-Derived, Protein-Bound Uremic Toxins in the Cardiovascular Complications of Acute Kidney Injury. Toxins 2022, 14, 336. [Google Scholar] [CrossRef]
  87. Popkov, V.A.; Zharikova, A.A.; Demchenko, E.A.; Andrianova, N.V.; Zorov, D.B.; Plotnikov, E.Y. Gut Microbiota as a Source of Uremic Toxins. Int. J. Mol. Sci. 2022, 23, 483. [Google Scholar] [CrossRef]
  88. Yang, J.; Kim, C.J.; Go, Y.S.; Lee, H.Y.; Kim, M.-G.; Oh, S.W.; Cho, W.Y.; Im, S.-H.; Jo, S.K. Intestinal Microbiota Control Acute Kidney Injury Severity by Immune Modulation. Kidney Int. 2020, 98, 932–946. [Google Scholar] [CrossRef]
  89. Noel, S.; Martina-Lingua, M.N.; Bandapalle, S.; Pluznick, J.; Hamad, A.R.A.; Peterson, D.A.; Rabb, H. Intestinal Microbiota-Kidney Cross Talk in Acute Kidney Injury and Chronic Kidney Disease. Nephron Clin. Pr. 2014, 127, 139–143. [Google Scholar] [CrossRef]
  90. Poesen, R.; Windey, K.; Neven, E.; Kuypers, D.; De Preter, V.; Augustijns, P.; D’Haese, P.; Evenepoel, P.; Verbeke, K.; Meijers, B. The Influence of CKD on Colonic Microbial Metabolism. J. Am. Soc. Nephrol. 2016, 27, 1389–1399. [Google Scholar] [CrossRef]
  91. Vaziri, N.D.; Wong, J.; Pahl, M.; Piceno, Y.M.; Yuan, J.; DeSantis, T.Z.; Ni, Z.; Nguyen, T.-H.; Andersen, G.L. Chronic Kidney Disease Alters Intestinal Microbial Flora. Kidney Int. 2013, 83, 308–315. [Google Scholar] [CrossRef]
  92. Rabb, H.; Griffin, M.D.; McKay, D.B.; Swaminathan, S.; Pickkers, P.; Rosner, M.H.; Kellum, J.A.; Ronco, C. Inflammation in AKI: Current Understanding, Key Questions, and Knowledge Gaps. J. Am. Soc. Nephrol. 2016, 27, 371–379. [Google Scholar] [CrossRef] [PubMed]
  93. Rydzewska-Rosołowska, A.; Sroka, N.; Kakareko, K.; Rosołowski, M.; Zbroch, E.; Hryszko, T. The Links between Microbiome and Uremic Toxins in Acute Kidney Injury: Beyond Gut Feeling—A Systematic Review. Toxins 2020, 12, 788. [Google Scholar] [CrossRef] [PubMed]
  94. Li, J.; Moturi, K.R.; Wang, L.; Zhang, K.; Yu, C. Gut Derived-Endotoxin Contributes to Inflammation in Severe Ischemic Acute Kidney Injury. BMC Nephrol. 2019, 20, 16. [Google Scholar] [CrossRef] [PubMed]
  95. Albenberg, L.; Esipova, T.V.; Judge, C.P.; Bittinger, K.; Chen, J.; Laughlin, A.; Grunberg, S.; Baldassano, R.N.; Lewis, J.D.; Li, H.; et al. Correlation Between Intraluminal Oxygen Gradient and Radial Partitioning of Intestinal Microbiota. Gastroenterology 2014, 147, 1055–1063.e8. [Google Scholar] [CrossRef]
  96. Singhal, R.; Shah, Y.M. Oxygen Battle in the Gut: Hypoxia and Hypoxia-Inducible Factors in Metabolic and Inflammatory Responses in the Intestine. J. Biol. Chem. 2020, 295, 10493–10505. [Google Scholar] [CrossRef]
  97. Kunst, C.; Schmid, S.; Michalski, M.; Tümen, D.; Buttenschön, J.; Müller, M.; Gülow, K. The Influence of Gut Microbiota on Oxidative Stress and the Immune System. Biomedicines 2023, 11, 1388. [Google Scholar] [CrossRef]
  98. Saranya, G.; Viswanathan, P. Gut Microbiota Dysbiosis in AKI to CKD Transition. Biomed. Pharmacother. 2023, 161, 114447. [Google Scholar] [CrossRef]
  99. Zhao, H.; Chen, L.; Yang, T.; Feng, Y.-L.; Vaziri, N.D.; Liu, B.-L.; Liu, Q.-Q.; Guo, Y.; Zhao, Y.-Y. Aryl Hydrocarbon Receptor Activation Mediates Kidney Disease and Renal Cell Carcinoma. J. Transl. Med. 2019, 17, 302. [Google Scholar] [CrossRef]
  100. LeBleu, V.S.; Taduri, G.; O’Connell, J.; Teng, Y.; Cooke, V.G.; Woda, C.; Sugimoto, H.; Kalluri, R. Origin and Function of Myofibroblasts in Kidney Fibrosis. Nat. Med. 2013, 19, 1047–1053. [Google Scholar] [CrossRef]
  101. Musiał, K.; Bargenda, A.; Zwolińska, D. Urine Survivin, E-Cadherin and Matrix Metalloproteinases as Novel Biomarkers in Children with Chronic Kidney Disease. Biomarkers 2015, 20, 177–182. [Google Scholar] [CrossRef]
  102. Rastaldi, M.P.; Ferrario, F.; Giardino, L.; Dell’Antonio, G.; Grillo, C.; Grillo, P.; Strutz, F.; Müller, G.A.; Colasanti, G.; D’Amico, G. Epithelial-Mesenchymal Transition of Tubular Epithelial Cells in Human Renal Biopsies. Kidney Int. 2002, 62, 137–146. [Google Scholar] [CrossRef] [PubMed]
  103. Robertson, H.; Ali, S.; McDonnell, B.J.; Burt, A.D.; Kirby, J.A. Chronic Renal Allograft Dysfunction: The Role of T Cell–Mediated Tubular Epithelial to Mesenchymal Cell Transition. J. Am. Soc. Nephrol. 2004, 15, 390–397. [Google Scholar] [CrossRef] [PubMed]
  104. Taki, K.; Nakamura, S.; Miglinas, M.; Enomoto, A.; Niwa, T. Accumulation of Indoxyl Sulfate in OAT1/3-Positive Tubular Cells in Kidneys of Patients with Chronic Renal Failure. J. Ren. Nutr. 2006, 16, 199–203. [Google Scholar] [CrossRef] [PubMed]
  105. Kim, S.H.; Yu, M.-A.; Ryu, E.S.; Jang, Y.-H.; Kang, D.-H. Indoxyl Sulfate-Induced Epithelial-to-Mesenchymal Transition and Apoptosis of Renal Tubular Cells as Novel Mechanisms of Progression of Renal Disease. Lab. Investig. 2012, 92, 488–498. [Google Scholar] [CrossRef]
  106. Sun, C.-Y.; Chang, S.-C.; Wu, M.-S. Uremic Toxins Induce Kidney Fibrosis by Activating Intrarenal Renin–Angiotensin–Aldosterone System Associated Epithelial-to-Mesenchymal Transition. PLoS ONE 2012, 7, e34026. [Google Scholar] [CrossRef]
  107. Chang, J.-F.; Hsieh, C.-Y.; Lu, K.-C.; Chen, Y.-W.; Liang, S.-S.; Lin, C.-C.; Hung, C.-F.; Liou, J.-C.; Wu, M.-S. Therapeutic Targeting of Aristolochic Acid Induced Uremic Toxin Retention, SMAD 2/3 and JNK/ERK Pathways in Tubulointerstitial Fibrosis: Nephroprotective Role of Propolis in Chronic Kidney Disease. Toxins 2020, 12, 364. [Google Scholar] [CrossRef]
  108. Milanesi, S.; Garibaldi, S.; Saio, M.; Ghigliotti, G.; Picciotto, D.; Ameri, P.; Garibotto, G.; Barisione, C.; Verzola, D. Indoxyl Sulfate Induces Renal Fibroblast Activation through a Targetable Heat Shock Protein 90-Dependent Pathway. Oxidative Med. Cell. Longev. 2019, 2019, 2050183. [Google Scholar] [CrossRef]
  109. Bolati, D.; Shimizu, H.; Higashiyama, Y.; Nishijima, F.; Niwa, T. Indoxyl Sulfate Induces Epithelial-to-Mesenchymal Transition in Rat Kidneys and Human Proximal Tubular Cells. Am. J. Nephrol. 2011, 34, 318–323. [Google Scholar] [CrossRef]
  110. Bolati, D.; Shimizu, H.; Niwa, T. AST-120 Ameliorates Epithelial-to-Mesenchymal Transition and Interstitial Fibrosis in the Kidneys of Chronic Kidney Disease Rats. J. Ren. Nutr. 2012, 22, 176–180. [Google Scholar] [CrossRef]
  111. Chen, J.-H.; Chao, C.-T.; Huang, J.-W.; Hung, K.-Y.; Liu, S.-H.; Tarng, D.-C.; Chiang, C.-K. Early Elimination of Uremic Toxin Ameliorates AKI-to-CKD Transition. Clin. Sci. 2021, 135, 2643–2658. [Google Scholar] [CrossRef]
  112. Motojima, M.; Hosokawa, A.; Yamato, H.; Muraki, T.; Yoshioka, T. Uremic Toxins of Organic Anions Up-Regulate PAI-1 Expression by Induction of NF-κB and Free Radical in Proximal Tubular Cells. Kidney Int. 2003, 63, 1671–1680. [Google Scholar] [CrossRef] [PubMed]
  113. Shimizu, H.; Yisireyili, M.; Nishijima, F.; Niwa, T. Indoxyl Sulfate Enhances P53-TGF-β1-Smad3 Pathway in Proximal Tubular Cells. Am. J. Nephrol. 2013, 37, 97–103. [Google Scholar] [CrossRef] [PubMed]
  114. Miyazaki, T.; Aoyama, I.; Ise, M.; Seo, H.; Niwa, T. An Oral Sorbent Reduces Overload of Indoxyl Sulphate and Gene Expression of TGF-β1 in Uraemic Rat Kidneys. Nephrol. Dial. Transplant. 2000, 15, 1773–1781. [Google Scholar] [CrossRef]
  115. Aoyama, I.; Shimokata, K.; Niwa, T. An Oral Adsorbent Downregulates Renal Expression of Genes That Promote Interstitial Inflammation and Fibrosis in Diabetic Rats. Nephron 2002, 92, 635–651. [Google Scholar] [CrossRef] [PubMed]
  116. Lekawanvijit, S.; Kompa, A.R.; Manabe, M.; Wang, B.H.; Langham, R.G.; Nishijima, F.; Kelly, D.J.; Krum, H. Chronic Kidney Disease-Induced Cardiac Fibrosis Is Ameliorated by Reducing Circulating Levels of a Non-Dialysable Uremic Toxin, Indoxyl Sulfate. PLoS ONE 2012, 7, e41281. [Google Scholar] [CrossRef]
  117. Shimoishi, K.; Anraku, M.; Kitamura, K.; Tasaki, Y.; Taguchi, K.; Hashimoto, M.; Fukunaga, E.; Maruyama, T.; Otagiri, M. An Oral Adsorbent, AST-120 Protects Against the Progression of Oxidative Stress by Reducing the Accumulation of Indoxyl Sulfate in the Systemic Circulation in Renal Failure. Pharm. Res. 2007, 24, 1283–1289. [Google Scholar] [CrossRef]
  118. Shimizu, H.; Saito, S.; Higashiyama, Y.; Nishijima, F.; Niwa, T. CREB, NF-κB, and NADPH Oxidase Coordinately Upregulate Indoxyl Sulfate-Induced Angiotensinogen Expression in Proximal Tubular Cells. Am. J. Physiol. Cell Physiol. 2013, 304, C685–C692. [Google Scholar] [CrossRef]
  119. Bolati, D.; Shimizu, H.; Yisireyili, M.; Nishijima, F.; Niwa, T. Indoxyl Sulfate, a Uremic Toxin, Downregulates Renal Expression of Nrf2 through Activation of NF-κB. BMC Nephrol. 2013, 14, 56. [Google Scholar] [CrossRef]
  120. Nakagawa, N.; Hasebe, N.; Sumitomo, K.; Fujino, T.; Fukuzawa, J.; Hirayama, T.; Kikuchi, K. An Oral Adsorbent, AST-120, Suppresses Oxidative Stress in Uremic Rats. Am. J. Nephrol. 2006, 26, 455–461. [Google Scholar] [CrossRef]
  121. Palm, F.; Nangaku, M.; Fasching, A.; Tanaka, T.; Nordquist, L.; Hansell, P.; Kawakami, T.; Nishijima, F.; Fujita, T. Uremia Induces Abnormal Oxygen Consumption in Tubules and Aggravates Chronic Hypoxia of the Kidney via Oxidative Stress. Am. J. Physiol. Ren. Physiol. 2010, 299, F380–F386. [Google Scholar] [CrossRef]
  122. Mutsaers, H.A.M.; Wilmer, M.J.G.; Reijnders, D.; Jansen, J.; Van Den Broek, P.H.H.; Forkink, M.; Schepers, E.; Glorieux, G.; Vanholder, R.; Van Den Heuvel, L.P.; et al. Uremic Toxins Inhibit Renal Metabolic Capacity through Interference with Glucuronidation and Mitochondrial Respiration. Biochim. Biophys. Acta BBA Mol. Basis Dis. 2013, 1832, 142–150. [Google Scholar] [CrossRef] [PubMed]
  123. Sun, C.-Y.; Cheng, M.-L.; Pan, H.-C.; Lee, J.-H.; Lee, C.-C. Protein-Bound Uremic Toxins Impaired Mitochondrial Dynamics and Functions. Oncotarget 2017, 8, 77722–77733. [Google Scholar] [CrossRef] [PubMed]
  124. Jeon, B.-J.; Yang, H.-M.; Lyu, Y.-S.; Pae, H.-O.; Ju, S.-M.; Jeon, B.-H. Apigenin Inhibits Indoxyl Sulfate-Induced Endoplasmic Reticulum Stress and Anti-Proliferative Pathways, CHOP and IL-6/P21, in Human Renal Proximal Tubular Cells. Eur. Rev. Med. Pharmacol. Sci. 2015, 19, 2303–2310. [Google Scholar] [PubMed]
  125. Kawakami, T.; Inagi, R.; Wada, T.; Tanaka, T.; Fujita, T.; Nangaku, M. Indoxyl Sulfate Inhibits Proliferation of Human Proximal Tubular Cells via Endoplasmic Reticulum Stress. Am. J. Physiol. Ren. Physiol. 2010, 299, F568–F576. [Google Scholar] [CrossRef]
  126. Kim, H.Y.; Kim, D.H.; Lee, S.J.; Kang, Y.J.; Kim, G.; Koh, H.B.; Ko, Y.E.; Shin, H.M.; Lee, H.; Yoo, T.-H.; et al. Uremic Toxin Indoxyl Sulfate Induces Trained Immunity via the AhR-Dependent Arachidonic Acid Pathway in ESRD. bioRxiv 2022. [Google Scholar] [CrossRef]
  127. Hopp, K.; Kleczko, E.K.; Gitomer, B.Y.; Chonchol, M.; Klawitter, J.; Christians, U.; Klawitter, J. Metabolic Reprogramming in a Slowly Developing Orthologous Model of Polycystic Kidney Disease. Am. J. Physiol. Ren. Physiol. 2022, 322, F258–F267. [Google Scholar] [CrossRef]
  128. Yu, Y.; Guan, X.; Nie, L.; Liu, Y.; He, T.; Xiong, J.; Xu, X.; Li, Y.; Yang, K.; Wang, Y.; et al. DNA Hypermethylation of sFRP5 Contributes to Indoxyl Sulfate-Induced Renal Fibrosis. J. Mol. Med. 2017, 95, 601–613. [Google Scholar] [CrossRef]
  129. Sun, C.-Y.; Chang, S.-C.; Wu, M.-S. Suppression of Klotho Expression by Protein-Bound Uremic Toxins Is Associated with Increased DNA Methyltransferase Expression and DNA Hypermethylation. Kidney Int. 2012, 81, 640–650. [Google Scholar] [CrossRef]
  130. Shimizu, H.; Bolati, D.; Adijiang, A.; Adelibieke, Y.; Muteliefu, G.; Enomoto, A.; Higashiyama, Y.; Higuchi, Y.; Nishijima, F.; Niwa, T. Indoxyl Sulfate Downregulates Renal Expression of Klotho through Production of ROS and Activation of Nuclear Factor-ĸB. Am. J. Nephrol. 2011, 33, 319–324. [Google Scholar] [CrossRef]
  131. Shimizu, H.; Bolati, D.; Adijiang, A.; Muteliefu, G.; Enomoto, A.; Nishijima, F.; Dateki, M.; Niwa, T. NF-κB Plays an Important Role in Indoxyl Sulfate-Induced Cellular Senescence, Fibrotic Gene Expression, and Inhibition of Proliferation in Proximal Tubular Cells. Am. J. Physiol. Cell Physiol. 2011, 301, C1201–C1212. [Google Scholar] [CrossRef]
  132. Shimizu, H.; Yisireyili, M.; Nishijima, F.; Niwa, T. Stat3 Contributes to Indoxyl Sulfate-Induced Inflammatory and Fibrotic Gene Expression and Cellular Senescence. Am. J. Nephrol. 2012, 36, 184–189. [Google Scholar] [CrossRef] [PubMed]
  133. Sun, C.-Y.; Hsu, H.-H.; Wu, M.-S. P-Cresol Sulfate and Indoxyl Sulfate Induce Similar Cellular Inflammatory Gene Expressions in Cultured Proximal Renal Tubular Cells. Nephrol. Dial. Transplant. 2013, 28, 70–78. [Google Scholar] [CrossRef] [PubMed]
  134. Yisireyili, M.; Takeshita, K.; Saito, S.; Murohara, T.; Niwa, T. Indole-3-Propionic Acid Suppresses Indoxyl Sulfate-Induced Expression of Fibrotic and Inflammatory Genes in Proximal Tubular Cells. Nagoya J. Med. Sci. 2017, 79, 477. [Google Scholar] [CrossRef]
  135. Ito, S.; Higuchi, Y.; Yagi, Y.; Nishijima, F.; Yamato, H.; Ishii, H.; Osaka, M.; Yoshida, M. Reduction of Indoxyl Sulfate by AST-120 Attenuates Monocyte Inflammation Related to Chronic Kidney Disease. J. Leukoc. Biol. 2013, 93, 837–845. [Google Scholar] [CrossRef] [PubMed]
  136. Barisione, C.; Garibaldi, S.; Furfaro, A.L.; Nitti, M.; Palmieri, D.; Passalacqua, M.; Garuti, A.; Verzola, D.; Parodi, A.; Ameri, P.; et al. Moderate Increase of Indoxyl Sulfate Promotes Monocyte Transition into Profibrotic Macrophages. PLoS ONE 2016, 11, e0149276. [Google Scholar] [CrossRef]
  137. Shimizu, H.; Bolati, D.; Higashiyama, Y.; Nishijima, F.; Shimizu, K.; Niwa, T. Indoxyl Sulfate Upregulates Renal Expression of MCP-1 via Production of ROS and Activation of NF-κB, P53, ERK, and JNK in Proximal Tubular Cells. Life Sci. 2012, 90, 525–530. [Google Scholar] [CrossRef]
  138. Hamano, H.; Ikeda, Y.; Watanabe, H.; Horinouchi, Y.; Izawa-Ishizawa, Y.; Imanishi, M.; Zamami, Y.; Takechi, K.; Miyamoto, L.; Ishizawa, K.; et al. The Uremic Toxin Indoxyl Sulfate Interferes with Iron Metabolism by Regulating Hepcidin in Chronic Kidney Disease. Nephrol. Dial. Transplant. 2018, 33, 586–597. [Google Scholar] [CrossRef]
  139. Ahmed, M.S.E.; Abed, M.; Voelkl, J.; Lang, F. Triggering of Suicidal Erythrocyte Death by Uremic Toxin Indoxyl Sulfate. BMC Nephrol. 2013, 14, 244. [Google Scholar] [CrossRef]
  140. Tozoni, S.S.; Dias, G.F.; Bohnen, G.; Grobe, N.; Pecoits-Filho, R.; Kotanko, P.; Moreno-Amaral, A.N. Uremia and Hypoxia Independently Induce Eryptosis and Erythrocyte Redox Imbalance. Cell Physiol. Biochem. 2019, 53, 794–804. [Google Scholar] [CrossRef]
  141. Gao, C.; Ji, S.; Dong, W.; Qi, Y.; Song, W.; Cui, D.; Shi, J. Indolic Uremic Solutes Enhance Procoagulant Activity of Red Blood Cells through Phosphatidylserine Exposure and Microparticle Release. Toxins 2015, 7, 4390–4403. [Google Scholar] [CrossRef]
  142. Rutkowski, P.; Maria Słomińska, E.; Szołkiewicz, M.; Aleksandrowicz, E.; Tomasz Smoleński, R.; Wołyniec, W.; Renke, M.; Wisterowicz, K.; Świerczyński, J.; Rutkowski, B. Relationship between Uremic Toxins and Oxidative Stress in Patients with Chronic Renal Failure. Scand. J. Urol. Nephrol. 2007, 41, 243–248. [Google Scholar] [CrossRef] [PubMed]
  143. Niwa, T.; Aiuchi, T.; Nakaya, K.; Emoto, Y.; Miyazaki, T.; Maeda, K. Inhibition of Mitochondrial Respiration by Furancarboxylic Acid Accumulated in Uremic Serum in Its Albumin-Bound and Non-Dialyzable Form. Clin. Nephrol. 1993, 39, 92–96. [Google Scholar] [PubMed]
  144. Park, J.S.; Kim, D.-H.; Choi, H.-I.; Kim, C.S.; Bae, E.H.; Ma, S.K.; Kim, S.W. 3-Carboxy-4-Methyl-5-Propyl-2-Furanpropanoic Acid (CMPF) Induces Cell Death through Ferroptosis and Acts as a Trigger of Apoptosis in Kidney Cells. Cell Death Dis. 2023, 14, 78. [Google Scholar] [CrossRef]
  145. Okada, A.; Nangaku, M.; Jao, T.-M.; Maekawa, H.; Ishimono, Y.; Kawakami, T.; Inagi, R. D-Serine, a Novel Uremic Toxin, Induces Senescence in Human Renal Tubular Cells via GCN2 Activation. Sci. Rep. 2017, 7, 11168. [Google Scholar] [CrossRef] [PubMed]
  146. Mihout, F.; Shweke, N.; Bigé, N.; Jouanneau, C.; Dussaule, J.-C.; Ronco, P.; Chatziantoniou, C.; Boffa, J.-J. Asymmetric Dimethylarginine (ADMA) Induces Chronic Kidney Disease through a Mechanism Involving Collagen and TGF-Β1 Synthesis. J. Pathol. 2011, 223, 37–45. [Google Scholar] [CrossRef]
  147. Schurek, H.J.; Jost, U.; Baumgartl, H.; Bertram, H.; Heckmann, U. Evidence for a Preglomerular Oxygen Diffusion Shunt in Rat Renal Cortex. Am. J. Physiol. Ren. Physiol. 1990, 259, F910–F915. [Google Scholar] [CrossRef]
  148. Welch, W.J.; Baumgärtl, H.; Lübbers, D.; Wilcox, C.S. Nephron pO2 and Renal Oxygen Usage in the Hypertensive Rat Kidney. Kidney Int. 2001, 59, 230–237. [Google Scholar] [CrossRef]
  149. Evans, R.G.; Gardiner, B.S.; Smith, D.W.; O’Connor, P.M. Intrarenal Oxygenation: Unique Challenges and the Biophysical Basis of Homeostasis. Am. J. Physiol. Ren. Physiol. 2008, 295, F1259–F1270. [Google Scholar] [CrossRef]
  150. Mimura, I.; Nangaku, M. The Suffocating Kidney: Tubulointerstitial Hypoxia in End-Stage Renal Disease. Nat. Rev. Nephrol. 2010, 6, 667–678. [Google Scholar] [CrossRef]
  151. Shoji, K.; Tanaka, T.; Nangaku, M. Role of Hypoxia in Progressive Chronic Kidney Disease and Implications for Therapy. Curr. Opin. Nephrol. Hypertens. 2014, 23, 161–168. [Google Scholar] [CrossRef]
  152. Orphanides, C.; Fine, L.G.; Norman, J.T. Hypoxia Stimulates Proximal Tubular Cell Matrix Production via a TGF-Β1-Independent Mechanism. Kidney Int. 1997, 52, 637–647. [Google Scholar] [CrossRef] [PubMed]
  153. Manotham, K.; Tanaka, T.; Matsumoto, M.; Ohse, T.; Inagi, R.; Miyata, T.; Kurokawa, K.; Fujita, T.; Ingelfinger, J.R.; Nangaku, M. Transdifferentiation of Cultured Tubular Cells Induced by Hypoxia. Kidney Int. 2004, 65, 871–880. [Google Scholar] [CrossRef] [PubMed]
  154. Samarakoon, R.; Dobberfuhl, A.D.; Cooley, C.; Overstreet, J.M.; Patel, S.; Goldschmeding, R.; Meldrum, K.K.; Higgins, P.J. Induction of Renal Fibrotic Genes by TGF-Β1 Requires EGFR Activation, P53 and Reactive Oxygen Species. Cell. Signal. 2013, 25, 2198–2209. [Google Scholar] [CrossRef]
  155. Han, W.-Q.; Zhu, Q.; Hu, J.; Li, P.-L.; Zhang, F.; Li, N. Hypoxia-Inducible Factor Prolyl-Hydroxylase-2 Mediates Transforming Growth Factor Beta 1-Induced Epithelial–Mesenchymal Transition in Renal Tubular Cells. Biochim. Biophys. Acta BBA Mol. Cell Res. 2013, 1833, 1454–1462. [Google Scholar] [CrossRef] [PubMed]
  156. Kim, J.; Seok, Y.M.; Jung, K.-J.; Park, K.M. Reactive Oxygen Species/Oxidative Stress Contributes to Progression of Kidney Fibrosis Following Transient Ischemic Injury in Mice. Am. J. Physiol. Ren. Physiol. 2009, 297, F461–F470. [Google Scholar] [CrossRef] [PubMed]
  157. Nezu, M.; Souma, T.; Yu, L.; Suzuki, T.; Saigusa, D.; Ito, S.; Suzuki, N.; Yamamoto, M. Transcription Factor Nrf2 Hyperactivation in Early-Phase Renal Ischemia-Reperfusion Injury Prevents Tubular Damage Progression. Kidney Int. 2017, 91, 387–401. [Google Scholar] [CrossRef] [PubMed]
  158. Ellis, R.J.; Small, D.M.; Vesey, D.A.; Johnson, D.W.; Francis, R.; Vitetta, L.; Gobe, G.C.; Morais, C. Indoxyl Sulphate and Kidney Disease: Causes, Consequences and Interventions: Indoxyl Sulphate and the Kidneys. Nephrology 2016, 21, 170–177. [Google Scholar] [CrossRef]
  159. Pieniazek, A.; Bernasinska-Slomczewska, J.; Gwozdzinski, L. Uremic Toxins and Their Relation with Oxidative Stress Induced in Patients with CKD. Int. J. Mol. Sci. 2021, 22, 6196. [Google Scholar] [CrossRef]
  160. Vida, C.; Oliva, C.; Yuste, C.; Ceprián, N.; Caro, P.J.; Valera, G.; González De Pablos, I.; Morales, E.; Carracedo, J. Oxidative Stress in Patients with Advanced CKD and Renal Replacement Therapy: The Key Role of Peripheral Blood Leukocytes. Antioxidants 2021, 10, 1155. [Google Scholar] [CrossRef]
  161. Suomalainen, A.; Battersby, B.J. Mitochondrial Diseases: The Contribution of Organelle Stress Responses to Pathology. Nat. Rev. Mol. Cell Biol. 2018, 19, 77–92. [Google Scholar] [CrossRef]
  162. Che, R.; Yuan, Y.; Huang, S.; Zhang, A. Mitochondrial Dysfunction in the Pathophysiology of Renal Diseases. Am. J. Physiol. Ren. Physiol. 2014, 306, F367–F378. [Google Scholar] [CrossRef] [PubMed]
  163. Aparicio-Trejo, O.E.; Avila-Rojas, S.H.; Tapia, E.; Rojas-Morales, P.; León-Contreras, J.C.; Martínez-Klimova, E.; Hernández-Pando, R.; Sánchez- Lozada, L.G.; Pedraza-Chaverri, J. Chronic Impairment of Mitochondrial Bioenergetics and β-Oxidation Promotes Experimental AKI-to-CKD Transition Induced by Folic Acid. Free Radic. Biol. Med. 2020, 154, 18–32. [Google Scholar] [CrossRef] [PubMed]
  164. Perry, H.M.; Huang, L.; Wilson, R.J.; Bajwa, A.; Sesaki, H.; Yan, Z.; Rosin, D.L.; Kashatus, D.F.; Okusa, M.D. Dynamin-Related Protein 1 Deficiency Promotes Recovery from AKI. J. Am. Soc. Nephrol. 2018, 29, 194–206. [Google Scholar] [CrossRef] [PubMed]
  165. Birk, A.V.; Liu, S.; Soong, Y.; Mills, W.; Singh, P.; Warren, J.D.; Seshan, S.V.; Pardee, J.D.; Szeto, H.H. The Mitochondrial-Targeted Compound SS-31 Re-Energizes Ischemic Mitochondria by Interacting with Cardiolipin. J. Am. Soc. Nephrol. 2013, 24, 1250–1261. [Google Scholar] [CrossRef]
  166. Hu, Q.; Ren, J.; Ren, H.; Wu, J.; Wu, X.; Liu, S.; Wang, G.; Gu, G.; Guo, K.; Li, J. Urinary Mitochondrial DNA Identifies Renal Dysfunction and Mitochondrial Damage in Sepsis-Induced Acute Kidney Injury. Oxidative Med. Cell. Longev. 2018, 2018, 8074936. [Google Scholar] [CrossRef]
  167. Gallazzini, M.; Pallet, N. Endoplasmic Reticulum Stress and Kidney Dysfunction: Endoplasmic Reticulum Stress. Biol. Cell 2018, 110, 205–216. [Google Scholar] [CrossRef]
  168. Mo, J.-S.; Choi, D.; Han, Y.-R.; Kim, N.; Jeong, H.-S. Morin Has Protective Potential against ER Stress Induced Apoptosis in Renal Proximal Tubular HK-2 Cells. Biomed. Pharmacother. 2019, 112, 108659. [Google Scholar] [CrossRef]
  169. Porter, A.W.; Nguyen, D.N.; Clayton, D.R.; Ruiz, W.G.; Mutchler, S.M.; Ray, E.C.; Marciszyn, A.L.; Nkashama, L.J.; Subramanya, A.R.; Gingras, S.; et al. The Molecular Chaperone GRP170 Protects against ER Stress and Acute Kidney Injury in Mice. JCI Insight 2022, 7, e151869. [Google Scholar] [CrossRef]
  170. Fan, Y.; Xiao, W.; Lee, K.; Salem, F.; Wen, J.; He, L.; Zhang, J.; Fei, Y.; Cheng, D.; Bao, H.; et al. Inhibition of Reticulon-1A–Mediated Endoplasmic Reticulum Stress in Early AKI Attenuates Renal Fibrosis Development. J. Am. Soc. Nephrolog. 2017, 28, 2007–2021. [Google Scholar] [CrossRef]
  171. Kim, Y.; Park, S.-J.; Manson, S.R.; Molina, C.A.F.; Kidd, K.; Thiessen-Philbrook, H.; Perry, R.J.; Liapis, H.; Kmoch, S.; Parikh, C.R.; et al. Elevated Urinary CRELD2 Is Associated with Endoplasmic Reticulum Stress–Mediated Kidney Disease. JCI Insight 2017, 2, e92896. [Google Scholar] [CrossRef]
  172. Popkov, V.A.; Silachev, D.N.; Zalevsky, A.O.; Zorov, D.B.; Plotnikov, E.Y. Mitochondria as a Source and a Target for Uremic Toxins. Int. J. Mol. Sci. 2019, 20, 3094. [Google Scholar] [CrossRef] [PubMed]
  173. Thome, T.; Kumar, R.A.; Burke, S.K.; Khattri, R.B.; Salyers, Z.R.; Kelley, R.C.; Coleman, M.D.; Christou, D.D.; Hepple, R.T.; Scali, S.T.; et al. Impaired Muscle Mitochondrial Energetics Is Associated with Uremic Metabolite Accumulation in Chronic Kidney Disease. JCI Insight 2021, 6, e139826. [Google Scholar] [CrossRef]
  174. Nishikawa, M.; Ishimori, N.; Takada, S.; Saito, A.; Kadoguchi, T.; Furihata, T.; Fukushima, A.; Matsushima, S.; Yokota, T.; Kinugawa, S.; et al. AST-120 Ameliorates Lowered Exercise Capacity and Mitochondrial Biogenesis in the Skeletal Muscle from Mice with Chronic Kidney Disease via Reducing Oxidative Stress. Nephrol. Dial. Transplant. 2015, 30, 934–942. [Google Scholar] [CrossRef] [PubMed]
  175. Curran, C.S.; Kopp, J.B. The Complexity of Nicotinamide Adenine Dinucleotide (NAD), Hypoxic, and Aryl Hydrocarbon Receptor Cell Signaling in Chronic Kidney Disease. J. Transl. Med. 2023, 21, 706. [Google Scholar] [CrossRef]
  176. Hasegawa, S.; Inagi, R. Harnessing Metabolomics to Describe the Pathophysiology Underlying Progression in Diabetic Kidney Disease. Curr. Diab. Rep. 2021, 21, 21. [Google Scholar] [CrossRef] [PubMed]
  177. Saxena, S.; Dagar, N.; Shelke, V.; Lech, M.; Khare, P.; Gaikwad, A.B. Metabolic Reprogramming: Unveiling the Therapeutic Potential of Targeted Therapies against Kidney Disease. Drug Discov. Today 2023, 28, 103765. [Google Scholar] [CrossRef]
  178. Schaub, J.A.; Venkatachalam, M.A.; Weinberg, J.M. Proximal Tubular Oxidative Metabolism in Acute Kidney Injury and the Transition to CKD. Kidney360 2021, 2, 355–364. [Google Scholar] [CrossRef]
  179. Li, Z.; Lu, S.; Li, X. The Role of Metabolic Reprogramming in Tubular Epithelial Cells during the Progression of Acute Kidney Injury. Cell. Mol. Life Sci. 2021, 78, 5731–5741. [Google Scholar] [CrossRef]
  180. Florens, N.; Calzada, C.; Lyasko, E.; Juillard, L.; Soulage, C. Modified Lipids and Lipoproteins in Chronic Kidney Disease: A New Class of Uremic Toxins. Toxins 2016, 8, 376. [Google Scholar] [CrossRef]
  181. Tanaka, T. Epigenetic Changes Mediating Transition to Chronic Kidney Disease: Hypoxic Memory. Acta Physiol. 2018, 222, e13023. [Google Scholar] [CrossRef]
  182. Rodríguez-Romo, R.; Berman, N.; Gómez, A.; Bobadilla, N.A. Epigenetic Regulation in the Acute Kidney Injury to Chronic Kidney Disease Transition. Nephrology 2015, 20, 736–743. [Google Scholar] [CrossRef] [PubMed]
  183. Mar, D.; Gharib, S.A.; Zager, R.A.; Johnson, A.; Denisenko, O.; Bomsztyk, K. Heterogeneity of Epigenetic Changes at Ischemia/Reperfusion- and Endotoxin-Induced Acute Kidney Injury Genes. Kidney Int. 2015, 88, 734–744. [Google Scholar] [CrossRef] [PubMed]
  184. Tampe, B.; Steinle, U.; Tampe, D.; Carstens, J.L.; Korsten, P.; Zeisberg, E.M.; Müller, G.A.; Kalluri, R.; Zeisberg, M. Low-Dose Hydralazine Prevents Fibrosis in a Murine Model of Acute Kidney Injury–to–Chronic Kidney Disease Progression. Kidney Int. 2017, 91, 157–176. [Google Scholar] [CrossRef] [PubMed]
  185. Bechtel, W.; McGoohan, S.; Zeisberg, E.M.; Müller, G.A.; Kalbacher, H.; Salant, D.J.; Müller, C.A.; Kalluri, R.; Zeisberg, M. Methylation Determines Fibroblast Activation and Fibrogenesis in the Kidney. Nat. Med. 2010, 16, 544–550. [Google Scholar] [CrossRef] [PubMed]
  186. Soofi, A.; Kutschat, A.P.; Azam, M.; Laszczyk, A.M.; Dressler, G.R. Regeneration after Acute Kidney Injury Requires PTIP-Mediated Epigenetic Modifications. JCI Insight 2020, 5, e130204. [Google Scholar] [CrossRef] [PubMed]
  187. Ko, Y.-A.; Mohtat, D.; Suzuki, M.; Park, A.; Izquierdo, M.; Han, S.; Kang, H.; Si, H.; Hostetter, T.; Pullman, J.M.; et al. Cytosine Methylation Changes in Enhancer Regions of Core Pro-Fibrotic Genes Characterize Kidney Fibrosis Development. Genome Biol. 2013, 14, R108. [Google Scholar] [CrossRef]
  188. Kang, S.W.; Shih, P.B.; Mathew, R.O.; Mahata, M.; Biswas, N.; Rao, F.; Yan, L.; Bouchard, J.; Malhotra, R.; Tolwani, A.; et al. Renal Kallikrein Excretion and Epigenetics in Human Acute Kidney Injury: Expression, Mechanisms and Consequences. BMC Nephrol. 2011, 12, 27. [Google Scholar] [CrossRef]
  189. Kölling, M.; Seeger, H.; Haddad, G.; Kistler, A.; Nowak, A.; Faulhaber-Walter, R.; Kielstein, J.; Haller, H.; Fliser, D.; Mueller, T.; et al. The Circular RNA ciRs-126 Predicts Survival in Critically Ill Patients with Acute Kidney Injury. Kidney Int. Rep. 2018, 3, 1144–1152. [Google Scholar] [CrossRef]
  190. Wing, M.R.; Devaney, J.M.; Joffe, M.M.; Xie, D.; Feldman, H.I.; Dominic, E.A.; Guzman, N.J.; Ramezani, A.; Susztak, K.; Herman, J.G.; et al. DNA Methylation Profile Associated with Rapid Decline in Kidney Function: Findings from the CRIC Study. Nephrol. Dial. Transplant. 2014, 29, 864–872. [Google Scholar] [CrossRef]
  191. Canaud, G.; Bonventre, J.V. Cell Cycle Arrest and the Evolution of Chronic Kidney Disease from Acute Kidney Injury. Nephrol. Dial. Transplant. 2015, 30, 575–583. [Google Scholar] [CrossRef]
  192. Crépin, T.; Legendre, M.; Carron, C.; Vachey, C.; Courivaud, C.; Rebibou, J.-M.; Ferrand, C.; Laheurte, C.; Vauchy, C.; Gaiffe, E.; et al. Uraemia-Induced Immune Senescence and Clinical Outcomes in Chronic Kidney Disease Patients. Nephrol. Dial. Transplant. 2020, 35, 624–632. [Google Scholar] [CrossRef] [PubMed]
  193. Yu, J.-B.; Lee, D.-S.; Padanilam, B.J.; Kim, J. Repeated Administration of Cisplatin Transforms Kidney Fibroblasts through G2/M Arrest and Cellular Senescence. Cells 2022, 11, 3472. [Google Scholar] [CrossRef] [PubMed]
  194. Castellano, G.; Franzin, R.; Sallustio, F.; Stasi, A.; Banelli, B.; Romani, M.; De Palma, G.; Lucarelli, G.; Divella, C.; Battaglia, M.; et al. Complement Component C5a Induces Aberrant Epigenetic Modifications in Renal Tubular Epithelial Cells Accelerating Senescence by Wnt4/Βcatenin Signaling after Ischemia/Reperfusion Injury. Aging 2019, 11, 4382–4406. [Google Scholar] [CrossRef] [PubMed]
  195. Yang, L.; Besschetnova, T.Y.; Brooks, C.R.; Shah, J.V.; Bonventre, J.V. Epithelial Cell Cycle Arrest in G2/M Mediates Kidney Fibrosis after Injury. Nat. Med. 2010, 16, 535–543. [Google Scholar] [CrossRef] [PubMed]
  196. Canaud, G.; Brooks, C.R.; Kishi, S.; Taguchi, K.; Nishimura, K.; Magassa, S.; Scott, A.; Hsiao, L.-L.; Ichimura, T.; Terzi, F.; et al. Cyclin G1 and TASCC Regulate Kidney Epithelial Cell G 2 -M Arrest and Fibrotic Maladaptive Repair. Sci. Transl. Med. 2019, 11, eaav4754. [Google Scholar] [CrossRef] [PubMed]
  197. Li, C.; Xie, N.; Li, Y.; Liu, C.; Hou, F.F.; Wang, J. N-Acetylcysteine Ameliorates Cisplatin-Induced Renal Senescence and Renal Interstitial Fibrosis through Sirtuin1 Activation and P53 Deacetylation. Free Radic. Biol. Med. 2019, 130, 512–527. [Google Scholar] [CrossRef]
  198. Kashani, K.; Al-Khafaji, A.; Ardiles, T.; Artigas, A.; Bagshaw, S.M.; Bell, M.; Bihorac, A.; Birkhahn, R.; Cely, C.M.; Chawla, L.S.; et al. Discovery and Validation of Cell Cycle Arrest Biomarkers in Human Acute Kidney Injury. Crit. Care 2013, 17, R25. [Google Scholar] [CrossRef]
  199. Lacroix, J.S.; Urena-Torres, P. Potentielle application de l’axe fibroblast growth factor 23-Klotho dans la maladie rénale chronique. Néphrologie Thérapeutique 2020, 16, 83–92. [Google Scholar] [CrossRef]
  200. Gewin, L.; Vadivelu, S.; Neelisetty, S.; Srichai, M.B.; Paueksakon, P.; Pozzi, A.; Harris, R.C.; Zent, R. Deleting the TGF-β Receptor Attenuates Acute Proximal Tubule Injury. J. Am. Soc. Nephrol. 2012, 23, 2001–2011. [Google Scholar] [CrossRef]
  201. Chung, S.; Overstreet, J.M.; Li, Y.; Wang, Y.; Niu, A.; Wang, S.; Fan, X.; Sasaki, K.; Jin, G.-N.; Khodo, S.N.; et al. TGF-β Promotes Fibrosis after Severe Acute Kidney Injury by Enhancing Renal Macrophage Infiltration. JCI Insight 2018, 3, e123563. [Google Scholar] [CrossRef]
  202. Yang, Q.; Ren, G.; Wei, B.; Jin, J.; Huang, X.R.; Shao, W.; Li, J.; Meng, X.; Lan, H.Y. Conditional Knockout of TGF-βRII /Smad2 Signals Protects against Acute Renal Injury by Alleviating Cell Necroptosis, Apoptosis and Inflammation. Theranostics 2019, 9, 8277–8293. [Google Scholar] [CrossRef] [PubMed]
  203. Gewin, L.S. Transforming Growth Factor-β in the Acute Kidney Injury to Chronic Kidney Disease Transition. Nephron 2019, 143, 154–157. [Google Scholar] [CrossRef] [PubMed]
  204. Hills, C.E.; Squires, P.E. The Role of TGF-β and Epithelial-to Mesenchymal Transition in Diabetic Nephropathy. Cytokine Growth Factor. Rev. 2011, 22, 131–139. [Google Scholar] [CrossRef] [PubMed]
  205. Ko, G.J.; Boo, C.-S.; Jo, S.-K.; Cho, W.Y.; Kim, H.K. Macrophages Contribute to the Development of Renal Fibrosis Following Ischaemia/Reperfusion-Induced Acute Kidney Injury. Nephrol. Dial. Transplant. 2007, 23, 842–852. [Google Scholar] [CrossRef]
  206. Fontecha-Barriuso, M.; Martín-Sánchez, D.; Martinez-Moreno, J.M.; Carrasco, S.; Ruiz-Andrés, O.; Monsalve, M.; Sanchez-Ramos, C.; Gómez, M.J.; Ruiz-Ortega, M.; Sánchez-Niño, M.D.; et al. PGC-1α Deficiency Causes Spontaneous Kidney Inflammation and Increases the Severity of Nephrotoxic AKI. J. Pathol. 2019, 249, 65–78. [Google Scholar] [CrossRef]
  207. Maekawa, H.; Inoue, T.; Ouchi, H.; Jao, T.-M.; Inoue, R.; Nishi, H.; Fujii, R.; Ishidate, F.; Tanaka, T.; Tanaka, Y.; et al. Mitochondrial Damage Causes Inflammation via cGAS-STING Signaling in Acute Kidney Injury. Cell Rep. 2019, 29, 1261–1273.e6. [Google Scholar] [CrossRef]
  208. Lerolle, N.; Nochy, D.; Guérot, E.; Bruneval, P.; Fagon, J.-Y.; Diehl, J.-L.; Hill, G. Histopathology of Septic Shock Induced Acute Kidney Injury: Apoptosis and Leukocytic Infiltration. Intensive Care Med. 2010, 36, 471–478. [Google Scholar] [CrossRef]
  209. Murashima, M.; Nishimoto, M.; Kokubu, M.; Hamano, T.; Matsui, M.; Eriguchi, M.; Samejima, K.; Akai, Y.; Tsuruya, K. Inflammation as a Predictor of Acute Kidney Injury and Mediator of Higher Mortality after Acute Kidney Injury in Non-Cardiac Surgery. Sci. Rep. 2019, 9, 20260. [Google Scholar] [CrossRef]
  210. Wang, J.; Liu, Y.; Wang, Y.; Sun, L. The Cross-Link between Ferroptosis and Kidney Diseases. Oxidative Med. Cell. Longev. 2021, 2021, 6654887. [Google Scholar] [CrossRef]
  211. Guo, R.; Duan, J.; Pan, S.; Cheng, F.; Qiao, Y.; Feng, Q.; Liu, D.; Liu, Z. The Road from AKI to CKD: Molecular Mechanisms and Therapeutic Targets of Ferroptosis. Cell Death Dis. 2023, 14, 426. [Google Scholar] [CrossRef]
  212. Xie, Y.; Hou, W.; Song, X.; Yu, Y.; Huang, J.; Sun, X.; Kang, R.; Tang, D. Ferroptosis: Process and Function. Cell Death Differ. 2016, 23, 369–379. [Google Scholar] [CrossRef] [PubMed]
  213. Wang, H.; Liu, C.; Zhao, Y.; Gao, G. Mitochondria Regulation in Ferroptosis. Eur. J. Cell Biol. 2020, 99, 151058. [Google Scholar] [CrossRef] [PubMed]
  214. Liu, Y.; Wang, J. Ferroptosis, a Rising Force against Renal Fibrosis. Oxidative Med. Cell. Longev. 2022, 2022, 7686956. [Google Scholar] [CrossRef] [PubMed]
  215. Zhou, L.; Xue, X.; Hou, Q.; Dai, C. Targeting Ferroptosis Attenuates Interstitial Inflammation and Kidney Fibrosis. Kidney Dis. 2022, 8, 57–71. [Google Scholar] [CrossRef] [PubMed]
  216. Balestrieri, B.; Di Costanzo, D.; Dwyer, D.F. Macrophage-Mediated Immune Responses: From Fatty Acids to Oxylipins. Molecules 2021, 27, 152. [Google Scholar] [CrossRef] [PubMed]
  217. Ide, S.; Kobayashi, Y.; Ide, K.; Strausser, S.A.; Abe, K.; Herbek, S.; O’Brien, L.L.; Crowley, S.D.; Barisoni, L.; Tata, A.; et al. Ferroptotic Stress Promotes the Accumulation of Pro-Inflammatory Proximal Tubular Cells in Maladaptive Renal Repair. eLife 2021, 10, e68603. [Google Scholar] [CrossRef]
  218. Proneth, B.; Conrad, M. Ferroptosis and Necroinflammation, a yet Poorly Explored Link. Cell Death Differ. 2019, 26, 14–24. [Google Scholar] [CrossRef]
  219. Wen, Q.; Liu, J.; Kang, R.; Zhou, B.; Tang, D. The Release and Activity of HMGB1 in Ferroptosis. Biochem. Biophys. Res. Commun. 2019, 510, 278–283. [Google Scholar] [CrossRef]
  220. Li, X.; Zou, Y.; Xing, J.; Fu, Y.-Y.; Wang, K.-Y.; Wan, P.-Z.; Zhai, X.-Y. Pretreatment with Roxadustat (FG-4592) Attenuates Folic Acid-Induced Kidney Injury through Antiferroptosis via Akt/GSK-3 β/Nrf2 Pathway. Oxidative Med. Cell. Longev. 2020, 2020, 6286984. [Google Scholar] [CrossRef]
  221. Belavgeni, A.; Meyer, C.; Stumpf, J.; Hugo, C.; Linkermann, A. Ferroptosis and Necroptosis in the Kidney. Cell Chem. Biol. 2020, 27, 448–462. [Google Scholar] [CrossRef]
  222. 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]
  223. Bissinger, R.; Bhuyan, A.A.M.; Qadri, S.M.; Lang, F. Oxidative Stress, Eryptosis and Anemia: A Pivotal Mechanistic Nexus in Systemic Diseases. FEBS J. 2019, 286, 826–854. [Google Scholar] [CrossRef]
  224. Dreischer, P.; Duszenko, M.; Stein, J.; Wieder, T. Eryptosis: Programmed Death of Nucleus-Free, Iron-Filled Blood Cells. Cells 2022, 11, 503. [Google Scholar] [CrossRef]
  225. Föller, M.; Lang, F. Ion Transport in Eryptosis, the Suicidal Death of Erythrocytes. Front. Cell Dev. Biol. 2020, 8, 597. [Google Scholar] [CrossRef]
  226. Li, D.; Zheng, X.; Zhang, Y.; Li, X.; Chen, X.; Yin, Y.; Hu, J.; Li, J.; Guo, M.; Wang, X. What Should Be Responsible for Eryptosis in Chronic Kidney Disease? Kidney Blood Press. Res. 2022, 47, 375–390. [Google Scholar] [CrossRef]
  227. Basile, D.P. The Endothelial Cell in Ischemic Acute Kidney Injury: Implications for Acute and Chronic Function. Kidney Int. 2007, 72, 151–156. [Google Scholar] [CrossRef] [PubMed]
  228. Basile, D.P.; Donohoe, D.; Roethe, K.; Osborn, J.L. Renal Ischemic Injury Results in Permanent Damage to Peritubular Capillaries and Influences Long-Term Function. Am. J. Physiol. Ren. Physiol. 2001, 281, F887–F899. [Google Scholar] [CrossRef]
  229. Sutton, T.A.; Fisher, C.J.; Molitoris, B.A. Microvascular Endothelial Injury and Dysfunction during Ischemic Acute Renal Failure. Kidney Int. 2002, 62, 1539–1549. [Google Scholar] [CrossRef]
  230. Sutton, T.A.; Mang, H.E.; Campos, S.B.; Sandoval, R.M.; Yoder, M.C.; Molitoris, B.A. Injury of the Renal Microvascular Endothelium Alters Barrier Function after Ischemia. Am. J. Physiol. Ren. Physiol. 2003, 285, F191–F198. [Google Scholar] [CrossRef]
  231. Yang, B.; Lan, S.; Dieudé, M.; Sabo-Vatasescu, J.-P.; Karakeussian-Rimbaud, A.; Turgeon, J.; Qi, S.; Gunaratnam, L.; Patey, N.; Hébert, M.-J. Caspase-3 Is a Pivotal Regulator of Microvascular Rarefaction and Renal Fibrosis after Ischemia-Reperfusion Injury. J. Am. Soc. Nephrol. 2018, 29, 1900–1916. [Google Scholar] [CrossRef]
  232. Gerhardt, H.; Betsholtz, C. Endothelial-Pericyte Interactions in Angiogenesis. Cell Tissue Res. 2003, 314, 15–23. [Google Scholar] [CrossRef] [PubMed]
  233. Schrimpf, C.; Xin, C.; Campanholle, G.; Gill, S.E.; Stallcup, W.; Lin, S.-L.; Davis, G.E.; Gharib, S.A.; Humphreys, B.D.; Duffield, J.S. Pericyte TIMP3 and ADAMTS1 Modulate Vascular Stability after Kidney Injury. J. Am. Soc. Nephrol. 2012, 23, 868–883. [Google Scholar] [CrossRef] [PubMed]
  234. Kramann, R.; Wongboonsin, J.; Chang-Panesso, M.; Machado, F.G.; Humphreys, B.D. Gli1+ Pericyte Loss Induces Capillary Rarefaction and Proximal Tubular Injury. J. Am. Soc. Nephrol. 2017, 28, 776–784. [Google Scholar] [CrossRef] [PubMed]
  235. Koller, G.M.; Schafer, C.; Kemp, S.S.; Aguera, K.N.; Lin, P.K.; Forgy, J.C.; Griffin, C.T.; Davis, G.E. Proinflammatory Mediators, IL (Interleukin)-1β, TNF (Tumor Necrosis Factor) α, and Thrombin Directly Induce Capillary Tube Regression. Arterioscler. Thromb. Vasc. Biol. 2020, 40, 365–377. [Google Scholar] [CrossRef]
  236. Sato, Y.; Yanagita, M. Immune Cells and Inflammation in AKI to CKD Progression. Am. J. Physiol. Ren. Physiol. 2018, 315, F1501–F1512. [Google Scholar] [CrossRef]
  237. Ohashi, R.; Shimizu, A.; Masuda, Y.; Kitamura, H.; Ishizaki, M.; Sugisaki, Y.; Yamanaka, N. Peritubular Capillary Regression during the Progression of Experimental Obstructive Nephropathy. J. Am. Soc. Nephrol. 2002, 13, 1795–1805. [Google Scholar] [CrossRef]
  238. Loganathan, K.; Salem Said, E.; Winterrowd, E.; Orebrand, M.; He, L.; Vanlandewijck, M.; Betsholtz, C.; Quaggin, S.E.; Jeansson, M. Angiopoietin-1 Deficiency Increases Renal Capillary Rarefaction and Tubulointerstitial Fibrosis in Mice. PLoS ONE 2018, 13, e0189433. [Google Scholar] [CrossRef]
  239. Kang, D.-H.; Joly, A.H.; Oh, S.-W.; Hugo, C.; Kerjaschki, D.; Gordon, K.L.; Mazzali, M.; Jefferson, J.A.; Hughes, J.; Madsen, K.M.; et al. Impaired Angiogenesis in the Remnant Kidney Model: I. Potential Role of Vascular Endothelial Growth Factor and Thrombospondin-1. J. Am. Soc. Nephrol. 2001, 12, 1434–1447. [Google Scholar] [CrossRef]
  240. Chen, J.; Hamm, L.L.; Kleinpeter, M.A.; Husserl, F.; Khan, I.E.; Chen, C.-S.; Liu, Y.; Mills, K.T.; He, C.; Rifai, N.; et al. Elevated Plasma Levels of Endostatin Are Associated with Chronic Kidney Disease. Am. J. Nephrol. 2012, 35, 335–340. [Google Scholar] [CrossRef]
  241. Potente, M.; Ghaeni, L.; Baldessari, D.; Mostoslavsky, R.; Rossig, L.; Dequiedt, F.; Haendeler, J.; Mione, M.; Dejana, E.; Alt, F.W.; et al. SIRT1 Controls Endothelial Angiogenic Functions during Vascular Growth. Genes Dev. 2007, 21, 2644–2658. [Google Scholar] [CrossRef]
  242. Ota, H.; Akishita, M.; Eto, M.; Iijima, K.; Kaneki, M.; Ouchi, Y. Sirt1 Modulates Premature Senescence-like Phenotype in Human Endothelial Cells. J. Mol. Cell. Cardiol. 2007, 43, 571–579. [Google Scholar] [CrossRef] [PubMed]
  243. Kida, Y.; Zullo, J.A.; Goligorsky, M.S. Endothelial Sirtuin 1 Inactivation Enhances Capillary Rarefaction and Fibrosis Following Kidney Injury through Notch Activation. Biochem. Biophys. Res. Commun. 2016, 478, 1074–1079. [Google Scholar] [CrossRef] [PubMed]
  244. Kuppe, C.; Ibrahim, M.M.; Kranz, J.; Zhang, X.; Ziegler, S.; Perales-Patón, J.; Jansen, J.; Reimer, K.C.; Smith, J.R.; Dobie, R.; et al. Decoding Myofibroblast Origins in Human Kidney Fibrosis. Nature 2021, 589, 281–286. [Google Scholar] [CrossRef] [PubMed]
  245. Wu, I.-W.; Hsu, K.-H.; Hsu, H.-J.; Lee, C.-C.; Sun, C.-Y.; Tsai, C.-J.; Wu, M.-S. Serum Free P-Cresyl Sulfate Levels Predict Cardiovascular and All-Cause Mortality in Elderly Hemodialysis Patients--a Prospective Cohort Study. Nephrology Dialysis Transplantation 2012, 27, 1169–1175. [Google Scholar] [CrossRef]
  246. Fan, P.-C.; Chang, J.C.-H.; Lin, C.-N.; Lee, C.-C.; Chen, Y.-T.; Chu, P.-H.; Kou, G.; Lu, Y.-A.; Yang, C.-W.; Chen, Y.-C. Serum Indoxyl Sulfate Predicts Adverse Cardiovascular Events in Patients with Chronic Kidney Disease. J. Formos. Med. Assoc. 2019, 118, 1099–1106. [Google Scholar] [CrossRef]
  247. Lin, C.-J.; Wu, V.; Wu, P.-C.; Wu, C.-J. Meta-Analysis of the Associations of p-Cresyl Sulfate (PCS) and Indoxyl Sulfate (IS) with Cardiovascular Events and All-Cause Mortality in Patients with Chronic Renal Failure. PLoS ONE 2015, 10, e0132589. [Google Scholar] [CrossRef]
  248. Tumur, Z.; Niwa, T. Indoxyl Sulfate Inhibits Nitric Oxide Production and Cell Viability by Inducing Oxidative Stress in Vascular Endothelial Cells. Am. J. Nephrol. 2009, 29, 551–557. [Google Scholar] [CrossRef]
  249. Tumur, Z.; Shimizu, H.; Enomoto, A.; Miyazaki, H.; Niwa, T. Indoxyl Sulfate Upregulates Expression of ICAM-1 and MCP-1 by Oxidative Stress-Induced NF-ĸB Activation. Am. J. Nephrol. 2010, 31, 435–441. [Google Scholar] [CrossRef]
  250. Six, I.; Gross, P.; Rémond, M.C.; Chillon, J.M.; Poirot, S.; Drueke, T.B.; Massy, Z.A. Deleterious Vascular Effects of Indoxyl Sulfate and Reversal by Oral Adsorbent AST-120. Atherosclerosis 2015, 243, 248–256. [Google Scholar] [CrossRef]
  251. Pletinck, A.; Glorieux, G.; Schepers, E.; Cohen, G.; Gondouin, B.; Van Landschoot, M.; Eloot, S.; Rops, A.; Van De Voorde, J.; De Vriese, A.; et al. Protein-Bound Uremic Toxins Stimulate Crosstalk between Leukocytes and Vessel Wall. J. Am. Soc. Nephrol. 2013, 24, 1981–1994. [Google Scholar] [CrossRef]
  252. Adijiang, A.; Goto, S.; Uramoto, S.; Nishijima, F.; Niwa, T. Indoxyl Sulphate Promotes Aortic Calcification with Expression of Osteoblast-Specific Proteins in Hypertensive Rats. Nephrol. Dial. Transplant. 2008, 23, 1892–1901. [Google Scholar] [CrossRef] [PubMed]
  253. Bouabdallah, J.; Zibara, K.; Issa, H.; Lenglet, G.; Kchour, G.; Caus, T.; Six, I.; Choukroun, G.; Kamel, S.; Bennis, Y. Endothelial Cells Exposed to Phosphate and Indoxyl Sulphate Promote Vascular Calcification through Interleukin-8 Secretion. Nephrol. Dial. Transplant. 2019, 34, 1125–1134. [Google Scholar] [CrossRef] [PubMed]
  254. Gondouin, B.; Cerini, C.; Dou, L.; Sallée, M.; Duval-Sabatier, A.; Pletinck, A.; Calaf, R.; Lacroix, R.; Jourde-Chiche, N.; Poitevin, S.; et al. Indolic Uremic Solutes Increase Tissue Factor Production in Endothelial Cells by the Aryl Hydrocarbon Receptor Pathway. Kidney Int. 2013, 84, 733–744. [Google Scholar] [CrossRef] [PubMed]
  255. Salyers, Z.R.; Coleman, M.; Balestrieri, N.P.; Ryan, T.E. Indoxyl Sulfate Impairs Angiogenesis via Chronic Aryl Hydrocarbon Receptor Activation. Am. J. Physiol. Cell Physiol. 2021, 320, C240–C249. [Google Scholar] [CrossRef]
  256. Esquivias-Motta, E.; Martín-Malo, A.; Buendia, P.; Álvarez-Lara, M.A.; Soriano, S.; Crespo, R.; Carracedo, J.; Ramírez, R.; Aljama, P. Hemodiafiltration with Endogenous Reinfusion Improved Microinflammation and Endothelial Damage Compared with Online-Hemodiafiltration: A Hypothesis Generating Study: Thoughts and Progress. Artif. Organs 2017, 41, 88–98. [Google Scholar] [CrossRef]
  257. Glorieux, G.; Vanholder, R.; Van Biesen, W.; Pletinck, A.; Schepers, E.; Neirynck, N.; Speeckaert, M.; De Bacquer, D.; Verbeke, F. Free p-Cresyl Sulfate Shows the Highest Association with Cardiovascular Outcome in Chronic Kidney Disease. Nephrol. Dial. Transplant. 2021, 36, 998–1005. [Google Scholar] [CrossRef]
  258. Jing, Y.J.; Ni, J.W.; Ding, F.H.; Fang, Y.H.; Wang, X.Q.; Wang, H.B.; Chen, X.N.; Chen, N.; Zhan, W.W.; Lu, L.; et al. P-Cresyl Sulfate Is Associated with Carotid Arteriosclerosis in Hemodialysis Patients and Promotes Atherogenesis in apoE−/− Mice. Kidney Int. 2016, 89, 439–449. [Google Scholar] [CrossRef]
  259. Lai, Y.-H.; Wang, C.-H.; Kuo, C.-H.; Lin, Y.-L.; Tsai, J.-P.; Hsu, B.-G. Serum P-Cresyl Sulfate Is a Predictor of Central Arterial Stiffness in Patients on Maintenance Hemodialysis. Toxins 2019, 12, 10. [Google Scholar] [CrossRef]
  260. Meijers, B.K.I.; Van Kerckhoven, S.; Verbeke, K.; Dehaen, W.; Vanrenterghem, Y.; Hoylaerts, M.F.; Evenepoel, P. The Uremic Retention Solute P-Cresyl Sulfate and Markers of Endothelial Damage. Am. J. Kidney Dis. 2009, 54, 891–901. [Google Scholar] [CrossRef]
  261. Han, H.; Chen, Y.; Zhu, Z.; Su, X.; Ni, J.; Du, R.; Zhang, R.; Jin, W. P-Cresyl Sulfate Promotes the Formation of Atherosclerotic Lesions and Induces Plaque Instability by Targeting Vascular Smooth Muscle Cells. Front. Med. 2016, 10, 320–329. [Google Scholar] [CrossRef]
  262. Han, H.; Chen, Y.; Zhu, J.; Ni, J.; Sun, J.; Zhang, R. Atorvastatin Attenuates P-Cresyl Sulfate-Induced Atherogenesis and Plaque Instability in ApoE Knockout Mice. Mol. Med. Rep. 2016, 14, 3122–3128. [Google Scholar] [CrossRef] [PubMed]
  263. Hung, S.-C.; Kuo, K.-L.; Huang, H.-L.; Lin, C.-C.; Tsai, T.-H.; Wang, C.-H.; Chen, J.-W.; Lin, S.-J.; Huang, P.-H.; Tarng, D.-C. Indoxyl Sulfate Suppresses Endothelial Progenitor Cell–Mediated Neovascularization. Kidney Int. 2016, 89, 574–585. [Google Scholar] [CrossRef] [PubMed]
  264. Wu, V.-C.; Young, G.-H.; Huang, P.-H.; Lo, S.-C.; Wang, K.-C.; Sun, C.-Y.; Liang, C.-J.; Huang, T.-M.; Chen, J.-H.; Chang, F.-C.; et al. In Acute Kidney Injury, Indoxyl Sulfate Impairs Human Endothelial Progenitor Cells: Modulation by Statin. Angiogenesis 2013, 16, 609–624. [Google Scholar] [CrossRef] [PubMed]
  265. Shen, W.-C.; Liang, C.-J.; Huang, T.-M.; Liu, C.-W.; Wang, S.-H.; Young, G.-H.; Tsai, J.-S.; Tseng, Y.-C.; Peng, Y.-S.; Wu, V.-C.; et al. Indoxyl Sulfate Enhances IL-1β-Induced E-Selectin Expression in Endothelial Cells in Acute Kidney Injury by the ROS/MAPKs/NFκB/AP-1 Pathway. Arch. Toxicol. 2016, 90, 2779–2792. [Google Scholar] [CrossRef]
  266. Matsumoto, T.; Takayanagi, K.; Kojima, M.; Taguchi, K.; Kobayashi, T. Acute Exposure to Indoxyl Sulfate Impairs Endothelium-Dependent Vasorelaxation in Rat Aorta. Int. J. Mol. Sci. 2019, 20, 338. [Google Scholar] [CrossRef]
  267. Matsumoto, T.; Takayanagi, K.; Kojima, M.; Katome, T.; Taguchi, K.; Kobayashi, T. Direct Impairment of the Endothelial Function by Acute Indoxyl Sulfate through Declined Nitric Oxide and Not Endothelium-Derived Hyperpolarizing Factor or Vasodilator Prostaglandins in the Rat Superior Mesenteric Artery. Biol. Pharm. Bull. 2019, 42, 1236–1242. [Google Scholar] [CrossRef]
  268. Tang, W.-H.; Wang, C.-P.; Yu, T.-H.; Tai, P.-Y.; Liang, S.-S.; Hung, W.-C.; Wu, C.-C.; Huang, S.-H.; Lee, Y.-J.; Chen, S.-C. Protein-Bounded Uremic Toxin p-Cresylsulfate Induces Vascular Permeability Alternations. Histochem. Cell Biol. 2018, 149, 607–617. [Google Scholar] [CrossRef]
  269. Zhang, M.; Malik, A.B.; Rehman, J. Endothelial Progenitor Cells and Vascular Repair. Curr. Opin. Hematol. 2014, 21, 224–228. [Google Scholar] [CrossRef]
  270. Wynn, T.A.; Vannella, K.M. Macrophages in Tissue Repair, Regeneration, and Fibrosis. Immunity 2016, 44, 450–462. [Google Scholar] [CrossRef]
  271. Chen, Y.; Lin, L.; Tao, X.; Song, Y.; Cui, J.; Wan, J. The Role of Podocyte Damage in the Etiology of Ischemia-Reperfusion Acute Kidney Injury and Post-Injury Fibrosis. BMC Nephrol. 2019, 20, 106. [Google Scholar] [CrossRef]
  272. Angeletti, A.; Cantarelli, C.; Petrosyan, A.; Andrighetto, S.; Budge, K.; D’Agati, V.D.; Hartzell, S.; Malvi, D.; Donadei, C.; Thurman, J.M.; et al. Loss of Decay-Accelerating Factor Triggers Podocyte Injury and Glomerulosclerosis. J. Exp. Med. 2020, 217, e20191699. [Google Scholar] [CrossRef] [PubMed]
  273. Niranjan, T.; Murea, M.; Susztak, K. The Pathogenic Role of Notch Activation in Podocytes. Nephron Exp. Nephrol. 2009, 111, e73–e79. [Google Scholar] [CrossRef] [PubMed]
  274. 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]
  275. Nishad, R.; Meshram, P.; Singh, A.K.; Reddy, G.B.; Pasupulati, A.K. Activation of Notch1 Signaling in Podocytes by Glucose-Derived AGEs Contributes to Proteinuria. BMJ Open Diab Res. Care 2020, 8, e001203. [Google Scholar] [CrossRef] [PubMed]
  276. Lin, X.; Jin, H.; Chai, Y.; Shou, S. Cellular Senescence and Acute Kidney Injury. Pediatr. Nephrol. 2022, 37, 3009–3018. [Google Scholar] [CrossRef] [PubMed]
  277. Eymael, J.; Sharma, S.; Loeven, M.A.; Wetzels, J.F.; Mooren, F.; Florquin, S.; Deegens, J.K.; Willemsen, B.K.; Sharma, V.; Van Kuppevelt, T.H.; et al. CD44 Is Required for the Pathogenesis of Experimental Crescentic Glomerulonephritis and Collapsing Focal Segmental Glomerulosclerosis. Kidney Int. 2018, 93, 626–642. [Google Scholar] [CrossRef] [PubMed]
  278. Roeder, S.S.; Stefanska, A.; Eng, D.G.; Kaverina, N.; Sunseri, M.W.; McNicholas, B.A.; Rabinovitch, P.; Engel, F.B.; Daniel, C.; Amann, K.; et al. Changes in Glomerular Parietal Epithelial Cells in Mouse Kidneys with Advanced Age. Am. J. Physiol. Ren. Physiol. 2015, 309, F164–F178. [Google Scholar] [CrossRef]
  279. Appel, D.; Kershaw, D.B.; Smeets, B.; Yuan, G.; Fuss, A.; Frye, B.D.; Elger, M.; Kriz, W.; Floege, J.D.; Moeller, M.J. Recruitment of Podocytes from Glomerular Parietal Epithelial Cells. J. Am. Soc. Nephrol. 2009, 20, 333–343. [Google Scholar] [CrossRef]
  280. Miesen, L.; Steenbergen, E.; Smeets, B. Parietal Cells—New Perspectives in Glomerular Disease. Cell Tissue Res. 2017, 369, 237–244. [Google Scholar] [CrossRef]
  281. Gelasco, A.K.; Raymond, J.R. Indoxyl Sulfate Induces Complex Redox Alterations in Mesangial Cells. Am. J. Physiol. Ren. Physiol. 2006, 290, F1551–F1558. [Google Scholar] [CrossRef]
  282. Owada, S.; Goto, S.; Bannai, K.; Hayashi, H.; Nishijima, F.; Niwa, T. Indoxyl Sulfate Reduces Superoxide Scavenging Activity in the Kidneys of Normal and Uremic Rats. Am. J. Nephrol. 2008, 28, 446–454. [Google Scholar] [CrossRef] [PubMed]
  283. Niwa, T.; Ise, M.; Miyazaki, T. Progression of Glomerular Sclerosis in Experimental Uremic Rats by Administration of Indole, a Precursor of Indoxyl Sulfate. Am. J. Nephrol. 1994, 14, 207–212. [Google Scholar] [CrossRef] [PubMed]
  284. Niwa, T.; Ise, M. Indoxyl Sulfate, a Circulating Uremic Toxin, Stimulates the Progression of Glomerular Sclerosis. J. Lab. Clin. Med. 1994, 124, 96–104. [Google Scholar] [PubMed]
  285. Watanabe, H.; Miyamoto, Y.; Honda, D.; Tanaka, H.; Wu, Q.; Endo, M.; Noguchi, T.; Kadowaki, D.; Ishima, Y.; Kotani, S.; et al. P-Cresyl Sulfate Causes Renal Tubular Cell Damage by Inducing Oxidative Stress by Activation of NADPH Oxidase. Kidney Int. 2013, 83, 582–592. [Google Scholar] [CrossRef]
  286. Schulman, G.; Berl, T.; Beck, G.J.; Remuzzi, G.; Ritz, E.; Arita, K.; Kato, A.; Shimizu, M. Randomized Placebo-Controlled EPPIC Trials of AST-120 in CKD. J. Am. Soc. Nephrol. 2015, 26, 1732–1746. [Google Scholar] [CrossRef]
  287. Yamaguchi, J.; Tanaka, T.; Inagi, R. Effect of AST-120 in Chronic Kidney Disease Treatment: Still a Controversy? Nephron 2017, 135, 201–206. [Google Scholar] [CrossRef]
  288. Bennis, Y.; Cluet, Y.; Titeca-Beauport, D.; El Esper, N.; Ureña, P.; Bodeau, S.; Combe, C.; Dussol, B.; Fouque, D.; Choukroun, G.; et al. The Effect of Sevelamer on Serum Levels of Gut-Derived Uremic Toxins: Results from In Vitro Experiments and A Multicenter, Double-Blind, Placebo-Controlled, Randomized Clinical Trial. Toxins 2019, 11, 279. [Google Scholar] [CrossRef]
  289. Cha, R.; Kang, S.W.; Park, C.W.; Cha, D.R.; Na, K.Y.; Kim, S.G.; Yoon, S.A.; Han, S.Y.; Chang, J.H.; Park, S.K.; et al. A Randomized, Controlled Trial of Oral Intestinal Sorbent AST-120 on Renal Function Deterioration in Patients with Advanced Renal Dysfunction. Clin. J. Am. Soc. Nephrol. 2016, 11, 559–567. [Google Scholar] [CrossRef]
  290. Montemurno, E.; Cosola, C.; Dalfino, G.; Daidone, G.; De Angelis, M.; Gobbetti, M.; Gesualdo, L. What Would You Like to Eat, Mr CKD Microbiota? A Mediterranean Diet, Please! Kidney Blood Press. Res. 2014, 39, 114–123. [Google Scholar] [CrossRef]
  291. Sirich, T.L.; Plummer, N.S.; Gardner, C.D.; Hostetter, T.H.; Meyer, T.W. Effect of Increasing Dietary Fiber on Plasma Levels of Colon-Derived Solutes in Hemodialysis Patients. Clin. J. Am. Soc. Nephrol. 2014, 9, 1603–1610. [Google Scholar] [CrossRef]
  292. Rossi, M.; Johnson, D.W.; Xu, H.; Carrero, J.J.; Pascoe, E.; French, C.; Campbell, K.L. Dietary Protein-Fiber Ratio Associates with Circulating Levels of Indoxyl Sulfate and p-Cresyl Sulfate in Chronic Kidney Disease Patients. Nutr. Metab. Cardiovasc. Dis. 2015, 25, 860–865. [Google Scholar] [CrossRef] [PubMed]
  293. Di Iorio, B.R.; Rocchetti, M.T.; De Angelis, M.; Cosola, C.; Marzocco, S.; Di Micco, L.; Di Bari, I.; Accetturo, M.; Vacca, M.; Gobbetti, M.; et al. Nutritional Therapy Modulates Intestinal Microbiota and Reduces Serum Levels of Total and Free Indoxyl Sulfate and P-Cresyl Sulfate in Chronic Kidney Disease (Medika Study). J. Clin. Med. 2019, 8, 1424. [Google Scholar] [CrossRef] [PubMed]
  294. Black, A.P.; Anjos, J.S.; Cardozo, L.; Carmo, F.L.; Dolenga, C.J.; Nakao, L.S.; De Carvalho Ferreira, D.; Rosado, A.; Carraro Eduardo, J.C.; Mafra, D. Does Low-Protein Diet Influence the Uremic Toxin Serum Levels from the Gut Microbiota in Nondialysis Chronic Kidney Disease Patients? J. Ren. Nutr. 2018, 28, 208–214. [Google Scholar] [CrossRef] [PubMed]
  295. Marzocco, S.; Dal Piaz, F.; Di Micco, L.; Torraca, S.; Sirico, M.L.; Tartaglia, D.; Autore, G.; Di Iorio, B. Very Low Protein Diet Reduces Indoxyl Sulfate Levels in Chronic Kidney Disease. Blood Purif. 2013, 35, 196–201. [Google Scholar] [CrossRef] [PubMed]
  296. Evenepoel, P.; Meijers, B.; Masereeuw, R.; Lowenstein, J. Effects of an SGLT Inhibitor on the Production, Toxicity, and Elimination of Gut-Derived Uremic Toxins: A Call for Additional Evidence. Toxins 2022, 14, 210. [Google Scholar] [CrossRef] [PubMed]
  297. Mishima, E.; Fukuda, S.; Kanemitsu, Y.; Saigusa, D.; Mukawa, C.; Asaji, K.; Matsumoto, Y.; Tsukamoto, H.; Tachikawa, T.; Tsukimi, T.; et al. Canagliflozin Reduces Plasma Uremic Toxins and Alters the Intestinal Microbiota Composition in a Chronic Kidney Disease Mouse Model. Am. J. Physiol. Ren. Physiol. 2018, 315, F824–F833. [Google Scholar] [CrossRef]
  298. Ho, H.; Kikuchi, K.; Oikawa, D.; Watanabe, S.; Kanemitsu, Y.; Saigusa, D.; Kujirai, R.; Ikeda-Ohtsubo, W.; Ichijo, M.; Akiyama, Y.; et al. SGLT-1-specific Inhibition Ameliorates Renal Failure and Alters the Gut Microbial Community in Mice with Adenine-induced Renal Failure. Physiol. Rep. 2021, 9, e15092. [Google Scholar] [CrossRef]
  299. Fang, C.-Y.; Lu, J.-R.; Chen, B.-J.; Wu, C.; Chen, Y.-P.; Chen, M.-J. Selection of Uremic Toxin-Reducing Probiotics in Vitro and in Vivo. J. Funct. Foods 2014, 7, 407–415. [Google Scholar] [CrossRef]
  300. De Faria Barros, A.; Borges, N.A.; Nakao, L.S.; Dolenga, C.J.; Do Carmo, F.L.; De Carvalho Ferreira, D.; Stenvinkel, P.; Bergman, P.; Lindholm, B.; Mafra, D. Effects of Probiotic Supplementation on Inflammatory Biomarkers and Uremic Toxins in Non-Dialysis Chronic Kidney Patients: A Double-Blind, Randomized, Placebo-Controlled Trial. J. Funct. Foods 2018, 46, 378–383. [Google Scholar] [CrossRef]
  301. Thongprayoon, C.; Kaewput, W.; Hatch, S.T.; Bathini, T.; Sharma, K.; Wijarnpreecha, K.; Ungprasert, P.; D’Costa, M.; Mao, M.A.; Cheungpasitporn, W. Effects of Probiotics on Inflammation and Uremic Toxins Among Patients on Dialysis: A Systematic Review and Meta-Analysis. Dig. Dis. Sci. 2019, 64, 469–479. [Google Scholar] [CrossRef]
  302. Saxena, A.; Srinivasa, S.; Veerappan, I.; Jacob, C.; Mahaldar, A.; Gupta, A.; Rajagopal, A. Enzobiotics—A Novel Therapy for the Elimination of Uremic Toxins in Patients with CKD (EETOX Study): A Multicenter Double-Blind Randomized Controlled Trial. Nutrients 2022, 14, 3804. [Google Scholar] [CrossRef] [PubMed]
  303. Caggiano, G.; Stasi, A.; Franzin, R.; Fiorentino, M.; Cimmarusti, M.T.; Deleonardis, A.; Palieri, R.; Pontrelli, P.; Gesualdo, L. Fecal Microbiota Transplantation in Reducing Uremic Toxins Accumulation in Kidney Disease: Current Understanding and Future Perspectives. Toxins 2023, 15, 115. [Google Scholar] [CrossRef] [PubMed]
  304. Barba, C.; Soulage, C.O.; Caggiano, G.; Glorieux, G.; Fouque, D.; Koppe, L. Effects of Fecal Microbiota Transplantation on Composition in Mice with CKD. Toxins 2020, 12, 741. [Google Scholar] [CrossRef] [PubMed]
  305. Wang, X.; Yang, S.; Li, S.; Zhao, L.; Hao, Y.; Qin, J.; Zhang, L.; Zhang, C.; Bian, W.; Zuo, L.; et al. Aberrant Gut Microbiota Alters Host Metabolome and Impacts Renal Failure in Humans and Rodents. Gut 2020, 69, 2131–2142. [Google Scholar] [CrossRef]
  306. Emal, D.; Rampanelli, E.; Stroo, I.; Butter, L.M.; Teske, G.J.; Claessen, N.; Stokman, G.; Florquin, S.; Leemans, J.C.; Dessing, M.C. Depletion of Gut Microbiota Protects against Renal Ischemia-Reperfusion Injury. J. Am. Soc. Nephrolog. 2017, 28, 1450–1461. [Google Scholar] [CrossRef]
  307. Nakade, Y.; Iwata, Y.; Furuichi, K.; Mita, M.; Hamase, K.; Konno, R.; Miyake, T.; Sakai, N.; Kitajima, S.; Toyama, T.; et al. Gut Microbiota–Derived D-Serine Protects against Acute Kidney Injury. JCI Insight 2018, 3, e97957. [Google Scholar] [CrossRef]
  308. Nazzal, L.; Soiefer, L.; Chang, M.; Tamizuddin, F.; Schatoff, D.; Cofer, L.; Aguero-Rosenfeld, M.E.; Matalon, A.; Meijers, B.; Holzman, R.; et al. Effect of Vancomycin on the Gut Microbiome and Plasma Concentrations of Gut-Derived Uremic Solutes. Kidney Int. Rep. 2021, 6, 2122–2133. [Google Scholar] [CrossRef]
  309. Devlin, A.S.; Marcobal, A.; Dodd, D.; Nayfach, S.; Plummer, N.; Meyer, T.; Pollard, K.S.; Sonnenburg, J.L.; Fischbach, M.A. Modulation of a Circulating Uremic Solute via Rational Genetic Manipulation of the Gut Microbiota. Cell Host Microbe 2016, 20, 709–715. [Google Scholar] [CrossRef]
  310. Saito, H.; Saigo, C.; Nomura, Y.; Yamamoto, Y.; Sagata, M.; Matsunaga, R.; Jono, H.; Nishi, K. Meclofenamate Elicits a Nephropreventing Effect in a Rat Model of Ischemic Acute Kidney Injury by Suppressing Indoxyl Sulfate Production and Restoring Renal Organic Anion Transporters. Drug Des. Dev. Ther. 2014, 8, 1073–1082. [Google Scholar] [CrossRef]
  311. Saito, H.; Yoshimura, M.; Saigo, C.; Komori, M.; Nomura, Y.; Yamamoto, Y.; Sagata, M.; Wakida, A.; Chuman, E.; Nishi, K.; et al. Hepatic Sulfotransferase as a Nephropreventing Target by Suppression of the Uremic Toxin Indoxyl Sulfate Accumulation in Ischemic Acute Kidney Injury. Toxicol. Sci. 2014, 141, 206–217. [Google Scholar] [CrossRef]
  312. Steinke, I.; Ghanei, N.; Govindarajulu, M.; Yoo, S.; Zhong, J.; Amin, R.H. Drug Discovery and Development of Novel Therapeutics for Inhibiting TMAO in Models of Atherosclerosis and Diabetes. Front. Physiol. 2020, 11, 567899. [Google Scholar] [CrossRef] [PubMed]
  313. Vanholder, R.C.; Eloot, S.; Glorieux, G.L.R.L. Future Avenues to Decrease Uremic Toxin Concentration. Am. J. Kidney Dis. 2016, 67, 664–676. [Google Scholar] [CrossRef] [PubMed]
  314. Paats, J.; Adoberg, A.; Arund, J.; Dhondt, A.; Fernström, A.; Fridolin, I.; Glorieux, G.; Leis, L.; Luman, M.; Gonzalez-Parra, E.; et al. Serum Levels and Removal by Haemodialysis and Haemodiafiltration of Tryptophan-Derived Uremic Toxins in ESKD Patients. Int. J. Mol. Sci. 2020, 21, 1522. [Google Scholar] [CrossRef] [PubMed]
  315. Meert, N.; Eloot, S.; Schepers, E.; Lemke, H.-D.; Dhondt, A.; Glorieux, G.; Van Landschoot, M.; Waterloos, M.-A.; Vanholder, R. Comparison of Removal Capacity of Two Consecutive Generations of High-Flux Dialysers during Different Treatment Modalities. Nephrol. Dial. Transplant. 2011, 26, 2624–2630. [Google Scholar] [CrossRef] [PubMed]
  316. Ma, Y.; Li, S.; Tonelli, M.; Unsworth, L.D. Adsorption-Based Strategies for Removing Uremic Toxins from Blood. Microporous Mesoporous Mater. 2021, 319, 111035. [Google Scholar] [CrossRef]
  317. Cabello-Alvarado, C.; Andrade-Guel, M.; Pérez-Alvarez, M.; Cadenas-Pliego, G.; Cortés-Hernández, D.A.; Bartolo-Pérez, P.; Ávila-Orta, C.A.; Cruz-Delgado, V.J.; Zepeda-Pedreguera, A. Graphene Nanoplatelets Modified with Amino-Groups by Ultrasonic Radiation of Variable Frequency for Potential Adsorption of Uremic Toxins. Nanomaterials 2019, 9, 1261. [Google Scholar] [CrossRef]
  318. De Loor, H.; Meijers, B.K.I.; Meyer, T.W.; Bammens, B.; Verbeke, K.; Dehaen, W.; Evenepoel, P. Sodium Octanoate to Reverse Indoxyl Sulfate and P-Cresyl Sulfate Albumin Binding in Uremic and Normal Serum during Sample Preparation Followed by Fluorescence Liquid Chromatography. J. Chromatogr. A 2009, 1216, 4684–4688. [Google Scholar] [CrossRef]
  319. Tao, X.; Thijssen, S.; Kotanko, P.; Ho, C.-H.; Henrie, M.; Stroup, E.; Handelman, G. Improved Dialytic Removal of Protein-Bound Uraemic Toxins with Use of Albumin Binding Competitors: An in Vitro Human Whole Blood Study. Sci. Rep. 2016, 6, 23389. [Google Scholar] [CrossRef]
  320. Madero, M.; Cano, K.B.; Campos, I.; Tao, X.; Maheshwari, V.; Brown, J.; Cornejo, B.; Handelman, G.; Thijssen, S.; Kotanko, P. Removal of Protein-Bound Uremic Toxins during Hemodialysis Using a Binding Competitor. Clin. J. Am. Soc. Nephrol. 2019, 14, 394–402. [Google Scholar] [CrossRef]
  321. Yamamoto, S.; Kazama, J.J.; Wakamatsu, T.; Takahashi, Y.; Kaneko, Y.; Goto, S.; Narita, I. Removal of Uremic Toxins by Renal Replacement Therapies: A Review of Current Progress and Future Perspectives. Ren. Replace. Ther. 2016, 2, 43. [Google Scholar] [CrossRef]
Figure 1. Tubular, endothelial, and glomerular mechanisms leading to the AKI-to-CKD transition and the involvement of UTs. After tubular, endothelial, or glomerular injury, necrosed cells stimulate the innate system and then the adaptive immune system. Imbalanced cytokine and TGF-β secretion leads to tubular maladaptive repair (mitochondrial dysfunction, ER stress, cell cycle arrest, metabolic reprogramming, tubular senescence, and EMT), endothelial senescence and rarefaction, hypoxia, and podocyte loss. Across several compartments, the overproduction of profibrotic factors stimulates extracellular matrix-secreting myofibroblasts, increases fibrosis, and hastens progression to CKD. AKI, acute kidney disease; CKD, chronic kidney disease; EMT, epithelial-to-mesenchymal transition; ER, endoplasmic reticulum. Created with BioRender.com.
Figure 1. Tubular, endothelial, and glomerular mechanisms leading to the AKI-to-CKD transition and the involvement of UTs. After tubular, endothelial, or glomerular injury, necrosed cells stimulate the innate system and then the adaptive immune system. Imbalanced cytokine and TGF-β secretion leads to tubular maladaptive repair (mitochondrial dysfunction, ER stress, cell cycle arrest, metabolic reprogramming, tubular senescence, and EMT), endothelial senescence and rarefaction, hypoxia, and podocyte loss. Across several compartments, the overproduction of profibrotic factors stimulates extracellular matrix-secreting myofibroblasts, increases fibrosis, and hastens progression to CKD. AKI, acute kidney disease; CKD, chronic kidney disease; EMT, epithelial-to-mesenchymal transition; ER, endoplasmic reticulum. Created with BioRender.com.
Ijms 24 16152 g001
Figure 2. Chemical structure of UTs. TMAO and urea belong to the group of water-soluble UTs with low molecular weight (MW < 500 Da). PTH belongs to the group of water-soluble UTs with high molecular weight (MW > 500 Da). IS, pCS, and IAA belong to the group of protein-binding UTs. IAA, indole-3-acetic acid; IS, indoxyl sulfate; pCS, para-cresyl sulfate; PTH, parathyroid hormone; TMAO, trimethylamine-N-oxide.
Figure 2. Chemical structure of UTs. TMAO and urea belong to the group of water-soluble UTs with low molecular weight (MW < 500 Da). PTH belongs to the group of water-soluble UTs with high molecular weight (MW > 500 Da). IS, pCS, and IAA belong to the group of protein-binding UTs. IAA, indole-3-acetic acid; IS, indoxyl sulfate; pCS, para-cresyl sulfate; PTH, parathyroid hormone; TMAO, trimethylamine-N-oxide.
Ijms 24 16152 g002
Table 1. Mechanisms of UT toxicity involved in the AKI-to-CKD transition in the tubulo-interstitial compartment.
Table 1. Mechanisms of UT toxicity involved in the AKI-to-CKD transition in the tubulo-interstitial compartment.
UTsMechanisms Underlying the AKI-to-CKD TransitionThe Tubulo-Interstitial Compartment
ModelsMain ResultsReferences
ISEMT, TGF-βIn vitroExtinction of the epithelial phenotype: alteration of the cubic epithelial aspect, ↓ ZO-1, ↓ E-cadherin
Mesenchymal expression: ↑ α-SMA, ↑ TGF-β, ↑ Smad2/3/4 pathway, ↑ Snail, ↑ fibronectin,
Inhibition of IS-induced EMT: probenecid ↓ EMT-induced MAPK/ERK pathway
[105,106,109,112,113]
In vivoExtinction of the epithelial phenotype: ↓ ZO-1, ↓ E-cadherin,
Mesenchymal expression: ↑ TGF-β, ↑ α-SMA, ↑ collagen, ↑ vimentin
↑ Fibrosis
Inhibition of UT-induced EMT:
- Losartan ↓ UTs, TGF-β and Snail and ↓ fibrosis
- AST-120 ↓ IS concentration, ↑ ZO-1, ↓ α-SMA, ↓ fibrosis
[106,107,109,110,111,114,115,116,117]
Oxidative stressIn vitro- ↑ ROS, ↑ NF-κB, ↓ Nrf2
- N-acetyl cysteine or NF-κB inhibitors or NADPH oxidase inhibitor → ↓ NF-κB, ↓ NOX4
[112,118,119]
In vivo- AST-120 ↓ urine and serum IS concentrations, ↑ kidney oxygenation, ↓ 8-OHdG, ↓ interstitial fibrosis, ↑ Nrf2, ↑ HO-1 and ↑ NQO1[119,120,121]
Mitochondria dysfunctionIn vitro- ↓ Glucuronidation, ↓ complex II activity, ↓ electron transport capacity
- ↓ Fusion, ↑ fission, ↑ autophagy
[122,123]
In vivo- ↓ Nitrogen metabolism, alteration of the inner membrane, ↑ fission, ↑ autophagy[123]
ER stressIn vitro- ↓ GRP78, ↑ CHOP[124,125]
In vivo- AST-120 ↓ CHOP[125]
Metabolic reprogrammingIn vitro- ↑ AhR-dependent arachidonic acid pathway-reprogramming monocytes[126]
In vivo- Alteration of metabolic pathways, including tryptophan metabolism[127]
Epigenetic alteration
Cycle cell arrest
Senescence
In vitro- Methylation of CpG islands of sFRP => Wnt/β-catenin pathway activation
- Activation of p53 and p21
- NF-κB inhibitors suppressed IS-induced p53 and p21
[128,129,130]
In vivo- Administration of recombinant sFRP5 alleviated IS-induced fibrosis
- Hypermethylation of Klotho
- AST-120 decreased p65, p53, p21, β-galactosidase activation, TGF-β, and α-SMA
- Stat3 siRNA suppressed IS-induced β-galactosidase activation and fibrosis
[128,129,130,131,132]
InflammationIn vitro- ↑ Expression of IL-1β, IL-6, IL-15, IL-6 and IL-15, TGF-β, NF-κB, Smad, Stat, B2m, Bax, and Bcl2
- Indole-3-propionic acid suppressed IS-induced MCP-1
- Correlation between CD14 + CD16+ monocytes and plasma IS concentration in AAA patients
- Plasma IS from AAA patients promotes IL-10, PPARγ, TGF-β, TIMP-1, IL-6, CCL2, and COX2
[133,134,135,136]
In vivo- ↑ MCP-1
- Indole-3-propionic acid ↓ IS-induced MCP-1
- AST-120 ↓ IS-induced Mac-1
[134,135,137]
Iron death pathwaysIn vitro - ↑ Hepcidin expression through AhR
- ↑ Intracellular Ca2+ concentration and ceramide concentration in erythrocytes
- ↑ Eryptosis and thrombosis
[138,139,140,141]
In vivo- AST-120 ↓ IS-induced hepcidin expression[138]
IAAIron death pathwaysIn vitro - ↑ Eryptosis and thrombosis[141]
pCSInflammationIn vitro- ↑ Expression of IL-1β, IL-6, IL-15, IL-6 and IL-15, TGF-β, NF-κB, Smad, Stat, B2m, Bax, and Bcl2[133,134]
Me2PY Me4PYOxidative stressClinical study- Correlation between Me2PY or Me4PY and the oxidative stress marker GSH[142]
CMPFMitochondrial dysfunctionIn vitro- Hemodialyzed patient serum with CMPF inhibited ADP-stimulated oxidation[143]
Iron death pathwaysIn vitro- ↓ GSH levels and GPX4, FHC, FLC, and ↑ intracellular iron concentration[144]
D-serineCycle cell arrestIn vitro- Activation of GCN2 → senescence[145]
ADMATGF-βIn vivo- ↑ TGF-β, ↑ α-SMA, ↑ collagen, ↑ fibronectin, ↑ fibrosis[146]
Abbreviations: 8-OHdG, 8-hydroxyguanosine; AAA, abdominal aortic aneurysm; ADMA, asymmetric dimethylarginine; AhR, aryl hydrocarbon receptor; α-SMA, α-smooth muscle actin; B2m, β-2 microglobulin; Bax, Bcl-2–associated X protein; Bcl2, B-cell lymphoma 2; CCL2, chemokine ligand 2; CHOP, C/EBP homologous protein; CMPF, 3-carboxy-4-methyl-5-propyl-2-furanpropionate; COX2, cyclooxygenase 2; EMT, epithelial-mesenchymal transition; ER, endoplasmic reticulum; FHC, ferritin heavy chains; FLC, ferritin light chains; GCN2, general control nonderepressible 2; GPX4, glutathione peroxidase 4; GRP78, glucose-regulated protein 78; GSH, reduced glutathione; HO-1, heme oxygenase-1; IAA, indole-3-acetic acid; IL, interleukin; IS, indoxyl sulfate; Mac-1, macrophage-1 antigen; MCP-1, monocyte chemoattractant protein 1; Me2PY, N-methyl-2-pyridone-5-carboxamide; Me4PY, N-methyl-4-pyridone-5-carboxamide; NF-κB, nuclear factor-κ B; NOX4, NADPH oxidase 4; NQO1, NAD(P)H quinone dehydrogenase 1; Nrf2, nuclear factor (erythroid-derived 2)-like 2; pCS, para-cresyl sulfate; PPARγ, peroxisome proliferator-activated receptor-γ; ROS, reactive oxygen species; sFRP, secreted frizzled-related protein; TGF-β, transforming growth factor β; TIMP-1, tissue inhibitor of metalloproteinase 1; UT, uremic toxin; ZO-1, zonula occludens-1.
Table 3. Mechanisms of UT toxicity involved in the AKI-to-CKD transition in the glomerular compartment.
Table 3. Mechanisms of UT toxicity involved in the AKI-to-CKD transition in the glomerular compartment.
UTsMechanism Underlying the AKI-to-CKD TransitionEndothelium
ModelsMain ResultsReferences
ISPodocyte loss and senescenceIn vitro- ↑ AhR, ↑ vimentin
- Podocyte effacement, with ↓ in Actn4, Cd2ap, Myh9, Nphs1, Nphs2, Podxl, Synpo, and Wt1 mRNA
- ↑ reduction rate, ↑ ROS, ↑ SOD sensitivity in mesangial cells
[69,281]
In vivo- ↑ AhR, ↑ vimentin
- Podocyte effacement, with ↓ in Actn4, Cd2ap, Myh9, Nphs1, Nphs2, Podxl, Synpo, and Wt1 mRNA
- AST-120 ↓ glomerular sclerosis
[69,283,284]
Abbreviations: AhR, aryl hydrocarbon receptor; IS, indoxyl sulfate; ROS, reactive oxygen species; SOD, superoxyde dismutase; UT, uremic toxin.
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

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. https://doi.org/10.3390/ijms242216152

AMA Style

André C, Bodeau S, Kamel S, Bennis Y, Caillard P. The AKI-to-CKD Transition: The Role of Uremic Toxins. International Journal of Molecular Sciences. 2023; 24(22):16152. https://doi.org/10.3390/ijms242216152

Chicago/Turabian Style

André, Camille, Sandra Bodeau, Saïd Kamel, Youssef Bennis, and Pauline Caillard. 2023. "The AKI-to-CKD Transition: The Role of Uremic Toxins" International Journal of Molecular Sciences 24, no. 22: 16152. https://doi.org/10.3390/ijms242216152

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